Вы находитесь на странице: 1из 351

Heme, Chlorophyll,

and Bilins
Methods and Protocols
EDITED BY

Alison G. Smith
Michael Witty

HUMANA PRESS
Heme, Chlorophyll, and Bilins
Heme, Chlorophyll,
and Bilins
Methods and Protocols

Edited by

Alison G. Smith
Department of Plant Sciences, University of Cambridge, Cambridge, UK

and

Michael Witty
Department of Biochemistry, University of Cambridge, Cambridge, UK

Humana Press Totowa, New Jersey


© 2002 Humana Press Inc.
999 Riverview Drive, Suite 208
Totowa, New Jersey 07512

www.humanapress.com

The content and opinions expressed in this book are the sole work of the authors and editors, who have
warranted due diligence in the creation and issuance of their work. The publisher, editors, and authors are
not responsible for errors or omissions or for any consequences arising from the information or opinions
presented in this book and make no warranty, express or implied, with respect to its contents.

This publication is printed on acid-free paper. ∞


ANSI Z39.48-1984 (American Standards Institute)
Permanence of Paper for Printed Library Materials.

Cover design by Patricia F. Cleary.


For additional copies, pricing for bulk purchases, and/or information about other Humana titles, contact
Humana at the above address or at any of the following numbers: Tel.: 973-256-1699; Fax: 973-256-8341; E-
mail: humana@humanapr.com; or visit our Website: www.humanapress.com

Photocopy Authorization Policy:

Authorization to photocopy items for internal or personal use, or the internal or personal use of specific
clients, is granted by Humana Press Inc., provided that the base fee of US $10.00 per copy, plus US $00.25
per page, is paid directly to the Copyright Clearance Center at 222 Rosewood Drive, Danvers, MA 01923. For
those organizations that have been granted a photocopy license from the CCC, a separate system of
payment has been arranged and is acceptable to Humana Press Inc. The fee code for users of the Transac-
tional Reporting Service is: [1-58829-111-1/02 (hardcover) $10.00 + $00.25].

Printed in the United States of America. 10 9 8 7 6 5 4 3 2 1

Library of Congress Cataloging in Publication Data

Heme, chlorophyll, and bilins: methods and protocols / edited by Alison G. Smith and Michael Witty
p. cm.
Includes bibliographical references (p.)
ISBN 1-58829-111-1 (alk. paper)
1. Chlorophyll. 2. Heme. 3. Tetrapyrroles. 4. Plant pigments. I. Smith, Alison G. II. Witty, Michael.
QK898.C5 H46 2001
572'.46–dc21 2001039604
Preface
The men of experiment are like the ant, they only collect and use; the
reasoners resemble spiders, who make cobwebs out of their own
substance. But the bee takes the middle course: it gathers its material
from the flowers of the garden and field, but transforms and digests it by
a power of its own. Not unlike this is the true business of philosophy
[science]; for it neither relies solely or chiefly on the powers of the mind,
nor does it take the matter which it gathers from natural history and
mechanical experiments and lay up in the memory whole, as it finds it,
but lays it up in the understanding altered and digested. Therefore, from
a closer and purer league between these two faculties, the experimental
and the rational (such as has never been made), much may be hoped.
Francis Bacon, Novum Organum, 1620
(Republished in 1960 by Liberal Arts Press, New York, p. 93)
Each time a new researcher joins a laboratory, there is a passing on of methods and
technical know-how from existing members, so that expertise is maintained and refined.
As long as the procedures are current, then the information remains easily accessible,
and can be transferred to other research groups by exchange visits, or when a researcher
moves labs. But it is seldom that the methods are published in anything other than an
abbreviated form, or with the inclusion of technical tips that can make the difference
between a method working or failing. With the handling and manipulation of
tetrapyrroles, a discipline that has been carried out over the last hundred years or so,
there have been a number of excellent handbooks published over the years that detail
the characteristics of these important compounds, and provide methods for their
preparation, analysis, and use. However, these books are now mostly out-of-print, and
in many cases had a theoretical rather than practical orientation. In the experience of
one of us (MW), as someone new moving into the area of tetrapyrrole research, despite
collecting all the methods from publications and colleagues, the knowledge was
disjointed and hard to put into practice. Furthermore, it seemed that although many
modern and state-of-the-art procedures were practiced, the simpler, more traditional
methods had been forgotten about, or lost with the retirement of older scientists.
Our goal in producing this book, therefore, was to ask scientists who routinely carry
out the experiments, to describe their basic protocols and technology for the study of
chlorophyll, heme, and related molecules, including technical tips and ways to avoid
common pitfalls. In the editing process, we have worked hard to ensure that the
contributions from each author provided a coherent and accessible introduction to their
topic, be it chemical, biophysical, or molecular biological, and that the protocols were
comprehensible to novices (us!). We are extremely grateful to all the contributors for

v
vi Preface

their willingness to modify their chapters as we requested, and for their forbearance in
the length of time it has taken to complete the project. We would also like to thank Tom
Lanigan at Humana Press Inc., for being prepared to take the project on, and Christine
McAndrew for all her help at a difficult time.
Alison G. Smith
Michael Witty
Contents

Preface ................................................................................................................... v
Contributors ....................................................................................................... ix
1 Laboratory Methods for the Study of Tetrapyrroles
Alison G. Smith and Michael Witty ................................................................ 1
2 Syntheses of Tetrapyrroles
Kevin M. Smith ................................................................................................ 13
3 General Laboratory Methods for Tetrapyrroles
Jerry C. Bommer and Peter Hambright ........................................................ 39
4 Enzymatic Preparation of Tetrapyrrole Intermediates
Martin J. Warren and Peter M. Shoolingin-Jordan .................................. 69
5 Analysis of Biosynthetic Intermediates, 5-Aminolevulinic Acid
to Heme
Chang Kee Lim ................................................................................................... 95
6 Analysis of Intermediates and End Products of the Chlorophyll
Biosynthetic Pathway
Constantin A. Rebeiz ......................................................................................111
7 Analysis of Heme and Hemoproteins
Angela Wilks ..................................................................................................157
8 Hemoproteins Purification and Characterization by Using Aqueous
Two-Phase Systems
Daniel Forciniti ............................................................................................. 185
9 Structural Study of Heme Proteins by Electron Microscopy
of 2-Dimensional Crystals
Terrence G. Frey ............................................................................................. 209
10 Analysis and Reconstitution of Chlorophyll–Proteins
Harald Paulsen and Volkmar H. R. Schmid ............................................. 235
11 Two-Dimensional Crystallization of Chlorophyll Proteins
Georgios Tsiotis ............................................................................................255
12 Biosynthesis and Analysis of Bilins
Matthew J. Terry ........................................................................................... 273

vii
viii Contents

13 Analysis and Reconstitution of Phytochromes


Michael T. McDowell and J. Clark Lagarias ............................................. 293
14 Analysis and Reconstitution of Phycobiliproteins: Methods for the
Characterization of Bilin Attachment Reactions
Wendy M. Schluchter and Donald A. Bryant ............................................ 311
Index................................................................................................................... 335
Contributors

JERRY C. BOMMER • Frontier Scientific/Porphyrin Products, Logan, UT, USA


DONALD A. BRYANT • Department of Biochemistry and Molecular Biology, The
Pennsylvania State University, University Park, PA, USA
DANIEL FORCINITI • Chemical Engineering Department, University of
Missouri-Rolla, Rolla, MO, USA
TERRENCE G. FREY • San Diego State University, San Diego, CA, USA
PETER HAMBRIGHT • Department of Chemistry, Howard University, Washington,
DC, USA
J. CLARK LAGARIAS • University of California–Davis, Davis, CA, USA
CHANG KEE LIM • MRC Bioanalytical Science Group, School of Biological and
Chemical Sciences, Birbeck College, University of London, London, UK
MICHAEL T. MCDOWELL • University of California–Davis, Davis, CA, USA
HARALD PAULSEN • Institut für Allgemeine Botanik der Johannes-Gutenberg,
Univerität Mainz, Mainz, Germany
CONSTANTIN A. REBEIZ • University of Illinois, Urbana, IL, USA
WENDY M. S CHLUCHTER • Department of Biological Sciences, University of
New Orleans, New Orleans, LA, USA
VOLKMAR H. R. SCHMID • Institut für Allgemeine Botanik der Johannes-
Gutenberg, Univerität Mainz, Mainz, Germany
PETER M. SHOOLINGIN-JORDAN • School of Biological Sciences, University of
Southampton, Southamton, UK
ALISON G. SMITH • Department of Plant Sciences, University of Cambridge,
Cambridge, UK
KEVIN M. SMITH • Department of Chemistry, University of California–Davis,
Davis, CA, USA
MATTHEW J. TERRY • University of Southampton, Southampton, UK
GEORGIOS TSIOTIS • Department of Chemistry, University of Crete, Heraklion,
Greece
MARTIN J. WARREN • School of Biological Sciences, Queen Mary Westfield
College, London, UK
ANGELA WILKS • University of Maryland, Baltimore, MD, USA
MICAHEL WITTY • Department of Biochemistry, University of Cambridge,
Cambridge, UK

ix
Laboratory Methods for the
1 Study of Tetrapyrroles

Alison G. Smith and Michael Witty


University of Cambridge, Cambridge, CB2 3EA, UK

1. TETRAPYRROLE STRUCTURE These metals have stabilized oxidation


AND FUNCTION states and solubility. Aside from these two
important properties, tetrapyrroles also
have a subtly substituted ring structure
1.1. Structure of Tetrapyrroles which alters the light absorbance properties
of the conjugated double bond system, the
Tetrapyrroles are a group of organic geometry of metal ion binding (and there-
molecules that includes chlorophyll (Figure fore the type of metal bound), and medi-
1), hemes (Figure 2), bilins (Figure 3), and ates interactions of the tetrapyrrole with
corrins, such as vitamin B12 (37). These proteins.
molecules are also often referred to as por- Most metals and metalloids in the peri-
phyrins, although strictly, these are only odic table have been incorporated into
those compounds with the same oxidation complexes with tetrapyrroles (27), and
state as heme. Chlorophyll, for example, many metals are observed in mineral por-
has one more saturated bond and is there- phyrins (10). However, because of the dif-
fore a chlorin (30). ferences in abundance and differential
A pyrrole is a 5-membered ring contain- stability of the complexes, nickel and vana-
ing one nitrogen, which is colorless, but dium are the most common ions in natur-
when four pyrroles are linked by unsaturat- al abiotic porphyrins, whereas the follow-
ed methine groups, the properties of the ing seven have been seen in living systems:
tetrapyrrole macrocycle are changed dra- Mg, Fe, Mn, Co, Zn, Ni, and V (6).
matically, and two extremely important
characteristics emerge. Tetrapyrroles con- 1.2. Distribution of Tetrapyrroles
tain a ring rich in conjugated double bonds
that absorb light strongly, and they have Porphyrins are spontaneous products of
four nitrogens oriented towards a cavity organic chemical reactions which can be
that may accommodate metal ions and synthesized in Urey-Miller type experi-
allow coordination of the metal ion above ments that mimic prebiotic atmospheric
or below the plane of the macrocycle. conditions: UV irradiation of 5-aminole-

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

1
A.G. Smith and M. Witty

vulinic acid (ALA) has produced pyrroles although most deposits contain only a few
(33), while electrical discharge in the pres- parts per million, some contain significant
ence of pyrrole and formaldehyde has pro- amounts of free or complexed porphyrins,
duced porphyrins (14), and they have been for example the Gibellina sedimentary
detected in sterile meteorites (14,15). deposits, which contain 24 mg/g copper
Porphyrins are chemically stable (30) and nickel porphyrins (29).
and can persist in the environment for Although they are found in abiotic sys-
many millions of years. Porphyrins are tems, most tetrapyrroles are biological, and
found in large fossils such as mollusk shells indeed they are the most conspicuous liv-
(17) and also in molecular fossil forms in ing molecule on earth. Chlorophylls can be
geological strata. The best examples of seen from satellites in space, where vegeta-
these are coal and oil deposits, where they tion types can be identified and used to
are found as mostly nickel(II) and vanadyl predict underlying geology (31). Even
complexes (9). Mineral porphyrins have when viewed from outside, the Earth looks
been detected in sedimentary deposits with enticing because of tetrapyrroles. If there
high organic content laid down as early as are Men from Mars, they would pick on
precambrian times (8). They may precipi- Earth for special interest, and they would
tate to form distinct bedding planes and, be right to do so (25).

Figure 1. The structure of chlorophyll a. Chlorophylls are present in protein complexes in the membrane of photosynthetic bac-
teria and the thylakoid membrane of chloroplasts, where they harvest and trap light energy during photosynthesis (Chapters 10
and 11).

2
Laboratory Methods for the Study of Tetrapyrroles

1.3. Importance of Tetrapyrroles in nature. Tetrapyrroles are also essential in


Nature many other biochemical processes. They
form the prosthetic groups of metalloen-
Although there are a large number of zymes such as sulfite reductase, nitrite
chemical types and ionic conjugates of reductase, peroxidase, and catalase, which
tetrapyrroles, only a few species and their carry out a wide range of oxidation and
derivatives are very abundant in nature: reduction reactions. Vitamin B12 is a
chlorophylls, hemes, and linear tetrapyr- cobalt tetrapyrrole complex that acts as a
roles, the bilins. Tetrapyrroles are important cofactor in methyltransferases, and factor
in living cells because of their physical prop- F430 is a nickel tetrapyrrole that is involved
erties. The tetrapyrrole macrocycle can be in methane formation in certain bacteria.
highly conjugated and absorb visible light Bilins are linear tetrapyrroles with no tight-
strongly, therefore many tetrapyrroles are ly bound metal and are important as the
photochemically active, the most important accessory pigments in algae and as phy-
interaction with light being the capture of tochromobilin, the red-light receptor of
energy by chlorophyll in photosynthesis. higher plants (Chapters 12–14) (21).
Chlorophylls are an essential part of the
photosynthetic apparatus, and the heme of
cytochromes is an essential part of electron 2. A COMMON BIOCHEMICAL
transfer chains in both respiration and PATHWAY
photosynthesis. These two tetrapyrrole
types are essential for the most significant As might be expected from their com-
reduction and oxidation processes in mon structure, all cellular tetrapyrroles are

Figure 2. The structure of protoheme IX.


Hemes are found in a wide range of differ-
ent proteins, including photosynthetic
and respiratory cytochromes involved in
electron transfer, the oxidative enzymes
catalase and peroxidase, cytochrome
P450s, which catalyze mono-oxygenase
reactions, and oxygen-carrying proteins
such as hemoglobin and myoglobin
(Chapters 7, 8, and 9).

3
A.G. Smith and M. Witty

made by a common biochemical pathway Reduced uroporphyrinogen III is


(Figure 4) from the central intermediate formed with methylene rather than meth-
uroporphyrinogen III (for a review see Ref- ine bridges to prevent photoactivity, pro-
erence 37). duction of singlet oxygen, and similar
The first committed precursor is ALA, damaging species. The porphyrinogen
which contains all of the carbon and nitro- form is maintained until the step preceding
gen atoms required by the tetrapyrrole metal ion insertion. Uroporphyrinogen III
nucleus. Two biosynthetic pathways that has two possible fates. On the corrin path-
lead to ALA have evolved (Chapter 4). The way, it is methylated and used to produce
first pathway to be discovered was the so- siroheme, the cofactor of sulfite and nitrite
called Shemin pathway, in which ALA is reductases, or vitamin B12, after the inser-
formed from glycine and succinyl CoA by tion of ferrous iron or cobalt, respectively.
ALA synthase (ALAS). This occurs in ani- Alternatively, uroporphyrinogen III is
mals, fungi, and some bacteria. However, oxidatively decarboxylated in three steps to
the ancestral pathway, characteristic of the form protoporphyrin IX, the last common
majority of bacteria, algae, and plants, is intermediate of heme and chlorophyll
the C5 pathway, in which ALA is formed (Chapters 4 and 5).
from glutamate in three steps involving Ferrochelatase catalyzes insertion of iron
glutamyl-tRNA as an intermediate. Mono- into protoporphyrin IX for heme biosynthe-
mers are formed by condensation of ALA sis, which is followed by insertion into pro-
by ALA dehydratase (Figure 5) to form tein complexes. Heme may be metabolized
porphobilinogen (PBG), which is in turn further to form bilins (Chapters 12–14) by
tetramerized by PBG deaminase (Figure 6) linearization and the loss of the iron atom,
to form the linear intermediate 1-hydrox- catalyzed by heme oxygenase. The insertion
ymethylbilane (or preuroporphyrinogen). of magnesium into protoporphyrin IX by
This is cyclized and isomerized by uropor- magnesium chelatase is the first step of
phyrinogen III synthase, to produce the chlorophyll biosynthesis and is followed by
common intermediate to all cellular further modification of the tetrapyrrole
tetrapyrroles. nucleus by esterification, methylation,

Figure 3. The structure of phytochromobilin. This is the chromophore of phytochromobilin, which is the red-light receptor of
higher plants (Chapter 13). Linear tetrapyrroles are also found as accessory light-harvesting pigments in cyanobacteria and many
algae (Chapter 14).

4
Laboratory Methods for the Study of Tetrapyrroles

Figure 4. The tetrapyrrole biosynthetic pathway, showing the different endproducts and the major intermediates (Chapters 4,
5, 6, and 12). Enzymes are shown in italics.

5
A.G. Smith and M. Witty

reduction of vinyl group, and formation of a only provides the means for photosynthet-
fifth ring to produce protochlorophyllide. In ic organisms to live, but also indirectly sup-
the presence of light, protochlorophyllide is ports almost all life on earth with carbohy-
reduced to form chlorophyllides, which drates and oxygen.
undergo esterification by phytyl diphos-
phate or geranylgeranyldiphosphate to pro- 3.2. Oxidation of Carbohydrates to
duce chlorophyll (see Chapter 6). Produce Usable Energy
Nonphotosynthetic cells obtain their
3. ROLES OF TETRAPYRROLES
energy by the oxidation of carbohydrates,
3.1. Light Harvesting which in aerobic organisms results in the
formation of CO2. This process involves a
Photosynthetic organisms contain a series of reduction-oxidation (redox) reac-
sophisticated system of several hundred tions whereby the large gap in oxidation
chlorophylls (or bacteriochlorophylls) and state between carbohydrate and carbon
other accessory pigments, which act as dioxide is released in a series of gentle and
antennae to absorb light and pass the ener- efficient steps, with oxygen as the final
gy to special chlorophylls in reaction cen- electron acceptor. Transition metals, such
ters. Here the light energy is trapped as as the iron found in heme (Figure 2), are
excited electrons, which are then trans- well suited to catalyze these reactions
ferred through an electron transfer chain to because they contain d-electron orbital sys-
generate ATP. In higher plants, algae, and tems with small differences in energy lev-
cyanobacteria, this process results in the els, thus allowing a range of oxidation
oxidation of water to evolve molecular oxy- states so that energy can be released in a
gen and the production of reduced nicoti- controlled and useful way (cf section 3.4).
namide adenine dinucleotide phosphate In the bacterial membrane, and the
(NADPH) (see Chapters 10 and 11 for mitochondria of eukaryotes, a series of pro-
more detail). The ATP and NADPH gen- tein complexes containing cofactors, which
erated by the light-dependent reactions are include heme (see Chapter 7), undergo a
used to fix CO2 into organic combination series of reversible redox reactions that gen-
via the Calvin cycle. Photosynthesis not erates ATP. In this respect, the process of

Figure 5. The formation of


a pyrrole. The reaction cat-
alyzed by ALA dehydratase.

6
Laboratory Methods for the Study of Tetrapyrroles

Figure 6. The formation of a tetrapyrrole. The reaction catalyzed by PBG deaminase. The holoenzyme (E) contains an active site
dipyrromethane cofactor. This is used to accept PBG monomers and form enzyme substrate complexes. A, acetate; P, propionate
moiety. Pyrrole rings are labeled A, B, C, and D.

7
A.G. Smith and M. Witty

oxidative phosphorylation is similar to that ply from one beat to the next. Intracellular
in photosynthesis (section 3.1), reflecting myoglobin is also found in bacteria, proto-
common evolutionary ancestors of some of zoans, plants, and invertebrates (32).
the components involved. Plants also contain leghemoglobin used
for the homeostasis of oxygen. The best
3.3. Transport and Homeostasis of studied example is in the legume root nod-
Oxygen ule, where symbiotic bacteria consume
large amounts of energy during the reduc-
Metabolism requires the consumption tion of atmospheric nitrogen to ammonia,
of large amounts of oxygen, and therefore which is then available to the host plant.
transport from the atmosphere to cells Leghemoglobin facilitates the maintenance
buried deep in animal bodies is necessary of high levels of oxygen needed for bacteri-
to allow a rapid rate of metabolism. While al respiration, while preventing poisoning
several kinds of oxygen transport proteins of the nitrogen fixing machinery by molec-
exist, including hemerythrins (non-heme ular oxygen (2). Unlike in animals, the
iron proteins) and hemocyanins (copper protein is not circulated, but rather simple
proteins), globins are the most abundant diffusion down an oxygen gradient created
and widespread oxygen transport protein by bacterial respiration promotes transport
(12). They use reversible oxygen coordina- of oxygen from the outside.
tion to protoheme IX iron (FeII) for trans-
port of oxygen from respiratory organs 3.4. Protection of Cellular Processes
throughout the animal body. from Reactive Oxygen Species
Hemoglobins transport oxygen in many
animal groups, and most have similar As well as the desired biological reaction
structures and functions (35). In circulat- of oxygen as a terminal acceptor of electrons
ing red blood cells, hemoglobins consist of in the controlled oxidation of carbohydrates
17 kDa polypeptides, each containing a in respiration, oxygen will also react with
heme group that can bind one oxygen mol- electrons encountered at random, to pro-
ecule. Hemoglobins are typically tetramers duce reactive oxygen species (ROS) such as
that allow for cooperative binding and oxygen radicals and superoxide. These are
release of oxygen, depending on the PO of very harmful to living systems, causing lipid
2
the surrounding tissue. peroxidation, membrane damage, and
In addition to oxygen transport, hemo- genetic mutation. Cells contain a number
globins are associated with other important of enzymes that act to remove these inter-
functions. For example, circulating hemo- mediates quickly. Many of these oxidases
globin I of the swamp clam Lucina pectina- are heme-containing proteins, including
ta is involved in sulfide transport. Sulfides peroxidases and catalase (7).
are bound with high affinity within a cage
of heme and three phenylalanine residues 3.5. Taking Advantage of Reactive
and are released to symbiotic bacteria upon Oxygen Species
reduction (3).
Myoglobin is a monomeric protein that Although ROS can be harmful, organ-
contributes to the transport of oxygen by isms have evolved systems in which they
diffusion, and large amounts are found in are useful. One important example of this
skeletal and cardiac muscles of mammals. is the biosynthesis and degradation of
In cardiac muscle, myoglobin acts as a lignin. Lignocellulose is a composite of
short-term oxygen buffer, smoothing sup- lignin and cellulose and probably the most
8
Laboratory Methods for the Study of Tetrapyrroles

abundant organic molecule in the bios- prosthetic group, tetrapyrroles also func-
phere, functioning as material for mechan- tion in key regulatory processes. These
ical strength in wood. Rather than a regular include control of gene expression, cellular
polymer such as cellulose, lignin is synthe- signaling pathways, and control of protein
sized by peroxidases, which oxidize phenol- transport within the cell.
propane units (such as coniferyl alcohol) to An example is heme-mediated feedback
form reactive radical species. These poly- control of its own synthesis, which appears
merize in an irregular fashion to form to occur in all groups of organisms, most
lignin, which thereby contains a wide array importantly at the production of the first
of chemical linkages (38). Because of this committed precursor ALA. In plants, there
unsymmetrical arrangement, lignin is is evidence that heme inhibits the enzyme
unusually resistant to enzymatic break- glutamyl-tRNA reductase (36). In animals,
down, and only a few microbes, such as the although heme inhibits the activity of
white rot fungi, have enzymes capable of ALAS, control is exerted at several other
doing so (20). Lignin is also an important points as well. In mammals, there are two
byproduct of the paper industry, which forms of the enzyme, constitutive and ery-
generates about 30 million tons of unused throid-specific. Liver ALAS is inhibited by
lignin per year, in a process involving harsh heme in a negative feedback loop (11) to
chemical treatments (13). Genetic modifi- maintain levels of heme production for the
cation of tree species to reduce lignin con- maintenance of cellular processes. This feed-
tent is being explored as a means of avoid- back regulation is achieved by a combina-
ing this costly and polluting process (34). tion of effects including inhibition of ALAS
A more general, and essential, role of gene expression, increased ALAS mRNA
these reactive species is found in both degradation, and inhibition of pre-ALAS
plants and animals, where ROS have been protein transport to the mitochondrion
shown to modulate a wide range of cellular (19), with only a minor contribution by
and physiological processes, acting as part inhibition of ALAS catalytic activity (28).
of the signal transduction pathway. For In contrast to the liver, in erythroid cells,
example, one of the earliest responses to transcription of the ALAS gene, together
pathogen attack is a marked increase in with genes for later enzymes in the pathway
ROS in the infected tissue, produced in and for globins, is stimulated by heme to
part by the activity of plasma membrane- produce the large amounts of heme needed
bound NADPH oxidase, a hemoprotein for hemoglobin in red blood cells.
complex. The ROS, in turn, act as a trigger In yeast, expression of the ALAS gene is
for defense responses, such as modification controlled by heme and mediated through
of membrane permeability and ion fluxes, the transcription factor HAP1, which
and systemic acquired resistance in plants binds heme for activity. The binding
(1). Similarly in animals, ROS influence domain contains multiple copies of a short
signaling cascades and transcriptional– motif, which is also found in the mito-
posttranscriptional control of gene expres- chondrial transit peptide of mammalian
sion, thereby playing an essential role in ALAS. This motif, involved in transient
processes such as apoptosis (23). binding of heme, is quite different to the
more stable heme-binding domain of
3.6. Control of Metabolic and Cellular cytochromes and globins (39).
Processes by Signaling There is accumulating evidence that
tetrapyrrole intermediates play a role in sig-
As well as functioning as an enzymic naling. In plants, there is coordination
9
A.G. Smith and M. Witty

between the chloroplast and the nucleus, rigid structures that they are able to form
such that nuclear-encoded genes for chlo- and the fact that they can coordinate a
roplast-targeted proteins are transcribed number of metal ions which are involved
only in cells with functional chloroplasts. in the catalysis. For example, using por-
Although the exact identity of the so-called phyrin molecular boxes and zinc coordina-
“plastid-factor” remains elusive, plant tion, it has been possible to influence the
mutants with defects in certain steps of the stereospecificity of reactions by the geo-
tetrapyrrole biosynthetic pathway have metrical constraints of a host cavity (26).
altered plastid-nuclear signaling (24). Other novel uses of tetrapyrroles have
been established in clinical medicine, in
3.7. Subtle Pigmentation particular for the treatment of cancer cells,
in a technique called photodynamic thera-
While plants are green because of the py (PDT). The rationale behind the meth-
presence of chlorophyll and animal tissues od is to load the cancerous cells with pho-
are largely red due to heme, some of the tosensitizing porphyrin mixtures, which,
more subtle animal colors are also conferred upon irradiation with visible light, cause
by tetrapyrroles. The cuticle of birds’ eggs the production of singlet oxygen, thereby
with colored shells contain tetrapyrroles leading to the destruction of the cells as
which contribute to their camouflage. Most described in section 3.4. Porphyrins are
markings and pigmentation are due to pro- ideal compounds for this technique, not
toporphyrin IX, which is associated with only because of their light absorption prop-
brown and black coloring. Blue eggshells erties, but also because there is some pref-
are associated with biliverdin IXα, and erential uptake of these molecules by
green eggshells are associated with zinc- tumor cells. Initially, in the 1960s and
biliverdin IXα with traces of copropor- 1970s, the major photosensitizers used
phryin III (18). The feathers of some birds were hematoporphyrins and related prepa-
also contain tetrapyrrole pigments. The rations derived from acid extraction of
feathers of Turocos contain red turacin (cop- blood (or hemoglobin), and therefore, are
per-uroporphyrin III) and green tura- not chemically defined compounds. How-
coverdin (22). Uroporphryin I is found in ever, since 1980, new sensitizers have been
many calcified mollusk tissues such as shells developed, including chlorins and phthalo-
(17) and pearls (16), though the function of cyanines, which have been chemically syn-
the tetrapyrrole is unknown. thesized (5).

3.8. Artificial Uses of Tetrapyrroles


4. LABORATORY METHODS FOR
In addition to their importance in biolo- THE STUDY OF TETRAPYRROLES
gy, tetrapyrroles are increasingly of interest
to a much wider range of researchers. For Tetrapyrroles are clearly a diverse and
example, chemists are able to create syn- important group of molecules, and re-
thetic molecules which mimic the recogni- searchers from a wide range of different
tion and catalytic properties of enzymes. A fields may wish to study them, whether it
particular aspect of this work is catalysis of be a clinician using them for PDT, an ecol-
reactions for which there are no known ogist studying the chlorophyll composition
natural enzymes, such as Diels-Alder reac- of leaves in a tropical forest, or a cell biolo-
tions (4). Porphyrins have proved very use- gist investigating the function of specific
ful for this sort of study because of the hemoproteins. However, as we know from
10
Laboratory Methods for the Study of Tetrapyrroles

our own laboratory experience, there are due to exocyclic ring size. Inorg. Chem. 35:199-209.
certain “tricks-of-the-trade” which are nec- 10.Dailey, K.K. and T.B. Rauchfuss. 1997. π-Complex-
es of metalloporphyrins as model intermediates in
essary to use in order to carry out success- hydrodemetallation (HDM) catalysis. Polyhedron
ful experiments. In this volume, we have 16:3129-3136.
selected articles written by people who 11.Granick, S. 1966. The induction in vitro of the syn-
thesis of δ-aminolevulinic acid synthase in chemical
actually carry out these procedures on a porphyria: a response to certain drugs, sex hormones,
daily basis in their own laboratories. Each and foreign chemicals. J. Biol. Chem. 241:1359-
chapter provides an overview of the topic 1375.
12.Hardison, R. 1998. Hemoglobins from bacteria to
with general information on the experi- man: evolution of different patterns of gene expression.
mental approach, as well as a number of J. Exp. Biol. 201:1099-1117.
13.Hartley, B.S., P.M.A. Broda, and P.J. Senior. 1987.
step-by-step procedures, which should pro- Technology in the 1990s: Utilization of Lignocellulosic
vide the basis for any novice tetrapyrrolo- Wastes. The Royal Society, London.
gist taking their first steps into this field. 14.Hodgson, G.W. and B.L. Baker. 1964. Evidence for
porphyrins in the orgueil meteorite. Nature 202:125-
131.
15.Hodgson, G.W. and B.L. Baker. 1967. Porphyrin
ABBREVIATIONS abiogenesis from pyrrole and formaldehyde under sim-
ulated geochemical conditions. Nature 216:29-32.
16.Iwahashi, Y. and S. Akamatsu. 1994. Porphyrin pig-
ALA, 5-aminolevulinic acid; ALAS, ment in black-lip pearls and its application to pearl
ALA synthase; PBG, porphobilinogen; identification. Fisheries Sci. 60:69-71.
17.Kennedy, G.Y. 1975. Porphyrins in invertebrates. Ann.
PDT, photodynamic therapy; ROS, reac- NY Acad. Sci. 244:662-673.
tive oxygen species. 18.Kennedy, G.Y. and H.G. Vevers. 1976. A survey of
avian eggshell pigments. Comp. Biochem. Physiol. B
55:117-123.
19.Lathrop, J.T. and M.P. Timko. 1993. Regulation by
REFERENCES heme of mitochondrial protein-transport through a
conserved amino-acid motif. Science 259:522-525.
1.Alvarez, M.E., R.I. Pennell, P.J. Meijer, A. Ishikawa, 20.Leonowicz, A., A. Matuszewska, J. Luterek, D.
R.A. Dixon, and C. Lamb. 1998. Reactive oxygen Ziegenhagen, M. Wojtas-Wasilewska, N.S. Cho, M.
intermediates mediate a systemic signal network in the Hofrichte, and J. Rogalski. 1999. Biodegradation of
establishment of plant immunity. Cell 98:773-784. lignin by white rot fungi. Fungal Genet. Biol. 27:175-
2.Appleby, C.A. 1984. Leghemoglobin and rhizobium 185.
respiration. Annu. Rev. Plant Physiol. 35:443-478. 21.McDonagh, A.F. 1979. Bile pigments: bilatrienes and
3.Bolognesi, M., C. Rosano, R. Losso, A. Borassi, M. 5,15-biladienes, p. 293-491. In D. Dolphin (Ed.), The
Rizzi, J.B. Wittenberg, A. Boffi, and P. Ascenzi. 1999. Porphyrins, Vol. 1. Academic Press, London.
Cyanide binding to Lucina pectinata hemoglobin I and 22.Nicholas, R.E.H. and C. Rimington. 1952. Isolation
to sperm whale myoglobin: an X-ray crystallographic of unequivocal uroporphyrin III, a further study of
study. Biophys. J. 77:1093-1099. turacin. Biochem. J. 50:194-201.
4.Bonarlaw, R.P., L.G. Mackay, C.J. Walter, V. Mar- 23.Nose K. 2000. Role of reactive oxygen species in the
vaud, and J.K.M. Sanders. 1994. Towards synthetic regulation of physiological functions. Biol. Pharmacol.
enzymes based on porphyrins and steroids. Pure Appl. Bull. 23:897-903.
Chem. 66:803-810. 24.Oster, U., H. Brunner, and W. Rudiger. 1996. The
5.Bonnett, R. 1999. Photodynamic therapy in historical greening process in cress seedlings. 5. Possible interfer-
perspective. Rev. Contemp. Pharmacother. 10:1-17. ence of chlorophyll precursors, accumulated after thu-
6.Buchler, J.W. 1975. Static coordination chemistry of japlicin treatment, with light-regulated expression of
metalloporphyrins, p. 157-231. In K.M. Smith (Ed.), Lhc genes. J. Photochem. Photobiol. B 36:255-261.
Porphyrins and Metalloporphyrins. Elsevier, Amster- 25.Sagan, C., W.R. Thompson, R. Carlson, D. Gurnett,
dam. and C. Hord. 1993. A search for life on earth from the
7.Cadenas, E. 1989. Biochemistry of oxygen toxicity. Galileo spacecraft. Nature 365:715-721.
Annu. Rev. Biochem. 58:79-100. 26.Sanders, J.K.M. 1998. Supramolecular catalysis in
8.Callot, H.J. 1991. Geochemistry of chlorophylls, p. transition. Chem. Eur. J. 4:1378-1383.
339-364. In H. Scheer (Ed.) Chlorophylls. CRC Press, 27.Sanders, J.K.M., N. Bampos, Z. Clyde-Watson, S.L.
Boca Raton. Darling, J.C. Hawley, H.J. Kim, C.C. Mak, and S.J.
9.Czernuszewicz, R.S., J.G. Rankin, and T.D. Lash. Webb. 2000. Axial coordination chemistry of metallo-
1996. Fingerprinting petroporphyrin structures with porphyrins, p. 349-390. In K.M. Kadish, K.M. Smith,
vibrational spectroscopy. 4. Resonance raman spectra and R. Guilard (Eds.), The Porphyrin Handbook. Aca-
of nickel(II) cycloalkanoporphyrins: structural effects demic Press, London.

11
A.G. Smith and M. Witty

28.Sassa, S. and T. Nagai. 1996. The role of heme in gene tors from Antirrhinum regulate phenylpropanoid and
expression. Int. J. Hematol. 63:167-178. lignin biosynthesis in transgenic tobacco. Plant Cell
29.Schaeffer, P., R. Ocampo, H.J. Callot, and P. 10:135-154.
Albrecht. 1993. Extraction of bound porphyrins from 35.Terwilliger, N.B. 1998. Functional adaptations of
suphur-rich sediments and their use for reconstruction oxygen-transport proteins. J. Exp. Biol. 201:1085-
of palaeoenvironments. Nature 364:133-136. 1098.
30.Smith, K.M. 1975. Porphyrins and Metallopor- 36.Vothknecht, U.C., C.G. Kannangara, and D. Wett-
phyrins, p. 829-836. Elsevier, Amsterdam. stein. 1998. Barley glutamyl tRNA(Glu) reductase:
31.Smith, M.O., S. Jacquemond, M. Verstraete, and Y. mutations affecting haem inhibition and enzyme activ-
Govaerts. 1999. Geobotany: vegetation mapping for ity. Phytochemistry 47:513-519.
earth sciences, p. 189-248. In Remote Sensing for the 37.Warren, M.J. and A.I. Scott. 1990. Tetrapyrrole
Earth Sciences, Manual of Remote Sensing, Vol. 3. assembly and modification into the ligands of biologi-
John Wiley & Sons, New York. cally functional cofactors. Trends Biochem. Sci.
32.Suzuki, T. and K. Imai. 1998. Evolution of myoglo- 15:486-491.
bin. Cell. Mol. Life Sci. 54:979-1004. 38.Whetten, R.W., J.J. MacKay, and R.R. Sedoroff.
33.Szutka, A. 1966. Formation of pyrrolic compounds by 1998. Recent advances in understanding lignin biosyn-
ultra-violet irradiation of δ-aminolevulinic acid. thesis. Annu. Rev. Plant Physiol. Plant Mol. Biol.
Nature 212:401-402. 49:585-609.
34.Tamagnone, L., A. Merida, A. Parr, S. Mackay, F.A. 39.Zhang, L. and L. Guarente. 1995. Heme binds to a
Culianez-Macia, K. Roberts, and C. Martin. 1998. short sequence that serves a regulatory function in
The AmMYB308 and AmMYB330 transcription fac- diverse proteins. EMBO J. 14:313-320.

12
Syntheses of Tetrapyrroles
2
Kevin M. Smith
Department of Chemistry, University of California–Davis,
Davis, CA, USA

1. INTRODUCTION tors. An up-to-date and highly detailed


description of the synthetic art of por-
This chapter addresses basic methodol- phyrin chemistry can be found in The Por-
ogy that can be used to obtain tetrapyrrole phyrin Handbook (39).
macrocycles in the porphyrin and chlorin At the outset it must be mentioned that
series from natural materials and some sim- a certain degree of expertise in experimen-
ple methods for the total chemical synthe- tal organic chemistry is essential for success
sis of typical pyrroles and porphyrins. The in the endeavors described herein; also
aim is to provide investigators with enough essential are the appropriate laboratory
information to decide whether to take on equipment (fume hoods, rotary evapora-
the task of preparing samples of useful tors, temperature controlled reaction mon-
porphyrin and chlorophyll derivatives or itors, chromatographic equipment, etc.)
whether to simply purchase them or col- and glassware. Since hazardous waste
laborate with other individuals more expert chemicals and solvents will also need to be
in the established synthetic procedures. disposed of, approved facilities for these
The procedures reported herein are usually responsibilities must also be available.
those which are easiest for the nonexpert to In terms of chemical technique and pro-
perform, while at the same time being suf- cedures, pyrrole and porphyrin derivatives
ficient to provide pure samples of the tend to be easy to work with. With the
required product. exception of porphyrinogens, they usually
The porphyrin field has a very rich his- do not require stringent exclusion of oxy-
tory; Hans Fischer’s books present a labo- gen and water vapor (as is the case with
ratory approach to synthesis of porphyrin much of the rest of organometallic chem-
compounds dating back from the 1930s istry), they are stable at room temperature
(20,22,24). In 1975, Porphyrins and Metal- and higher temperatures, and they can be
loporphyrins was published (64); this con- purified by recrystallization and chro-
tained a fairly detailed laboratory methods matography in the air at room tempera-
section, which was useful at that time and ture. As might be expected with any col-
is probably still useful to many investiga- ored compound (which will be absorbing

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

13
K.M. Smith

light of various wavelengths and therefore a well-characterized so-called Diels-Alder


will be accessing excited electronic reaction is known to take place in the pres-
states—porphyrins fluoresce strongly), ence of oxygen and light to afford a mix-
attempts should be made routinely to keep ture of photoprotoporphyrin and isopho-
porphyrin and chlorin compounds out of toprotoporphyrin IX dimethyl ester [3 and
the light; this is not difficult, and alu- 4, respectively] (7,34); this represents the
minum foil wrapped around a sample flask extreme of normal porphyrin photolability
or around a chromatography column usu- and is caused by the presence of the 3- or
ally suffices. In the particular case of proto- 8-vinyl groups. If you can successfully han-
porphyrin IX [1] or its dimethyl ester [2], dle protoporphyrin IX without continually

14
Syntheses of Tetrapyrroles

generating two polar green bands upon 3. PREPARATION OF PORPHYRINS


chromatography, you should do just fine. AND CHLORINS BY DEGRADA-
Further advice on the specific requirements TION OF NATURAL PIGMENTS
for handling these molecules can be found
in Chapter 3 by Bommer and Hambright. It is truly fortunate that massive amounts
of natural products containing both hemin
[11] and chlorophylls a and b [12,13] can
2. NOMENCLATURE be accessed. Fischer’s three volumes
(20,22,24), Die Chemie des Pyrrols, report
Over the years, two different schemes for an astonishing array of procedures for
nomenclature of porphyrin systems have obtaining tetrapyrrole compounds from
been used. The Fischer system for porphyrin natural sources. Thus, large volumes of
nomenclature [structure 5] provides a link blood can be processed to provide hemin in
back to the rich history of porphyrin chem- kilogram quantities. From hemin, a large
istry mentioned above—many trivial names number of porphyrins and derivatives can
were generated which, particularly in the be obtained (see later). Similarly, chloro-
field of chlorophyll chemistry, are almost phyll derivatives in the a and b series can be
impossible to do without. Likewise, in the obtained by extraction of leaves, usually
porphyrin field, there are some names that spinach. But if only chlorophyll a deriva-
are indispensable (e.g., protoporphyrin IX, tives are desired, one can take advantage of
the “first” porphyrin, and deuteroporphyrin the fact that certain algae, such as Spirulina,
IX, the “second” porphyrin); the “IX” given produce only chlorophyll a; thus, a labori-
after the porphyrin name refers to the (sec- ous separation of the chlorophyll a and b
ondary) type-IX arrangement of the por- series can be avoided. If chlorophyll b deriv-
phyrin substituents. When there are only atives are required, there used to be no
two types of substituent, for example methyl option but to extract plant chlorophylls and
and ethyl, with one methyl and one ethyl on perform the chromatographic separation,
each pyrrole ring, only four “primary type- either by preparative scale high-perfor-
isomers” [6–9] of the so-called “etio”por- mance liquid chromatography (HPLC) or
phyrins are possible. When there are three by gravity column chromatography on
kinds of substituent (as with the methyl, sucrose. Some years ago, a chemical deriva-
vinyl, and propionic substituents in proto- tization approach was developed to make
porphyrin IX), no less than fifteen “sec- the chromatographic separations more
ondary type-isomers” are possible (provided palatable, and that will be discussed later.
there is one methyl on each pyrrole sub-
unit), and the type-IX isomer is the biologi- 3.1. Porphyrins from Hemoglobin
cally significant one. In the primary type
isomer series, type-III is the biologically sig- 3.1.1. Hemin [11]
nificant arrangement. But all that said, and
given the near impossibility of naming some Because of the relative ease with which
porphyrin and chlorophyll derivatives with- hemin can be obtained from blood, it can
out the use of Fischer’s trivial names, the be purchased from a number of chemical
International Union of Pure and Applied companies at costs around a few dollars per
Chemistry (IUPAC) system of nomencla- gram. The method of choice (19) for prepa-
ture [structure 10] is the officially adopted ration of hemin from blood involves addi-
nomenclature system, and this will have to tion of heparinized, citrated, or defibrinated
be used in this chapter. blood to hot acetic acid containing sodium
15
K.M. Smith

chloride. After cooling and removal of coag- (16,44). Hemes [iron(II) porphyrins] can be
ulated protein (usually with a wooden obtained from hemins [iron(III) chloride
stick), the hemin separates and can be col- porphyrins] most commonly by reduction
lected by filtration. Alternatively, the messy with sodium dithionite under nitrogen or
protein can be precipitated by addition of a argon. Since autoxidation of iron(II) to
solution of strontium chloride, followed by iron(III) porphyrins is very facile in air, use
concentration to give hemin as above of nitrogen or (preferably the heavier) argon

16
Syntheses of Tetrapyrroles

is absolutely essential. Chromatographic 3.1.3. Mesoporphyrin IX [15]


purification of hemins is best accomplished
on the corresponding (usually methyl) Mesoporphyrin IX [15] is related to
esters; but hemins [e.g., 11] bearing car- protoporphyrin IX [1] with the important
boxylic acid groups should not be esterified difference that the sensitive 3- and 8-vinyl
with diazomethane—a side-reaction takes groups in [1] are replaced with durable
place with the iron atom. For methyl esters ethyl groups—hence, mesoporphyrin IX
(the simplest and best ester to use under does not undergo the photo-oxygenation
normal circumstances), 5% sulfuric acid in reaction mentioned above for protopor-
methanol is the best mixture to use (CAU- phyrin. Early biosynthetic investigations of
TION: take care to gently add the acid to the the metabolism of protoporphyrin IX often
stirred and cooled alcohol) (66). Hemin used the easy to handle mesoporphyrin IX
esters can be hydrolyzed to the correspond- [15], and so incorporated a hydrogenation
ing free carboxylic acids using base (66). step to accomplish reduction of the 3- and
8-vinyl groups in protoporphyrin IX (9);
3.1.2. Protoporphyrin IX [1] the method of choice (22) is catalytic
hydrogenation over palladium in formic
Protoporphyrin IX [1] is the product acid. Either protoporphyrin IX, its ester, or
obtained by removal of iron from hemin hemin are used, and the iron in [11] is
[11], but acid alone does not accomplish removed concomitantly during the reac-
this result because iron(III) is very difficult tion. Mesohemin IX [16], the iron(III)
to eject from a porphyrin. Commercial chloride of mesoporphyrin IX, is best
samples of protoporphyrin IX are usually obtained by the introduction of iron into
not very pure because of the sensitivity of [15] rather than by reduction of hemin
protoporphyrin to photo-oxygenation at [11]. Esterification of mesoporphyrin IX
the vinyl groups (see above). The best can be carried out using either diazo-
method for obtaining protoporphyrin IX is methane or sulfuric acid acid–alcohol.
to treat hemin [11] with ferrous sulfate in
hydrochloric acid (46,51,52); the hemin is 3.1.4. Hematoporphyrin IX [14]
reduced to heme, and the iron(II), in strict
contrast to iron (III), is readily removed by Hematoporphyrin IX [14] was the first
the acid. Commercial hematoporphyrin IX porphyrin to be isolated (in 1867) (69); it
[14] is often very pure (unlike protopor- was obtained by treatment of blood with
phyrin IX), so a method for the preparation concentrated sulfuric acid. Nominally,
of [1] by double dehydration of hematopor- hematoporphyrin IX [14] is obtained
phyrin IX [14] has been reported (40). This chemically from protoporphyrin by hydra-
involves brief heating of [14] with toluene tion of both of the 3- and 8-vinyl groups.
p-sulfonic acid in 1,2-dichlorobenzene. The Since the 31- and 81-carbon atoms are chi-
dimethyl ester [2] of protoporphyrin IX can ral in [14], a mixture of four optical iso-
be obtained by esterification with either dia- mers (enantiomers and diastereomers) is
zomethane (CAUTION: diazomethane can be obtained, and these can be separated by
explosive under certain circumstances) or HPLC. Porphyrin [14] can also be pur-
with methanol–sulfuric acid (CAUTION) as chased from commercial sources.
mentioned above for hemin. The very use- Using protoporphyrin IX [1] as the
ful Grinstein method (33) can be used to starting material, hematoporphyrin IX is
prepare protoporphyrin IX dimethyl ester best prepared by treatment with hydrogen
[2] in one step from hemin [11]. bromide in acetic acid, followed by hydrol-
17
K.M. Smith

ysis of the resulting 3,8-di(1-bromoethyl)- [e.g., Reference 66]. 3,8-Diacetyldeutero-


derivative [17] with water (22). If a com- porphyrin IX [21] can be prepared by oxi-
mon alcohol (R1OH) such as methanol dation of hematoporphyrin IX (62), or by
(R1 = CH3) is used in this last stage, then Friedel-Crafts acetylation of deuterohemin
the 3,8-di(1-alkoxyethyl) analogue [18] is IX [22] using acetic anhydride and pyri-
produced. Alternatively, reduction of 3,8- dine, followed by removal of the iron (66).
diacetyldeuteroporphyrin IX dimethyl Use of sulfuric acid and methanol to ester-
ester [19] with sodium borohydride affords ify the propionic acids in [14] is not
hematoporphyrin IX dimethyl ester [20] advised because acid-catalyzed dehydra-

18
Syntheses of Tetrapyrroles

tion, or ether formation, at the 3,8-(1- [11] (29). Deuterohemin [22] can be
hydroxyethyl) groups is a problem; it is obtained from “proto” hemin by brief heat-
best to use diazomethane in methanol to ing of [11] in a resorcinol melt (60), via the
obtain the dimethyl ester [20] (CAUTION). so-called Schumm reaction in which the
vinyl groups are replaced by hydrogen
3.1.5. Deuteroporphyrin IX [23] atoms (10,12,17,42). Demetalation, as
reported above for the preparation of pro-
Deuteroporphyrin IX [23] is of signifi- toporphyrin IX from hemin, then affords
cant historical importance because it was deuteroporphyrin IX [23].
the first porphyrin isolated in Fischer’s Numerous 3,8-disubstitution products
Nobel Prize winning synthesis of hemin (and 3- or 8-monosubstitution analogues)

19
K.M. Smith

of deuteroporphyrin IX and its esters can 3.2 Porphyrins and Chlorins from Plants
be prepared, usually by way of aromatic and Algae
electrophilic substitution on the hemin
or its copper(II) complex. A typical In this section, some simple degradation
example is 3,8-diacetyldeuteroporphyrin reactions, which furnish porphyrins and
IX [21] (see above), which was also an chlorins in useful quantities from plants
intermediate in the Fischer’s hemin total and algae, will be described.
synthesis. The traditional source for chlorophylls a
[12] and b [13], usually present in a ratio
3.1.6. Coproporphyrin III [24] of about 3:1, was leaf tissue, usually
spinach (25,68). A very useful chemical
Coproporphyrin III [24] is a biologically adjunct for simplification of the mandato-
significant porphyrin because its hexahydro- ry chromatographic separation of the
derivative, coproporphyrinogen III [25], is a chlorophyll a and b pigments has been
colorless intermediate on the pathway reported (41); it employs the Girard
between uroporphyrinogen III [26], proto- reagent T as a means of dramatically
porphyrinogen IX [27], and protoporphyrin increasing the polarity of the series b com-
IX [1] in normal porphyrin metabolism. ponent in the mixture. For example, reac-
Under normal circumstances, the amount of tion of methyl pheophorbide a [28] and b
[25] present at steady state is small. Howev- [29] mixture (see above) with Girard’s
er, biological oxidation of coproporphyrino- reagent T gives a mixture consisting of
gen III yields the colored coproporphyrin unreacted a series compound, i.e., methyl
III, which takes it out of the normal meta- pheophorbide a [28], and the salt [30]
bolic sequence. Hence, certain diseases of from the b series. Column chromatography
porphyrin metabolism can result in a then achieves a very simple separation in
buildup of excess photochemically active which [30] remains adsorbed to the top of
porphyrins in tissues; such diseases are the column, whereas the relatively nonpo-
known collectively as porphyrias. Chemical- lar a series compound [28] is eluted quick-
ly, porphyrinogens can be oxidized very effi- ly. Use of a polar solvent then elutes the b
ciently to porphyrins by use of 2,3-dichloro- series salt, which can be hydrolyzed to give
5,6-dicyanobenzoquinone (DDQ). If pure methyl pheophorbide b [29].
biosynthetic work using porphyrinogens is Investigators wishing only to deal with
to be carried out, the corresponding por- chlorophyll derivatives in the a series were
phyrin can usually be reduced to por- advantaged when it was shown that Spir-
phyrinogen using sodium amalgam or cat- ulina maxima (from Mexico) or S. pacifica
alytic hydrogenation (15). When vinyl (from Hawaii) contain only the chloro-
groups are present on the porphyrin macro- phyll a series of pigments. On account of
cycle, of course, only the sodium amalgam the fairly drastic extraction conditions,
route is recommended—catalytic hydro- chlorophyll a itself is usually not obtained
genation will probably reduce the vinyls to directly from the alga, but large quantities
ethyls. It must be kept in mind when han- of pheophytin a [31] and methyl pheo-
dling porphyrinogens, that oxygen and light phorbide a [28] (up to 0.4% measured by
can efficiently oxidize the hexahydro mater- dry weight) can be obtained (67).
ial to the porphyrin level, which will make it Treatment of the plant chlorophylls
inactive in biosynthetic investigations—the (either separately or as a mixture) with
first true porphyrin in porphyrin biosynthe- mild acid gives the metal-free pheophytins
sis is protoporphyrin IX itself. a [31] and b [32]; this, as a dried paste, is
20
Syntheses of Tetrapyrroles

usually the form in which the pigments are pheophorbides still contain one ester, and
stored prior to further degradation to use- that hydrolysis of this ester will cause con-
ful materials. Hydrolysis of the pheo- comitant decarboxylation on ring E).
phytins gives the corresponding pheophor- Alternatively, and preferably (for ease of
bides a [33] and b [34]; (note that the handling), methanolysis of pheophytin a

21
K.M. Smith

22
Syntheses of Tetrapyrroles

or b provides the corresponding methyl 18 [38], which bears a very useful anhy-
pheophorbides a or b [28 or 29, respective- dride ring [45]. On the other hand, dia-
ly]—these contain two methyl esters. zomethane esterification (CAUTION) yields
Transesterification of the phytyl ester for purpurin 7 trimethyl ester [39] (26–
methyl, without removal of the magnesium 28,45). Heating of [39] in collidine gives a
atom, can be accomplished to afford the diversely substituted porphyrin, 3-vinyl-
methyl chlorophyllides [35] and [36] (26). rhodoporphyrin XV dimethyl ester [40]
A number of simple to perform but (28). If the so-called “meso” (i.e., 3-ethyl
mechanistically complex reactions can be instead of 3-vinyl) series of pigments is
carried out on chlorophyll derivatives. For used, another porphyrin, rhodoporphyrin
example, oxidation of pheophytin a [31] XV dimethyl ester [41], is obtained.
under highly alkaline conditions accom- The isocyclic ring (E) in chlorophylls
plishes cleavage the 131-132 bond in the β- and their derivatives contains a β-ketoester
ketoester ring E, with hydrolysis of the of function which imparts a high degree of
phytyl ester, to give Fischer’s “unstable chemical reactivity upon the compounds
chlorin” [37] (28). Simple evaporation of containing it. Such lability is often a disad-
the solution affords the so-called purpurin vantage in the use of chlorophyll derivatives

23
K.M. Smith

for specific purposes; the spectrum of are much less reactive than are conjugated
chemical reactivity in the ring E portion of ketoesters. Thus, heating of methyl pheo-
the pigments can be dramatically decreased phorbide a [28] (or b [29]) in collidine (30)
by removal of the 132-CO2Me group. gives methyl pyropheophorbide a [42] (or
When the 132-CO2Me group is removed, b [43]) in virtually quantitative yield; use of
the so-called “pyro” series of chlorophyll collidine is a yield-enhancing improvement
derivatives are obtained. Basically, ketones upon the classical method (28) which uti-

24
Syntheses of Tetrapyrroles

lized pyridine. Identical demethoxycar- infra] can be treated with alkali to give,
bonylation reactions take place with the so- after esterification with diazomethane,
called meso- (3-ethyl) series of compounds. chlorin e6 trimethyl ester [45] and chloro-
The 5-membered isocyclic ring in the porphyrin-e6 trimethyl ester [46], respec-
pyro-series of chlorophylls cannot be tively (24). Methanolysis of pheophorbide
cleaved, but the ring E in its β-ketoester a also affords [45]. This reaction can be
form can be readily opened since the high- reversed, and ring E is reformed either by
ly reactive conjugated functionality pro- treatment with methoxide (24), with tert-
vides a handle for chemical elaboration of butoxide (65), or best of all using tri-
ring E. For example, pheophorbide a [33] phenylphosphine and bis(trimethylsilyl)
and 3-vinylpheoporphyrin a5 [44, vide amide (31).

25
K.M. Smith

Although the chlorophyll b series of pig- 4. The green filtrate is evaporated and
ments is less accessible than those from then purified by flash chromatography
chlorophyll a (and indeed, as mentioned on Grade V neutral alumina, eluting
above, Spirulina algae contains no chloro- first with n-hexane to remove a fast run-
phyll b) a series of reactions parallel to ning yellow band, with dichlormethane
those described above also occurs in the b to remove the major blue-gray pheo-
series; the analogue of chlorin e6 trimethyl phytin a band, and finally with 97:3
ester in the b series is called rhodin g7 dichloromethane:tetrahydrofuran to
trimethyl ester [47] and of chloropor- remove some bright green magnesium-
phyrin e6 trimethyl ester is rhodinpor- containing pigments.
phyrin g7 trimethyl ester [48]. 5. The evaporated pheophytin a fraction is
Chlorins can be converted into por- treated with 500 mL of 5% sulfuric acid
phyrins by using DDQ as a dehydrogena- in methanol (degassed by bubbling with
tion agent. The β-ketoester functionality nitrogen gas) for 12.5 hours at room
does not take kindly to oxidative stress, so temperature in the dark (aluminum
methyl pheophorbide a [28] gives only a foil) under nitrogen, followed by dilu-
low yield of 3-vinylpheoporphyrin a5 di- tion with dichloromethane, and rinsing
methyl ester [44]. Using a “sledgehammer” with water.
approach to preparation of porphyrins 6. The mixture is rinsed with 10% saturat-
from chlorophyll derivatives, chlorophyll a ed aqueous sodium bicarbonate, the
under very vigorous basic conditions fol- organic layer is dried over anhydrous
lowed by esterification (methanolysis), sodium sulfate, followed by evapora-
affords phylloporphyrin XV methyl ester tion and crystallization of the residue
[49] and pyrroporphyrin XV methyl ester from dichloromethane:methanol. This
[50] (23). gives methyl pheophorbide a [28]
(average yield 1.8 g).
❖ Procedure 1. Isolation of Methyl Pheo-
phorbide a [28] from S. maxima (67)
4. CHEMICAL SYNTHESES OF
1. About 500 g of dried S. maxima alga is PORPHYRINS
slurried in 2 L of acetone, and then liq-
uid nitrogen is added to form a frozen Porphyrin chemical synthesis will be dis-
slush. cussed here in connection with two series
2. After transferring to a 5-L 3-necked of compounds: (i) those porphyrins which
round-bottom flask, the mixture is have been most often used in connection
heated at reflux with mechanical stir- with model studies, e.g., 5,10,15,20-tetra-
ring for 2 hours. The supernatant is fil- phenylporphyrin (TPP) [51] and 2,3,7,8,
tered through a Whatman filter paper 12,13,17,18-octaethylporphyrin (OEP)
(Whatman, Clifton, NJ, USA) using a [52]; and (ii) those related to protopor-
Buchner funnel, and more acetone is phyrin IX [1]. Simply based on the sym-
added to the solid debris. metry in the substituent arrays of [51] and
3. The extraction process, with refluxing, [52] and the lack of symmetry in [1], it is
is repeated twice more—note that the obvious that it would be a waste of time to
debris retains its deep green color, but approach the synthesis of both series of
amounts of additional chlorophyll compounds using the same strategy. To
obtained are marginal. attempt the synthesis of OEP [52] by labo-

26
Syntheses of Tetrapyrroles

rious multistep construction of an open- give pyrrole [54]. Simply pouring the
chain tetrapyrrolic intermediate would be cooled reaction mixture into iced water
plainly unwise—such symmetrically sub- causes precipitation of the product pyrrole.
stituted compounds are most efficiently The reaction works with a variety of sub-
obtained by tetramerization of a suitable stituents on the central (i.e., 2-) carbon of
monopyrrole (see below). On the other the 1,3-dione and with a variety of esters
hand, there is no way (in the absence of on the acetoacetate. The reaction described
enzymes) that protoporphyrin IX [1] can above (using acetoacetates) is the Johnson
be synthesized chemically by monopyrrole version, while the Kleinspehn modification
chemical self-condensation, so a more employs oximinomalonic esters in place of
sophisticated chemical approach is essen- the acetoacetates.
tial. As it happens, porphyrins [51] and Compared with the above synthesis of
[52] can be synthesized by self-condensa- pyrroles, methodology for preparing
tion of monopyrroles, while protopor- pyrroles such as [58] is relatively new. A
phyrin IX [1] can be accessed by a number major advance in the field was made when
of routes, the most simple (and the one to the Barton–Zard pyrrole synthesis was
be used as an example in this chapter) reported (8); the importance of this route
being from dipyrroles. was related to the substituent patterns
which could be accessed using it. Thus,
4.1. Syntheses of Pyrroles treatment of a nitroalkene [59a] or its syn-
thetic precursor, an acetoxynitroalkane
For both series of compounds men- [e.g., 59b], with an isocyanoacetate [e.g.,
tioned above, it is first essential to 60] affords excellent yields of pyrroles such
synthesize monopyrroles. Pyrrole itself as [58].
[53] is commercially available. Syntheses
of two common examples of useful
pyrroles (from the many dozens in the ❖ Procedure 2. Synthesis of Ethyl 3,4-
literature) (5,20,32,37,38) will be illus- Diethylpyrrole-2-Carboxylate [58]
trated here. (55)
Pyrroles bearing peripheral substituents
1. A mixture of 3-acetoxy-4-nitrohexane
are those which are most useful for appli-
[59b] (8)(16.3 g), ethyl isocyanoacetate
cation to dipyrrole and porphyrin synthe-
[60] (9.8 g; Sigma, St. Louis, MO,
sis. The Johnson–Kleinspehn synthesis
USA), and 1,8-diazabicyclo[5.4.0]
(11,43) is perhaps the most useful for tetra-
undec-7-ene (26.4 g; Sigma) in tetrahy-
substituted pyrroles. For example, pyrrole
drofuran (100 mL) is stirred at 20°C
[54], bearing very useful methyl and pro-
for 12 hours.
pionate groups, is prepared by the reaction
of dione [55] with benzyl oximinoacetoac- 2. The mixture is poured into water con-
etate [56]—compound [56] is in turn taining 1 M HCl and then extracted
obtained by the reaction of benzyl acetoac- with ethyl acetate.
etate [57] with sodium nitrite in the pres- 3. The extracts are washed with water and
ence of acetic acid. Slow admixture of dried over anhydrous magnesium sul-
equimolar amounts of [55] and [56] and fate.
excess zinc powder and sodium acetate in 4. After evaporation of the solvents, the
hot acetic acid results in reduction of the residue is chromatographed on a col-
oximinoderivative [56] to the amine, fol- umn of silica gel eluted with hexane:
lowed by in situ condensation with [55] to dichloromethane mixtures.

27
K.M. Smith

28
Syntheses of Tetrapyrroles

5. Evaporation of the eluates containing advantage of using the clay is that it can be
the red band will give the required pyr- removed simply by filtration after the reac-
role [58], with an average yield of 14.2 g. tion is complete. Compound [62] is
obtained from the corresponding methyl-
4.2. Syntheses of Dipyrromethanes pyrrole [64] simply by treatment with lead
tetra-acetate. Note that pyrrole [64] pos-
Unsymmetrically substituted dipyrro- sesses the same “symmetry pattern” as does
methanes, [e.g., 61], can be prepared by pyrrole [54]; its synthesis is relatively
condensation of 2-acetoxymethylpyrroles straightforward (21). Pyrrole [63] can be
[62] with 2-unsubstituted pyrroles [63] in obtained from [64] by following a
acetic acid containing a catalytic amount sequence of reactions as shown in Scheme
(<0.1 equiv.) of toluene p-sulfonic acid 1. The dipyrromethane [61] will form
(13). Montorillonite K-10 clay has also rings A and B of protoporphyrin IX
been shown to be a very useful acid catalyst dimethyl ester [2] in the synthesis, which
in dipyrromethane syntheses (30,36); the will be described later. The 1- and 9-car-

29
K.M. Smith

boxylate substituents are differentially pro- 4.3. Porphyrins via Monopyrrole


tected, and the future vinyl groups are pro- Tetramerization
tected as 2-chloroethyls.
Symmetrically substituted dipyrrometh- By definition, tetramerization of mono-
pyrroles must result either in a single sym-
anes [e.g., 65] are best prepared in one step
metrically substituted porphyrin (if the 3-
by self-condensation of bromomethyl-
and 4-substituents are identical) or in a
pyrroles [e.g., 66] in hot methanol (21), or mixture of porphyrins (if the 3- and 4-sub-
by heating 2-acetoxymethylpyrroles [e.g., stituents are different—see later). By far,
67] in methanol-hydrochloric acid (50). the easiest way to prepare a porphyrin
The 1- and 9-benzyl esters can be cleaved involves the reaction of pyrrole [53] with
using catalytic hydrogenation with hydro- benzaldehyde. The product is the almost
gen gas and 5% (or 10%) palladium–car- legendary TPP [51]. This simple route was
bon as catalyst. The resulting 1,9-dicar- first reported by Rothemund (57,58) and,
boxylic acid [68] can then be formylated after modification by Adler, Longo and col-
using the Vilsmeier reagent (phosphoryl leagues (involving use of refluxing propi-
chloride or benzoyl chloride mixed with onic acid instead of sealed tube chemistry)
equimolar amounts of dimethylforma- (1), was finally optimized as a two-step
mide) to give [69]. The 1- and 9-formyl procedure by Lindsey’s group (48). The
groups serve as the bridging carbons in the nonexpert procedure that is easiest to fol-
MacDonald porphyrin macrocyclization, low for the synthesis of [51] involves addi-
which will be described later. tion of equimolar amounts of crude (undis-

30
Syntheses of Tetrapyrroles

tilled) pyrrole [53] and benzaldehyde to be recovered by distillation and then


refluxing propionic acid. After heating for reused. Higher yields of TPP can be
about 30 minutes, the mixture is allowed to obtained by use of more elaborate and
cool, and the TPP is filtered off, usually in expensive chemistry, but, for TPP, quick
20% to 22% yield. The propionic acid can and dirty seems to work well. The product

31
K.M. Smith

32
Syntheses of Tetrapyrroles

from the Rothemund and Adler–Longo 6. The alumina is washed with dichloro-
(propionic acid) approaches is somewhat methane (200 mL), and the combined
impure (though highly crystalline) and filtrates are concentrated to approxi-
contains (4) about 5% or less of meso- mately 200 mL before addition of 200
tetraphenylchlorin [70]. Brief treatment (6) mL of methanol.
of the crude product with DDQ accom- 7. Filtration results in the chlorin-free
plishes transformation of [70] into [51]; product as glistening purple crystals,
earlier methodology involved the separa- with an average yield of 19.2 g.
tion of these two components on a chro-
matography column, but transformation of Approaches to so-called octaalkylpor-
[70] into [51] instead of separation of [51] phyrins (such as OEP [52]) are a little more
from [70] is much more sensible. Using complicated, but only with regard to the
this kind of methodology, literally kilo- difficulty of preparing the pyrrole starting
grams of TPP can be prepared. TPP can, in materials. In this case, the future meso-
any case, be purchased either “chlorin-free” (i.e., interpyrrolic) carbons can either be
or crude. Additionally, with only relatively present already on the pyrrole, or as in the
few exceptions, the reaction tolerates sub- case with TPP (wherein the meso-carbons
stitution of other arylaldehydes for ben- were provided by the formyl carbon in
zaldehyde, and good yields of a variety of benzaldehyde), the meso-carbons can be
tetra-arylporphyrins can be obtained (47). added separately from the pyrrole. A prima-
ry stricture, as mentioned above, is that a
❖ Procedure 3. Synthesis of Chlorin-Free monopyrrole tetramerization approach can
TPP [51] (6) only be used to give structurally unique
product if the substituents at positions 3-
1. Benzaldehyde (66.5 mL) and pyrrole and 4- in the monopyrrole are identical.
(46.5 mL) are simultaneously added to Thus, fully symmetrical porphyrins such
refluxing propionic acid (2.5 L), and as octaethylporphyrin [52] can be prepared
the mixture is refluxed for a further 30 easily using two major routes. The first
minutes before being allowed to cool approach, which chronologically was devel-
overnight to room temperature. oped first, involves the tetramerization of
2. The crude TPP is filtered off, washed pyrroles [71] bearing 2-CH2R substituents;
with hot water, and then washed with the “R” group must be a good leaving
methanol until the filtrate is colorless, group, and the methylene carbon of the 2-
to give 20.4 g (20% yield) of purple substituent will eventually be the source of
glistening crystals. the 5,10,15, and 20-carbons of the product
porphyrin. After the condensation reaction,
3. Concentration of the propionic acid
an oxidation step is necessary to afford good
filtrate affords a second crop of crystals.
yields of symmetrical porphyrin. Useful
4. The crude TPP (20 g) is dissolved in examples for attachment of CH2R groups
refluxing ethanol-free chloroform (2.5 to pyrroles are (i) the Mannich reaction of
L) before addition of DDQ (5 g) in pyrrole [72] with formaldehyde and
dry toluene (150 mL). dimethylamine [or better, with commercial-
5. The mixture is refluxed for 3 hours ly available (N,N-dimethylmethylene)am-
before filtration of the yellowish solu- monium iodide, Eschenmoser’s reagent
tion, under suction, through a sintered (56,59)] to give the 2-(N,N-dimethy-
glass funnel containing Grade I alumina laminomethyl)pyrrole [73]; heating of this
(300 g). in acetic acid gives a 52% yield of [52]
33
K.M. Smith

(18,70); and (ii) hydrolysis of the pyrrole 6. Evaporation gives a residue which is
[74] to give pyrrole [75] which is tetramer- chromatographed on silica gel, eluted
ized to give [52] in 44% yield by heating in with dichloromethane to give OEP
acetic acid containing potassium ferri- [52], with an average 55% yield (240
cyanide (35,63). Most recently, the Bar- mg).
ton–Zard pyrrole synthesis (see above) (8) Alternatively, tetramerization of 2,5-di-
has greatly simplified preparative approach- unsubstituted pyrroles [e.g., 72] in the
es to monopyrroles of the type [58]; lithium presence of reagents that can provide the
aluminum hydride (CAUTION: reacts vio- four meso-methine carbons of the product
lently with moisture) reduction, followed by can be used. Cyclization of 3,4-diethylpyr-
tetramerization of the resulting pyrrole-2- role [72] with formaldehyde affords
carbinol [76] under acidic conditions, gives 55%–75% yields of OEP [52] (61).
[52] in 55% yield (2,55). If the 3- and 4-substituents on the
monopyrrole component are not identical,
❖ Procedure 4. Synthesis of OEP [52] mixtures will usually result due to acid cat-
from Pyrrole [58] (54) alyzed pyrrole ring scrambling reactions
(49). Thus, acid catalyzed tetramerization
1. Ethyl 3,4-diethylpyrrole-2-carboxylate of pyrrole [77] will result in production of
[58] (see above, 657 mg) is added drop- a mixture of the four etioporphyrin type
wise at 0°–5°C to a stirred solution of isomers [6–9]. However, a method has
lithium aluminum hydride (320 mg; been devised which does produce only
Sigma) in dry tetrahydrofuran (15 mL). etioporphyrin I [6] from tetramerization of
The mixture is stirred for 2 hours at a pyrrole related to [77]; treatment of 2-
0°–5°C before destroying the excess (N,N-dimethylaminomethyl)pyrroles [e.g.,
lithium aluminum hydride by addition 78] with methyl iodide gives [79], which
of ethyl acetate. has a leaving group that is labile even under
2. It is then poured into saturated aqueous neutral conditions (i.e., no acid), which
ammonium chloride, extracted with would cause pyrrole ring scrambling, is
ethyl acetate (3 times 10 mL), washed present (53,54). This, quaternized in meth-
with aqueous sodium chloride, and dried anol containing potassium ferricyanide (to
over anhydrous magnesium sulfate. accomplish rapid in situ oxidation of the
labile porphyrinogen intermediate), gives a
3. The solution is evaporated to dryness
good yield of pure etioporphyrin I [6].
under vacuum before addition of
dichloromethane (15 mL).
❖ Procedure 5. Synthesis of
4. To this solution is added dimethoxy- Etioporphyrin I [6] from Pyrrole [78]
methane (0.7 mL; Sigma) and toluene
p-sulfonic acid (110 mg), and the mix- 1. Benzyl 4-ethyl-5-(N,N-dimethylamino-
ture is stirred for 12 hours at room tem- methyl)-3-methylpyrrole-2-carboxylate
perature. Aerial oxidation occurs under (53,54) (1.88 g) is dissolved in tetrahy-
these conditions, but chloranil (Sigma) drofuran (600 mL), and 10% palladi-
can also be used, although without any um on carbon (500 mg; Sigma) is
improvement in yield. added.
5. The mixture is washed with aqueous 2. The resulting mixture is stirred under
sodium bicarbonate, and the organic hydrogen at room temperature for 12
layer is dried over anhydrous magne- hours before the catalyst is removed,
sium sulfate. and the solvent is evaporated to dryness.
34
Syntheses of Tetrapyrroles

3. Recrystallization from dichlorometh- heated under reflux for 15 minutes.


ane/hexane affords 4-ethyl-5-(N,N-di- 5. Potassium ferricyanide (3.8 g) is added,
alkyl-aminomethyl)-3-methylpyrrole-2- and the reaction is continued at reflux
carboxylic acid as an off-white powder for another 10 hours.
in quantitative yield. Note that because 6. After removal of the solvent, the residue
of spontaneous decarboxylation at is redissolved in chloroform, the insolu-
room temperature to give [78], this ble material filtered off, and the red solu-
must be used immediately. tion is passed through a short column of
4. The pyrrole carboxylic acid (1.29 g) is silica gel (eluted with chloroform).
dissolved in a solution of methanol 7. The solvent is evaporated, and the
(200 mL) and triethylamine (2 mL) and residual porphyrin is recrystallized from

35
K.M. Smith

dichloromethane/methanol to afford either of the two individual dipyrrometh-


etioporphyrin-I in 36% yield (284 mg). anes from reacting with themselves) and
dipyrromethane [69] is symmetrical about
4.4. Porphyrins via Dipyrromethane its 5-carbon. Conversion of the 3,8-bis(2-
Intermediates chloroethyl)porphyrin [83] into protopor-
phyrin IX dimethyl ester [2] is accom-
If two dipyrromethane units with plished simply by treatment with base
appropriate bridging carbons are con- (40)—just in case this base treatment also
densed together, there are three possible accomplished hydrolysis of the methyl
products because the dipyrromethanes can esters, the product is then set aside in meth-
either react with themselves or with each anol containing 5% sulfuric acid (CAUTION:
other. If the dipyrromethanes individually add the acid to the methanol, cooled and
possess an unsymmetrical array of sub- slowly). Workup and chromatography
stituents, even greater mixtures can occur [NOTE: protoporphyrin is photolabile (see
because there is no control over which end above), so the column should be run in the
of one dipyrromethane reacts with the end dark or with aluminum foil wrapped
of another. These symmetry limitations are around it] then produces the product, [2].
common with all so-called “2 + 2” synthe- It has been my intention to provide a
ses; in a porphyrin synthesis involving an summary of fairly simple procedures that
A-B and a C-D dipyrromethane is to be can be used, given a certain competence in
condensed, the symmetry problems can be synthetic organic chemistry, to obtain by
avoided if the A-B or C-D dipyrromethane extraction or by total synthesis some useful
unit is symmetrical about the interpyrrolic porphyrins with defined symmetry and
(5-) carbon atom. Arsenault, MacDonald, structural characteristics. But competence in
and coworkers showed (3) that a 1,9-difor- organic chemistry is not easily earned. If the
myldipyrromethane, [e.g., 69], can be con- procedures still look too complex, or (more
densed with a 1,9-di-unsubstituted dipyrr- likely) if you do not have the basic laborato-
omethane or its 1,9-dicarboxylic acid [80] ry equipment with which to carry out the
in the presence of an acid catalyst to give procedures described, then the best bet is to
pure porphyrin [e.g., 81], often in high collaborate. Just remember, most organic
yields. MacDonald used hydriodic acid, chemists would not know where to start if
but since that time, toluene p-sulfonic acid they needed to run a gel or if they needed
has been shown (14,15) to be a much bet- to do a northern blot. They will want to col-
ter choice and more convenient too. laborate also if they need these things.
This 2 + 2 route using dipyrromethanes
is probably the most widely used pathway
to synthetic porphyrins. Thus, for example, ABBREVIATIONS
treatment of dipyrromethane [82]
(obtained from [61] by catalytic debenzyla- DDQ, 2,3-dichloro-5,6-dicyanobenzo-
tion followed by treatment with trifluo- quinone; OEP, 2,3,7,8,12,13,17,18-octa-
roacetic acid) with 1,9-diformyldipyrro- ethylporphyrin; TPP, 5,10,15,20-tetra-
methane [69] gives a good yield of phenylporphyrin.
porphyrin [83] after oxidation of the inter-
mediate porphodimethene [84]; no mix-
tures are produced because both of the ACKNOWLEDGMENTS
future linking meso-carbons are sited on
the same dipyrromethane (preventing Some of the work reported herein was
36
Syntheses of Tetrapyrroles

developed within my own research group. I of haemoproteins. J. Chem. Soc. [Perkin 1] 1771-
would like to thank all of my collaborators, 1781.
15.Cavaleiro, J.A.S., G.W. Kenner, and K.M. Smith.
over the years, for their important contri- 1974. Biosynthesis of protoporphyrin-IX from copro-
butions to our group effort; their names porphyrinogen-III. J. Chem. Soc. [Perkin 1] 1188-
can be found in the various literature cita- 1194.
16.Chu, T.C. and E.J.-H. Chu. 1955. Paper chromatog-
tions. I would also like to thank the raphy of iron complexes of porphyrins. J. Biol. Chem.
National Institutes of Health (HL 22252) 212:1-7.
and the National Science Foundation 17.DiNello, R.K. and D.H. Dolphin. 1981. Evidence
for a fast (major) and slow (minor) pathway in the
(CHE 99-04076) for uninterrupted sup- Schumm devinylation reaction of vinylporphyrins. J.
port of our efforts over the past 24 years. Org. Chem. 46:3498-3502.
18.cf. Eisner, U., A. Lichtarowicz, and R.P. Linstead.
1957. Chlorophylls and related compounds. Part VI.
REFERENCES The synthesis of octaethylchlorin. J. Chem. Soc. 733-
739.
1.Adler, A.D., F.R. Longo, J.D. Finarelli, J. Goldmach- 19.Fischer, H. 1955. Preparation of hemin. Org. Synth.
er, J. Assour, and L. Korsakoff. 1967. A simplified 3:442.
synthesis for meso-tetraphenylporphyrin. J. Org. 20.Fischer, H. and H. Orth. 1934. Die Chemie des
Chem. 32:476. Pyrrols. Vol. 1. Akademische Verlagsgesellschaft,
2.Aoyagi, K., T. Haga, H. Toi, Y. Aoyama, T. Mizutani, Leipzig.
and H. Ogoshi. 1997. Electron deficient porphyrins. 21.Fischer, H. and H. Orth. 1934. Die Chemie des
III. Facile syntheses of perfluoroalkylporphyrins Pyrrols. Vol. 1, p. 333. Akademische Verlagsge-
including water soluble porphyrin. Bull. Chem. Soc. sellschaft, Leipzig.
Jpn. 70:937-943. 22.Fischer, H. and H. Orth. 1937. Die Chemie des
3.Arsenault, G.P., E. Bullock, and S.F. MacDonald. Pyrrols. Vol. 2, part 1. Akademische Verlagsge-
1960. Pyrromethanes and porphyrins therefrom. J. sellschaft, Leipzig.
Am. Chem. Soc. 82:4384-4389. 23.Fischer, H. and H. Orth. 1937. Die Chemie des
4.Badger, G.M., R.A. Jones, and R.L. Laslett. 1964. Pyrrols. Vol. 2, part 1, p 358. Akademische Verlagsge-
Porphyrins. VII. The synthesis of porphyrins by the sellschaft, Leipzig.
Rothemund reaction. Aust. J. Chem. 17:1028-1035. 24.Fischer, H. and A. Stern. 1940. Die Chemie des
5.Baltazzi, E. and L.I. Krimen. 1963. Recent advances Pyrrols. Vol. 2, part 2. Akademische Verlagsge-
in the chemistry of pyrrole. Chem. Rev. 63:511. sellschaft, Leipzig.
6.Barnett, G.H., M.F. Hudson, and K.M. Smith. 1975. 25.Fischer, H. and A. Stern. 1940. Die Chemie des
Concerning meso-tetraphenylporphyrin purification. J. Pyrrols. Vol. 2, part 2, p. 48. Akademische Verlagsge-
Chem. Soc. [Perkin 1] 1401-1403. sellschaft, Leipzig.
7.Barrett, J. 1959. Detection of hydroxyl groups in por- 26.Fischer, H. and A. Stern. 1940. Die Chemie des
phyrins and chlorins. Nature 183:1185-1186. Pyrrols. Vol. 2, part 2, p. 52. Akademische Verlagsge-
8.Barton, D.H.R., J. Kervagoret, and S.Z. Zard. 1990. sellschaft, Leipzig.
A useful synthesis of pyrroles from nitroolefins. Tetra- 27.Fischer, H. and A. Stern. 1940. Die Chemie des
hedron 46:7587-7598. Pyrrols. Vol. 2, part 2, p. 73. Akademische Verlagsge-
9.Battersby, A.R. and E. McDonald. 1975. Biosynthe- sellschaft, Leipzig.
sis of porphyrins, chlorins, and corrins. In K.M. Smith 28.Fischer, H. and A. Stern. 1940. Die Chemie des
(Ed.), Porphyrins and Metalloporphyrins. Elsevier, Pyrrols. Vol. 2, part 2, p. 108. Akademische Verlagsge-
Amsterdam. sellschaft, Leipzig.
10.Bonnett, R., I.H. Campion-Smith, and A.J. Page. 29.Fischer, H. and K. Zeile. 1929. Synthese des haemato-
1977. Protiodevinylation: the Schumm reaction of porphyrins, protoporphyrins und haemins. Liebigs
vinylporphyrins. J. Chem. Soc. [Perkin 1] 68-71. Ann. Chem. 468:98-116.
11.Bullock, E., A.W. Johnson, E. Markham, and K.B. 30.Freeman, B.A. and K.M. Smith. 1995. Novel uses of a
Shaw. 1958. A synthesis of coproporphyrin III. J. naturally occurring heterogeneous catalyst to improve
Chem. Soc. 1430-1440. organic syntheses. Proc. NOBCChE 22:227-242.
12.Burbidge, P.A., G.L. Collier, A.H. Jackson, and G.W. 31.Gerlach, B., S.E. Brantley, and K.M. Smith. 1998.
Kenner. 1967. Syntheses and spectra of some meso- Novel synthetic routes to 8-vinyl chlorophyll deriva-
methylated porphyrins. J. Chem. Soc. B 930-937. tives. J. Org. Chem. 63:2314-2320.
13.Cavaleiro, J.A.S., A.M.d’A.R. Gonsalves, G.W. Ken- 32.Gossauer, A. 1974. Die Chemie der Pyrrole. Springer,
ner, and K.M. Smith. 1973. Synthesis of Berlin.
pyrromethanes and a tripyrrane. J. Chem. Soc. [Perkin 33.Grinstein, M. 1947. Studies of protoporphyrin. J. Biol.
1] 2471-2478. Chem. 167:515-519.
14.Cavaleiro, J.A.S., A.M.d’A.R. Gonsalves, G.W. Ken- 34.Iakovides, P. and K.M. Smith. 1996. Syntheses of oxy-
ner, and K.M. Smith. 1974. Total synthesis of deuteri- gen analogues of sulfhemes-A and -C. Tetrahedron
ated derivatives of protoporphyrin-IX for NMR studies 52:1123-1148.

37
K.M. Smith

35.Inhoffen, H.H., J.-H. Fuhrhop, H. Voigt, and H. 52.Morell, D.B. and M. Stewart. 1956. Removal of iron
Brockmann, Jr. 1966. Formylierung der meso- from hemins. Aust. J. Exp. Biol. Med. Sci. 34:211-
Kohlenstoffeatome von alkylsubstituierten porphyri- 218.
nen. Liebigs Ann. Chem. 695:133. 53.Nguyen, L.T., M.O. Senge, and K.M. Smith. 1996.
36.Jackson, A.H., R.K. Pandey, K.R.N. Rao, and E. Simple methodology for syntheses of porphyrins pos-
Roberts. 1985. Reactions on solid supports, part II: a sessing multiple peripheral substituents with an ele-
convenient method for synthesis of pyrromethanes ment of symmetry. J. Org. Chem. 61:998-1003.
using a Montmorillonite clay as catalyst. Tetrahedron 54.Nguyen, L.T. and K.M. Smith. 1996. Syntheses of
Lett. 26:793-796. type-I porphyrins via monopyrrole tetramerization.
37.Jackson, A.H. and K.M. Smith. 1973. In J.W. ApSi- Tetrahedron Lett. 37:7177-7180.
mon (Ed.), Total Synthesis of Natural Products, Vol. 1, 55.Ono, N., H. Kawamura, M. Bougauchi, and K.
p. 143-278. John Wiley & Sons, New York. Maruyama. 1990. Porphyrin synthesis from nitrocom-
38.Jackson, A.H. and K.M. Smith. 1984. In J.W. ApSi- pounds. Tetrahedron 46:7483-7496.
mon (Ed.)Total Synthesis of Natural Products Vol. 6, 56.Ono, M., R. Lattmann, K. Imomota, C. Lehmann, T.
p. 237-280. John Wiley & Sons, New York. Früh, and, A. Eschenmoser. 1985. Monopyrrole pre-
39.Kadish, K., K.M. Smith, and R. Guilard. (Eds.). cursors for the synthesis of uroporphyrinogenoctani-
1999. The Porphyrin Handbook. Academic Press, trile. Croat. Chem. Acta 58:627.
Boston. 57.Rothemund, P. 1935. Formation of porphyrins from
40.Kenner, G.W., S.W. McCombie, and K.M. Smith. pyrrole and aldehydes. J. Am. Chem. Soc. 57:2010-
1973. Protection of porphyrin vinyl groups. A synthe- 2011.
sis of coproporphyrin-III from protoporphyrin-IX. 58.Rothemund, P. and A.R. Menotti. 1941. Porphyrin
Liebigs Ann. Chem. 1329-1338. studies. IV. The synthesis of α,β,γ,δ-tetraphenylpor-
41.Kenner, G.W., S.W. McCombie, and K.M. Smith. phyrin. J. Am. Chem. Soc. 63:267-270.
1973. Separation and oxidative degradation of chloro- 59.Schreiber, J., H. Maag, N. Hashimoto, and A.
phyll derivatives. J. Chem. Soc. [Perkin 1]. 2517-2523. Eschenmoser. 1971. Dimethyl(methylene)ammonium
42.Kenner, G.W., J.M.E. Quirke, and K.M. Smith. 1976. iodide. Angew. Chem. Int. Ed. Engl. 10:330-331.
Transformations of protoporphyrin-IX into harderopro- 60.Schumm, O.Z. 1928. Preparation of hemin derivatives
phyrin, pemptoporphyrin, chlorocruoroporphyrin and by pyro reactions. Z. Physiol. Chem. 178:1-18.
their isomers. Tetrahedron 32:2753-2756. 61.Sessler, J.L., A. Mozaffari, and M.R. Johnson. 1991.
43.Kleinspehn, G.G. 1955. Pyrrole series XXVII. Novel 3,4-Diethylpyrrole and 2,3,7,8,12,13,17,18-octaethyl-
route to certain 2-pyrrolecarboxylic esters and nitriles. porphyrin. Org. Synth. 70:68-78.
J. Am. Chem. Soc. 77:1546-1548. 62.Shiau, F.-Y., R.K. Pandey, S. Ramaprasad, T.J.
44.Labbe, R.F. and G. Nishida. 1957. A new method of Dougherty, and K.M. Smith. 1990. The isomeric
hemin isolation. Biochim. Biophys. Acta 26:437. mono-acetyl-mono-(1-hydroxyethyl)-deuteroporphyrins:
45.Lee, S.-J.H., N. Jagerovic, and K.M. Smith. 1993. Use synthesis, characterization and use for syntheses of
of the chlorophyll derivative, purpurin-18, for synthe- regioselectively methyl- and vinyl-deuterated hemins.
ses of sensitizers for photodynamic therapy. J. Chem. J. Org. Chem. 55:2190-2195.
Soc. [Perkin 1] 2369-2377. 63.cf. Siedel, W. and F. Winkler. 1943. Oxydation von
46.Lemberg, R., B. Bloomfield, P. Caiger, and W. Lock- pyrrolderivaten mit bleitetraacetat. Liebigs Ann.
wood. 1955. Porphyrins with formyl groups. V. Isola- Chem. 554:162-201.
tion of porphyrin a from heart muscle and determina- 64.Smith, K.M. (Ed.). 1975. Porphyrins and Metallopor-
tion of its heme a content. Aust. J. Exp. Biol. Med. Sci. phyrins. Elsevier, Amsterdam.
33:435-450. 65.Smith, K.M., G.M.F. Bisset, and M.J. Bushell. 1980.
47.Lindsey, J.S. 1999. Synthesis of meso-substituted por- Partial syntheses of optically pure methyl bacterio-
phyrins. In K. Kadish, K.M. Smith, and R. Guilard pheophorbides c and d from methyl pheophorbide a. J.
(Eds.), The Porphyrin Handbook, Chapter 2. Acade- Org. Chem. 45:2218-2224.
mic Press, Boston. 66.Smith, K.M., E.M. Fujinari, K.C. Langry, D.W.
48.Lindsey, J.S., I.C. Schreiman, H.C. Hsu, P.C. Kear- Parish, and H.D. Tabba. 1983. Manipulation of vinyl
ney, and A.M. Marguerettaz. 1987. Rothemund and groups in protoporphyrin-IX: introduction of deuteri-
Adler-Longo reactions revisited: synthesis of tetraphenyl- um and carbon-13 labels for spectroscopic studies. J.
porphyrins under equilibrium conditions. J. Org. Am. Chem. Soc. 105:6638-6646.
Chem. 52:827-836. 67.Smith, K.M., D.A. Goff, and D.J.Simpson. 1985.
49.Mauzerall, D. 1960. The thermodynamic stability of Meso-substitution of chlorophyll derivatives: direct
porphyrinogens. J. Am. Chem. Soc. 82:2601-2605. route for transformation of bacteriopheophorbides-d
50.Mironov, A.F., T.R. Ovsepyan, R.P. Evstigneeva, and into bacteriopheophorbides-c. J. Am. Chem. Soc.
N.A. Preobrazenskii. 1965. Synthetic studies in the 107:4946-4954.
dipyrrylmethane series. Synthesis of 4,4′-dimethyl- 68.Strain, H.H. and W.A. Svec. 1966. In L.P. Vernon and
3,3′-bis(β-carbomethoxyethyl)dipyrrylmethane. Zh. G.R. Seely (Eds.), p. 21. The Chlorophylls, Academic
Obshch. Khim. 35:324-328. Press, New York.
51.Morell, D.B., J. Barrett, and P.S. Clezy. 1961. The 69.Thudichum, J.L.W. 1867. Rpt. Med. Off. Privy
prosthetic groups of cytochrome oxidase. 1. Purifica- Council, App. 7. 10:152.
tion as porphyrin a and conversion into haemin a. 70.Whitlock, H.W. and R. Hanauer. 1968. Octaethyl-
Biochem. J. 78:793-797. porphyrin. J. Org. Chem. 33:2169-2171.

38
General Laboratory Methods
3 for Tetrapyrroles

Jerry C. Bommer1 and Peter Hambright2


1Frontier Scientific/Porphyrin Products, Logan, UT, and 2Department of

Chemistry, Howard University, Washington, DC, USA

1. INTRODUCTION tion states, and certain porphyrins form


large supramolecular aggregates.
There are thousands of porphyrins and A number of books are important in the
metalloporphyrins, and hundreds of new biological porphyrin area. The classics are
derivatives appear each year. This variety Die Chemie des Pyrroles in 1934 (38) and
arises because the cyclic conjugated tetra- 1937 (39) by Fischer and Orth, the 1940
pyrrole nucleus (Figure 1) can have Fischer and Stern (41), and the 1949
different substituents at the eight β-pyrrole Hematin Compounds and Bile Pigments by
positions, at the four meso [5,10,15,20] car- Lemberg and Legge (74). The monograph
bon atoms, and N-alkyl groups can be by Lascelles on Tetrapyrrole Biosynthesis and
added to the four central nitrogen atoms. Its Regulation appeared in 1964 (70). Fol-
Since its synthesis in 1972, over 8000 liter- lowing the publication of Porphyrins and
ature references have appeared on 5,10,15, Metalloporphyrins by Falk in 1964 (35),
20-tetrakis(N-methyl-4-pyridyl)porphyrin Smith edited a new expanded edition of
compounds, and a similar number on its the same title in 1974 (106). The last chap-
precursor, 5,10,15,20-tetrakis(4-pyridyl) ter on “Laboratory Methods” gives infor-
porphyrin and its derivatives. Most metals mation on basic laboratory manipulations
and metalloids in the periodic table form and transformations of porphyrin macro-
metalloporphyrins, and iron porphyrins cycles (44). Dolphin edited an eight vol-
have been prepared in oxidation states ume series entitled The Porphyrins in 1978
ranging from 0 to +5. In addition, the por- (30a), followed by The Chemistry and Bio-
phyrin ring itself can be oxidized, reduced, chemistry of N-Substituted Porphyrins by
and cleaved. In solution, metallopor- Lavallee in 1987 (71). Kalyansundaram’s
phyrins can exist as monomers, homo- and book Photochemistry of Polypyridine and
heteronuclear dimers and higher polymers, Porphyrin Complexes appeared in 1991
they can form molecular complexes and (63). The Colours of Life: An Introduction to
exist in various protonation and axial liga- the Chemistry of Porphyrins and Related

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

39
J.C. Bommer and P. Hambright

Compounds by Milgrom in 1998 (84) is range and is valuable for the separation of
suggested reading both for beginners and compounds having similar retention factor
experienced workers desiring a broader (Rf ) values. High-pressure liquid chroma-
view of the field. The Porphyrin Handbook, tography (HPLC) can separate and quanti-
a ten volume series edited by Kadish, tate samples on the microgram scale. Many
Smith, and Guilard was published in 2000 porphyrins and diamagnetic metallopor-
(61a). Dupré maintains “Porphynet, the phyrins fluoresce under UV irradiation,
porphyrin site” (www. porphyrin.net), and a hand-held long wavelength 366 nm
which lists 40 currently available mono- UV lamp is an aid in monitoring the sepa-
graphs on porphyrins. This site is a source rations. Porphyrin solution should always
of general information to those working in be filtered before application to columns or
all aspects of the porphyrin field, and lists plates, and one should avoid “overloading”
vendors, e-mail addresses of porphyrin the media with too high a concentration of
chemists, future conferences, etc. The Jour- pigment. A small amount of the column
nal of Porphyrins and Phthalocyanines began material often accompanies the purified
publication in 1997. The Chemical Ab- porphyrin and tends to complicate subse-
stracts Service “CA Selects: Porphyrins” quent CHN determinations. Filtering the
gives biweekly coverage of books, disserta- solution through a 47 mm 0.2- or 0.45-µm
tions, reports, reviews, patents, and journal nylon filter (Millipore, Bedford, MA, USA)
abstracts dealing with most areas of por- aids in the removal of small particles. Such
phyrin chemistry. The free Medline filters are also useful for the collection of
(www.ncbi.nih.gov/PubMed) has over small quantities of a valuable precipitate.
23 000 listings under “porphyrins”. Our seventy years experience at the bench
The chapter by Smith in this volume indicates that accidents do happen, and we
describes the nomenclature of the por- have tried to indicate throughout this chap-
phyrin nucleus, and the preparations of ter where accidental loss of a valuable por-
numerous 3,8-disubstituted deuteropor- phyrin can be avoided. A clean heavy-wall
phyrins, chlorophylls, and recent advances empty flask should be between the water
in porphyrin synthesis. We intend to cover aspirator and the filtration flask, and the fil-
laboratory techniques useful for the syn- ter flask should always be securely clamped
thesis, purification, and analysis of water- to a ring stand. It is worthwhile to date all
soluble and insoluble porphyrins and met- sample vials containing solid porphyrins.
alloporphyrins, and general aspects of the This is an aid in rapidly locating in your
solution chemistry of such macrocyclic laboratory book at a later time the synthet-
species. While only a few examples are ic or isolation method used for the particu-
mentioned, such procedures can usually be lar preparation, and also because certain
applied to whole classes of compounds. compounds have unexpectedly short shelf-
lives. Columns and TLC work should be
done under the hood to minimize exposure
2. CHROMATOGRAPHY to the often hazardous solvents, and it is
worthwhile to wear disposable gloves in all
Most porphyrins are purified by some porphyrin manipulations. Lim has edited a
form of chromatography. Column chro- volume on porphyrin chromatography to
matography is used for isolation of 100 mg appear in 2000 (75a). White, Bachman,
to multigram quantities of material. Thin and Burnham (116) have an excellent chap-
layer chromatography (TLC) on coated ter on chromatography of porphyrins and
glass plates is employed in the 1 to 100 mg metalloporphyrins, and Varaldi, Longo and
40
General Laboratory Methods for Tetrapyrroles

Adler (114) have summarized nonchro- methylene chloride, acetone, ethyl acetate,
matographic methods (electrophoresis, tetrahydrofuran, acetonitrile, pyridine, n-
countercurrent distribution, sublimation, propanol, ethanol, methanol, acetic acid,
extraction, precipitation, and crystalliza- and water. Numerous examples of useful
tion) for the purification of porphyrins. solvent mixtures and sorbents are men-
tioned in later sections of this chapter for
2.1. Column Chromatography particular compounds. For silica gel or alu-
mina chromatography, we find that ethyl
Most column chromatography involves acetate in various concentrations (1%–
glass columns of varying lengths, although 15%) mixed with CHCl3 or CH2Cl2, in
some prefer plastic columns that can be cut combination with increasing amounts of
to isolate the porphyrin bands. Others methanol, is effective for the separation of
claim that better separations occur in pear- a wide range of synthetic porphyrins and
shaped separating funnels. In contrast to for porphyrin esters in general.
gravity techniques, some groups prefer Many groups prefer to evaporate a con-
“flash chromatography”, in which a low centrated organic solution of the impure
pressure (5–15 psi) of an inert gas is porphyrin(s) in the presence of the packing
applied to force the sample and solvent material. This solid is then placed on top of
rapidly through the sorbent. A glass-wool the column, and elution is begun with a
plug and sand are placed on the bottom of relatively nonpolar solvent. One rationale
open columns, or commercial columns for this procedure is that many porphyrins
with glass frits overlaid with a layer of sand are relatively insoluble in solvents of low
are employed. The “dry column” technique polarity. For example, the room temperature
(116) in which the column is packed with solubility of nickel(II) etioporphyrin I
dry absorbent is sometimes useful, but in (µg/mL) in various solvents is: chloroform
general, better separations are achieved (2.2 × 103), methylene chloride (1.6 × 103),
when the column is loaded with a slurry of ethyl acetate (2.2 × 102), cyclohexane
the packing material, which is prepared by (approximately 7), followed by acetonitrile
stirring the solid into a large volume of the cyclohexane, ethanol, methanol, and
liquid. A layer of sand is added to the top hexane at approximately 2 µg/mL. Solu-
of the column so that the bed is not dis- bility depends on both the type of por-
turbed by solvent addition. It is best to dis- phyrin ring and the identity of the coordi-
solve the impure porphyrin mixture in a nated metal; for example, the solubilities
relatively nonpolar solvent, so that the in chloroform are Ni-Etio I (2.2 × 103),
majority of the porphyrin and impurities are Ni-octaethylporphyrin (5.1 × 103), VO-
initially adsorbed near the top of the col- Etio I (6.8 × 103), and VO- octaethylpor-
umn. This is followed by selective elution phyrin (OEP) (8.2 × 103). OEP has eight
with a solvent, solvent mixture, or successive ethyl groups in the β- pyrrole positions,
solvent mixtures of higher polarity, which while etioporphyrin I has four methyl and
move the desired porphyrin away from the four ethyl substituents.
adsorbed impurities, or separate a mixture Commercial CHCl3 is often stabilized
of porphyrins one from another. One with approximately 0.75% ethanol, and
elutropic series of solvents useful in por- the polar ethanol affects the separations. If
phyrin work are n-hexane, petroleum necessary, the ethanol can be removed by
ether, cyclohexane, carbon tetrachloride, shaking the CHCl3 with an equal volume
toluene, benzene, diethyl ether, trichloroeth- of water, drying with CaCl2, and distilling
ylene, 1,2-dichloroethane, chloroform, over P2O5, collecting only the middle frac-
41
J.C. Bommer and P. Hambright

tions. Some groups directly use CHCl3 sta- water, and grade V contains 15% water.
bilized with amylenes. CHCl3 and Other adsorbents include Florisil (60–
CH2Cl2 (both carcinogenic to laboratory 100 mesh), calcium carbonate (for proto-
animals) often contain HCl, which can porphyrin IX dimethyl ester [DME]),
protonate the porphyrin free base H2-P sugar (for chlorophylls), talc (to remove
into the charged diacid H4-P2+, leading to reduced porphyrin impurities), 80% mag-
poor separations. To test for HCl, the inex- nesol-20% cellulose (for magnesium por-
pensive and readily available tetraphenyl- phyrin esters) microcrystalline cellulose,
porphyrin (H2-TPP) is dissolved in ben- Kieselguhr, Fullers earth, and diatoma-
zene, producing a red solution of the free ceous earth (for sulfonated deuteropor-
base. Several milliliters of CHCl3 or phyrins). Bachmann and Burnham (8) sep-
CH2Cl2 are added to a similar volume of arated, on the macro or micro scale, the
the red reagent, and a green solution of the methyl esters of meso, proto, copro I, and
porphyrin diacid forms if HCl is present. uro I by gel filtration on Sephadex LH-20
The acid can be removed by shaking these (Amersham Pharmacia Biotech, Piscat-
solvents with an aqueous solution of 0.1 M away, NJ, USA), with 1/1 CHCl3-MeOH
NaOH or 0.1 M ammonia until the test (containing 1 g Tris base/L) as the eluant.
solution remains red. The solvents are then Sephadex has been used to remove unde-
dried over solid anhydrous CaCl2, sired salts from porphyrin solutions.
Na2SO4, or K2CO3 before use. For the Polyamide columns are useful for iron por-
separation of a metalloporphyrin that is phyrin carboxylic acids and acid-labile
stable in acid from small amounts of the metalloporphyrins (29). Reverse phase
unmetalated free base, the mixture is often octadecyl (C18) columns using methanol-
dissolved in chloroform containing 2% tri- buffered water mixtures can separate water-
fluoroacetic acid to purposely protonate soluble anionic porphyrins by total por-
the undesired H2-P into H4-P2+. The phyrin charge (110). Cationic porphyrins
green diacid will tend to stick near the top of differing charge can be resolved on
of the column, resulting in a better separa- silica gel plates developed with saturat-
tion than can be obtained simply with the ed KNO 3:water:acetonitrile in a 1:1:8
metalloporphyrin and its free base. mixture (10). Cation exchange columns
A variety of packing materials have been (-SO3-H+ or -SO 3-Na+) are used to separ-
used in column techniques. Silica gel ate divalent metal cations from aqueous
(100–200 mesh) and alumina (80–200 solutions of anionic water-soluble por-
mesh) are perhaps the most common phyrins. For cationic porphyrins, anion
-
choices. Activated alumina can be pur- exchange columns [-N(CH3)3+Cl ] replace
chased in acidic, neutral, or basic forms, tosylates, or the precipitating agents iodide,
and these activated (Grade I) materials or perchlorate in solution with chloride.
often adsorb the porphyrin so strongly that TLC or absorption spectrophotometry
it becomes difficult to remove from the (350–800 nm) are used to monitor the
column. The alumina can be deactivated fractions finally coming off of columns and
by adding water to the adsorbent (wt/wt) also to determine what impurities are pre-
in a stoppered Erlenmeyer flask. The mix- sent prior to running a column. In many
ture is shaken to remove all lumps and cases, a given fraction must be rechro-
allowed to cool to room temperature matographed many times, filtered, and
before use. Activity grade I has no added then recrystallized to obtain pure material.
water, grade II contains 3% water, grade III It is advantageous to use a long spatula or
contains 6% water, grade IV contains 10% aspirator to remove the obvious impurity
42
General Laboratory Methods for Tetrapyrroles

band(s) from the top of the column once and tetra-aryl porphyrins on alumina. With
development has begun. Sometimes, it is sterically hindered compounds not allowing
best to carefully empty the column and dimerization, such as tetrakis(2,4,6-tri-
slice out the bands of interest for further methyl-phenyl)porphyrin, the monohyd-
treatment. It is prudent to empty the col- roxy Fe(III) porphyrin would be obtained.
umn of packing material after the separa- To isolate the solid OEP dimer, the
tion, as some sorbents swell and will break solution was filtered through a 0.45-µm
the column upon prolonged standing. filter, the solvent removed on a rotary evap-
Chromatography is an art, and often trial orator at 60°C, and the purple solid dried
and error is the only means of discovering overnight at 60°C in a vacuum oven. To
an efficient separation technique for new transform the dimer into the Fe(OEP)Cl
and unusual compounds. The following monomer, the CH2Cl2 solution was fil-
procedure illustrates a typical column chro- tered, saturated with HCl(g) followed by
matographic purification of a water insolu- N2(g) to remove excess HCl(g), and the
ble iron porphyrin, crude iron(III) OEP solvent was then removed under the hood
chloride (section 10.2, Procedure 4). on a steam bath. The monomer was dried
TLC of this iron porphyrin in HCl-free overnight at 70°C in a vacuum oven. The
CH2Cl2 on an alumina plate showed one HCl(g) could come from a lecture bottle
major band with an Rf value of 0.6 and a with a trap between the cylinder and solu-
moderate amount of substance that did not tion, or blown into the CH2Cl2 from the
move from the origin. The absorption spec- head-space of a reagent bottle of HCl.
tra of the impure compound in the same
solvent gave bands (and relative peak ❖ Procedure 1. Column Chromato-
heights) at 633.5 nm (1.0), 535.5 nm (1.9), graphy of Iron-Octaethylporphyrin
and 506 nm (1.8), indicating that the
major form in solution was the Fe(OEP)Cl 1. Wearing gloves and under the hood, a
monomer. If present, the metal-free H2- 3- × 50-cm glass column is fitted with
OEP would be the first species to elute, glass wool, tamped down with a meter
having bands in CH2Cl2 at 619.5 nm stick, and overlaid with approximately
(1.0), 566.0 nm (1.5), 531.5 nm (2.7), and 2 cm of acid-washed sea sand.
497.5 nm (2.9). In this particular example,
no H2-OEP was found, and approximately 2. The bottom of the column is placed in
500 mL of the red metalloporphyrin solu- a small diameter circular metallic
tion was collected. An approximately 2-cm clamp (to prevent the heavy column
thick impurity band was present at the ori- from sliding into the collection
gin, which did not move in CH2Cl2. The beaker), and the upper portion is held
addition of ethanol-stabilized chloroform vertical with a vinyl covered extension
eluted an unidentified minor band from the clamp (not overly tightened such that
origin material, and thus chromatography the column cracks).
in CHCl3 would have been undesirable. 3. A beaker is placed under the column,
The spectra of the eluted metalloporphyrin which is then loaded with 250 g of
in CH2Cl2 showed peaks at 588.0 nm (1.0) Grade IV 80 to 200 mesh alumina (A-
and 560.0 nm (1.7) indicating the 540; Fisher Scientific, Pittsburgh, PA,
Fe(OEP)Cl monomer had been trans- USA) that has been slurried in 400 mL
formed into the u-oxo dimer (OEP)FeIII- CH2Cl2. The slurry is poured into the
O-FeIII(OEP), which is typical behavior for column with the Teflon stopcock
most iron(III) protoporphyrin type esters open, taking care that solvent is always
43
J.C. Bommer and P. Hambright

present above the solid phase. If the the zinc invariably incorporates into a frac-
upper layer runs dry, more solvent tion or all of the H2-P on the plate forming
should be added and the upper layer the unwanted Zn-P complex. For fairly
stirred with a glass rod until no more basic porphyrins, shaking the organic sol-
bubbles percolate up through the slurry. vent containing the Zn-P with an approxi-
4. After settling, sand is added to a depth mately 3 M solution of HCl, followed by a
of approximately 2 cm. The alumina water wash, is often sufficient to remove
fills 64% of the column, leaving room the zinc from Zn-P. Another technique for
such that a reasonable amount of solu- less basic porphyrins involves adding triflu-
tion or solvent can be added at one oroacetic acid to a chloroform solution
time. containing the Zn-P and stirring until the
5. Two grams of impure iron OEP is dis- absorption spectrum of neutralized samples
solved in 350 mL of CH2Cl2, filtered, indicate that the two bands between 500
and slowly poured onto the column and 700 nm, due to Zn-P, have been trans-
with the stopcock in the open position. formed into the four-banded spectra char-
acteristic of many H2-P derivatives. The
6. Once all of the metalloporphyrin acidic mixture should be extracted repeat-
reaches the sand layer, small portions of edly with deionized water before shaking
solvent are added until the entire com- with a base. Immediate neutralization with a
pound has been absorbed, and then the dilute ammonia or NaOH often forms reac-
upper space is filled with methylene tive Zn(NH3)×2+ or Zn(OH)+/Zn(OH)3-
chloride. complexes, which then reintroduce zinc
7. The void volume of the solid phase is back into the porphyrin.
approximately 200 mL, and since the All TLC manipulations should be done
metalloporphyrin runs near the solvent under the hood and in dim light, as certain
front, about 200 mL of pure solvent is porphyrins and metalloporphyrins have a
collected before the porphyrin enters tendency to photodecompose on the plates.
the collection beaker. A line of closely spaced spots resolves better
than material simply streaked onto the
2.2. Thin Layer Chromatography plates, and the walls of the developing
chamber can be lined with absorbent paper
To purify small amounts of porphyrins towels to saturate the atmosphere in the
in the 1 to 100 mg range, or larger enclosure. A single- edged razor blade is
amounts if necessary, TLC plates usually useful for scraping off the various fractions,
give better resolutions than do columns, which should then be ground into a fine
with the resolution inversely proportional powder. Methanol often must be added to
to the thickness of the plate. Commercial- deactivate the packing material before the
ly, 200-, 250-, 500-, and 1000-µm silica porphyrin can be eluted from the powder
gel plates are available, and 2000-µm or with an organic solvent. With low Rf mate-
thicker preparative plates can be prepared rial, pretreatment with acetic acid is some-
in house. Alumina, microcrystalline cellu- times necessary.
lose, RP-C18-silica gel, Kieselguhr, An excellent TLC method for determin-
polyamide and Florisil (Mg silicate) plates ing the purity of methyl esters of natural
can be purchased. Fluorescent TLC plates porphyrins is one originally developed by
(and fluorescent column material) are to be Chu and Chu (25) and modified by Elder
avoided. These plates often contain Zn2+ (33). The system consists of chloroform,
salts as a component of the phosphor, and kerosene, and methanol in a volume ratio
44
General Laboratory Methods for Tetrapyrroles

of 200:100:7 on silica plates. This TLC ❖ Procedure 2. TLC Separation of the


system with modifications in the methanol Atropisomers of Tetrakis(2-amino-
content is useful over a broad array of the phenyl)porphyrin
more polar organic soluble porphyrins and
metalloporphyrins including many syn- 1. Under the hood, 100 mL of 1:1
thetic derivatives. There is something (vol/vol) benzene-diethylether is placed
about the kerosene that cannot be dupli- in a 27.0 × 26.5 × 7.0 cm developing
cated by substitution with any pure chamber, which is covered and allowed
hydrocarbon. to stand for 20 minutes.
If only two or three bands of slightly dif- 2. A saturated solution of tetrakis(2-
ferent Rf values are present, we have had aminophenyl)porphyrin (26) is pre-
success using radial chromatography on a pared in benzene and filtered.
commercial Chromatotron (Harrison 3. Eight individual spots are made
Research, Palo Alto, CA, USA) with an approximately 2 cm above the bottom
automated sample collector to isolate 50 to of a 5 × 20 cm 250 µm, 60 Å pore size
100 mg amounts of pure material. Circu- general purpose TLC plate (Aldrich
lar silica gel 1-, 2-, and 4-mm thick disks Chemical, Milwaukee, WI, USA),
are readily prepared. The same disk can be using an open capillary tube for spot-
reused many times. A typical TLC proce- ting and allowing the solvent to evapo-
dure is outlined below for the separation of rate.
the atropisomers of tetrakis(2-amino- 4. The plate is placed in the chamber and
phenyl)porphyrin. developed until the solvent is 5.0 cm
In this example, the Rf values were cal- above the origin.
culated as 0.12 (0.60/5.0) for the α,α,α,α 5. The plate is removed from the cham-
isomer, 0.6 for α,α,α,β, 0.8 for α,α,β,β, ber, and a pencil is used to mark quick-
and 0.88 for the least polar α,β,α,β com- ly the four band positions (0.60, 3.0,
pound. The notation α,α,α,α is for the cis- 4.0, and 4.4 cm) and the height of the
isomer with all four amino groups on the solvent front (5.0 cm).
same side of the porphyrin plane; α,α,α,β
is for three amino groups on one side and 2.3. High-Pressure Liquid
the other in the opposite direction, and so Chromatography
on. The Rf s from the literature are 0.04,
0.43, 0.64, and 0.77, respectively (26), HPLC seems an especially attractive
indicating the variability often found method for analysis of tetrapyrroles. How-
between laboratories. Statistically, the rela- ever, like many analytical methods, it can
tive concentration of each isomer should be deceiving without input from other ana-
be 4 for α,α,α,β, 2 for α,α,β,β, 1 for lytical techniques. Similar to analysis of
α,α,α,α, and 1 for α,β,α,β. The observed just about any material by HPLC, truly
intensities of the spots were in qualitative accurate analytical results must rely on
agreement with predictions. In some cases, comparing the response in the chro-
investigators isolate each band and use matogram of the sample being tested to the
absorption spectrophotometry to deter- response of a standard sample of known
mine the relative amounts of each com- purity and concentration. There are some
pound. A small amount of unidentified points to be aware of when doing qualita-
material was found at the origin, which is tive or semiquantitative work by HPLC.
usually the case for impure porphyrins. Many detection systems for tetrapyrrole

45
J.C. Bommer and P. Hambright

work are chosen for their sensitivity it is injected, will often change the relative
towards the compounds of interest, e.g., size of the eluted peaks to give a good indi-
wavelengths near the Soret maxima for cation that artifacts are indeed a problem.
UV/VIS absorbance detection systems. Generally, we have found that normal
This can give a good idea of purity relative phase HPLC of porphyrins tends to be less
to contamination by other tetrapyrroles, reproducible than reverse phase chro-
but tells one nothing about what other matography. Very precise conditioning of
types of dissimilar organic or inorganic the column seems to be required, and
compounds may reside in the sample. The therefore, we have moved to reverse phase
same can be said for fluorescence detection, systems exclusively. Systems that seem to
except in this case, another class of conta- work well for monocarboxylic through
minant, nonfluorescent or weakly fluores- tetracarboxylic tetrapyrroles use C-18
cent metallo-tetrapyrroles, may be missed columns with eluant consisting of tetrabut-
or under-represented along with other ylammonium phosphate (0.002–0.01 M)
nonfluorescent compounds. Also, only at about pH 2.7 and methanol or acetoni-
substances that are actually eluted from the trile as organic modifiers. Some systems for
column are detected. In the case of more polar tetrapyrroles rely on sodium or
tetrapyrroles, there are a number of conta- potassium phosphate buffers at near neu-
minants that will stick more or less irre- tral pH with concentrations from 0.01 to
versibly on the column, such as partially 0.1 M containing methanol or acetonitrile.
esterified porphyrins in the instance of sili- The higher salt concentrations are most
ca chromatography of porphyrin car- useful for the most polar tetrapyrroles,
boxylic acid esters and black polypyrroles, such as highly sulfonated species or uro-
etc., from synthesis of meso-substituted porphyrins. It is best to use some type of
porphyrins. Thus, initially, TLC is useful gradient system unless analyzing very pure
in determining whether such contaminants samples, so that less polar contaminants are
are present in the HPLC samples. eluted from the column in a reasonable
HPLC chromatography of tetrapyrroles period of time.
is particularly prone to artifacts. This prob- As an example, the system we use to
ably arises from the relative insolubility of evaluate a mixture of porphyrin carboxylic
most of these compounds and their ten- acids, which contain from 2 to 8 carboxyl
dency to aggregate. This is especially pro- groups is as follows: Hamilton PRP-1 poly-
nounced in reverse phase chromatography meric supported C-18 column with a gra-
of some of the less aqueous soluble por- dient from 0% acetonitrile in 0.01 M sodi-
phyrins such as mono- or dicarboxylic acid um phosphate buffer at pH 6.85 and flow
porphyrins or chlorins derived from natur- 2 mL/minute to 25% acetonitrile over 14
al sources and some of the derivatives of minutes, then a further gradient to 80%
meso-tetraphenylporphine. Systems that acetonitrile over 2 minutes.
tend to minimize this problem usually
involve ion-pairing reagents such as tetra-
butylammonium ion and a relatively low 3. RECRYSTALLIZATION
pH. Sometimes, multiple peaks appear on
the chromatogram, which seem to be due As with many organic compounds,
to dimers and higher aggregates that are recrystallization can be a powerful tool for
relatively stable under the chromatograph- the purification of tetrapyrroles. It is gen-
ic conditions. Changing the concentration erally only practical however when more
of injected sample, or the solvent in which than a few milligrams of compound are
46
General Laboratory Methods for Tetrapyrroles

available. It has been our experience that As noted in Section 4, tetrapyrroles are
for most porphyrins, a fairly high level of notorious for retaining solvents, especially
purity, perhaps 80% or better for tetrapyr- if crystallized from a high boiling solvent
role impurities and somewhat less if impu- like N,N-dimethylformamide (DMF) and
rities are not tetrapyrroles, is necessary a certain amount of DMF decomposes
before satisfactory results can be obtained upon heating into dimethylamine, which
by recrystallization. It has also been our binds to certain metals in metallopor-
experience that certain types of porphyrins phyrins. Sometimes these molecules can
do not crystallize well, and recrystallization only be removed by heating the solid near
as a purification step is not productive. the boiling point of the solvent, if the sta-
Type III isomers of the biologically impor- bility of the tetrapyrrole allows. The re-
tant porphyrins for instance are very diffi- moval can be followed by monitoring the
cult to crystallize, but type I isomers crys- increase in absorbance at the Soret peak for
tallize nicely with improvement in isomeric a standard concentration by weight over a
purity as well as removal of other por- period of hours or days. Lower tempera-
phyrin contaminants. Some porphyrins tures can be employed if the substance is
can be recrystallized as the dihydrochlo- placed under vacuum in a vacuum oven or
rides by dissolution in concentrated HCl drying pistol.
and refluxing until constant boiling HCl is
achieved then adding water slowly to the ❖ Procedure 3. Recrystallization of
refluxing solution until a lower normality Protoporphyrin IX Dimethyl Ester
of HCl is achieved in which the porphyrins
have reduced solubility. In general, dissolu- 1. Crude protoporphyrin IX DME (ap-
tion in a minimum amount of low boiling proximately 7 g) is obtained by remov-
solvent in which they have high solubility, ing the iron from 10 g of hemin with
then adding an equal or greater amount of concurrent esterification [Grinstein
a higher boiling solvent in which they have method (29)] and partially purified by
only slight solubility can recrystallize small flash chromatography over silica or alu-
quantities of porphyrins or other tetra- mina eluting with 10% ethyl acetate in
pyrroles. The solution in an open contain- dichloromethane.
er is then left to evaporate at room temper-
ature or slightly warmed on a trivet until 2. The solution containing the porphyrin
the desired degree of crystal formation is (about 300–500 mL) is placed in a
complete. Filtering and washing with the L-rotary evaporating flask, and the
higher boiling solvent completes the opera- removal of solvent under vacuum is
tion. Larger amounts of tetrapyrroles can commenced using a 50° to 60°C bath
be handled more efficiently and rapidly by with continuous addition of 500 mL of
recrystallization on a rotary evaporator. In ethyl acetate. The rate of addition is
this case, the tetrapyrrole is dissolved in a controlled such that the volume in the
low boiling solvent, placed in a round bot- flask approaches about 300 mL con-
tom flask on the rotary evaporator fitted currently with complete addition of the
with a continuous feed tube, and the sol- ethyl acetate.
vent removed under reduced pressure while 3. Completion of the crystallization occurs
adding the higher boiling solvent in which upon standing in the refrigerator for sev-
the compound is less soluble, until the eral hours, at which time the crystals are
desired degree of crystallization is reached. collected by filtration on a Buchner fun-
A typical procedure is described below. nel and washed with ethyl acetate.
47
J.C. Bommer and P. Hambright

4. The product is analyzed by the Elder monomer and 15% of the µ-oxo-bridged
kerosene TLC technique (33), and the P-Fe-O-FeP dimer (91). The CHN analy-
procedure can be repeated as desired if ses and corresponding ratios are always
further purification is necessary by dis- helpful, but the moles of water are usually
solving in minimum dichloromethane determined by calculation based on the
and again exchanging with ethyl acetate. observed values. Crystal structures of
5. The yield is about 5 g, and an overall water-insoluble porphyrins usually contain
purity of about 97% to 98% can be several molecules of the crystallizing sol-
expected after one or two recrystalliza- vent, and tetra-arylporphyrin crystals have
tions. channels throughout the solid that occlude
solvent molecules and act as “porphyrin
sponges” (19). On new compounds, most
4. ANALYSIS OF PORPHYRINS AND authors report CHN and metal analyses,
METALLOPORPHYRINS the absorption spectra and extinction coef-
ficients, infrared (IR) band positions, the
It is often important to know the con- analyzed 1H nuclear magnetic resonance
centration in solution of a particular por- (NMR) data, and the major peaks obtained
phyrin or metalloporphyrin. Since this by mass spectrometry.
class of molecules often contains various It is better to determine solution con-
kinds and amounts of nonporphyrin mate- centrations based on known extinction
rial that are isolated along with the por- coefficients, than to rely on calculations
phyrin itself, simply weighing out a given based on the weight of the sample. Due to
amount of compound may give inaccurate aggregation effects, the spectra should be
results. With water-soluble porphyrins, measured at concentration levels similar to
water molecules are usually indicated by the value quoted in the literature. DiNello
chemical analysis of the isolated solids. The and Chang (29) give band positions and
anionic water-soluble H2-TPPS4 (Figure 1) extinction coefficients in CHCl3 for the
analyzes as Na4H2-TPPS4·10 H2O when free bases of modified natural porphyrin
oven dried at 110°C (83), the cationic pyri- derivatives, and for the corresponding
dinium compound as H2-TMPyP(4)Cl4·4 Fe(II) pyridine hemochromes, measured in
H2O, various phenyl/(4-sulfonatophenyl), 4 M pyridine-0.2 M KOH with the addi-
and phenyl/(N-methyl-4-pyridyl) por- tion of small amounts of the reducing
phyrins contain from 2 to 11 moles of agent sodium dithionite. Caughey and
water, often in nonstoichiometric amounts, coworkers (23) have spectra on other free
as Zn-TPPS4 analyzes for 16.6 H2O. base 3,8-disubstituted deuteroporphyrin
Thermo-gravimetric work (53) indicated DMEs and metallo derivatives (22).
that the latter compound gradually lost Caughey et al. have compiled data on iso-
weight up to 200°C, a constant-weight mers of uroporphyrins and copropor-
region was found in the 220° to 400°C phyrins and other biologically important
range, while rapid decomposition was molecules (21). Fuhrhop and Smith (44)
noted above 400°C. Compounds prepared have five tables of extinction coefficients of
at different times sometimes contain differ- various porphyrins and metalloporphyrins
ing amounts of water. The Fe(III)- in organic solvents and in aqueous sodium
TMPyP(4) is a monomer in acid, but dodecyl sulfate solutions. The spectra of
when precipitated from acid solution by centrally N-alkylated porphyrins and met-
the addition of 1 M HClO4, Mossbauer alloporphyrins are found in Lavallee’s
spectra on the solid indicated 85% monograph (71), and James and coworkers
48
General Laboratory Methods for Tetrapyrroles

(83) give data on well-characterized water- Finally, there are a number of laboratory
soluble porphyrins and their precursors. procedures to determine porphyrin con-
Methods for the determination of por- centrations. In Drabkin’s method, iron
phyrins in biological material have been protoporphyrin IX is digested with hydro-
reviewed (103). gen peroxide in basic solution, the liberat-

Figure 1. The structures of some water-soluble porphyrins and several of their precursors. Each compound is the porphyrin
with the indicated substituents on the four meso (5, 10, 15 and 20) positions. H2-TPP: meso-tetraphenylporphyrin; H2-
TPyP(4), meso-tetrakis(4-pyridyl)porphyrin; H2-TMPyP(X): the ortho (2), meta (3) and para (4) isomers of meso-tetrakis(N-
methyl-X-pyridyl)porphyrin; H2-TPPC4, meso-tetrakis(4-carboxyphenyl)porphyrin; H2-TPPS4: meso-tetrakis(4-sul-
fonatophenyl)porphyrin; H2-TAPP, meso-tetrakis(4-N,N,N-trimethylanilinium)porphyrin. A fuller explanation of the
nomenclature of porphyrins can be found in Chapter 2.

49
J.C. Bommer and P. Hambright

ed ferric iron reduced with ascorbate, and in the delivery of oligonucleotides into the
the Fe(II) spectrophotometrically analyzed nucleus (43). Four coordinate Cu(II),
with o-phenanthroline (32). With metallo- Ni(II), and Au(III) TMPyP(4) complexes
TPPS4 complexes, approximately 10-mg intercalate into certain DNAs at low ionic
samples were digested with a 3:1:1 mixture strengths, and show external groove bind-
of H2SO4-HNO3-HClO4, and the metal ing at higher salt levels (89). The N-methyl
then determined by titration with EDTA groups in the ortho position of metallo
(53). Brisbin and coworkers (13) spectro- TMPyP(2) complexes provide a steric bar-
photometrically determined the concentra- rier to intercalation, and the bulky tetraca-
tion of protoporphyrin IX DME to ± 2%. tionic tetrakis(4-N,N,N-trimethylanilini-
To a constant amount of porphyrin um)porphyrin, H2-TAPP4+ also does not
(approximately 10-5 M) in acetic acid, high- intercalate (81). Fe(III) and Mn(III)-
er and lower concentrations of standard- TMPyP(X) compounds are excellent super-
ized metal acetate solutions (divalent Zn, oxide dismutase mimics (10–12), and they
Co, Ni, Fe, and Cu) were added. After also transform the cytotoxic peroxynitrite
heating to completely form the 1:1 com- (ONOO-) into NO2 and the O = Mn(IV)
plex, appropriate plots of absorbance ver- porphyrin (36,73). Mn(III)-tetra(N-ethyl-
sus metal ion concentration indicate the 2-pyridyl)porphyrin is superior to oxyhe-
point at which the concentration of the moglobin for the determination of NO
metal is equal to the concentration of the concentrations. Water-soluble Fe(III),
porphyrin. This method can also be Mn(III), and Gd(III) porphyrins have been
applied to determine the concentration of explored as liver and tumor imaging agents
the metals (2–5 × 10-5 M) by titration with using magnetic resonance imaging (MRI)
a known concentration of porphyrin. Petro techniques (79), and radioactive sulfonated
and Marzilli (93) determined the molar metalloporphyrins have potential as tumor
extinction coefficients of a series of cation- localizing agents in nuclear medicine. Nat-
ic porphyrins by adding excess standard- ural and synthetic water-soluble porphyrins
ized Zn(II) to aqueous solutions of the por- act against the human immunodeficiency
phyrins at pH 12.0. After the reaction was virus (30), and H2-TMPyP(2) and H2-
complete, the pH was brought to 9.0, and TMPyP(4) both inhibit human telomerase
excess zinc was determined with the colori- (51,58). Porphyrins that produce singlet
metric zinc reagent, zincon. The precision dioxygen within tumors are valuable in
of this method was approximately 3%. photodynamic therapy (108). The struc-
tures and common abbreviations used for
certain water-soluble compounds are
5. WATER-SOLUBLE PORPHYRINS shown in Figure 1.

There are a variety of synthetic por-


phyrins and metalloporphyrins with posi- 6. PRECURSORS FOR CATIONIC
tive and negatively charged substituents AND ANIONIC PORPHYRINS
that are of biochemical and biomedical
interest. The meso-tetrakis(N-methyl-4- From health considerations, we mention
pyridyl)porphyrin, H2-TMPyP(4)4+ and its again that one should always wear inexpen-
3 and 2 isomers (Figure 1) are soluble in sive disposable gloves when working with
water the full pH range and monomeric porphyrins and the reagents and solvents
below 10-4 M in aqueous solution involved in porphyrin synthesis. Most work
(20,49,62). These cationic compounds aid should be done in a well-ventilated hood.
50
General Laboratory Methods for Tetrapyrroles

The H2-TMPyP(X)4+ compounds are DDQ (100). The cooled solution is


made from the meso-tetrakis(4-pyridyl)por- extracted with a 1% NaOH solution con-
phyrin, H2-TPyP(4), and its isomers. These taining sodium dithionite, and the organic
are prepared by refluxing, for 45 minutes, a phase then washed with water. The toluene
propionic acid solution which is 0.24 M in is removed under vacuum, and the chlorin-
pyrrole and 0.24 M in the 4(3 or 2)-pyri- free porphyrin is crystallized from
dine-carboxaldehyde (78). In contrast to CH2Cl2-MeOH. The solubilities in 17
tetraphenylporphyrin, H2-TPP, which is solvents of the H2-TPyP(X) compounds
prepared in the same manner and precipi- have been tabulated (109), and the
tates from solution (3), the pyridyl por- observed order 3 >>2 >4, was also noted
phyrins are soluble in propionic acid. One for other tetraaryl porphyrin isomers (42).
method of isolation involves adding a The Adler-Longo propionic acid method
large volume of water to the cooled mix- (3) and the Smith DDQ oxidation proce-
ture, and then stirring in solid sodium dure outlined above are general techniques
acetate to bring the pH to approximately for the preparation and purification of an
3.0 (48). The pyridyl groups are deproto- array of meso-substituted porphyrins.
nated at this pH, and the purple por- More complicated meso-substituted
phyrin filtered off and washed with compounds can be prepared by the “mixed
methanol, DMF, and water. Another aldehyde” approach (6). For example, 0.1
method is to evaporate off all of the pro- moles each of benzaldehyde and 4-pyridine
pionic acid (78) and titurate the resulting carboxaldehyde are mixed with 0.2 mole of
tar with DMF. Most of the impurities are pyrrole and refluxed in propionic acid
soluble in DMF, and the porphyrin is fil- (105). TLC on silica gel plates developed
tered from the cooled solution. Other with 97.5/2.5 chloroform-methanol show
groups neutralize the tar with NaOH, dis- six bands for the product, with Rf values of
solve the material in CH2Cl2, and run 0.97 for H2-TPP, 0.94 for the monopy-
column chromatography on alumina ridyl, 0.86 for trans, 0.75 for cis, 0.66 for
(slurried in acetone), eluting the por- the tri(4-pyridyl)-mono-phenyl, and 0.60
phyrin fraction with CH2Cl2 containing for H2-TPyP(4). The compounds can be
5% to 10% pyridine (62). The yields isolated on a preparative scale from Florisil
approach 16% with the para- and meta- columns eluted with CH2Cl2 mixed with a
isomers, while less than 1% of H2-TPyP(2) more polar solvent (101). Thus, the mono-
can be isolated under such conditions. 4-pyridyl requires 1% to 5% acetone, the
To obtain pure compounds, the crude trans requires 5% to 15% acetone, the cis
porphyrin is dissolved in HCl-free chloro- requires 20% to 50% acetone, the tri(4-
form, and refluxed with 2,3-dichloro-5,6- pyridyl)-mono-phenyl requires 2% MeOH
dicyano-1,4-quinone (DDQ) in order to and 10% MeOH for H2-TPyP(4). The ini-
oxidize the 5% to 10% chlorin impurity tial aldehyde ratio can be adjusted to pro-
that is always formed from the porphyrin duce more or less of a given component.
itself during the synthesis (9). The warm Sterically hindered tetraaryl porphyrins
mixture is then chromatographed on a col- containing 2,6-dichloro, 2,6-dibromo, or
umn of dry alumina, the porphyrin eluted 2,6-dimethylphenyl, and 2,4,6-trimethyl-
with CH2Cl2, and finally recrystallized phenyl groups are produced in low yield
from CHCl3-MeOH. To avoid chro- from the Adler-Longo propionic acid pro-
matography on a prepurified porphyrin cedure, but are often readily synthesized
contaminated with its chlorin, the mixture with the Lindsay room temperature
can be refluxed in toluene containing method (77). The aldehyde and pyrrole in
51
J.C. Bommer and P. Hambright

a 1:1 ratio (each approximately 10-2 M) are reaction is complete. To ascertain the
mixed in CH2Cl2 or CHCl3 containing degree of N-alkylation, a sample from the
0.75% EtOH and approximately 10-3 M pot is spotted on a silica gel TLC plate,
BF3-OEt2 (as the acid catalyst) and stirred and the plate is developed with a 1:1:8
for several hours at 25°C. The cyclic por- (vol/vol/vol) mixture of saturated aqueous
phyrinogen formed is then oxidized in the KNO3-water-acetonitrile (10). During
same pot to the porphyrin with DDQ or the course of the reaction, six bands are
p-chloranil at reflux, and the impurities are observed, with the slowest moving and
removed by chromatography. last remaining the tetra (N-alkylated)-por-
phyrin. Other workers use 3:3:1:2:1 iso-
propanol-H2O-acetone-acetic acid-con-
7. N-ALKYLATIONS TO PREPARE centrated NH3 for the separation of
CATIONIC PORPHYRINS differently charged cationic porphyrins
and metalloporphyrins. The sterically hin-
It should be noted that all alkylating dered H2-TPyP(2) was also tetra-N-
agents are hazardous, and extreme caution methylated in neat dimethyl sulfate at
should be taken when working with these 110°C. The same N-methylation tech-
substances. Many workers methylate the niques in DMF are used to prepare the
H2-TPyP(X) compounds in hot or reflux- tetrakis[N-methyl-4 (or 3) quinolyl]por-
ing chloroform in the presence of excess phyrins (1), and the popular tetra
CH3I, and the solid iodide salts of H2- (4-N,N,N-trimethylanilinium)porphyrin,
TMPyP(X) precipitate from solution. Since H2-TAPP from tetra(4-N,N-dimethyl-
the iodides are not very soluble in water, the anilinium)porphyrin (65). Evaporating
product is stirred with the chloride form of the water from an aqueous solution of
an ion-exchange resin either in water, or in M-TAPP in the oven leads to loss of the
water–methanol, and warmed until the N-methyl groups.
solid dissolves. After filtration, the solution Several examples of water-soluble “pick-
is slowly passed through a long column of et-fence” porphyrins have been prepared.
chloride resin, and the water is removed by The starting material, tetra(2-nitro-
lyophilization (90). In some cases, both the phenyl)porphyrin, is synthesized by the
tri- and tetra-N-methylated iodides precipi- Adler-Longo technique in acetic acid (26).
tate from chloroform, as indicated by elec- This compound is dissolved in concentrat-
trophoresis studies on the products (16), ed HCl and reduced to the tetra(2-
and thus full tetra-N-methylation is not aminophenyl)porphyrin with SnCl2 at
always achieved in chloroform with CH3I. 70°C. The H2-T(2-NH2P)P is a mixture
The N-methylation is perhaps best done in of four atropisomers, with the amino
DMF with methyl-p-toluenesulfonate groups above and below the porphyrin
(MTS). In a typical procedure, 0.5 g of por- plane. A TLC method to separate these iso-
phyrin is added to 50 mL of DMF in a mers is given in section 2.2.1. An 8- × 30-
100-mL flask (83). The solution is warmed, cm column filled with a silica gel-chloro-
and before boiling, 2 mL of MTS is added. form slurry was used on the preparative
The solution is refluxed for 4 hours, and the scale. The column was loaded with a chlo-
tosylate salt of the porphyrin is removed roform solution of the atropisomers, and
from the cooled solution by filtration and the three undesired and less polar com-
ion-exchanged into the chloride form. In pounds removed with 1:1 benzene:diethyl
some instances, the N-alkylated porphyrins ether. The target and most polar cis-
decompose if not isolated soon after the α,α,α,α isomer was then eluted with 1:1
52
General Laboratory Methods for Tetrapyrroles

acetone:diethyl ether. The other three isomers per with concentrated H2SO4. While most
were re-equilibrated at 100°C in CHCl3- water-soluble manganese porphyrins are pro-
toluene, forming more of the desired duced in the 3+ oxidation state, the Mn(II)-
α,α,α,α species. More efficiently, the isomer β-Br8-TMPyP(4) is the stable form of this
mixture is refluxed overnight in benzene in electron deficient porphyrin having a
the presence of silica gel. As it forms, the deformed nuclear structure (11). One to
α,α,α,α is preferentially adsorbed on the four chlorine atoms can be added to the
solid and can be removed with 1:1 acetone: β-pyrroles of H2-TPyP(2) by refluxing the
ether (76). The reaction of nicotinic anhy- compound in CHCl3 with N-chlorosuccin-
dride at room temperature in CH2Cl2-pyri- imide (60). The products are separated by
dine with the amino compound forms the chromatography, and the H2-β-ClxTEt-
α,α,α,α-tetrakis(o-nicotinamidophenyl)por- PyP(4) are then formed in DMF by the
phyrin (85). This species can be gently N- addition of ethyl-p-toluenesulfonate. The
methylated in dry trimethyl phosphate by the sterically hindered 2,6-dichloro-TMPyP(4)
addition of methyl trifluoromethylsulfonate, has been prepared (57), as well as an octaca-
with added 2,6-lutidine to scavenge protons. tionic derivative (54). A series of compound
The ortho-isonicotinamido compound has containing (N-methyl-4-pyridyl) groups on
also been prepared (50,113). The four atro- the β-pyrrole positions have also been synthe-
pisomers of the water-soluble Cu(II)- sized (34). Tetraphenyl type porphyrins with
TMPyP(2) could be separated on silica gel -CH2X substituents in the para positions,
TLC plates developed with 2-butanone-con- with X = N+Et3, N+Ph3, NH2, and PO32- are
centrated NH3-NH4PF6-n-butylamine. The known, and porphyrins have been made
Zn(II) and Ni(II) isomers, but not those of water-soluble by the addition of sugar
the metal-free H2-TMPyP(2) or its Mn(III) residues (47). Other compounds contain
complex, could also be separated under such three (N-methyl-4-pyridyl) groups for water
conditions (64). solubility, and the fourth phenyl or pyridyl is
Refluxing the cobalt(II) complex of the derivatized with substituents that can interact
meso-tetrakis(pentafluorophenyl) porphy- with nucleic acids (75). Four moles of ethyl-
rin overnight in DMF (61) leads to the enediamine (and related diamines) have been
production of meso-tetrakis-[2,3,5,6,tetra- added to protoporphyrin IX DME to form
fluoro-4-(dimethy lamino)phenyl]porphy- acid-soluble compounds (117), and similar
rinato cobalt(II). This complex can be con- species containing two moles of ethylenedi-
verted into the water-soluble triflate salt amine can be prepared from meso or
(68,69) using methyl trifluoromethanesul- deuteroporphyrin IX DME. These protopor-
fonate in trimethyl phosphate overnight at phyrin derivatives are soluble over a wider pH
60°C under N2. The metallo triflate salts range if the -NH-(CH2)2-N+Me3 forms are
are stable at room temperature, while the prepared, using techniques similar to those
solid chlorides decompose within days. described above. Then an octacationic
The electron withdrawing tetrafluo- tetrakis[2,4,6-trimethyl-3,5-bis(-CH2N+Me3)
rophenyl groups reduce the electron densi- phenyl]porphyrin is known (5).
ty at the central nitrogen atoms, and a larg-
er effect can be achieved by halogenations
at the β-pyrrole positions (31). Thus, 8. NEGATIVELY CHARGED
Cu(II)-TMPyP(4) dissolved in DMF can PORPHYRINS
be β-octabromonated (96) by addition of 8.1. Synthetic Derivatives
Br2(l), and the metal-free H2-β-Br8TM-
PyP(4) is prepared by removal of the cop- The synthetic tetranegatively charged
53
J.C. Bommer and P. Hambright

tetra(4-carboxyphenyl)porphyrin, H2-TPPC4 is to add monoprotonated o-phenanthro-


is prepared by the Adler-Longo method line to a pH approximately 4.0 solution of
in propionic acid, and is water-soluble H2-TPPS4. The insoluble (H-Phen+)4/H2-
above pH 7.0 due to ionization of the car- TPPS44-.2 H2O salt precipitates, and can
boxylic acid groups (78). Porphyrins with be washed with water to remove extraneous
carboxylic acids in the meta- and ortho- ions. The solid is then slurried with an ion
phenyl positions are also known. It is often exchange resin in the Na+ form until dis-
best to prepare these compounds as their solved and passed through a sodium ion-
methyl esters, which can be purified by exchange column to remove the protonated
chromatography, and hydrolyze the esters o-phenanthroline. The partially sulfonated
in base at a later stage (28). An enormous compounds can be isolated using low-pres-
amount of work has been done with sure liquid chromatography columns
tetrakis(4-sulfonatophenyl)porphyrin, H2- packed with LiChroprep RP-18 silica gel
TPPS4 and its metal complexes. This por- and eluted with mixtures of MeOH/phos-
phyrin is soluble in water down to pH phate-buffered water (110).
approximately 2.0, and, at lower pHs, Using neat chlorosulfonic acid at 100°C
appears colloidal in solution. To prepare with the tetra(2,6-dichlorophenyl)-por-
this compound, H2-TPP is added to con- phyrin, the 3-SO2Cl species was isolated,
centrated H2SO4, and the mixture is heat- and hydrolysis produced the 2,6-dichloro-
ed at 100° to 110°C (66). To monitor the 3-SO3-phenyl derivative (45). Sulfonation
extent of sulfonation, a sample is neutral- of the tetrakis(pentafluorophenyl) por-
ized (110) and spotted on a reverse phase phyrin (7) with oleum for 10 hours at
KC-18 TLC plate (Whatman, Clifton, NJ, 140°C leads to four -SO3- groups on the β-
USA), and developed with 80/20 MeOH- pyrrole positions, while a 3,5-disulfonated
H2O (pH approximately 7.4, 0.01 M product was found for the octabromonated
phosphate buffer). The Rf values are 0.94 tetrakis(2,4,6-trimethylphenyl)porphyrin (54).
for the fully sulfonated H2-TPPS4, 0.88 for With compounds containing both phenyl
the trisulfonated H2-TPPS3, 0.74 for trans and 4-pyridyl groups, only the phenyl rings
-H2-TPPS2, 0.59 for cis-TPPS2, and 0.12 sulfonate (83).
for H2-TPPS1. When the reaction is com-
plete, ice is added to the green solution, and 8.2. Anionic Compounds from Natural
the H2SO4 is carefully neutralized with Porphyrins
concentrated NaOH, adding more ice as
needed. The transformation of the por- Anionic porphyrins, metalloporphyrins,
phyrin from the green diacid (H4-TPPS42-) and their derivatives from natural sources
into the red free base begins at pH approxi- have found a wide variety of usage in mod-
mately 5.0. When the pH reaches about ern medicine and biochemistry including
9.0, the water is evaporated in the oven, the field of photodynamic therapy for vari-
and after pulverizing the resulting solid, it ous disease states, heme oxygenase inhibi-
is extracted with methanol in a Soxhlet tion for prevention of jaundice, and inhi-
apparatus. The sodium salt of H2-TPP4 is bition and induction of this enzyme as a
soluble in MeOH, and the Na2SO4 tool for biochemical research. Some metal-
remains in the cup. For further purification, loporphyrins have been used as dioxygen
some groups use dialysis techniques, while detectors in fluids or air via phosphores-
others add acetone to a concentrated solu- cence quenching and as MRI contrast
tion of H2-TPPS4 in methanol to precipi- agents (47). Of course the porphyrins
tate a brown solid. A useful procedure (59) along the heme and chlorophyll biosyn-
54
General Laboratory Methods for Tetrapyrroles

thetic pathways are employed as standards or alumina and chromatography must be


for intermediates excreted in various dis- carried out rapidly. The chloroform,
ease states and for biomedical research of kerosene, and methanol system in a volume
these diseases. ratio of 200:100:7 on silica plates men-
Isolation of many of the porphyrins and tioned earlier is extremely useful for deter-
chlorins from natural sources has been mining the purity of these compounds.
described in Chapter 2 by Smith. The most Many of the porphyrins, which occur as
common anionic chlorins one encounters porphyrinogens along the biosynthetic
in the laboratory are derived from chloro- pathways can be isolated from the natural
phyll a or b. Pheophorbide a or b each have sources such as protoporphyrin from
a single free propionic acid group and as hemin, coproporphyrin I from the urine or
such have very limited water solubility. feces of animals or humans having certain
They can be handled in aqueous solutions types of porphyria (20,97,118), copropor-
containing 50% or more water-miscible phyrin III from bacterial sources (70,86),
organics such as methanol and can be puri- and uroporphyrin I from the urine of cattle
fied by chromatography on C-18 silica (118) or humans (98) having congenital
adsorbents using sodium phosphate-buf- porphyria. Porphyrins excreted from these
fered aqueous–organic eluants. Purity can natural sources can usually be concentrat-
be checked with TLC on C-18 silica plates ed at a neutral pH by collection on a
(Si-C-18; J.T. Baker, Phillipsburg, NJ, reverse phase adsorbent such as Sep-pak C-
USA), eluting with 85% methanol, 15% 18 cartridges (Waters, Milford, MA, USA)
0.01 M sodium phosphate buffer at pH for small quantities or bulk C-18 packing
6.85. The Rf values are approximately 0.44 in a Buchner funnel for large volumes. The
for pheophorbide a and approximately porphyrins may then be partially purified
0.30 for pyropheophorbide a. by careful elution with methanol–buffer or
Chlorin e6 can be obtained from pheo- acetonitrile–buffer solutions. A note of
phorbide a or pheophytin a by basic hydro- caution when working with biological sam-
lysis of the refluxing alcoholic solutions ples that may contain porphyrinogens:
using NaOH or KOH (27,37,40) and One should not make the solutions strong-
purified on C-18 silica packing as the free ly acidic before oxidation to the por-
carboxylic acid form similar to the proce- phyrins, which can be accomplished with
dure for pheophorbides but with higher addition of iodine in ethanol, since even at
aqueous content of the eluant. The TLC room temperature, we have noted that a
system to check for purity is as above, but substantial amount of scrambling to the
with the eluant 75% methanol-25% buffer. isomer mixtures can occur. The porphyrins
The Rf values are approximately 0.76 for can be isolated from the above solutions
chlorin e6 and 0.66 for chlorin e4, the through removal or the organic solvent by
meso-acetic acid decarboxylation product rotary evaporation, then flocculation at pH
of chlorin e6. In all cases the Rf’s for the 4.0 followed by collection by centrifuga-
chlorophyll b derivatives are slightly greater tion and washing with water adjusted to
than found for the corresponding chloro- pH 3.0 to 4.0 with acetic acid. Further
phyll a products. The methyl esters of the purification can be achieved by reverse
above chlorins can be purified by silica or phase chromatography or esterification to
alumina column chromatography using the methyl esters and silica or alumina
CHCl3 or CH2Cl2 containing varying chromatography. The methyl esters can be
amounts of ethyl acetate. Pheophorbide a , checked by the Elder TLC system
however, is unstable in the presence of silica described above, and the free carboxylic
55
J.C. Bommer and P. Hambright

acid forms can be evaluated on C-18 plates tion, then twice more with the same vol-
with 70% to 80% methanol–sodium phos- ume of deionized water. The volume is
phate buffer system for porphyrins with reduced on a rotary evaporator, and the
four or fewer carboxy groups and 50% to porphyrin mixture is applied to a silica or
60% methanol/1 or 2 M ammonium alumina column to effect separation and
acetate for porphyrins with four or more purification of the components.
carboxyl groups. If porphyrin esters are not desired, the
porphyrins may be collected from the
8.3. Isolation of Natural Porphyrins decanted and filtered growth medium
from Bacterial Cultures directly onto the bulk C-18 silica reverse
phase packing such as that available in 55
Many bacteria, especially photosynthet- to 105 µm size from Waters or Millipore
ic bacteria, produce substantial amounts of (Bedford, MA, USA) activating first with
porphyrins, porphyrinogens, and bacteri- methanol then washing with water. The
ochlorophyll, or can be made to do so packing is then washed with water, and the
under certain conditions of stress. In gen- porphyrins eluted with 90% methanol and
eral, the porphyrins or porphyrinogens are water vol/vol. The solvent is removed by
mostly excreted into the growth media and rotary evaporation, and the porphyrins
can be treated separately from the bacteri- taken up in water, filtered, and either col-
ochlorophyll in the case of photosynthetic lected by flocculation at pH 4.0, or applied
bacteria that remain within the cellular to a reverse phase column for further
structure of the bacterium. purification. These procedures are applica-
The cells are separated from the medium ble to most tetrapyrroles found in any
by centrifugation at a minimum 2000× g. aqueous-based solution whether of mam-
The medium is decanted and stirred or malian origin, such as urine and extracted
shaken while adding 5 mL of 5% iodine in feces, or aqueous extracts of plant material.
ethanol per liter of the medium. The solu- One must be careful of course of treating
tion is allowed to stand for 1 hour protect- some tetrapyrroles of biological origin with
ed from light to complete oxidation of any strong acids such as in the esterification
porphyrinogens to the corresponding por- steps described. Such porphyrin may
phyrins. If porphyrin esters are desired, the require slightly different handling tech-
medium is passed through a layer of diethy- niques.
laminoethyl (DEAE) cellulose (about 100
mL of aqueous gravity packed adsorbent
per liter of medium) on a Buchner funnel, 9. PORPHYRINS AND METALLO-
which binds the anionic porphyrin tightly. PORPHYRINS IN SOLUTION
The packing is washed with water, dried in
the oven, or preferably air-dried, or dried 9.1. Behavior in Solution at the
under vacuum. The porphyrins are eluted Molecular Level
from the cellulose with 5% wt/vol sulfuric
acid in methanol or methanol saturated Under certain conditions, porphyrins
with HCl until color ceases and allowed to and metalloporphyrins undergo intermole-
stand protected from light for 24 hours at cular association in solution. In water at
room temperature. The esterifying mixture pH 7.5 in 0.01 M Tris buffer, plots of
is diluted with an equal volume of absorbance versus concentration for H2-
dichloromethane, and washed first with an TPPS3 follow Beers law from approximate-
equal volume of 1 M sodium acetate solu- ly 5 × 10-7 M to 1.4 × 10-5 M, and the
56
General Laboratory Methods for Tetrapyrroles

compound is considered monomeric (90). acid converts the absorbed free base into
In the presence of 0.1 M KNO3 at this the more weakly adsorbed green diacid H4-
pH, however, increasingly negative devia- TMPyP6+. For low porphyrin concentra-
tions from Beers law are observed as the tion work, many workers prefer to use a
concentration of the porphyrin increases, new plastic cuvette for each measurement
consistent with a monomer–dimer equilib- on freshly prepared solutions. The ampho-
rium, where the absorbance of the dimer is teric compound tetrakis[N-(2-sulfoethyl)-
less than that of the monomer. Equations 4-pyridyl]porphyrin is monomeric in the
have been developed to determine the pH range 2.0 to 12.0, and does not adsorb
dimerization constant, KD, from such on glass surfaces (55).
absorbance–concentration data. The position of substituents on the por-
phyrin periphery influence the extent of
2H2-TPPS3 [H2-TPPS3]2 KD [Eq. 1] aggregation. While H2-TPPS4, sulfonated
Since dimerization increases with ionic para-substituted TPP species and H2-TPPC
strength, overlay spectra of a given concen- associate, the ortho-and di-ortho-substituted
tration of porphyrin measured at differing sulfonated TPP derivatives, as well as tetra(2-
salt concentrations also provides evidence carboxyphenyl)porphyrin are monomeric
for the extent of porphyrin aggregation. (110,111). The electron-rich natural por-
Another method is to obtain the spectra of phyrins such as meso and protoporphyrin
the porphyrin at a given salt concentration IX examined by fluorescence techniques
in a 0.10-cm cell. The solution is diluted have high KD values of 2.7 × 106 M-1 and
1/10 and the spectra then taken in a 1.0 1.9 × 107 M-1, respectively (80), while the
cm cell, followed by another 1/10 dilution, octanegative uroporphyrin I shows no evi-
run in a 10.0-cm cell. If the compound is dence of dimerization at moderate ionic
monomeric, the overlay spectra should be strengths above pH 7.0. Dimerization also
superimposable. If dimers form, the most depends on the nature of the coordinated
dilute solution produces the highest ab- metal ion. The five or six coordinate
sorbance. Isosbestic points are often noted. Zn(II), VO(IV), Cr(III), Mn(III), and
Temperature–jump relaxation methods Co(III) complexes of TPPS4 are monomer-
allow the determination of the rate con- ic under conditions in which the four coor-
stants for dimer formation (kf) and dissoci- dinate Cu(II), Pd(II), and Ag(II)-TPPS4
ation (kd), and for H2-TPPS3, KD = 4.8× complexes are dimers (67). In nonaqueous
104 M-1, kf = 2.2 × 108 M-1 s-1, and kd = solutions, electron spin resonance studies
4.6 × 103 s-1. Under the same conditions, on paramagnetic metalloporphyrins and
both with and without added electrolyte, concentration-dependent NMR work on
H2-TMPyP(4) follows Beers law and metal-free compounds are used to access
shows no kinetic relaxation behavior, and monomer–dimer behavior (115).
thus behaves as a monomer. The electron An example of the practical conse-
withdrawing pyridinium groups remove quences of dimerization and larger aggre-
electron density from the porphyrin ring, gate formation is the sometimes anomalous
disfavoring the van der Waals interactions behavior of porphyrins during HPLC
leading to dimerization. Many water-solu- analysis. We have noted, for instance, the
ble porphyrins, such as H2-TMPyP(4), are reverse phase HPLC of H2-TPPS4 in phos-
adsorbed strongly on glassware. A flask that phate buffer systems can show a series of
once contained this compound when peaks eluting at rather regular intervals in
washed with water appears clean, but 0.1 what has been shown to be an essentially
M HCl added to the flask turns green, as pure sample by other chromatographic
57
J.C. Bommer and P. Hambright

means. This can be explained as a separa- with KD values in the 105-106 M-1 range
tion of the monomeric, dimeric, and high- (88). Addition of salts such as NaClO4 lead
er aggregated species, which are not rapidly to dimer dissociation. Job’s law mole-
dissociated under these HPLC conditions. ratio spectrophotometric studies indicated
Changing the concentration of the injected that the double decker porphyrin CeIII-
sample or the solvent composition of the [TMPyP(4)]27+ reacted with two moles of
injection media changes the relative size of Ni-TPPS4- or VO-TPPS4-, presumably with
the eluting peaks. one porphyrin on either face of the cerium
In aqueous solution, certain porphyrin dimer (18). Also, two moles of CeIV-[TAPP]28+
systems exhibit supramolecular behavior. complexed with one mole of either Ni(II) or
The diacid H4-TPPS2- has a Soret band at VO-TPPS4-.
433 nm. The new narrow peak at approxi- Only 1:1 molecular complexes were
mately 489 nm, which appears below pH formed between indigo di-, tri-, and tetrasul-
2.0 at high ionic strengths, is attributed to fonates (18) with Cu(II) and Zn(II)-TAPP4+
the presence of J-aggregates, edge-to-edge and Zn-TMPyP(4)4+. The magnitudes of the
ribbon-like zwitterionic electronically cou- association constants for molecular complexes
pled species in which the central diacid involving H2-TPPC44-, H2-TAPP4+, and
protons of one porphyrin interact with the tetrakis(N-propyl-4-pyridyl)porphyrin with
sulfonic acid groups of another (87). At various ligands that bind in a face-to-face fash-
higher porphyrin concentrations, another ion can be predicted (102). Molecular com-
peak at 422 nm appears, due to even larg- plexes between uroporphyrin-I and a variety
er H-aggregates, which are face-to-face of large organic cations and neutral hetero-
associations of the J-species, and involve cyclic molecules such as caffeine, o-phenan-
hundreds of thousands of interacting throline, methyl viologen, nicotinamide, and
porphyrin units (95). Resonance light adenine have been investigated (82).
scattering experiments (92) indicate that Porphyrins show acid base behavior, and
the trans diphenyl/di(4-sulfonatophenyl) the first two acid dissociation constants are
porphyrin, as the free base at pH 6.0 and defined as follows, where the charges of the
as the diacid at pH 3.0 show supramolecu- peripheral groups are neglected:
lar behavior, as does the diacid H4-(β-Br8-
TPPS)42- at pH approximately 1.0 and the H4-P2+ H3-P+ + H+ K4 [Eq. 2]
free base and Cu(II) complex of trans H3-P +
H2-P + H +
K3 [Eq. 3]
diphenyl/di(N-methyl-4-pyridyl)porphyrin.
Supramolecular chiral H- and J-aggregates The equilibria are measured spectrophoto-
of H4-TPPS42- are formed on poly-L- metrically, and it is important to make sure
glutamate at pH 2.9, but only in the pres- that the porphyrin is monomeric under the
ence of the cationic Zn(II), Mn(III), and pH titration conditions, and that the
Au(III)-TMPyP(4) species. The positive buffers used do not complex with the por-
porphyrins act as spacers, allowing the anion- phyrins (56). For example, H2-
ic porphyrins to approach the polypeptide TMPyP(4)4+ shows pK4 = 0.8, pK3 = 1.4,
and gain chirality (94). while for the more basic H2-TAPP4+, pK4
Heteronuclear dimers are formed = 3.95 and pK3 = 4.11. The pK2 and pK1
between oppositely charged porphyrins and values for most porphyrins are above 12,
metalloporphyrins in solution. Thus, 1:1 although the H2-[β-Br8-TMPyP(4)]4+
complexes in acetone–water were found with eight electron-withdrawing bromines
between H2-TAPP4+ (and Zn-TAPP4+) with on the β-pyrrole positions (96) gives pK2 =
H2-TPPS44-, Cu-TPPS44-, and Zn-TPPS44- 6.5 and pK1 = 10.2. The magnitudes of
58
General Laboratory Methods for Tetrapyrroles

these acid dissociation constants depend on DME. Partial potential values could be
the ionic strength and temperature. The assigned to substituent groups, such that
pK3 values are used to rank the relative when added to the potential of the refer-
basicities of water-soluble porphyrins (47), ence compound porphin, allow the calcu-
as in the series H2-TMPyP(2) [-0.9], H2- lation of E1/2(1) for new derivatives. For a
TMPyP(4) [1.4], H2-TMPyP(3) [1.8], series of porphyrins, a variety of spectro-
H2-TAPP [4.11], H2-TPPS4 [4.76], H2- scopic, kinetic, and equilibrium data can
TPPC4 [5.5], uroporphyrin I [6.0], and be correlated with either pK3 or E1/2 (1).
hematoporphyrin IX [6.1]. The cationic
porphyrins are usually less basic than the 9.2. Practical Aspects of Porphyrin
anionic compounds. Dissolution
Detergents have been used to bring 3,8-
disubstituted deuteroporphyrin IX DME As noted in the previous section, most
compounds into aqueous solution (46). porphyrins exist in an aggregated state in
With cationic detergents such as cetyltri- aqueous solution. This can translate into
methylammonium bromide, the monoca- the more fundamental problem of how to
tion H3-P+ is destabilized, and only the successfully dissolve some of the less hydro-
diacid–free base equilibria (K3K4) can be philic porphyrins and stabilize solutions
observed. In 2.5% sodium laurel sulfate, long enough to do meaningful biological
both pK3 and pK4 can be obtained, and experiments. This problem is generally
typical pK3 values for these esters are 5.8 for associated with mono- and dicarboxylic
mesoporphyrin, 5.5 for deuteroporphyrin, porphyrins and chlorins and their divalent
4.8 for protoporphyrin, and 3.3 for the 3,8 metallo derivatives, which lack hydrophilic
diacetyldeuteroporphyrin (23). Electron substituents other than the carboxylic acid
withdrawing groups decrease the proton groups. Protoporphyrin IX is an example of
affinity of the central nitrogen atoms. this type of porphyrin in that it is colloidal
The reduction potential of the free base in aqueous alkali (21). As a general rule,
porphyrin into its radical anion, E1/2 (1), these porphyrins can be dissolved by treat-
have been measured under the same condi- ing them with dilute base (0.01 to 0.1 M
tions in DMF for over a hundred water- NaOH, KOH, or better yet, ammoniacal
insoluble porphyrins (119,120), and such bases such as Tris or NH4OH if usage will
constants are also a measure of relative allow), then diluting to about 50% with
basicities that allows comparisons between aqueous soluble organic solvents such as
meso and β-pyrrole substituted com- ethanol, DMF, dimethylsulfoxide, etc. Rea-
pounds. sonably concentrated stock solutions on the
order of several millimolar can usually be
H2-P + e- H2-P- E1/2(1) [Eq. 4]
prepared in this way. The stock solution can
The more basic the porphyrin, the less ten- then be diluted into a large volume of
dency it has to add an electron, and the buffered aqueous solution or media to
more negative its reduction potential. A adjust the pH and minimize contribution
very basic porphyrin is OEP with E1/2(1) = from the organic solvent and base. Both
-1.85 V, followed by -1.82 for meso DME, solutions are likely to be unstable with time
-1.77 for deutero DME, -1.70 for proto resulting in increasing aggregation and pre-
DME, -1.66 for the unsubstituted por- cipitation, and should be prepared freshly
phyrin porphin, -1.56 for H2-TPP, -1.48 and used immediately for each experiment.
for the 3,8-diacetyl deutero DME and, Although the porphyrins likely exist as very
-1.32 for the less basic 3,8-dicyano deutero large aggregates in the final buffered solu-
59
J.C. Bommer and P. Hambright

tion, they apparently become absorbed by The most common incorporation


lipophilic membranes and are monomer- method involves simply refluxing the por-
ized in some manner during the course of phyrin in water with a water-soluble metal
cellular and in vivo experiments. salt. The diacid and monocations usually
do not incorporate metal ions, so the pH
should be kept high enough such that an
10. METALATION METHODS appreciable amount of H2-P is present. It is
best to run the reaction until all of the por-
10.1. Water-Soluble Porphyrins phyrin is metalated, as it is difficult to
remove a small amount of H2-P from M-P
Herrmann and coworkers have devel- at later times. With anionic porphyrins, the
oped a novel heterogeneous procedure to solution is filtered and slowly run through
incorporate many metal ions into an ion exchange column in the Na+ form to
water-soluble porphyrins (53). Using H2- remove uncomplexed metal ions. The elu-
TPPS4 as an example, 50 mg of porphyrin ate is lyophilized, and the metalloporphyrin
are added to 100 mL distilled water con- is purified from the salts by procedures
taining 0.5 to 1.0 gram of an insoluble mentioned above for the metal-free deriva-
metal oxide or acid-etched metal, and the tives, i.e., recrystallization, passage through
mixture is brought to reflux. The reaction Sephadex resins, precipitation with
is followed spectrophotometrically, and the HPhen+, etc. For positive porphyrins, sodi-
four H2-P bands between 700 and 500 mn um iodide or sodium perchlorate are often
are replaced by one or two peaks character- added to precipitate the cationic porphyrin
istic of the metalloporphyrin. The reac- salts. (CAUTION: Porphyrin perchlorates are
tions typically occur in 1 hour, and the potentially explosive, and iodide sometimes
cooled solution is filtered through a reduces a fraction of a trivalent metallopor-
0.22-µm filter, the aqueous metallopor- phyrin to the divalent state). The solids are
phyrin filtrate evaporated under reduced slurried with a chloride cation exchange
pressure, and the solid is briefly dried resin (heating is often required) and slowly
under vacuum at 120°C. Metal and CHN passed through a column of the resin, fol-
analyses were presented for Cu(II), Zn(II), lowed by lyophilization. A safer and more
Co(II), Ni(II), and Cd-TPPS4, and a vari- elegant method for small quantities of
ety of other ions were shown to incorporate cationic porphyrins involves the addition of
under these conditions. Certain oxides NH4PF6 as the precipitating agent, wash-
(Cr2O3, Mn2O3, PdO) and metals (Ti, V, ing the solid with 1:1 2-propanol-ether, and
Ru, Ag) were unreactive, and in other cases vacuum drying at room temperature (98).
(HgO, PbO, MnO, CaO) irreversible The PF6- salt is then dissolved in acetone,
absorption of the porphyrin on the oxide filtered, and the chloride salt of the por-
surface limited the amount of product phyrin precipitated with tetrabutyl-ammo-
isolated. Others have noted that the final nium chloride, washed with acetone, and
solid can be contaminated with metal ions dried in vacuo.
bound to the peripheral -SO3- groups, Divalent cadmium (104), lead (52), and
and further treatment is required to magnesium ions are in pH-dependent equi-
obtain pure compounds (45). Cationic libria with the corresponding metallopor-
porphyrins such as H2-TMPyP(4) can be phyrin in aqueous solution, for example:
metalated by this method, but the reac-
Cd2+ + H2-P Cd-P + 2H+ KCd [Eq. 5]
tions are often slower than found with
2+ 2+
anionic derivatives. Cd-P + Cu → Cu-P + Cd [Eq. 6]

60
General Laboratory Methods for Tetrapyrroles

Typical values for KCd are 7.9 × 10-7 M for Ce(IV)-[TPPC4]2 was produced by basic
TMPyP(2) and 4.2 × 10-11 M for TPPS4. hydrolysis of the tetramethyl ester. As
The deformed Cd-P reacts with Cu2+ (and noted, the oxidation state of the coordinat-
Fe2+, Zn2+, Mn2+ ions) approximately 102 ed metal may or may not change during
to 103 times faster than Cu2+ incorporates the reactions. A list of the metal ions that
into H2-P itself. Such room temperature have been incorporated into water-soluble
metal-catalyzed electrophilic substitution porphyrins has been compiled (47).
reactions have been used to insert metal
ions into picket-fence type porphyrins, 10.2. Water-Insoluble Porphyrins
where refluxing the solution would lead to
atropisomerization (85,113). Mercury(II) Adler’s DMF method is often employed
in acid forms Hg2-P2+ complexes, and sim- for insertion of various metal ions into
ilar displacement reactions occur after ini- water-insoluble porphyrins (4). The free
tial loss of a mercury ion (99). Lithium base porphyrin and a metal salt (acetate,
ions are in equilibrium with Li-P- com- chloride) are refluxed in DMF until the
plexes of TMPyP(X) (56) and β-Br8- absorption spectra indicates that metala-
TMPyP(4) (96) in base. Deformed central- tion is complete. The addition of water to
ly mono-N-alkylated porphyrins react with the cooled solution precipitates the metal-
metal ions several orders of magnitude loporphyrin, which can then be purified by
faster than do the parent compounds (71). chromatography. An example of this proce-
This fact has been used for the rapid prepa- dure is given below. One or two molecules
ration of short half-life radiolabeled por- of dimethylamine are often found bound
phyrins of divalent Cu, Co, and Pd, where to trivalent complexes. Buchler has devel-
the central N-benzyl group is lost upon oped techniques of incorporation of high
metalation (72). oxidation state metal ions in which the
In cases where high temperatures in reactions are run in imidazole or phenol
nonaqueous solvents are necessary for melts, and he has reviewed other useful
metalation with water-insoluble or metalation systems (15). These include
organometallic reagents, it is often best to reactions in acetic acid–sodium acetate, in
first metalate the water-insoluble precursor, pyridine and benzonitrile for acid labile
which can usually be purified by chro- complexes, and the uses of metallo acety-
matography. The water-soluble metallo- lacetonates, phenoxides, and organometal-
porphyrin is then formed in a subsequent lic reagents as metal carriers. Buchler’s “sta-
step. For example, H2-TPyP(4) in bility index” Si (the product of the Pauling
trichlorobenzene was reacted with n-BuLi electronegativity and cation charge divided
at room temperature to form Li-TPyP(4)-, by the ionic radius in picometers) is a guide
and after addition of Ce(acac)3.H2O, the to the tendency of a metalloporphyrin to
solution was refluxed until metalation was be demetalated by acids of various concen-
complete (17). The Ce(IV)-[TPyP(4)]2 trations (14), and relationships between
sandwich complex was purified by chro- the acid-catalyzed demetalation rate con-
matography on alumina, and after reaction stants for a series of M-TAPP complexes
in DMF with MTS (100°C for 5 days), the and Si have been explored (2). The loss of
water-soluble Ce(III)-[TMPyP(4)]2 was the metal ion by acid solvolysis reactions is
formed. The Ce(IV)-[TAPP]2 was made usually first-order in metalloporphyrin and
from the cerium(IV)-N.N-dimethylanilin- second-order in (H+).
ium precursor by N-methylation in The incorporation of many metals
CHCl3/EtOH with CH3I, and the requires high temperatures, which can be
61
J.C. Bommer and P. Hambright

problematic for most anionic porphyrins sis provided the ester has some solubility in
derived from natural sources. These por- this mixture (44).
phyrins and chlorins often have peripher- Porphyrins having acetic acid side chains
al groups that are labile or reactive with are prone to decarboxylate or undergo
the solvents at high temperature. In the other types of degradation if attempts are
case of vinyl or other unsaturated groups, made to metalate even the ester forms at
this can be as low as 80°C depending on high temperature. Thus, porphyrins such
the solvent, but in most cases, tempera- as uroporphyrin are usually not successful-
tures in excess of 150°C tend to cause the ly metalated in refluxing solvents such as
most difficulty. Synthetic procedures phenol, benzonitrile, dichlorobenzene, and
involving protection and regeneration of imidazole.
vinyl groups on porphyrins have been Insertion of such metals as Al, the lan-
described by Smith et al. (107). Metala- thanides, Pt, Sc, VO, TiO, and Zr into
tion of hematoporphyrin even at room these porphyrins is generally not successful.
temperature generally results in some Cobalt incorporation into porphyrins con-
dehydration of the hydroxyethyl groups to taining free carboxyl groups, even at room
vinyl groups, and if not during the meta- temperature, usually results in predomi-
lation, then certainly during the isolation nantly insoluble black polymer-like prod-
and drying process. ucts. This can sometimes be avoided by the
In general, it is best to do metal incor- addition of large amounts of pyridine to
porations on the ester form of porphyrins the metalating solution or in some cases by
with carboxyl groups. This tends to protect starting with the porphyrin ester and
these groups from decarboxylation, anhy- hydrolyzing the purified product. Use of
dride formation, and unwanted interac- pyridine may result in a product with one
tions with solvents or metalating agents. or two pyridines coordinated to the central
Purification of metalloporphyrin esters is metal ion. These ligands can usually be
generally easier than the free acid forms removed by washing with strong acid, but
using chromatographic and crystallization often this results in the formation of insol-
techniques. The resulting products can be uble polymer-like materials, or in certain
hydrolyzed with strong base, e.g., a stirred cases, loss of the coordinated metal
mixture of 2 to 4 M NaOH or KOH (24) through acid hydrolysis reactions. The pro-
with the metalloporphyrin ester dissolved cedure below is an example of the incorpo-
in an equal volume of tetrahydrofuran. ration of iron into OEP, using the DMF
Complete hydrolysis is usually accom- method of Adler (4). With its eight ethyl
plished in 12 to 24 hours at room temper- groups on the β-pyrrole positions, OEP is
ature and can be ascertained by reverse the most widely used model compound for
phase TLC. Hydrolysis is usually marked the natural protoporphyrins, which have
by precipitation of the product as the Na+ eight β-pyrrole substituents.
or K+ salt or the observed transfer of the
compound from the tetrahydrofuran ❖ Procedure 4. Incorporation of Iron
(THF) into the aqueous part of the two into Octaethylporphyrin
phase system. Removal of the THF, which
is dissolved in the aqueous layer by rotary 1. Under a well ventilated hood and wear-
evaporation, allows collection of the free ing gloves, pour 1.2 L of DMF and
acid metalloporphyrin by flocculation at approximately 10 mL of acetic acid
pH 4.0. Methanol and 1% KOH with a into a 4-L beaker containing 9.0 g
trace of water can also be used for hydroly- (16.8 mM) of OEP and stirring bar.
62
General Laboratory Methods for Tetrapyrroles

The absorption spectra of the metal- (4), meso-tetrakis(N-ethyl-4-pyridyl)por-


-free H2-OEP in this solution has phyrin with X chlorines on the β-pyrroles;
bands (and relative peak heights) at DDQ, 2,3-dichloro-5,6-dicyano-1,4-ben-
651.5 nm (1.0), 593.0 nm (1.41), zoquinone; DME, dimethylester; DMF,
533.5 nm (1.56), and 518.0 nm (3.45). N,N-dimethylformamide; EDTA, ethyl-
2. The beaker is placed on a stirrer–hot enediaminetetraacetic acid; ETIO-I, etio-
plate and slowly heated to approxi- porphyrin-I; H-PHEN+, monoprotonated
mately 100°C. At this stage, 13.4 g 1,10-phenanthroline; HPLC, high-pres-
of iron(II) chloride tetrahydrate sure liquid chromatography; MTS, methyl
(64.3 mM) are carefully added in por- para-toluenesulfonate; OEP, octaethylpor-
tions to the hot solution, and the tem- phyrin; TAPP, meso-tetrakis(4-N,N,N-
perature is raised until the mixture trimethylanilinium)porphyrin; TMPyP-
refluxes. (X), meso-tetrakis(N-methyl-X-pyridyl)
3. Heating is continued until the spectra porphyrin, X = 2, 3, or 4; T(2-NH2P)P,
of an aliquot in DMF indicates the meso-tetrakis(2-aminophenyl)porphyrin;
complete disappearance of the metal- TPP, meso-tetraphenylporphyrin; TPPC4,
free peaks (especially the 651.5 nm meso-tetrakis(4-carboxyphenyl)porphyrin;
absorbance), with the appearance of TPPS4, meso-tetrakis(4-sulfonatophen-
new bands due to the Fe(III) porphyrin yl)porphyrin; TPPS3, monophenyl-tri(4-
at 629.0 nm (1.0), 532.5 nm (1.92), sulfonatophenyl)porphyrin; TPPS2, di-
and 504.5 nm (1.90). phenyl-di(4-sulfonatoaphenyl)porphyrin;
TPPS1, triphenyl-mono(4-sulfonatophen-
4. While H2-OEP is not terribly soluble
yl)porphyrin; TPyP(X) meso-tetrakis(X-
in hot DMF, the porphyrin goes into
pyridyl)porphyrin, X = 2, 3, or 4.
solution as the more soluble FeIII-OEP
forms. The incorporation usually takes
20 minutes, and small amounts of ACKNOWLEDGMENTS
DMF are occasionally added to keep
the volume at approximately 1 L. P.H. thanks the Howard University
5. The solution is then allowed to come CSTEA project (NASA Contract No.
to room temperature and Buchner fil- NCC S-184) for financial support. We
tered, and then 2 L of 0.1 M HCl are thank Sabrina L. Bailey and Jeff Yearyean
added to essentially quantitatively pre- for helpful discussions.
cipitate the metalloporphyrin, which is
collected by filtration. REFERENCES
6. The brown solid is washed with 0.1 M
1.Adeyemo, A., Shamim, P. Hambright, and R.F.X.
HCl, then water, and dried in an oven Williams. 1982. meso-Tetrakis[N-methyl-4(or 3)quin-
at 70°C overnight. olyl]porphyrins: metallation rate/basicity correlations.
Indian J. Chem. 21A:763-766.
7. The purification of this crude Fe(III)- 2.Adeyemo, A., A. Valiotti, C. Burnham, and P. Ham-
OEP Cl on an alumina column is bright. 1981. Acid solvolysis kinetics of copper and
described in Procedure 1. nickel porphyrins: a rate-stability index correlation.
Inorg. Chim. Acta Lett. 54:L63-L65.
3.Adler, A.D., F.R. Longo, J.D. Finarelli, J. Goldmach-
er, J. Assour, and L. Korsakoff. 1967. A simplified
ABBREVIATONS synthesis for meso-tetraphenylporphyrin. J. Org.
Chem. 32:476-477.
4.Adler, A.D., F.R. Longo, F. Kampas, and J. Kim.
Br8-TMPyP(4), TMPyP(4) with 8 1970. On the preparation of metalloporphyrins. J.
bromines on the β-pyrroles; ClX-TEPyP- Inorg. Nucl. Chem. 32:2443-2445.

63
J.C. Bommer and P. Hambright

5.Almarsson, O., H. Adalsteinsson, and T.C. Bruice. 19.Byrn, M.P., C.J. Curtis, I. Goldberg, Y. Hsiou, S.J.
1995. Synthesis and characterization of an octacation- Khan, P.A. Sawin, S.K. Tendick, and C.E. Strouse.
ic iron tetraphenylporphyrin, which is soluble in water 1991. Porphyrin sponges: structural systematics of the
and non-m-oxo dimer forming. J. Am. Chem. Soc. host lattice. J. Am. Chem. Soc. 113:3617-3621.
117:4524-4532. 20.Cardinal, R.A., I. Bossenmaier, Z.J. Petryka, L. John-
6.Anton, J.A. and P.A. Loach. 1975. Synthesis of some son and C.J. Watson. 1968. Isolation of porphyrins
mono, di and trisubstituted tetraarylporphyrins. J. from porphyria urine by preparative thin-layer chro-
Heterocyclic. Chem. 12:573-576. matography. J. Chromatogr. 38:100-105.
7.Artaud, I., H. Grennberg, and D. Mansuy. 1992. 21.Caughey, W. S., A. Adler, B.F. Burnham, D. Dolphin,
Selective oxidative cleavage of 1,2-dimethoxyarenes to S.F. MacDonald, D.C. Mauzerall, and Z.J. Petyka.
muconic diesters catalyzed by an iron -sulfonated- 1972. Porphyrins, p. 185-216. In Specification and
tetrakis(pentafluorophenyl)porphyrin. J. Chem. Soc., Criteria for Biochemical Compounds, 3rd ed. Nation-
Chem. Commun. 1036-1038. al Academy of Sciences, Washington, D.C.
8.Bachmann, R.C. and B.F. Burnham. 1969. Purifica- 22.Caughey, W.S., R.M. Deal, C. Weiss, and M. Gouter-
tion of porphyrin esters by gel-filtration. J. Chromatogr. man. 1965. Electronic spectra of substituted metal
41:394-399. deuteroporphyrins. J. Mol. Spectrosc. 16:451-463.
9.Barnett, G.H., M.F. Hudson, and K.M. Smith. 23.Caughey, W.S., W.Y. Fujimoto, and B. Johnson. 1966.
1975. Concerning meso-substituted tetraphenylpor- Substituted deuteroporphyrins. II. Substituent effects
phyrin purification. J. Chem. Soc. [Perkins 1] 1401- on electronic spectra, nitrogen basicities, and ligand
1403. affinities. Biochemistry 5:3830-3843.
10.Batinic-Haberle, I., L. Benov, I. Spasojevic, and I. 24.Cavaleiro, J.A.S., G.W. Kenner, and K.M. Smith.
Fridovich. 1998. The ortho effect makes 1974. Pyrroles and related compounds. Part XXXII.
manganese(III) meso-tetrakis(N-methylpyridinium-2- Biosynthesis of protoporphyrin IX from copropor-
yl)porphyrin a powerful and potentially useful super- phyrininogen III. J. Chem. Soc. [Perkins I]. 1188-
oxide dismutase mimic. J. Biol. Chem. 273:24521- 1194.
24528. 25.Chu, T.C. and E.J.-H. Chu. 1966. Thin-layer chro-
11.Batinic-Haberle, I., S.I. Liochev, I. Spasojevic, and matography of methyl esters of porphyrins, chlorins
I. Fridovich. 1997. A potent superoxide dismutase and related compounds with Eastman “chromagram”.
mimic: manganese-β-octabromo-meso-tetrakis-(N- J. Chromatogr. 21:46-51.
methylpyridinium-4-yl)porphyrin. Arch. Biochem. 26.Collman, J.P., R.R. Gagne, C.A. Reed, T.P. Halbert,
Biophys. 343:225-233. G. Lang, and W.T. Robinson. 1975. “Picket fence por-
12.Batinic-Haberle, I., I. Spasojevic, P. Hambright, L. phyrins.” Synthetic models for oxygen binding hemo-
Benov, A.L. Crumbliss, and I. Fridovich. 1999. The proteins. J. Am. Chem. Soc. 97:1427-1439.
relationship between redox potentials, proton dissocia- 27.Conant, J.B. and W.W. Moyer. 1930. Studies in the
tion constants of pyrroic nitrogens and in vivo super- chlorophyll series. III. Products of the phase test. J.
oxide dismutase activities of manganese(III) and Am. Chem. Soc. 52:3013-3023.
iron(III) cationic and anionic porphyrins. Inorg. 28.Datta-Gupta, N. and T.J. Bardos. 1966. Synthetic
Chem. 38:4011-4022. porphyrins. I. Synthesis and spectra of some para-sub-
13.Brisbin, D.A. and J.O. Asgill. 1974. The micro-deter- stituted meso-tetraphenylporphines. J. Heterocyclic.
mination of porphyrins. An intergrated laboratory Chem. 3:495-502.
experiment. J. Chem. Ed. 51:211-213. 29.DiNello, R.K. and C.K. Chang. 1978. Isolation and
14.Buchler, J.W. 1975. Static coordination chemistry of modification of natural porphyrins, p. 289-339. In D.
metalloporphyrins, p. 157-231. In K.M. Smith (Ed.), Dolphin (Ed.), The Porphyrins, Vol. I, Part A. Acade-
Porphyrins and Metalloporphyrins, Elsevier, Amster- mic Press, New York.
dam. 30.Dixon, D.W., R. Schinazi, and L.G. Marzilli. 1990.
15.Buchler, J.W. 1979. Synthesis and properties of metal- Porphyrins as agents against the human immunodefi-
loporphyrins, p. 389-483. In D. Dolphin (Ed.), The ciency virus. Ann. NY Acad. Sci. 616:511-513.
Porphyrins, Vol. I, Part A. Academic Press, New York. 30a. Dolphin, D., (Ed.). 1978. The Porphyrins, Vol. I-VII.
16.Buchler, J.W., F.M. Kunzel, U. Mayer, and M. Nawra. Academic Press, New York.
1994. Metal complexes with tetrapyrrole ligands. Part 31.Dolphin, D., T.G. Traylor, and L.Y. Xie. 1997. Poly-
64. Electrophoresis of water-soluble porphyrins and haloporphyrins: unusual ligands for metals and metal
their metal complexes. Fresenius J. Anal. Chem. catalyzed oxidations. Acc. Chem. Res. 30:251-259.
348:371-376. 32.Drabkin, D.L. 1941. Spectrophotometric studies VIII.
17.Buchler, J.W. and M. Nawra. 1994. Metal complex- The microdetermination of iron in cytochrome c and
es with tetrapyrrole ligands. 67. Synthesis and hemin preparations. J. Biol. Chem. 140:387-396.
spectroscopic properties of water-soluble cerium 33.Elder, G. 1971. Separation of porphyrin methyl esters
bisporphyrinate double-decker ions. Inorg. Chem. by two-dimensional thin layer chromatography. J.
33:2830-2837. Chromatogr. 59:234-236.
18.Buchler, R.A. and M. Nawra. 1996. Metal complexes 34.Endisch, C., J.-H. Fuhrhop, J. Buschmann, P. Luger,
with tetrapyrrole ligands. 71. Heteroaggregation of and U. Siggel. 1996. β-Tetraethyl-β′-tetrapyridin-4-yl
cationic metal mono- and bisporphyrinates with anion porphyrins, their N-methylated tetracations, and het-
metal porphyrinates or indigosulfonates. Inorg. Chim. erodimers with ms-tetraphenylsulfonato porphyrins. J.
Acta. 251:227-237. Am. Chem. Soc. 118:6671-6680.

64
General Laboratory Methods for Tetrapyrroles

35.Falk, J.E. 1964. Porphyrins and Metalloporphyrins. 52.Haye, S.E. and P. Hambright. 1984. Equilibrium con-
Elsevier, Amsterdam. stants and electrophylic exchange reactions of lead(II)
36.Ferrer-Sueta, G., I. Batinic-Haberle, I. Spasojevic, I. porphyrins. Inorg. Chem. 23:4777-4779.
Fridovich, and R. Radi. 1999. Catalytic scavengening 53.Herrmann, O., S.H. Mehdi, and A. Corsini. 1978.
of peroxynitrite by isomeric Mn(III) N-methylpyridyl- Heterogeneous metal-insertion: a novel reaction with
porphyrins in the presence of reductants. Chem. Res. porphyrins. Can. J. Chem. 56:1084-1089.
Toxicol. 12:442-449. 54.Hoffmann, P., G. Labat, A. Robert, and B. Meunier.
37.Fischer, H. and R. Baumler. 1929. Uber phao und 1990. Highly selective bromination of tetramesitylpor-
phyllerythro-porphyrine. Ann. Chem. 474:65-120. phyrin: an easy access to robust metallo-porphyrins,
38.Fischer, H. and H. Orth. 1934. Die Chemie des M-Br8-TMP and M-Br8-TMPS. Tetrahedron Lett.
Pyrroles. Vol 1. Akademische Verlagsgellschaft, Liepzig. 31:1991-1994.
39.Fischer, H. and H. Orth. 1937. Die Chemie des 55.Igarashi, S. and T. Yotsuyanagi. 1984. New “ampho-
Pyrroles. Vol 2, part 1. Akademische Verlagsgellschaft, teric ion type” water-soluble porphines as a spec-
Liepzig. trophotometric agent. Chem. Lett. 1871-1874.
40.Fischer, H. and H. Siebel. 1932. Uber phaeophorbide 56.Islam, M.Q. and P. Hambright. 1998. Lithium com-
a, chorin e and chlorophyll a. Ann. Chem. 499:84- plexes and the kinetics of interactions of zinc ions with
108. tetra(N-methyl-4-pyridyl)porphyrins in basic solution.
41.Fischer, H. and A. Stern. 1940. Die Chemie des Trans. Met. Chem. 23:727-733.
Pyrroles. Vol 2, part 2. Akademische Verlagsgellschaft, 57.Ito, Y., K. Kunimoto, S. Miyachi, and T. Kato. 1991.
Liepzig. Photocatalytic cleavage of 1,2-diols by a cofacial hin-
42.Fleischer, E.B., R. Foust, D. Jeter, and R. Near. 1973. dered water-soluble iron(III) porphyrin. Tetrahedron
The solubilities of tetra(monofluorophenyl)porphines. Lett. 32:4007-4010.
Inorg. Nucl. Chem. Lett. 9:1219-1220. 58.Izbicka, E., R.T. Wheelhouse, E. Raymond, K.K.
43.Flynn, S.M., S.T. George, L. White, W. Devonish, Davidson, R.A. Lawrence, D. Sun, B.E. Windle, L.H.
and G.B. Takle. 1999. Water-soluble, meso-substitut- Hurley, and D.D. Von Hoff. 1999. Effect of cationic
ed cationic porphyrins—a family of compounds for porphyrins as G-quadruplex interactive agents in
cellular delivery of oligonucleotides. BioTechniques human tumor cells. Cancer Res. 59:639-644.
26:736-740. 59.Jimenez, H.R., M. Julve, J.M. Moratal and J. Fuas.
44.Fuhrhop, J.-H. and K.M. Smith. 1975. In K.M. 1987. Stability constants of metalloporphyrins. a study
Smith (Ed.), Porphyrins and Metalloporphyrins, of the protonation, deprotonation and formation of
Chapter 19. Elsevier, Amsterdam. copper(II) and zinc(II) complexes of meso-tetra(p-
45.Gonsalves, A.M.d’A.R., R.W.A. Johnstone, M.M. sufonatophenyl)porphyrin. J. Chem. Soc. Chem.
Pereira, A.M.P. de SantAna, A.C. Serra, A.J.F.N. Sor- Commun. 910-911.
bal, and P.A. Stocks. 1996. New procedures for the 60.Kachadourian, R., I. Batinic-Haberle, and I.
synthesis and analysis of 5,10,15,20-tetrakis(sul- Fridovich. 1999. Synthesis and superoxide dismuting
fophenyl)-porphyrins and derivatives through chloro- activities of partially (1-4) β-chloronated derivatives of
sulfonation. Heterocycles 43:829-838. manganese(III) meso-tetrakis(N-ethylpyridinium-3-
46.Hambright, P. 1975. Dynamic coordination chemistry yl)porphyrin. Inorg. Chem. 38:391-396.
of metalloporphyrins, p. 233-278. In K.M. Smith 61.Kadish, K.M., C. Araullo-McAdams, B.C. Han, and
(Ed.), Porphyrins and Metalloporphyrins, Elsevier, M.M. Franzen. 1990. Synthesis and spectroscopic
Amsterdam. characterization of (T(p-Me2N)F4PP)H2 and (T(p-
47.Hambright, P. 2000. The chemistry of water-soluble Me2N)F4PP)M where T(p-Me2N)F4PP is the dianion
porphyrins, Ch. 18. In K. Kadish, K.M. Smith and R. of meso-tetrakis(o,o,m,m,-tetrafluoro-p-(dimethy-
Guilard (Eds.), The Porphyrin Handbook. Academic lamino)phenyl)porphyrin and M = Co(II), Cu(II) or
Press, New York. Ni(II). Structures of (meso-tetrakis(pentafluo-
48.Hambright, P., A. Adeyemo, A. Shamim, and S. rophenyl)porphyrinato)cobalt(II), (TF5PP)Co. J. Am.
Lemelle. 1985. [[4,4′,4′′,4′′′-porphyrin-5,10,15,20- Chem. Soc. 112:8364-8368.
tetrayltetrakis(1-methylpyridiniumato](2-)]-indium(III) 61a. Kadish, K., K.M. Smith, and R. Guilard (Eds.).
pentaperchlorate. Inorg. Synth. 23:55-59. 2000. The Porphyrin Handbook. Academic Press,
49.Hambright, P. and E.B. Fleischer. 1970. The acid-base New York.
equilibria, kinetics of copper ion incorporation and 62.Kalyanasundaram, K. 1984. Photochemistry of water-
acid-catalyzed zinc ion displacement from the water- soluble porphyrins: comparative study of isomeric
soluble α,β,γ,δ-tetra(4-N-methylpyridyl)porphyrin. tetrapyridyl and tetraksi(N-methylpyridiniumyl)-por-
Inorg. Chem. 9:1757-1761. phyrins. Inorg. Chem. 23:2453-2459.
50.Hambright, P., A. Turner, J.S. Cohen, R.C. Lyon, A. 63.Kalyansundaram, K. 1991. Photochemistry of Poly-
Katz, P. Neta, and A. Adeyemo. 1987. An iron(III) pyridine and Porphyrin Complexes. Academic Press,
porphyrin that exhibits minimal dimerization in aque- New York.
ous solution. Inorg. Chim. Acta 128:L11-L14. 64.Kaufmann, T., B. Shamsai, R.S. Lu, R. Bau, and
51.Han, F.X., R.T. Wheelhouse, and L.H. Hurley. 1999. G.M. Miskelly. 1995. Separation of the rotational iso-
Interactions of TMPyP4 and TMPyP2 with quadru- mers of tetrakis(N-methyl-2-pyridiniumyl)-porphyrin
plex DNA. Structural basis for differential effects on and the crystal structure of α,α,α,β-(tetrakis(N-
telomerase inhibition. J. Am. Chem. Soc. 121:3561- methyl-2-pyridiniumyl)porphyrin copper hexacyano-
3570. ferrate. Inorg. Chem. 34:5073-5079.

65
J.C. Bommer and P. Hambright

65.Krishnamurthy, M. 1977. Synthesis and characteriza- 83.Meng, G.G., B.R. James, K.A. Skov, and M. Korbelik.
tion of a new water-soluble porphyrin. Indian J. 1994. Porphyrin chemistry pertaining to the design of
Chem. 15B:964-966. anti-cancer drugs; part 2, the synthesis and in vitro test
66.Krishnamurthy, M. 1977. Kinetics of anation reac- of water-soluble porphyrins containing, in the meso
tions of a water-soluble rhodium(III) porphyrin. Inorg. positions, the functional groups: 4-methylpyridinium,
Chim. Acta. 25:215-218. or 4-sulfonatophenyl, in combination with phenyl, 4-
67.Krishnamurthy, M., J.R. Sutter, and P. Hambright. pyridyl, 4-nitrophenyl, or 4-aminophenyl. Can. J.
1975. Monomer-dimer equilibration of water-soluble Chem. 72:2447-2457.
porphyrins as a function of the coordinated metal ion. 84.Milgrom, L.R. 1997. The Colours of Life: An Intro-
J. Chem. Soc. Chem. Commun. 13-14. duction to the Chemistry of Porphyrins and Related
68.La, T., R.A. Richards, R.S. Lu, R. Bau, and G.M. Compounds. Oxford Press, New York.
Miskelly. 1995. Solution chemistry and crystal struc- 85.Miskelly, G.M., W.S. Webley, C.R. Clark, and D.A.
ture of nickel tetrakis(2,3,5,6-tetrafluofo-N,N,N- Buckingham. 1988. Acidity and dimerization of three
trimethyl-4-aniliniumyl)porphyrin trifluoromethane- water-soluble iron(III) porphyrin cations: (meso-
sulfonate (NiTF4TMAP(CF3SO3)4). Inorg. Chem. α,β,α,β-tetrakis(o-(Nmethylnicotinamido) phenyl)
34:5632-5640. porphyrinato)iron(III), (meso α,α,α,α-tetrakis(o-N-
69.La, T., R.A.Richards, and G.M. Miskelly. 1994. Syn- methylnicotinamido) phenyl)porphyrinato)iron(III),
thesis and characterization of the cationic porphyrin and (meso-tetrakis(1-methylpyridinium-4-yl)porphyri-
meso-tetrakis(2,3,5,6-tetrafluoro-N,N,N-trimethyl-4- nato)iron(III). Inorg. Chem. 27:3773-3781.
aniliniumyl)porphyrin. Inorg. Chem. 33:3159-3163. 86.Neilands, J.B. and J.A. Garibaldi. 1960. Copropor-
70.Lascelles, J. 1964. Tetrapyrrole biosynthesis and its reg- phyrin III tetramethyl ester. Biochem. Prep. 7:36-38.
ulation. Benjamin, New York. 87.Ohno, O., Y. Kaizu, and H. Kobayashi. 1993. J-aggre-
71.Lavallee, D.K. 1987. The Chemistry and Biochemistry of gate formation of a water-soluble porphyrin in acidic
N-Substituted Porphyrins. VCH Publishers, New York. aqueous media. J. Chem. Phys. 99:4128-4439.
72.Lavallee, D.K., A. White, A. Diaz, J.-P. Battioni, and 88.Ojadi, E., R. Selzer, and H. Linschitz. 1985. Properties
D. Mansuy. 1986. Efficient metalloporphyrin synthe- of porphyrin dimers, formed by pairing cationic and
sis under mild conditions using N-benzylporphyrins. anionic porphyrins. J. Am. Chem. Soc. 107:7783-7784.
Tetrahedron Lett. 27:3521-3524. 89.Pasternack, R.F. and E.J. Gibbs. 1996. Porphyrin and
73.Lee, J., J.A. Hunt, and J.A. Groves. 1998. Manganese metalloporphyrin interactions with nucleic acids. In A.
porphyrins as redox-coupled peroxynitrite reductases. Sigel and H. Sigel (Eds.), Metal Ions In Biological Sys-
J. Am. Chem. Soc. 120:6053-6061. tems 33. Marcel Decker, New York.
74.Lemberg, R. and J.W. Legge. 1949. Haematin Com- 90.Pasternack, R.F., P.R. Huber, P. Boyd, G. Engasser, L.
pounds and Bile Pigments. Interscience, New York. Francesconi, E. Gibbs, P. Fasella, G. Cerio Venturo,
75.Li, H. and L. Czuchajowski. 1994. Ribofuransoides and L. deC. Hinds. 1972. On the aggregation of
N-substituted with meso-porphyrin as nucleoside-like meso-substituted water-soluble porphyrins. J. Am.
compounds. Tetrahedron Lett. 35:1629-1630. Chem. Soc. 94:4511-4517.
75a.Lim, C.K. (Ed.). 2001. Porphyrins. In Methods of 91.Pasternack, R.F., H. Lee, P. Malek, and C. Spenser.
Chromatography, Vol. 2. World Scientific Publishers, 1977. Solution properties of tetrakis-(4-N-methyl)
New York. pyridylporphineiron(III). J. Inorg. Nucl. Chem. 39:
76.Lindsey, J. 1980. Increased yield of a desired isomer by 1865-1870.
equilibria displacement of binding to silica gel, applied 92.Pasternack, R.A., K.F. Schaefer, and P. Hambright.
to meso-tetrakis(o-aminophenyl)-porphyrin. J. Org. 1994. Resonance light scattering studies of porphyrin
Chem. 45:5215. diacid aggregates. Inorg. Chem. 33:2062-2065.
77.Lindsey, J.S. and R.W. Wagner. 1989. Investigation of 93.Petho, G. and L.G. Marzilli. 1994. A new spectropho-
the synthesis of ortho-substituted tetraphenylpor- tometric method for determination of cationic por-
phyrins. J. Org. Chem. 54:828-836. phyrinis. Microchem. J. 50:178-183.
78.Longo, F. R., M. G. Finarelli, and J. Kim. The synthe- 94.Purrello, R., L. Monsu’ Scolaro, E. Bellacchio, S. Gur-
sis and some physical properties of ms-tetra(pentafluo- rieri, and A. Romer. 1998. Chiral H- and J-type aggre-
rophenyl)porphin and ms-tetra(pentachloro-phenyl) gates of meso-tetrakis(4-sulfonatophenyl)porphine on
porphin. 1969. J. Heterocycl. Chem. 6:927-931. α-helical polyglutamic acid induced by cationic por-
79.Lyon, R.C., P.J. Faustino, J.S. Cohen, A. Katz, F. phyrins. Inorg. Chem.37:3647-3648.
Mornex, D. Colcher, C. Baglin, S.H. Koenig and P. 95.Ribo, J.M., J. Crusats, J.-A. Farrera, and M.L. Valero.
Hambright. 1987. Tissue distribution and stability of 1994. Aggregation in water solutions of tetrasodium
metalloporphyrin MRI contrast agents. Magn. Reson. diprotonated meso-tetrakis(4-sulfonato-phenyl)por-
Med. 4:24-33. phyrin. J. Chem. Soc. Chem. Commun. 681-682.
80.Margalit, R. and M. Rotenberg. 1984. Thermody- 96.Richards, R.A., K. Hammons, M. Joe, and G.M.
namics of porphyrin dimerization in aqueous solution. Miskelly. 1996. Observation of a stable water-soluble
Biochem. J. 219:445-450 lithium porphyrin. Inorg. Chem 35:1940-1944.
81.Marzilli, L.G. 1990. Medical aspects of DNA-por- 97.Rimington, C. 1952. Haems and porphyrins in health
phyrin interactions. New J. Chem. 14:409-420. and disease. II. Acta Med. Scand. 143:177-196.
82.Mauzerall, D. 1965. Spectra of molecular complexes of 98.Rimington, C. and C.A. Miles. 1951. A study of the
porphyrins in aqueous solution. Biochemistry. 4:1801- porphyrins excreted in urine by a case of congenital
1810. porphyria. Biochem. J. 50:202-206.

66
General Laboratory Methods for Tetrapyrroles

99.Robinson, L.R. and P. Hambright. 1992. Mer- olytic reduction of porphyrins and metalloporphyrins
cury(II) reactions with water-soluble porphyrins. to chlorins or phlorins. J. Chem. Soc. Faraday Trans.
Inorg. Chem. 31:652-656. 89: 495-502.
100.Rousseau, K. and D. Dolphin. 1974. A purification 111.Turay, J., P. Hambright and N. Datta-Gupta. 1978.
of meso-tetraphenyl-porphyrin. Tetrahedron Lett. 48: Intermolecular association of natural and synthetic
4251-4254. water-soluble porphyrins, J. Inorg. Nucl. Chem.
101.Sari, M.A., J.P. Battioni, D. Dupre, D. Mansuy, 40:1687-1688.
and J.B. Le Pecq. 1990. Interaction of cationic por- 112.Turk, H. and W.T. Ford. 1991. Epoxidation of
phyrins with DNA: importance of the number and styrene with aqueous hypochlorite catalyzed by a
position of the charges and minimum structural manganese(III) porphyrin bound to collodial anion-
requirements for interaction. Biochemistry 29:4205- exchange particles. J. Org. Chem. 56:1253-1260.
4215. 113.Valiotti, A., A. Adeyemo, R.F.X. Williams, L.
102.Schneider, H.-J. and M. Wang. 1994. Ligand-por- Ricks, J. North and P. Hambright. 1981. A water-
phyrin complexes: quantitative evaluation of stacking soluble “picket fence” porphyrin and its isomers. J.
and ionic contributions. J. Org. Chem. 59:7464- Inorg. Nucl. Chem. 43:2653-2658.
7472. 114.Varadi, V., F.R. Longo, and A.D. Adler. Nonchro-
103.Schwartz, S., M.H. Berg, I. Bossenmaier, and H. matographic methods of purification of porphyrins.
Dinsmore. 1960. Determination of porphyrins in 1978, p. 581-588. In D. Dolphin (Ed.), The Por-
biological material, p. 221-293. In D. Glick (Ed.), phyrins, Vol. I, Part A. Academic Press, New York.
Methods of Biochemical Analysis, Vol. VIII. Wiley & 115.White, W.I. 1978. Aggregation of porphyrins and
Sons, New York. metalloporphyrins, p. 303-339. In D. Dolphin (Ed.),
104.Shamim, A. and P. Hambright. 1980. An equilibri- The Porphyrins, Vol. V. Chapter 7. Academic Press,
um and kinetic study of water-soluble cadmium por- New York.
phyrins. Inorg. Chem. 19:564-566. 116.White, W.I., R.C. Bachman, and B.F. Burnham.
105.Shamim, A., P. Worthington, and P. Hambright. 1978. Chromatography of porphyrins and metallo-
1981. Synthesis and characterization of phenyl/4- porphyrins, p 553-580. In D. Dolphin (Ed.), The
pyridyl meso substituted porphyrins. J. Chem. Soc. Porphyrins, Vol. I, Part A. Academic Press, New York.
Pakistan 3:1-3. 117.White, W.I. and R.A. Plane. 1974. A homologous
106.Smith, K.M. (Ed.) 1975. Porphyrins and Metallo- series of water-soluble porphyrins and metallopor-
porphyrins. Elsevier, Amsterdam. phyrins: synthesis, dimerization, protonation and self-
107.Smith, K.M., E.M. Fujinara, K.C. Langrey, D.W. complexation. Bioinorg. Chem. 4:21-35.
Parish, and H.D. Tabba. 1983. Manipulation of 118.With, T.K. 1958. Preparation of crystalline porphyrin
vinyl groups in protoporphyrin IX: introduction of esters from bovine porphyria urine. Biochem. J.
deuterium and carbon-13 labels for spectroscopic 68:717-720.
studies. J. Am. Chem. Soc. 105:6638-6646. 119.Worthington, P., P. Hambright, R.F.X. Williams,
108.Sternberg, E.D., D. Dolphin, and C. Bruckner. M.R. Feldman, K.M. Smith, and K.C. Langry.
1998. Porphyrin based photosensitizers for use in 1980. Reduction potentials of beta-substituted free
photodynamic therapy. Tetrahedron 54:4151-4202. base porphyrins. Inorg. Nucl. Chem. Lett.16:441-
109.Sugata, S., S. Yamanouchi, and Y. Matsushima. 447.
1977. Meso-tetrapyridyl-porpyrins and their metal 120.Worthington, P., P. Hambright, R.F.X. Williams, J.
complexes. Synthesis and physico-chemical proper- Reid, C, Burnham, A. Shamim, D.M. Bell, R.
ties. Chem. Pharm. Bull. 25:884-889. Kirkland, R.G. Little, N. Datta-Gupta, and U. Eis-
110.Sutter, T.P.G., R. Rahimi, P. Hambright, J. Bom- ner. 1980. Reduction potentials of seventy-five free
mer, M. Kumar, and P. Neta. 1993. Steric and base porphyrin molecules: reactivity correlations and
inductive effects on the basicity of porphyrins and on the production of potentials. J. Inorg. Biochem.
the site of protonation of porphyrin dianions: radi- 12:281-291.

67
Enzymatic Preparation of Tetrapyrrole
4 Intermediates

Martin J. Warren1 and Peter M. Shoolingin-Jordan2


1School of Biological Sciences, Queen Mary Westfield College, London,
England, UK; 2School of Biological Sciences, University of Southampton,
Southampton, England, UK

1. INTRODUCTION and modification. Despite their prime


metabolic significance, tetrapyrroles and
Tetrapyrroles are intensely colored natur- their derivatives are biosynthesized in sur-
al products of vital importance in the bios- prisingly small quantities and, prior to the
phere for essential processes such as respira- age of genetic engineering, it was difficult
tion and photosynthesis and are also of key to isolate large quantities of pathway inter-
importance as cofactors in a number of mediates and even more challenging to
other enzyme reactions. Tetrapyrroles may study the enzymes themselves. As a result,
either be linear in nature, as found in the many investigations prior to the 1980s
bilins, or cyclic as in the hemes, chloro- were carried out with isotopic tracers to
phylls, and corrins. In the cyclic tetrapyrrole enable biosynthetic conversions to be fol-
group, the four centrally located pyrrole lowed. The advent of molecular biology
nitrogen atoms of the macrocyclic ring offer has had a dramatic effect in the tetrapyrrole
a range of possibilities for metal chelation. field, allowing milligrams of recombinant
Modulation of the properties of the metal- enzymes to be prepared that can be used to
lotetrapyrrole prosthetic groups by individ- manufacture substantial amounts of
ual proteins give rise to a remarkably versa- tetrapyrrole products as well as permitting
tile family of powerful bio-organic reagents. detailed structural investigations of the
The structural complexity of tetra- enzymes. Central to any of these studies is
pyrroles is reflected in a highly intricate the availability of the encoded gene or
branched biosynthetic pathway. For organ- cDNA specifying the enzyme of interest
isms such as Rhodobacter spheroides and and suitable bacterial hosts for their expres-
Pseudomonas aeruginosa, which can biosyn- sion. In this chapter, we have confined our-
thesize four different classes of modified selves to methods for the enzymatic syn-
tetrapyrrole, there are over 40 separate thesis of intermediates along the porphyrin
enzymes dedicated to tetrapyrrole synthesis and siroheme biosynthetic pathways, most

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

69
M.J. Warren and P.M. Shoolingin-Jordan

Table 1. List of Strains and Plasmids Described in this Chapter

Strain Plasmid Properties Reference

JMA18 = (JM109/pA19) R. spheroides hemA cloned into 4


pUC19. Constitutive expression of
ALAS in E. coli

MS1 = (TB1/pMS1) E. coli hemB cloned into pUC19. 20


Constitutive expression of ALAD in E. coli.

BM3 = (TB1/pBM3) E. coli hemC cloned into pUC18. 32


Constitutive expression of PBGD.

SD2 = BL21(DE3)pLysE/ E. coli hemD cloned into pET14b. Raux, Davlin, and
pET14b-HemD Inducible expression of His-tagged UROS. Warren, unpublished.

ER293 = BL21(DE3)pLysS/ B. stearothermophilus cobA cloned into Raux and Warren,


pER291 pET14b. Inducible expression of His- unpublished.
tagged uroporphyrinogen methylase.

ER262 = BL21(DE3)pLysS/ S. cerevisiae MET8 cloned into pET14b. 25


pER259 Inducible expression of His-tagged Met8p.

SW500 = BL21(DE3)pLysS/ E. coli cysG cloned into pEt14b. Woodcock and


pET14b-CysG Inducible expression of His-tagged CysG. Warren, unpublished

BL21(DE3)pLysS/pHT#77 Human cDNA for UROD cloned into pAED4. 24


Inducible expression of His-tagged UROD.

JM109/pHHCPO Human cDNA for CPO cloned into a 22


modified pBTac-1 plasmid. Expression of
His-tagged CPO.

JM109/pMx-PPO M. xanthus hemG cloned into pTF20E, 6


a derivative of pBTac-1. Allows constitutive
expression of His-tagged PPO.

JM109/pLUG18e2 B. subtilis hemH cloned into pUC18. 11


Constitutive expression of ferrochelatase.

of which utilize recombinant proteins. For 2. OVERVIEW OF THE TETRA-


brevity, we have identified one enzyme for PYRROLE BIOSYNTHESIS
each stage of the pathway from a source PATHWAY
that we believe is the easiest to obtain and
handle. The clones for these various The heme biosynthetic pathway togeth-
enzymes can be obtained by contacting the er with the bifurcation points for the syn-
relevant authors as referenced in Table 1. thesis of the other modified tetrapyrroles is
More comprehensive information on each outlined in Figure 1. This chapter is struc-
enzyme, as well as on the history of the tured around the various enzymes high-
pathway elucidation, may be sourced from lighted in the diagram, and considers the
a recent review (28). synthesis of the following compounds:
70
Enzymatic Preparation of Tetrapyrrole Intermediates

• 5-aminolevulinic acid heme from uroporphyrinogen III


• porphobilinogen • coproporphyrinogen III
• preuroporphyrinogen • protoporphyrinogen IX
• uroporphyrinogen III, using multiple • protoporphyrin IX from copropor-
enzymes phyrinogen III, using multiple enzymes
• precorrin-2, sirohydrochlorin and siro- • protoheme

Figure 1. Biosynthesis of heme from ALA. The figure also highlights uroporphyrinogen III as the branchpoint for siroheme and
cobalamin synthesis. Abbreviations used: A, acetate side chain; p, propionate side chain.

71
M.J. Warren and P.M. Shoolingin-Jordan

3. THE ENZYMATIC SYNTHESIS OF ALAS is the rate-determining step in mam-


5-AMINOLEVULINIC ACID malian and fungal heme synthesis, and
intracellular levels of the enzyme are tight-
5-Aminolevulinic acid (ALA) is formed ly regulated. Two enzymes exist in mam-
by two different biosynthetic pathways malian systems; a ubiquitous enzyme,
(Figure 2). One, found in plants, algae, and ALAS1, which is encoded on chromosome
most bacteria, originates from glutamate, 13 and which is subject to tight control in
with glutamyl-tRNA and glutamate 1- all tissues, and the erythroid enzyme,
semialdehyde as intermediates (18), and is ALAS2, which is encoded on the X-chro-
traditionally referred to as the C5 pathway. mosome and expressed constitutively in
The other pathway, found in mammals, developing erythrocytes (9). The photosyn-
fungi, and some photosynthetic bacteria, thetic bacterium, R. spheroides, used for the
involves a single enzymatic step catalyzed isolation of the enzyme also has two genes,
by 5-aminolevulinic acid synthase (ALAS) hemA and hemT (23).
(17). This latter route, often referred to as Aminolevulinic acid can be synthesized
the Shemin, or C4, pathway, involves con- using purified ALAS and the procedure can
densation between glycine and succinyl- be adapted to prepare isotopically labeled
CoA in a reaction in which the carboxyl ALA for enzyme synthesis of labeled later
group of glycine is lost by decarboxylation. pathway intermediates. The ease of using

Figure 2. The biosynthesis of ALA. (a) ALA can be synthesized from glutamate by the C-5 pathway or (b) from glycine and suc-
cinyl-CoA by the Shemin route. In the case of the latter, it is known that the proR-hydrogen of glycine is removed in the overall
transformation into ALA.

72
Enzymatic Preparation of Tetrapyrrole Intermediates

ALAS has been greatly enhanced by the um phosphate buffer, pH 7.2, contain-
availability of the recombinant enzyme ing 0.5 mM pyridoxal 5′ phosphate, 2
from R. spheroides arising from the cloned mM EDTA, 10% glycerol, 5 mM 2-
and overexpressed hemA gene (4). mercaptoethanol, and 100 µM phenyl-
methanesulfonyl fluoride (PMSF). All
3.1. Enzyme Purification of subsequent stages are carried out at 4°C.
R. spheroides ALAS 4. The suspension is sonicated by placing
Expressed in Escherichia coli a large sonicator probe (e.g., a SANYO
Soniprep 150 Ultrasonic Disintegrator,
ALAS can be purified from wild-type R. Integrated Services TCP, Palisades Park,
spheroides (NCIB) according to published NJ, USA) about one third of the way
methods (27). However, preparing the into the bacterial suspension and soni-
media for the growth of this organism is cating the solution at medium ampli-
tedious, and the yield of purified enzyme is tude (10–12 µm) for 4 1-minute bursts
low. To overcome these problems, we have with 2 minutes cooling in between.
produced a recombinant strain of Escher- Cooling is achieved by placing the ves-
ichia coli (JMA19) that overexpresses the R. sel containing the bacterial solution in
spheroides ALAS (HemA) (Table 1), an ice-water slurry.
derived from strain JM109 that had been
5. After sonication, the extract is cen-
transformed with the plasmid pA19. The
trifuged at 15 000× g for 20 minutes to
plasmid (pA19) was constructed from a
remove cell debris. The clarified straw-
HindIII/EcoRI fragment containing the
colored supernatant contains the active
hemA gene from R. spheroides, which had
soluble enzyme.
been modified at the 5′ end by polymerase
chain reaction (PCR) and cloned into To those unfamiliar with the procedures
pUC19 (Table 1) (4). of protein purification, they are encour-
aged to read an excellent account of the
common procedures employed in protein
❖ Procedure 1. Preparation of E. coli
isolation (26).
Lysate Containing Recombinant
R. spheroides ALAS
❖ Procedure 2. Purification of
1. Bacterial growth: From an agar plate of Homogeneous Recombinant
recombinant E. coli harboring the R. R. spheroides ALAS
spheroides hemA gene (JMA18), a single
bacterial colony is removed and used to 1. Ammonium sulfate fractionation: Frac-
inoculate a starter culture (5 mL) of tionation with solid ammonium sulfate
Luria-Bertani (LB) medium containing is the first step of the purification
50 µg/mL ampicillin. process. This procedure is sometimes
2. The culture is grown for between 5 to referred to as salting out and is depen-
10 hours at 37°C and then used to dent upon the concentration of the
inoculate a larger (1 L) culture, which protein solution and the amount of salt
is grown overnight at 37°C with rotary that is added. In the case of ALAS, the
shaking (160–180 rpm) for 18 hours. enzyme is known to precipitate from
3. Harvesting and cell lysis: The cells are solution when the solution is saturated
harvested by centrifugation at 3000× g with 60% ammonium sulfate. To the
for 20 minutes, and the cell pellet is clarified bacterial extract, solid ammo-
resuspended in 10 mL of 20 mM sodi- nium sulfate is added to a saturation of
73
M.J. Warren and P.M. Shoolingin-Jordan

30% by adding 16.6 g of ammonium cedure that separates proteins according


sulfate per 100 mL of extract; to ease to their negative charge. The ALAS
the speed of solubility, the ammonium solution is applied to a diethylamino-
sulfate may be finely powdered in a ethyl (DEAE)-Sephacel chromatogra-
pestle and mortar. phy column (Amersham Pharmacia
2. After stirring for 10 minutes, the solu- Biotech) (25 × 2.7 cm) that had been
tion is clarified by centrifugation at pre-equilibrated in 10 mM sodium
10 000× g for 15 minutes, and the pel- phosphate buffer, pH 7.2, containing
let is discarded. The supernatant is 0.5 mM pyridoxal 5′ phosphate, 2 mM
then made 60% with respect to ammo- EDTA, 10% glycerol, 5 mM 2-
nium sulfate by the addition of a fur- mercaptoethanol, and 100 µM PMSF.
ther 18.4 g of solid ammonium sulfate The ALAS is eluted from the column by
per 100 mL of extract. the application of a linear gradient
3. After stirring for a further 10 minutes, extending from 0 to 500 mM NaCl in a
the suspension is centrifuged again, but total volume of 500 mL using the same
this time the supernatant is discarded, buffer.
and the protein pellet is retained. The 7. Fractions containing ALAS activity,
pellet is resuspended in 5 to 10 mL of which normally elutes between 30 to
the above buffer, but without PMSF, 50 mM NaCl, are pooled and dialyzed
and the extract is dialyzed overnight overnight against the same buffer.
against 5 L of the same buffer. 8. Hydroxyapatite chromatography: The
4. Gel filtration chromatography: The dialyzed enzyme preparation is applied
dialyzed extract is further purified by to a hydroxyapatite column (25 × 2.7
Sepharose S-200 chromatography cm) prepared from hydroxyapatite
(Amersham Pharmacia Biotech, Piscat- (HTP) (Bio-Rad Laboratories, Hercules,
away, NJ, USA). This is a size exclu- CA, USA). The column is washed with
sion procedure, which separates the 100 mL of the same buffer as above, and
protein mixture on the basis of native the enzyme is eluted by the application
molecular mass. As a homodimer, of a linear gradient extending from 0 to
ALAS has a native molecular mass of 500 mM NaCl in the same buffer. Frac-
about 90 kDa. Using a column (100 × tions eluting at about 25 mM NaCl
5 cm) that had been pre-equilibrated (total volume 50 mL) containing the
with the same buffer, the dialyzed pure ALAS are pooled and dialyzed
ammonium sulfate fraction is carefully against 20 mM sodium phosphate
placed on the top of the column, and buffer, pH 7.2, containing 0.5 mM
the system is developed at a flow rate pyridoxal 5′ phosphate, 2 mM EDTA,
of about 1 mL/minute. 10% glycerol, 5 mM 2-mercap-
5. Fractions containing ALAS are deter- toethanol, and 100 µM PMSF.
mined by the presence of ALAS activi- 9. Storage: The purified enzyme is con-
ty (see below) and are pooled. PMSF is centrated to 10 mL under nitrogen
added to give a final concentration of using an Amicon concentration cell
200 µM, and the extract is diluted 2- fitted with a PM-10 membrane (Milli-
fold with distilled water. pore, Bedford, MA, USA) and is stored
6. Anion exchange chromatography: The at -20°C, where it is known to remain
diluted ALAS solution is next subject to active for at least 3 months. The puri-
anion exchange chromatography, a pro- fied protein can be visualized after
74
Enzymatic Preparation of Tetrapyrrole Intermediates

sodium dodecyl sulfate polyacrylamide The reaction is terminated by the addi-


gel electrophoresis (SDS-PAGE), where tion of 150 µL of 10% (wt/vol) trichloro-
it migrates as a single polypeptide with acetic acid, and any protein precipitate is
a molecular mass of about 45 kDa. removed by centrifugation. A known vol-
One liter of culture should produce ume (300 µL) of the supernatant is trans-
about 5 mg of purified enzyme. ferred to a fresh tube containing 300 µL of
1 M sodium acetate buffer (pH 4.6), and
25 µL of acetylacetone is then added. The
3.2. Enzyme Assay and Incubation mixture is heated to 100°C for 10 minutes
Protocol and, after cooling, an equal volume of
ALA may be generated, using ALAS, modified Ehrlich’s reagent (prepared by dis-
with the following incubation mixture and solving 1 g of p-dimethylaminobenzalde-
substrates. hyde in 42 mL of acetic acid and 8 mL of
perchloric acid [62% wt/vol]) is added.
1. Incubation mixture: Stock reaction
After allowing 10 minutes for the color
buffer (100 µL) consisting of 20 mM
(pink) to develop fully, the absorbance of
potassium phosphate buffer, pH 7.2,
the resultant solution is measured at 553
containing 0.5 mM pyridoxal 5′ phos-
nm in a spectrophotometer. Enzyme rates
phate, and 250 mM glycine is mixed
are calculated using an extinction coeffi-
with 5 µL of purified ALAS, and the
cient of 6.04 × 104 M-1 cm-1.
reaction is initiated by the addition of
25 µL of 10 mM succinyl-CoA. Incu-
bation is carried out at 37°C for up to 3.2.2. Continuous Assay of ALAS
30 minutes. The incubation can be
A more convenient, though less sensi-
scaled up as required for the synthesis
tive, enzyme-linked spectrophotometric
of ALA. The yield of ALA should be in
assay can also be employed to monitor the
excess of 90%, and lower yields are nor-
activity of ALAS, in which the liberated
mally associated with an underestima-
CoA is coupled to the formation of acetyl-
tion in the amount of succinyl-CoA.
CoA and reduced nicotinamide adenine
2. Preparation of succinyl-CoA: Succinyl- dinucleotide (NADH) with the enzyme
CoA can either be purchased commer- pyruvate dehydrogenase (27).
cially (e.g., Sigma, St. Louis, MO,
USA) or may be prepared freshly by Glycine + succinyl-CoA → ALA + CoASH + CO2
reacting 8 mg of CoA, 1 mg of freshly CoASH + pyruvate + NAD → acetyl-CoA + CO2 +
powdered succinic anhydride, and 4.5 NADH + H+
mg of sodium bicarbonate in 1 mL of An alternative enzyme-linked continuous
distilled water on ice for 30 minutes assay using 2-oxoglutarate dehydrogenase
with stirring. More concentrated suc- involves the regeneration of succinyl-CoA
cinyl-CoA solutions can be obtained from liberated CoA and 2-oxoglutarate, also
by using less water. forming NADH that can be monitored
spectroscopically (12).
3.2.1. Discontinuous Assay of ALAS
3.3. Preparation of Isotopically Labeled
The ALAS may be quantified using the ALA
discontinuous chemical assay of Mauzerall
and Granick (21). In this case, the above An adaptation of the above assay
reaction is made to a final volume of 175 µL. method can be used to generate isotopical-
75
M.J. Warren and P.M. Shoolingin-Jordan

ly labeled ALA. For instance, either [13C] 4. THE ENZYMATIC SYNTHESIS OF


or [14C]-label at the C5 position of ALA PORPHOBILINOGEN
may be introduced from glycine, appropri-
ately labeled at C2. 2RS-[3H2]-glycine may ALAD catalyzes the first of three steps
be used to label 5S-[3H] ALA, where the for the transformation of ALA into uropor-
label is stereospecifically located on the phyrinogen III, which are found in all liv-
aminomethyl methylene carbon atom. ing organisms that synthesize tetrapyrroles
Labeled succinyl-CoA may be used for (13). The enzymes exist as homo-octamers
introducing label at ALA positions C1 with subunit molecular masses of 35 to 45
through C4. ALA, randomly or stere- kDa, depending on the source organism,
ospecifically tritiated at the C2 and C3 and catalyze the condensation of two mole-
positions, may be generated from 2-oxo-
cules of ALA into the pyrrole PBG (Figure
glutarate using 2-oxoglutarate dehydrogen-
3). Comparisons between the amino acid
ase, followed by decarboxylation to
succinate, and chemical conversion to suc- sequences derived from nucleotide sequenc-
cinyl-CoA. However, because of the insta- ing indicate that the enzyme structure is
bility of ALA, it is essential to transform strongly conserved, and this is confirmed
the ALA synthesized with ALAS rapidly by crystallographic studies that show that
into porphobilinogen (PBG), using puri- both prokaryotic and eukaryotic dehy-
fied 5-aminolevulinic acid dehydratase dratases have a similar (αβ)8 barrel subunit
(ALAD), in order to stabilize any labeled structure (7,8). The active site is located at
hydrogen atoms (1). The procedure for the center of the barrel with two juxtaposed
coupling ALAS to ALAD is covered in the lysines and an aspartic acid playing essential
next section. Labeled PBG prepared from roles in catalysis. One of the lysines, K247
stereospecifically tritiated or deuterated in the E. coli enzyme, forms a Schiff base
ALA in this way has proved important for with the substrate molecule at the P-site, so
mechanistic studies on ALAS and ALAD, called because it binds the ALA molecule
as well as on enzymes further along the that ultimately becomes the propionic acid
heme pathway (28). side chain of the product PBG. Pairs of

Figure 3. The biosynthesis of porphobilinogen from 2 molecules of ALA. It has been established that the proR hydrogen of the
ALA molecule occupying the P (propionate) site is stereoselectively removed during the reaction.

76
Enzymatic Preparation of Tetrapyrrole Intermediates

subunits are arranged as dimers, held ❖ Procedure 3. Purification of


together by long N-terminal arms, with Recombinant ALAD from E. coli
four dimers arranged in D4 symmetry to 1. Bacterial growth: E. coli strain TB1
form the octamer. The conservation of the containing the plasmid pUC19 har-
quaternary structure through evolution boring the E. coli hemB gene in a mod-
may, in part, be as a consequence of a sec- ified EcoRI-BamHI fragment (Table 1)
ond and somewhat surprising function of as constructed by Li et al. (20) is grown
the protein, namely, as the inhibitory com- in 500 mL of LB medium containing
plex of the proteasome (10). ampicillin (50 µg/mL) for 24 hours
ALADs differ in their requirement for after inoculation from a starter culture.
divalent metal ions. Those found in ani- 2. Harvesting and cell lysis: The cells are
mals require only zinc for activity, those harvested by centrifugation at 3000× g
found in plants require only magnesium, for 30 minutes and washed to remove
and others require zinc but are activated by excess medium. Approximately 4 g of
magnesium (14). E. coli ALAD, used for cell paste are then suspended in 20 mL
the methods below, is of the magnesium- of 50 mM potassium phosphate buffer,
activated zinc type. The metal ion in the pH 6.0, containing 100 µM ZnSO4,
zinc-dependent enzymes is chelated to a and 20 mM 2-mercaptoethanol. The
triple cysteine motif at the active site and cells are disrupted by sonication as out-
appears to be an essential part of the active lined in section 1.1 and the cell debris
site that binds the second molecule of ALA is removed by centrifugation at
at the A-site. The zinc-dependent dehy- 10 000× g for 20 minutes.
dratases are exceptionally sensitive to low
levels of lead, which displaces the zinc ion 3. Ammonium sulfate fractionation: The
and inactivates the enzyme. Although resulting supernatant is treated with
ALADs may be purified from a variety of solid ammonium sulfate to bring the
natural sources, the most convenient purifi- saturation to 30% by the addition of
cation (31) is from a recombinant strain of 16.6 g of solid ammonium sulfate per
E. coli harboring the E. coli hemB gene. 100 mL of extract, and the precipitate
is discarded. Addition of a further 5.7 g
of ammonium sulfate per 100 mL of
4.1. Purification of ALAD from E. coli extract is added to the supernatant to
Traditionally, ALADs have been isolated bring the saturation to 40%, and the
from sources that make large quantities of precipitate containing the enzyme is
either heme or chlorophyll, such as liver, collected by centrifugation. The pellet
erythrocytes, and plants. However, more is subsequently resuspended in 3 mL of
recent cloning strategies have led to the the above buffer.
production of large quantities of recombi- 4. Gel filtration chromatography: The
nant forms of the enzyme. In this section, enzyme is further purified by chro-
we will detail the purification of a recombi- matography using a Sephacryl S-300
nant version of the E. coli ALAD. Because gel filtration column (Amersham Phar-
of the way the protein is folded, it is not macia Biotech), previously equilibrated
possible to tag the enzyme, for instance, in the same buffer. Fractions contain-
with a polyhistidine epitope to enable ing the majority of the ALAD activity
affinity purification. Thus, overproduced (for assay see below) are collected from
ALAD has to be purified using conven- the column, concentrated to 20 mg/
tional chromatographic procedures. mL, and dialyzed against 50 mM
77
M.J. Warren and P.M. Shoolingin-Jordan

potassium phosphate buffer, pH 7.0, and to precipitate the thiol and protein.
containing 100 µM ZnSO4, and 20 After centrifugation, an aliquot of the
mM 2-mercaptoethanol. supernatant is mixed with an equal volume
5. High resolution anion exchange chro- of modified Ehrlich’s reagent (21), and the
matography: Final purification may be absorbance is measured at 555 nm (E555 =
achieved by chromatography using a 6.02 × 104 M-1 cm-1). Most ALADs are
Mono Q 5HR FPLC column susceptible to end-product inhibition, a
(Amersham Pharmacia Biotech) equili- factor that tends to limit the yields of PBG.
brated with the same buffer. The Typically a yield of 80% is achieved.
enzyme is eluted in buffer with a linear
gradient from 0 to 1 M KCl, and active 4.2.2. Large-Scale Preparation of PBG
fractions are collected and pooled.
ALAD (500 U) is incubated in a stop-
6. Storage: The pooled active fractions are
pered conical flask at 37°C in 1.9 L of 5
concentrated to about 2 mg/mL, and
mM potassium phosphate buffer, pH 6.8,
the purified enzyme is filter-sterilized
containing 5 µM ZnSO4, 5 mM 2-mercap-
for storage at 4°C in 50 mM potassium
toethanol, and ALA. The 5-aminolevulinic
phosphate buffer, pH 6.0, containing
acid hydrochloride (1 g; Sigma) is dissolved
100 µM ZnSO4, and 20 mM 2-mer-
in about 95 mL of the same buffer, adjust-
captoethanol. Activity is maintained
ed carefully to pH 6.8 with 0.1 M NaOH,
for 2 weeks. From a 0.5-L culture,
and made up to 100 mL before adding to
between 10 to 20 mg of purified
the above flask. Incubation is carried out
ALAD is obtained.
under nitrogen, typically, for 10 hours or
until the rate of PBG production has
4.2. Enzyme Assay and Incubation ceased. The reaction is followed by remov-
Protocol ing 10 µL of the incubation mixture at
Different protocols may be employed intervals and adding to 490 µL of 10%
for the enzymatic synthesis of PBG, trichloroacetic acid containing 0.1 M
depending on whether a small- or large- HgCl2 to precipitate the thiol. After cen-
scale preparation is required. Indeed, the trifugation, 0.4 mL of the supernatant is
small-scale synthesis is identical to that mixed with an equal volume of modified
used for the assay of the enzyme. Ehrlich’s reagent, and the absorbance is
measured as above. The PBG is purified
4.2.1. Assay and Small-Scale Enzymatic from the reaction mixture by adjusting the
Synthesis of PBG pH to 7.5 and passing the incubation mix-
ture slowly through a column (2 × 12 cm)
Purified E. coli ALAD (1–10 µg) is of Dowex 1 × 8 acetate (200–400 mesh).
preincubated in a total volume of 500 µL The column is first washed with 1 L of dis-
of 50 mM potassium phosphate buffer, pH tilled water, and the PBG is eluted with 1
8.0, containing 50 µM ZnSO4, and 10 M acetic acid and collected by freeze-dry-
mM 2-mercaptoethanol. The reaction is ing the solution or rapid-flash evaporation
initiated by the addition of ALA to give a below 30°C. The PBG is recrystallized by
final concentration of 5 mM. Incubation is dissolving it in a minimum volume of 1 M
carried out at 37°C for 3 minutes, after ammonia and adding 1 M acetic acid to
which time an equal volume (500 µL) of bring the pH to the isoelectric point of 5.5.
10% trichloroacetic acid containing 0.1 M After allowing the crystallization to proceed
HgCl2 is added to terminate the reaction for 1 hour, the crystals are filtered off and
78
Enzymatic Preparation of Tetrapyrrole Intermediates

washed with a minimum volume of ice- ly or stereospecifically located label at C2


cold methanol, followed by dry ether, and and C3 of ALA. This is accomplished by
stored desiccated in vacuo at -20°C. The labeling 2-oxoglutarate with either tritium
overall yield of purified PBG is about 50% or deuterium, either nonenzymically or
after recrystallization. using 2-oxoglutarate dehydrogenase, fol-
lowed by nonenzymatic decarboxylation to
4.2.3. Labeled PBG Synthesis succinate, cyclization to succinic anhydride
with dicyclohexylcarbodiimide, and conver-
For the preparation of radioactive PBG sion to succinyl-CoA as described above (see
from 5-amino[4-14C]levulinic acid, the also Reference 10). The succinyl-CoA is
concentration of potassium phosphate then transformed into PBG in 5 mM Tris-
buffer should be reduced to 5 mM and suf- HCl buffer, pH 6.8, containing 80 mM
ficient E. coli ALAD units used to ensure glycine, 10 µM pyridoxal 5′ phosphate,
quantitative conversion within 20 to 30 ALAS (20 U), and ALAD (35 U). The reac-
minutes. It is essential to adjust the pH of tion is started by adding labeled succinyl-
the ALA prior to addition to the enzyme, CoA to give a final concentration of 10 mM
particularly if it is dissolved in 0.1 M HCl. and a total volume of 1 mL, and the incu-
After synthesis of the PBG, the volume of bation is continued until the reaction is
the solution is reduced, for example using a complete, typically in 30 to 60 minutes.
Speedivac (centrifugation under reduced PBG is then separated from any ALA
pressure) or by lyophilization, and the PBG and glycine by adjusting the mixture to
is purified by chromatography using prepar- pH 7.2 and application to a Dowex 2 × 8
ative cellulose glass plates developed in n- acetate (400 mesh) column (2 × 10 cm).
butanol:acetic acid:water (4:1:1 vol/vol). ALA is removed by washing the column
After carefully drying the plates in a cool with water (50 mL), and PBG is eluted
nitrogen stream, PBG is eluted from the cel- with 20 mL of 1 M acetic acid and
lulose with water and lyophilized for storage purified by cellulose chromatography as
in liquid nitrogen. The PBG can be detect- above.
ed on the plate by spraying the edge of the Glycine labeled with 13C, 14C, 3H, or
plate with modified Ehrlich’s reagent. 2H label at C2 may be used to label ALA at
the C5 position. Thus, 2RS-[3H2]-glycine
4.2.4. Coupled Enzymatic Synthesis of incubated with ALAS generates stere-
Labeled PBG Samples from ALA ospecifically labeled 5S-[3H] ALA, which
Generated from Glycine and can be transformed by ALAD into 11S-
Succinyl-CoA by ALAS [3H]-PBG. In this case, the reaction mix-
ture is prepared at 0°C in a volume of 1
The difficulty of isolating labeled ALA, mL containing 10 mM Tris-HCl buffer,
prepared either from labeled glycine or suc- pH 6.8, 3 mM 2RS-[3H2]-glycine, 5 µM
cinyl-CoA, may be overcome by coupling pyridoxal 5′-phosphate, ALAS (20 U), and
the reaction to ALAD to convert rapidly ALAD (30 U). The reaction is started by
any ALA formed into PBG. The latter is the addition of 50 µL of succinyl-CoA (1
more stable and easier to isolate and purify µmol) and by raising the temperature to
and fulfills the additional requirement that 37°C. Further aliquots of succinyl-CoA
any labeled hydrogen atoms are located in may be added at 10-minute intervals. After
stable positions. incubation for 30 minutes, the PBG is
Tritiated or deuterated succinyl-CoA purified using a Dowex 2 × 8 acetate col-
may be used for introducing either random- umn as above.
79
M.J. Warren and P.M. Shoolingin-Jordan

5. SYNTHESIS OF PREURO- expressed from strain BM3 (Table 1), con-


PORPHYRINOGEN sisting of E. coli TB1 harboring a plasmid
(pBM3) constructed by cloning a 1.68 kb
Porphobilinogen deaminase (PBGD) also BamHI-SalI DNA fragment, containing
known as hydroxymethylbilane synthase the E. coli hemC gene from pST48, into
(HMBS) and incorrectly as uroporphyrino- pUC18 (32).
gen I synthase, catalyzes the formation of
preuroporphyrinogen from 4 molecules of ❖ Procedure 4. Purification of
PBG (Figure 1). Preuroporphyrinogen is a Recombinant E. coli PBGD
highly unstable 1-hydroxymethylbilane that
acts as the substrate for uroporphyrinogen 1. Bacterial growth: Sterilized bacterial
III synthase to yield uroporphyrinogen III, medium (4 L) containing 50 mg/mL
the common tetrapyrrole precursor for ampicillin is inoculated from a starter
other tetrapyrroles. PBGDs have been iso- culture and incubated at 37°C
lated from a number of sources, and their overnight in 4 baffled flasks (2 L).
properties have been well established (for a 2. Harvesting and cell lysis: The cells are
review see Reference 28). All PBGDs exist collected by centrifugation at 3000× g
as monomeric species with molecular mass for 30 minutes and resuspended (3–4
values between 33 and 45 kDa. The mL/g of cells) in 0.1 M potassium
nucleotide sequences of genes/cDNAs spec- phosphate buffer, pH 8.0, containing
ifying the deaminases from bacterial, plant, 14 mM 2-mercaptoethanol. PMSF,
and animal sources show considerable con- dissolved in ethanol, is added to give a
servation in the deduced protein sequences, final concentration of 0.1 mM. The
suggesting that all the enzymes are likely to bacteria are broken by sonication as
be structurally related to one another. Inves- described in Procedure 1, and the soni-
tigations with the deaminase from E. coli cated extract is clarified by centrifuga-
have identified a novel prosthetic group, tion at 15 000× g for 15 minutes.
named the dipyrromethane cofactor (16), 3. Heat treatment: PBGDs are thermo-
made up of 2 PBG-derived units linked stable enzymes, and this property is
together and covalently attached to the utilized during the purification proce-
enzyme. The cofactor acts as a primer for dure. The sonicated sample is heat-
the synthesis of the linear tetrapyrrole treated by placing the sample in a water
(bilane) chain that is built onto the free α- bath for 10 minutes at 60°C, followed
position of the cofactor. This occurs by the immediately by cooling to 0°C in an
sequential condensation of 4 PBG mole- ice–salt bath. The precipitated protein
cules with the holoenzyme through enzyme is removed by centrifugation at
intermediate complexes, termed ES, ES2, 10 000× g for 20 minutes at 4°C.
ES3, and ES4. The product, preuropor-
phyrinogen, is liberated from ES4 by 4. Ammonium sulfate fractionation: Solid
hydrolysis, regenerating the holoenzyme ammonium sulfate is added slowly to
with the cofactor still covalently attached. the above extract (protein 30 mg/mL)
to give 30% saturation by the addition
5.1. Enzyme Purification of 16.6 g of ammonium sulfate per 100
mL of extract. The solution is allowed
The deaminase is conveniently isolated to equilibrate with stirring for 10 min-
from a variety of sources (for reviews see utes at 4°C, and the supernatant is
Reference 1). In this case, PBGD is removed by centrifugation at 10 000× g
80
Enzymatic Preparation of Tetrapyrrole Intermediates

for 20 minutes at 4°C. Further The incubation mixture is equilibrated at


ammonium sulfate is added to give 37°C in a water bath, and the reaction is
60% saturation by the addition of a started by the addition of 100 µL of 1 mM
further 19.8 g per 100 mL of extract. PBG. After 10 minutes at 37°C the reac-
The pellet containing the enzyme is tion is terminated by the addition of 200
collected by centrifugation and resus- µL of 5 N HCl. A further 10 µL of benzo-
pended in 30 mL of 0.1 M potassium quinone (1 mg/mL in methanol) is added,
phosphate buffer, pH 8.0, containing and the mixture allowed to oxidize for a
14 mM 2-mercaptoethanol. The sam- further 20 minutes under bright light. The
ple is then dialyzed against 2 L of the absorbance is determined at 405 nm (E405
same buffer at 4°C for at least 4 hours = 5.48 × 105 M-1 L), and reading should
with stirring. fall between 0 to 1 OD units on the
5. Ion exchange chromatography: Ion spectrophotometer. It may be necessary to
exchange chromatography is per- dilute the sample 10-fold in 1 N HCl to
formed using a DEAE-Sephacel col- achieve such readings. One unit is defined
umn (2.5 × 20 cm) equilibrated and as the amount of PBGD enzyme needed to
eluted in an isocratic fashion by the consume 1 µmol of PBG per hour.
passage of a 2 L of 0.1 M potassium
phosphate buffer, pH 8, containing 14 5.3. Preparation of Preuroporphyrinogen
mM 2-mercaptoethanol, through the
column. The column fractions con- Preuroporphyrinogen (0.1 µmol) is gen-
taining the deaminase enzyme are erated from 0.5 µmol of PBG in a final
located by assay (see below) and by volume of 1 mL of degassed Tris-HCl
SDS-PAGE. buffer, pH 9.1, using 100 mg of purified
6. Storage: Active fractions found to be PBGD over a period of 1 minute at 37°C.
free from any major contaminating The reaction is performed at a higher pH
proteins are concentrated to 10 to 15 than the assay to help stabilize the preuro-
mL by ultrafiltration, using a 100-mL porphyrinogen. The sample is rapidly
ultrafiltration cell fitted with a PM-10 cooled to 0°C in liquid nitrogen, and the
membrane, and the deaminase is preuroporphyrinogen is separated from the
desalted into distilled water using a holo-deaminase by ultrafiltration through
small gel filtration system such as a a PM-10 membrane fitted to a 5-mL con-
PD-10 column. The purified enzyme centration cell under nitrogen at 4°C in a
(specific activity 30–40 U/mg) is then cold room. The preuroporphyrinogen is
lyophilized to yield a white solid that is used at once or frozen in liquid nitrogen
stable for several months when stored for up to 1 hour, under nitrogen, until
at -20°C under nitrogen. This protocol required. The yield of preuroporphyrino-
generates about 10 mg of purified gen is in excess of 80%.
PBGD per liter of culture.
6. MULTIENZYME SYNTHESIS OF
5.2. Assay of Enzyme UROPORPHYRINOGEN III

PBGD is assayed using a stopped assay. The enzyme uroporphyrinogen III syn-
To 750 µL of 0.1 M Tris-HCl buffer, pH thase (UROS) (also known as uropor-
8.0, is added 40 µL of enzyme containing phyrinogen III cosynthase) catalyzes a
between 0.1 to 1 µg of purified enzyme. remarkable reaction in which preuropor-
81
M.J. Warren and P.M. Shoolingin-Jordan

phyrinogen is rearranged and cyclized to porphyrinogen I, a physiologically unim-


yield uroporphyrinogen III (Figure 4). portant isomer (Figure 4). Uroporphyrino-
Uroporphyrinogen III is the common pre- gen III, however, represents an important
cursor for hemes, chlorophylls, vitamin transitory intermediate in the synthesis of
B12, and all other tetrapyrroles (for a the modified tetrapyrroles, since it repre-
review see Reference 28). The UROS sub- sents the first branchpoint in the pathway
strate, preuroporphyrinogen, is generated of cobalamin, siroheme, or coenzyme F430,
by the preceding enzyme of the tetrapyr- while decarboxylation of the 4 acetate side
role pathway, PBGD (see above section) by chains of uroporphyrinogen III by the
a reaction that involves the polymerization enzyme uroporphyrinogen III decarboxy-
of 4 molecules of the monopyrrole precur- lase produces coproporphyrinogen. The
sor PBG. Preuroporphyrinogen has a half- ability to produce uroporphyrinogen III in
life of less than 5 minutes at neutral pH good yields is therefore important for the
values (15) cyclizing spontaneously to uro- study of these branchpoint enzymes. Uro-

Figure 4. The synthesis of uroporphyrinogen I and III from preuroporphyrinogen. Note the action of UROS, which is able to
invert the orientation of ring d during the macrocyclic ring closure process.

82
Enzymatic Preparation of Tetrapyrrole Intermediates

porphyrinogens can be generated either final concentration of 0.4 mM, and the
non-enzymically, by the reduction of uro- cells are grown for another 2 hours.
porphyrin or, in situ, using a coupled 2. Harvesting and cell lysis: The bacteria
enzyme system. The non-enzymic reduc- are collected by centrifugation (10 000×
tion of uroporphyrin is achieved by the use g at 4°C). The bacterial pellet is resus-
of sodium amalgam. Although the reaction pended in 10 mL of binding buffer
is easy to perform, problems can arise from (5 mM imidazole, 0.5 M NaCl, 20 mM
the pH of the solution, which becomes Tris-HCl, pH 7.9). The bacterial sus-
very high by the end of the reductive pension is sonicated as described in
process. For these reasons, we have favored Procedure 1, and the solution is cen-
the generation of uroporphyrinogen by trifuged (10 000× g at 4°C) to remove
enzymatic transformation of PBG, the cellular debris.
employing the actions of the enzymes 3. His-bind column: The His-tag sequence
PBGD and UROS. of the fusion protein can bind to diva-
lent metal cations such as Co2+ and
6.1. Purification of UROS Ni2+ immobilized on to His-bind resin
(Novagen, Madison, WI, USA; howev-
UROS can be purified from a recombi- er, many suppliers make different forms
nant version of the E. coli hemD that has of metal chelate resin and readers are
been modified to incorporate a 6-histidine encouraged to browse the multitude of
(His) tag at the N terminus of the protein. catalogues available). After unbound
The E. coli hemD was amplified by PCR
proteins are washed away, the His-
with appropriately designed primers such
tagged protein is eluted with imidazole.
that the gene was cloned into the NdeI and
The resin (poured into a small column,
BamHI sites of pET14b, giving the plas-
1 × 2.5 cm) is initially prepared by rins-
mid pET14b-HemD (Table 1). When
ing with 15 mL of water, charged with
transformed into E. coli BL21(DE3)pLysE,
25 mL of a 50 mM divalent cation solu-
the strain was found to overproduce the
tion (normally Ni2+) (charge buffer),
His-tagged version of the protein, which
and equilibrated with 15 mL binding
has a molecular mass of 29 kDa. The strain
buffer. The supernatant is loaded onto
harboring the plasmid is comparatively
the charged His-bind column. The col-
unstable, and fresh transformants are
required when cultures are to be grown. umn is washed with 10 column vol-
The following protocol can be adapted for umes of binding buffer, 6 column vol-
purification of all the His-tagged enzymes umes of wash buffer (100 mM
described in this chapter. imidazole, 0.5 M NaCl, 20 mM Tris-
HCl, pH 7.9), and finally the protein is
eluted in 6 column volumes of elution
❖ Procedure 5. Purification of His- buffer (400 mM imidazole, 0.5 M
Tagged UROS from E. coli NaCl, 20 mM Tris-HCl, pH 7.9). The
1. Bacterial growth: The bacteria are grown protein eluting from the column can be
from a starter culture in 2-L baffled detected by the use of the Bio-Rad pro-
flasks containing 1 L of LB media (with tein assay and SDS-PAGE.
appropriate antibiotics) at 37°C with 4. Storage: Fractions containing the mod-
vigorous shaking until an A600 = 0.6 is ified UROS are pooled and desalted by
reached, at which point isopropyl-β-D- passing through a PD-10 column, pre-
thiogalactoside (IPTG) is added to a viously equilibrated in 50 mM Tris-
83
M.J. Warren and P.M. Shoolingin-Jordan

HCl, pH 7.8. The protein is lyo- tion mixture) the solution can be filtrated
philized and is stable in this form for in an ultrafiltration unit fitted with a PM-
up to 1 year. In comparison to some of 10 membrane. The filtrate should be kept
the other enzymes described in this under argon to help prevent any oxidation.
chapter, UROS is poorly expressed, The yield of uroporphyrinogen III from
and a yield of about 2 mg/L of culture PBG is normally in excess of 95%.
is normally achieved. The uroporphyrinogen I isomer can also
be synthesized by this method simply by
6.2. Enzymatic Preparation of omitting UROS from the incubation.
Uroporphyrinogen III
Uroporphyrinogen III can be synthe- 7. SYNTHESIS OF PRECORRIN-2
sized in vitro using PBG and purified (DIHYDROSIROHYDRO-
PBGD and UROS. The reaction can be CHLORIN), SIROHYDRO-
undertaken in a range of buffers between CHLORIN, AND SIROHEME
pH 7.5 and 9.0, although the uropor-
phyrinogen III is generally more stable at Enzymatic transformations of uropor-
the higher pH values. To prevent oxidation phyrinogen III into precorrin-2 are depen-
of the product, the buffers are normally dent upon the presence of the enzyme uro-
thoroughly degassed by freeze–thawing porphyrinogen III methyltransferase
under a vacuum of less than 1 mbar. For (Figure 5), which requires S-adenosyl-L-
efficient transformation of PBG into uro- methionine (SAM) as a methyl donor (3).
porphyrinogen III, the reaction mixture There are a number of sources of this
should contain PBGD at 10 µg/mL, enzyme including Pseudomonas denitrifi-
UROS at 2 µg/mL, and PBG at 100 µM. cans, Bacillus megaterium, and Bacillus
The reaction is effectively quantitative, stearothermophilus. The CysG enzyme from
thus producing uroporphyrinogen III at a both E. coli and Salmonella typhimurium
concentration approaching 25 µM. This can also be used, although CysG is, in fact,
can be verified by taking 50 µL of the incu- a multifunctional enzyme responsible for
bation, mixing with 950 µL of 1 N HCl, the conversion of precorrin-2 into siroheme
and leaving under a bright light for 20 (30). However, in the presence of only
minutes. After centrifugation in an Eppen- SAM and uroporphyrinogen III, the en-
dorf model microfuge at 13 000 rpm for 5 zyme will effectively transform uropor-
minutes, the absorbance of the solution at phyrinogen III into precorrin-2. The uro-
405 nm can be measured, and the concen- porphyrinogen methyltransferases are
tration of porphyrin can be determined normally homodimers with a subunit
using the extinction coefficient of 5.48 × molecular mass of about 30 kDa, while the
105 M-1 L. CysG proteins, which are also homodimers,
So long as the enzymatic incubation is have a subunit molecular mass of 50 kDa.
kept in an anaerobic environment under
reduced light, the uroporphyrinogen III is 7.1. Purification of Uroporphyrinogen
stable for several hours. The solution Methyltransferases
should appear colorless, but if it starts to
turn pink then this is diagnostic of the Although the uroporphyrinogen methyl-
solution starting to oxidize. To isolate the transferases can be purified from recombi-
uroporphyrinogen III from the incubation nant sources, the preparations are often
(i.e., to remove the enzymes from the reac- laborious and in low yields. We have favored
84
Enzymatic Preparation of Tetrapyrrole Intermediates

the use of His-tagged enzymes, including

Figure 5. The biosynthesis of siroheme from uroporphyrinogen. Uroporphyrinogen III is methylated at positions 2 and 7 to give precorrin-2 by the enzyme uroporphyrinogen methyltrans-
the B. stearothermophilus CobA and the E.
coli CysG, since these can be purified easily
in a couple of hours by metal chelate chro-
matography. For instance, the B. stearother-
mophilus CobA can be purified from strain
ER262 (Table 1), which is BL21(DE3)
pLysS transformed with pER259 (cobA
cloned into pET14b). As for all His-tagged
enzymes, the isolation procedure is very
similar to that described in Procedure 5
(Section 6.1). About 15 mg of purified
enzyme can be obtained per liter of culture.

7.2. Assay of Uroporphyrinogen


Methyltransferase
The enzyme is very difficult to assay.
Accurate activity for uroporphyrinogen
methyltransferases can be obtained by mea-
ferase, while dehydrogenation of precorrin-2 gives sirohydrochlorin and finally ferrochelation produces siroheme.

suring the incorporation of label from


[methyl-3H]SAM into the uroporphyrino-
gen III framework as previously described
(3). The enzyme is incubated in 50 mM
Tris-HCl buffer containing 50 µM SAM
(10 µCi.µmol-1) and 5 µM uroporphyrino-
gen III at either 30° or 37°C for up to 1
hour in a final volume of 1 mL. After incu-
bation, the mixture is quickly applied to a
small column (e.g., 0.5 mL bed volume) of
DEAE Sephacel. After washing the column
with 10 column volumes of buffer, the
tetrapyrrole compounds were eluted in 3
mL of 1 M HCl. After mixing with an
appropriate scintillant, the amount of
radioactivity transferred to uroporphyrino-
gen III can be determined.

7.3. Generation of Product by


Incubation of Recombinant
Enzyme
Since many of the uroporphyrinogen III
methyltransferases display substrate inhibi-
tion, uroporphyrinogen III is normally
incubated with the enzyme at a final con-
centration of 5 µM, with SAM at a concen-
tration of 50 µM (3). The high concentra-
85
M.J. Warren and P.M. Shoolingin-Jordan

tion of SAM helps to overcome inhibition ferred to a small plastic column. The resin is
with S-adenosyl-L-homocysteine. The reac- washed with buffer, and buffer containing
tion should be undertaken at pH 8.0 in 50 250 mM NaCl, to remove the more loosely
mM Tris-HCl buffer at either 30° or 37°C. bound proteins, and the precorrin-2 is elut-
As precorrin-2 is so unstable, we recom- ed in buffer containing 2 M NaCl. Precor-
mend that a high concentration of the uro- rin-2 is highly unstable with a tendency to
porphyrinogen methyltransferase be used in form mono- and dilactones. The com-
the reaction at a concentration of about 50 pound is difficult to store and should be
µg/mL. This ensures a rapid synthesis of used immediately. The uroporphyrinogen
precorrin-2, which can be monitored visu- methyltransferases are very susceptible to
ally since the solution turns a bright yellow feedback inhibition by S-adenosyl-L-homo-
color. In fact, precorrin-2 has a broad cysteine, and therefore, to achieve high
absorption maximum around 350 to 400 yields of precorrin-2 (in excess of 90%), a
nm. The newly synthesized precorrin-2 can high concentration of enzyme and SAM are
be separated from the other components of required in the incubation mixture.
the incubation mixture by ion exchange Sirohydrochlorin can be synthesized
chromatography. After mixing in a few mil- from precorrin-2 by the inclusion of either
liliters of ion exchange resin such as DEAE CysG or Met8p together with NAD+ to
Sephacel, the solution is slowly stirred for the above incubation (25). These enzymes
about 1 minute. Once the resin has settled, are purified in the same manner as
the majority of the supernatant can be described for the CobA (above) from the
decanted, and the resin slurry can be trans- appropriate strains shown in Table 1. The

Figure 6. Spectra of precorrin-2, sirohydrochlorin, and cobalt-sirohydrochlorin. The spectrum of precorrin-2 (large dashed line)
has a broad absorption maximum around 350 to 400 nm. The spectrum of sirohydrochlorin (filled line) has a more defined
absorption maximum at 378 nm, while cobalt sirohydrochlorin (dashed line) has defined maxima at 410 and 595 nm.

86
Enzymatic Preparation of Tetrapyrrole Intermediates

CysG or Met8p should be added at a con- function of UROD is manifested as the


centration of 50 µg/mL with NAD+ at 25 most common form of porphyria, porphyr-
µM. Sirohydrochlorin is characterized by ia cutanea tarda (19).
the appearance of a new absorption maxi-
mum at 378 nm (Figure 6). 8.1. Purification of UROD
Siroheme can be synthesized by the
inclusion of ferrous iron with the incuba- A His-tagged recombinant form of
tion. However, this reaction is difficult to UROD has been described recently (24),
follow, and a clearer spectral difference can in which the His-tag does not appear to
be obtained by the use of cobalt, which interfere with the catalytic activity of the
produces a spectrum with absorption max- enzyme. In this case, the cDNA corre-
ima at 410 and 595 nm (Figure 6). The sponding to human UROD was cloned
metal ions should be added to a concentra- into a T7 inducible plasmid with an N-ter-
tion no higher than 10 µM, otherwise the minal His-tag (Table 1). Expression of the
enzymes become inactivated. enzyme is achieved by transformation into
E. coli BL21(DE3)pLysS. The purification
of the His-tagged UROD is essentially sim-
8. SYNTHESIS OF COPRO-
ilar to that described in Procedure 5 (Sec-
PORPHYRINOGEN
tion 6.1), with yields in excess of 15 mg of
Decarboxylation of the 4 acetate side purified enzyme per liter of culture.
chains of uroporphyrinogen III leads to the
synthesis of coproporphyrinogen III. The 8.2. Assay of UROD
enzyme that catalyzes this reaction is uro-
porphyrinogen III decarboxylase (UROD). The simplest way to monitor the activity
The best characterized enzyme is that from of UROD is to employ a fluorometric
human, which can be expressed to high lev- method that relies on the difference in fluo-
els in E. coli cells as a His-tagged recombi- rescence between uroporphyrin and copro-
nant enzyme. The enzyme does not require porphyrin (29). The reaction mixture (3
any metal ions or cofactors for activity, mL) is stopped by the addition of
since it most likely catalyzes the reaction by trichloroacetic acid (to a final concentration
forming a protonated pyrrole within the of 5%), and the porphyrinogens are then
porphyrinogen substrate, which acts as an oxidized to their corresponding porphyrins
electron sink. The enzyme is prone to acy- by the addition of 60 µL of H2O2 (30%).
lation of cysteine residues and also to oxi- After 20 minutes, the amount of copropor-
dation from bound porphyrinogens. The phyrin can be estimated from its emission
enzyme would appear to be dimeric with a fluorescence at 610 nm after excitation at
subunit molecular mass of around 40 kDa. 406 nm. The fluorescence is compared to a
The overproduction of the human enzyme standard curve made from commercially
as a recombinant protein has allowed its obtained coproporphyrin. This technique
crystallization, and a detailed 3-dimension- can only be used as a rough guide to the
al structure is now available (33). In activity of the enzyme. More accurate assays
humans, a number of mutations within the rely on the exact quantities of porphyrin
UROD gene are known to cause hereditary isomers that are formed during the assay.
forms of porphyria, while the enzyme is This is generally achieved after esterification
also prone to inactivation by a number of of the reaction products and separation by
porphyrinogenic compounds. The dys- HPLC (see Chapter 5).

87
M.J. Warren and P.M. Shoolingin-Jordan

8.3. Synthesis of Coproporphyrinogen a requirement for metal ions for activity, the
human enzyme appears functional in the
Coproporphyrinogen III can be effi- absence of any metal or cofactors. Indeed,
ciently generated by the following proto- the simplest source of the enzyme is a His-
col. An incubation mixture containing 50 tagged version of the human enzyme,
mM Tris-HCl buffer, pH 8.0, 2 mM which is easily overproduced in E. coli,
dithiothreitol (DTT), and 5 µM uropor- yielding in excess of 10 mg/L.
phyrinogen III is prepared. The uropor-
phyrinogen III is made as described in Sec- 9.1. Purification of CPO
tion 4. The buffer should be thoroughly
degassed by freeze–thawing under reduced Although the human CPO is thought to
pressure. The reaction is started by the be associated with the outer surface of the
addition of purified UROD (5 µg/mL), inner membrane of the mitochondrion,
and the incubation is performed at 37°C when expressed in E. coli it is easily solubi-
under dim light. The coproporphyrinogen lized in the presence of 0.5% n-octyl-β-D-
III can be removed from the enzyme mix- glucopyranoside (22). Recombinant
ture by ultrafiltration through a PM-10 expression of the human CPO was
membrane in an ultrafiltration unit. The achieved by cloning the cDNA into the
solution should appear colorless, and any expression vector pBTac such that the
appearance of reddish coloration should be cDNA was cloned in-frame with a 6-histi-
taken as a sign of oxidation. The copropor- dine tag at the 5′ end (Table 1). The result-
phyrinogen should be used immediately, ing plasmid, termed pHHCPO, was trans-
although it may be possible to freeze the formed into E. coli JM109. Purification of
solution so long as it is kept under argon. the enzyme is essentially as described in
The yield of coproporphyrinogen from Procedure 5 (Section 6.1), except that the
uroporphyrinogen is in excess of 95%. resuspension buffer for the cell pellet (step
2) is 50 mM NaH2PO4, 300 mM NaCl,
0.5% n-octyl-β-D-glucopyranoside, and
9. SYNTHESIS OF 100 mM Tris-HCl, pH 8.0, containing 1
PROTOPORPHYRINOGEN mM PMSF. The Ni-column is washed with
resuspension buffer containing 20 mM
The synthesis of protoporphyrinogen imidazole, and the CPO is eluted from the
requires the decarboxylation of the two pro- column in resuspension buffer plus 250
pionate side chains on rings a and b of the mM imidazole. After dialysis against resus-
coproporphyrinogen III isomer by the pension buffer to remove the imidazole, the
enzyme coproporphyrinogen oxidase enzyme can be stored frozen at -20°C for
(CPO). There are two independent enzyme several months. The yield of purified
systems that achieve this transformation, enzyme is in excess of 10 mg/L of culture.
representing aerobic (encoded by hemF)
and anaerobic processes (encoded by 9.2. Assay of CPO and Synthesis of
hemN). However, the aerobic enzyme is Protoporphyrinogen
much better characterized, where purified
recombinant hemF-encoded CPO has been The incubation for the synthesis of pro-
shown to require two molecules of oxygen toporphyrinogen is undertaken in 50 mM
during the reaction with the release of two Tris-HCl, pH 8.0, containing 0.2%
molecules of carbon dioxide (22). Although Tween 20 and 2.5 mM glutathione.
some reports have suggested the enzyme has Coproporphyrinogen III is generated by
88
Enzymatic Preparation of Tetrapyrrole Intermediates

the reduction of coproporphyrin III dihy- introducing three new double bonds. In
drochloride (Porphyrin Products, Logan, aerobic organisms, the enzyme would
UT, USA) with 3% sodium amalgam. The appear to require the services of a flavin
reduction itself should be undertaken in cofactor and passes the electrons onto mol-
100 Tris-HCl, pH 8.0, and once the solu- ecular oxygen. The corresponding anaero-
tion turns colorless, or nearly colorless, the bic oxidation of protoporphyrinogen
solution is passed through a small 10-mL remains poorly understood, but in E. coli
column of glass wool. This not only serves it would appear to be a multiprotein com-
to remove the amalgam and mercury, but plex that is coupled to the respiratory chain
the glass wool also appears to bind the por- of the cell. From a commercial standpoint,
phyrin while allowing the porphyrinogen the enzymatic oxidation of protopor-
to pass through (Dailey, personal commu- phyrinogen represents an important target
nication). The pH of the solution is then for a number of herbicides, diphenyl ether
adjusted back to around 7.0 to 8.0 by addi- derivatives, which selectively inhibit the
tion of 2 M morpholinepropanesulfonic enzyme. Defects in the human enzyme are
acid (MOPS), pH 7.0. The copropor- associated with variegate porphyria, the
phyrinogen is added to the incubation form of porphyria that is particularly com-
mixture at a final concentration of 5 µM mon in South Africa (19).
and incubated with CPO at a concentra-
tion of 10 µg/mL. The synthesis of proto- 10.1. Purification of PPO
porphyrin can be followed by coupling a
small portion of the incubation with pro- Perhaps the simplest recombinant
toporphyrinogen oxidase (PPO) (see Sec- source of this enzyme is the PPO from
tion 8). Alternatively, the conversion of Myxococcus xanthus, as described by Dailey
coproporphyrinogen to protoporphyrino- and Dailey (6). This is a PPO that uses
gen can be determined by analysis of the molecular oxygen as the terminal electron
oxidized methyl esters and quantified using acceptor and is a single subunit enzyme. In
an HPLC system (see Chapter 5). The eukaryotes, the enzyme is found on the
incubation should be performed at 37°C cytosolic side of the inner mitochondrial
under dim light. The enzyme can be membrane or associated with chloroplast
removed from the incubation mixture by membranes, while in bacteria, it is a
ultrafiltration through a PM-10 membrane peripheral membrane protein. The gene
attached to an ultrafiltration cell. As with corresponding to the M. xanthus PPO was
the other porphyrinogens, protopor- amplified and modified such that the N
phyrinogen should be used immediately. terminus encodes for a 6-histidine tag. The
The yield of protoporphyrinogen from construct was subsequently cloned into a
coproporphyrinogen is in excess of 95%. Tac-driven derivative of pBTac-1, yielding
the plasmid pMx-PPO (Table 1).

10. SYNTHESIS OF PROTO- ❖ Procedure 6. Purification of


PORPHYRIN Recombinant PPO from M. xanthus
The conversion of protoporphyrinogen 1. Bacterial growth: E. coli cells harboring
into protoporphyrin is mediated by the pMx-PPO are grown, and the harvest-
enzyme PPO. The enzyme catalyzes the ed cells are sonicated as described
removal of six electrons and six protons above for CPO overproduction.
from the porphyrinogen ring, thereby 2. Membrane preparation: The lysed cells
89
M.J. Warren and P.M. Shoolingin-Jordan

are centrifuged at 100 000× g, and the fluorescence at 635 nm after excitation at
supernatant discarded, then this mem- 405 nm (29). Coproporphyrin emits at
brane fraction is resuspended in 60 mL 610 nm, so it is important to make sure
of NaH2PO4, pH 7.4, 300 mM NaCl, that the auto-oxidation of copropor-
and 0.5% n-octyl-β-D-glucopyrano- phyrinogen is not being observed. The
side. The suspension is centrifuged increase in protoporphyrin fluorescence is
again at 100 000× g to separate the sol- measured over a 10-minute period, and the
ubilized enzyme from the remaining enzyme activity can be deduced with refer-
membranes. ence to a calibration curve for the fluores-
3. His-bind column: This is carried out as cence of a standard solution of protopor-
for CPO, except that PPO is eluted in phyrin. The yield of protoporphyrin from
buffer containing 150 mM imidazole. protoporphyrinogen is in excess of 95%.
The recombinant protein can be
detected by SDS-PAGE, migrating
with a molecular mass of about 50 000 11. SYNTHESIS OF PROTOHEME
Da. The purified protein is yellow in
color due to the presence of the flavin The final step in the synthesis of proto-
cofactor and has a characteristic flavo- heme is the insertion of ferrous iron in a
protein UV/VIS spectrum. reaction that is catalyzed by ferrochelatase.
4. Storage: The protein can be stored In eukaryotes, this enzyme is normally
frozen at -20°C for several months. peripherally associated with the inner mem-
Purified PPO is obtained in excess of brane of the mitochondrion. Quite surpris-
10 mg/L of culture. ingly, the human enzyme contains an iron
sulphur center, although no immediate role
10.2. Synthesis of Protoporphyrin and has been forwarded for its presence. Defects
Assay in the human enzyme are associated with
erythropoietic protoporphyria, a relatively
The synthesis of protoporphyrin can be severe form of porphyria that can cause
achieved either by use of a coupled enzyme severe liver damage (19). In B. subtilis, fer-
system from coproporphyrinogen III or by rochelatase exists as a soluble protein and
chemical reduction of protoporphyrin by represents one of the simplest sources of the
sodium amalgam. The use of a coupled enzyme (11). The increased solubility of the
enzyme system is perhaps more attractive Bacillus enzyme was a major expedient in
and will be discussed here. The incubation the crystallization of the enzyme (2).
is set up as described for coproporphyrino-
gen synthesis above. The incubation for the 11.1. Enzyme Purification
synthesis of protoporphyrinogen is under-
taken in 50 mM Tris-HCl, pH 8.0, con- The B. subtilis hemH is cloned into
taining 0.2% Tween 20, and 2.5 mM glu- pUC18 under control of the lac promoter
tathione. Coproporphyrinogen is added to to give plasmid pLUG18e2. When trans-
the incubation mixture at a final concen- formed into E. coli JM109 cells, the plas-
tration of 5 µM and incubated with CPO mid causes the bacteria to constitutively
at a concentration of 10 µg/mL and PPO overproduce the enzyme to a level of about
at 20 µg/mL. The synthesis of protopor- 10 mg/L of culture (Table 1). The strain
phyrin can be followed fluorometrically by harboring pLUG18e2 is somewhat unsta-
making a 1:10 dilution of the incubation ble, and fresh transformants need to be
mixture with buffer and determining the used for new cultures.
90
Enzymatic Preparation of Tetrapyrrole Intermediates

❖ Procedure 7. Purification of mL bed volume) and the column is


Recombinant Ferrochelatase from washed with one bed volume of buffer.
B. subtilis The ferrochelatase is eluted from the
column by application of a linear gra-
1. Bacterial growth: The strain is grown dient of 0 to 0.6 M NaCl in 20 mM
in LB media in 2-L flasks containing 1 Tris-HCl. The enzyme elutes at
L of media supplemented with ampi- approximately 0.3 M NaCl.
cillin at 100 µg/mL at 37°C with vig- 6. Gel filtration chromatography: Frac-
orous shaking. tions containing the enzyme are pooled
2. Harvesting: The cells are collected by and concentrated to approximately 5
centrifugation (10 000× g for 10 min), mL in an ultrafiltration unit fitted with
and the cell pellet is suspended in 25 a PM-10 membrane. The concentrated
mL of 30 mM Tris-HCl, pH 8.0, con- sample is then applied to a column of
taining 20% (wt/vol) sucrose, lysozyme Sephacryl S-100 HR (2.6 × 100 cm).
(0.25 mg/mL), and EDTA (15 mM). The ferrochelatase elutes from the col-
After incubation at 25°C for 30 min- umn as a single peak in a homogeneous
utes, the resulting spheroplasts are har- form.
vested by centrifugation at 7000× g for 7. Storage: The enzyme can be concen-
15 minutes. The pellet is resuspended trated and stored at -20°C for several
in 12 mL of 50 mM Tris-HCl, pH 7.4, months without loss of activity. The
containing 5 mM MgSO4. purified ferrochelatase is obtained in a
3. Sonication: The spheroplast suspension yield of about 0.5 mg/L of culture.
is sonicated as described in Procedure
1, and the lysate is centrifuged at 11.2. Incubation Protocol and Assay
48 000× g for 30 minutes at 4°C. The
pellet is discarded, and the supernatant Ferrochelatase activity is best monitored
is retained. by recording the disappearance of proto-
porphyrin (5,11). This can be monitored
4. Ammonium sulfate fractionation: The by a decrease in fluorescence as a divalent
supernatant is made 70% with respect metal ion (normally zinc in assays) is
to ammonium sulfate by the addition chelated into the porphyrin macrocycle.
of 44.2 g/100 mL of extract. After 45 The reaction is normally undertaken in a
minutes at 0°C, the solution is cen- 3-mL cuvette with a 2.5-mL standard reac-
trifuged at 10 000× g for 10 minutes at tion mixture consisting of: 100 mM Tris-
4°C. The pellet is discarded, and the HCl, pH 7.2, 0.3 mg/mL Tween 80, 100
supernatant is made 90% with respect µM ZnCl2, and 1 to 5 µg of purified fer-
to ammonium sulfate by the addition rochelatase. The reaction is normally start-
of a further 13.6 g/100 mL of extract. ed by the addition of 1.5 µM protopor-
After 45 minutes at 0°C, the solution phyrin, prepared as described below, to the
is centrifuged at 10 000× g for 10 min- incubation, and the reaction is monitored
utes at 4°C, and the 70%–90% pellet for up to 10 minutes. The excitation wave-
is kept and resuspended in 4 mL of 20 length is 407 nm, and the emission of fluo-
mM Tris-HCl, pH 7.4. rescence at 635 nm is recorded.
5. Anion exchange chromatography: Protoheme can be synthesized from pro-
After dialysis against 5 L of the same toporphyrin IX and ferrous iron using the
buffer, the enzyme fraction is applied following procedure. The incubation mix-
to a column of DEAE Sephacel (40- ture contains 100 mM Tris-HCl, pH 7.2,
91
M.J. Warren and P.M. Shoolingin-Jordan

protoporphyrin IX at 2 µM, 0.3 mg/mL 6.Dailey, H.A. and T.A. Dailey. 1996. Protoporphyrino-
Tween 80, 20 µM Fe2+, 6 mM DTT, 5 gen oxidase of Myxococcus xanthus. J. Biol. Chem.
271:8714-8718.
mM sodium dithionite, and 2 µg/mL fer- 7.Erskine, P.T., N. Senior, S. Awan, R. Lambert, G.
rochelatase. Fe2+ is prepared daily as a stock Lewis, I.J. Tickle, M. Sarwar, P. Spencer, P. Thomas,
solution of 50 mM (NH4)2Fe(SO4)2 in 0.3 M.J. Warren et al. 1997. X-ray structure of 5-amino-
laevulinic acid dehydratase, a hybrid aldolase. Nat.
M DTT. Protoporphyrin IX is prepared as Struct. Biol. 4:1025-1031.
a stock of 100 µM disodium protopor- 8.Erskine, P.T., E. Norton, J.B. Cooper, R. Lambert, A.
phyrin dissolved in water containing 15 Coker, G. Lewis, P. Spencer, M. Sarwar, S.P. Wood,
M.J. Warren, and P.M. Shoolingin-Jordan. 1999. X-
mg/mL Tween 80. The insertion of ferrous Ray structure of 5-aminolevulinic acid dehydratase
iron can also be followed spectrofluoromet- from Escherichia coli complexed with the inhibitor lev-
ulinic acid at 2.0 A resolution. Biochemistry 38:4266-
rically by measuring the rate of protopor- 4276.
phyrin disappearance, as described above. 9.Ferreira, G.C. and J. Gong. 1995. 5-Aminolaevuli-
The yield of protoheme is in excess of 90%. nate synthase and the first step of heme biosynthesis. J.
Bioenerg. Biomembr. 27:151-159.
10.Guo, G.G., M. Gu, and J.D. Etlinger. 1994. 240-kDa
proteasome inhibitor CF-2. is identical to delta-
ABBREVIATIONS aminolevulinic acid dehydratase. J. Biol. Chem.
269:12399-12402.
11.Hansson, M. and L. Hederstedt. 1994. Purification
ALA, 5-aminolevulinic acid; ALAS, 5- and characterisation of a water-soluble ferrochelatase
aminolevulinic acid synthase; PBG, por- from Bacillus subtilis. Eur. J. Biochem. 220:201-208.
12.Hunter, G.A. and G.C. Ferreira. 1995. A continuous
phobilinogen; PBGD, porphobilinogen spectrophotometric assay for 5-aminolevulinate syn-
deaminase; CPO, coproporphyrinogen thase that utilizes substrate cycling. Anal. Biochem.
oxidase; Da, Dalton molecular mass unit; 226:221-224.
13.Jaffe, E.K. 1995. Porphobilinogen synthase, the first
LB medium, Luria-Bertani medium; source of heme's asymmetry. J. Bioenerg. Biomembr.
PMSF, phenylmethanesulfonyl fluoride; 27:169-179.
PPO, protoporphyrinogen oxidase; SAM, 14.Jaffe, E.K. 2000. The porphobilinogen synthase family
of metalloenzymes. Acta Crystallogr. D 56:115-128.
S-adenosyl-L-methionine; SDS-PAGE, 15.Jordan, P.M., G. Burton, H. Nordlöv, M.M. Schnei-
sodium dodecyl sulfate polyacrylamide gel der, L. Pryde, and A.I. Scott. 1979. J. Chem. Soc.,
electrophoresis. Chem. Commun. 204-205.
16.Jordan, P.M. and M.J. Warren. 1987. Evidence for a
dipyrromethane cofactor at the catalytic site of E. coli
porphobilinogen deaminase. FEBS Lett. 225:87-92.
REFERENCES 17.Jordan, PM. 1991. The biosynthesis of 5-aminolae-
vulinic acid and its transformation into uropor-
1.Akhtar, M. and C. Jones. 1986. Preparation of stere- phyrinogen III, p. 1-66. In A. Neuberger and L.L.M.
ospecifically labelled porphobilinogens. Methods van Deenen (Eds.), and P.M. Jordan (Vol. Ed.), New
Enzymol. 123:375-383. Comprehensive Biochemistry, Vol. 19, Biosynthesis of
2.Al-Karadaghi, S., M. Hansson, S. Nikonov, B. Jons- Tetrapyrroles. Elsevier, Amsterdam.
son, and L. Hederstedt. 1997. Crystal structure of fer- 18.Kannangara, C.G., R.V. Andersen, B. Pontoppidan,
rochelatase: the terminal enzyme in heme biosynthesis. R. Willows, and D. von Wettstein. 1994. Enzymic
Structure 5:1501-1510. and mechanistic studies on the conversion of gluta-
3.Blanche, F., L. Debussche, D. Thibaut, J. Crouzet, mate to 5-aminolaevulinate, p. 3-25. In D.J. Chad-
and B. Cameron. 1989. Purification and characteriza- wick, and K. Ackrill (Eds.), The Biosynthesis of
tion of S-adenosyl-L-methionine:uroporphyrinogen Tetrapyrrole Pigments, Ciba Foundation Symposium
methyltransferase from Pseudomonas denitrificans. J. 180. John Wiley & Sons, New York.
Bacteriol. 171:4222-4231. 19.Kappas, A., S. Sassa, R.A. Galbraith, and Y. Nord-
4.Bolt, E.L., L. Kryszak, J. Zeilstra-Ryalls, P.M. mann. 1995. The porphyrias, p. 2103-2160. In C.R.
Shoolingin-Jordan, and M.J. Warren. 1999. Char- Scriver, A.L. Beaudet, W.S. Sly, and D. Valle (Eds.),
acterisation of the R. sphaeroides 5-aminolevulinic The Metabolic and Molecular Basis of Inherited Dis-
acid synthase isoenzymes, HemA and HemT, isolated ease, 7th ed. McGraw Hill, New York.
from recombinant Escherichia coli. Eur. J. Biochem. 20.Li, J.M., C.S. Russell, and S.D. Cosloy. 1989. The
265:1-11. structure of the E. coli hemB gene. Gene 75:177-184.
5.Dailey, H.A. 1977. Purification and characterisation of 21.Mauzerall, D. and S. Granick. 1956. The occurrence
the membrane bound ferrochelatase from Spirillum and determination of δ-aminolevulinic acid and por-
itersonii. J. Bacteriol. 132:302-307. phobilinogen in urine. J. Biol. Chem. 219:435-446.

92
Enzymatic Preparation of Tetrapyrrole Intermediates

22.Medlock, A.E. and H.A. Dailey. 1996. Human proto- Nakanishi, and O. Meth-Cohn (Eds.), and J.W. Kelly
porphyrinogen oxidase is not a metalloprotein. J. Biol. (Vol. Ed.), Comprehensive Natural Products Chem-
Chem. 271:32507-32510. istry, Vol. 4, Amino Acids, Peptides, Porphyrins and
23.Neidle, E.L. and S. Kaplan. 1993. Expression of Alkaloids. Elsevier, Amsterdam.
Rhodobacter sphaeroides hemA and hemT genes encod- 29.Smith, A.G. and W.T. Griffiths. 1993. Enzymes of
ing two 5-aminolaevulinic acid synthase isoenzymes. J. chlorophyll and heme biosynthesis. Methods Plant
Bacteriol. 175:2292-2303. Biochem. 9:299-343.
24.Phillips, J., F.G. Whitby, J.P. Kushner, and C.P. Hill. 30.Spencer, J.B., N.J. Stolowich, C.A. Roessner, and A.I.
1997. Characterisation and crystallization of human Scott. 1993. The Escherichia coli cysG gene encodes the
uroporphyrinogen decarboxylase. Prot. Sci. 6:1343- multifunctional protein, siroheme synthase. FEBS
1346. Lett. 335:57-60.
25.Raux, E., T. McVeigh, S.E. Peters, T. Leustek, and 31.Spencer, P. and P.M. Jordan. 1993. Purification and
M.J. Warren. 1999. The role of Saccharomyces cerevisi- characterisation of 5-aminolaevulinic acid dehydratase
ae Met1p and Met8p in siroheme and cobalamin from E. coli and a study of reactive thiols at the metal
biosynthesis. Biochem. J. 338:701-708. binding domain. Biochem. J. 290:279-287.
26.Scopes, R.K. 1987. Protein Purification, Principles and 32.Thomas, S.D. and P.M. Jordan. 1986. Nucleotide
Practice, 2nd ed. Springer Verlag, Basel. sequence of the hemC locus encoding porphobilinogen
27.Shoolingin-Jordan, P.M., J.E. LeLean, and A.J. Lloyd. deaminase of Escherichia coli K12. Nucleic Acids Res.
1997. Continuous coupled assay for 5-aminolevulinate 14:6215-6226.
synthase. Methods Enzymol. 281:309-316. 33.Whitby, F.G., J.D. Phillips, J.P. Kushner, and C.P.
28.Shoolingin-Jordan, P.M. and K.-M. Cheung. 1999. Hill. 1998. Crystal structure of human uropor-
Biosynthesis of heme, p. 61-107. In D.H.R. Barton, K. phyrinogen decarboxylase. EMBO J. 17:2463-2471.

93
Analysis of Biosynthetic Intermediates,
5 5-Aminolevulinic Acid to Heme

Chang Kee Lim


MRC Bioanalytical Science Group, School of Biological and Chemical
Sciences, Birkbeck College, University of London, London, England, UK

1. INTRODUCTION matography, converted into the p-dimeth-


ylamino-benzaldehyde derivatives, and
Chromatographic techniques are widely then determined spectrophotometrically at
used for the analysis of heme and its pre- 553 nm (16). The procedures, widely
cursors. Recent and continuing improve- described in textbooks, are also available,
ments in column packing materials for with technical instructions, from ion
high-performance liquid chromatography exchange resins suppliers, e.g., Bio-Rad
(HPLC) have led to much better column Laboratories (Hercules, CA, USA). The
efficiency and resolution. There have also method is recommended for the routine
been great advances in the direct coupling qualitative and quantitative measurement
of liquid chromatography (LC), including of ALA and PBG.
capillary electrophoresis (CE), to mass ALA and PBG have been separated by
spectrometry (MS) to provide highly sensi- HPLC (11) and micellar electrokinetic capi-
tive and specific methods of analysis. llary chromatography (13). They were detec-
The separation and detection of the bio- ted with a UV detector at 220 to 240 nm.
synthetic intermediates from 5-aminolevu- A simple CE method has been developed
linic acid (ALA) to heme are described in for the separation of PBG. The compound
detail in this chapter. The emphasis is in was effectively separated on a 75-cm fused-
HPLC and CE, and the well-established silica capillary (75 µm inner diameter) with
thin-layer chromatography will not be 50 mM ammonium acetate buffer (pH
included. 5.16 adjusted with acetic acid) as the run-
ning buffer and 20 kV and 30°C as the run-
ning voltage and temperature, respectively.
2. 5-AMINOLEVULINIC ACID AND PBG was detected at 220 nm with a detec-
PORPHOBILINOGEN tion limit of 1 µg/mL. Under the CE condi-
tions, the charged PBG molecule could also
ALA and porphobilinogen (PBG) are be detected at 400 to 420 nm, although the
usually separated by ion exchange chro- detection was less sensitive than at 220 nm.

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

95
C.K. Lim

The above CE method has been modi- applications where high sensitivity and
fied by inclusion of 10% (vol/vol) acetoni- specificity are required.
trile in the running buffer (50 mM ammo-
nium acetate, pH 5.20) and coupled on-line
to electrospray ionization mass spectrome- 3. ANALYSIS OF PORPHYRINS
try (ESIMS) to provide an extremely sensi-
tive and specific analytical method for ALA The naturally occurring porphyrins exist
and PBG (12). The detection limits were in complex mixtures including isomeric
estimated to be 100 and 10 amol of ALA forms. Effective analysis, therefore, requires
and PBG on column, respectively. The sen- high resolution coupled with sensitive
sitivity could be further improved by the use detection. To date, the best technique for
of selected ion recording (SIR) scans or the separation of porphyrins and their iso-
nanospray ionization, or both. mers is HPLC. The resolution achieved by
Figure 1 shows the separation and detec- HPLC has not been reproduced by other
tion of ALA and PBG by CE-ESIMS. The separation methods.
protonated ion of ALA is at m/z 132 and
that of PBG is at m/z 227. However, the 3.1. Extraction of Porphyrins from
protonated PBG was found to lose ammo- Biological Materials for HPLC
nia (NH3) easily in the electrospray source Analysis
to give an intense ion at m/z 210, corre-
sponding to a methylenepyrrolenine ion. Sample preparation is an important and
PBG was, therefore, detected at m/z 210 integrated part of the successful application
for the methylenepyrrolenine ion and mul- of HPLC to the analysis of porphyrins in
tiple reaction monitoring (MRM) acquisi- biological materials. A good sample prepara-
tions could be used for PBG by monitor- tion procedure minimizes quantitative errors
ing the transition from m/z 227 to m/z and places less demand on the chromatogra-
210. This method is recommended for phy, allowing faster and better analysis.

Figure 1. CE-ESIMS of ALA and PBG. Capillary, 70 cm × 75 µm i.d.; running buffer, 50 mM ammonium acetate, pH 5.2:ace-
tonitrile (90:10, vol/vol); running voltage, 20 kV; ESI voltage, 3.5 kV.

96
Analysis of Heme and Its Precursors

It is recommended that, whenever possi- insoluble material at the interface, and


ble, porphyrins should be extracted and a lower layer of aqueous acid.
analyzed as the free acids. The separation of 6. Discard the ether layer that contains
porphyrin free acids is superior to that of the unwanted carotenoid pigments and
the corresponding methyl esters (7). chlorophyll derivatives.
Methyl esterification of porphyrins may 7. Record the volume (usually 4.5 mL) of
also cause structural modification of the the aqueous acid layer, which contains
parent compounds. The deconjugation the extracted porphyrins, and transfer
and transmethylation of protoporphyrin about 2 mL into a clean tube using a
glycoconjugates following esterification Pasteur pipet.
and extraction of porphyrins from rat 8. Filter the solution, e.g., through a
Harderian gland is a typical example. The syringe filter assembly, to remove any
procedure led to the incorrect identifica- particulate material. The solution may
tion of protoporphyrin glycoconjugates as be used for HPLC or spectrophoto-
the unconjugated protoporphyrin (9). metric analysis.
3.1.1. Preparation of Urine Samples The above procedure should be carried
out in subdued light, e.g., red safety light,
Fresh urine (200 to 500 µL) may be in order to minimize undue alteration to
injected after centrifugation into the HPLC light sensitive porphyrins, especially proto-
for analysis. Sediments or precipitates are porphyrin.
often seen in stored urine, and these may
adsorb porphyrins. The urine (1 mL) 3.1.3. Extraction of Plasma and Red Cell
should be thoroughly mixed with concen- Porphyrins
trated HCl (40 µL) to dissolve the precipi-
tated material before HPLC separation. For the extraction of porphyrins in plas-
ma (19), the sample (0.5 mL) is vortex
3.1.2. Extraction of Porphyrins from mixed with 5 mL of ether:acetic acid (4:1,
Feces vol/vol) followed by centrifugation to
remove the precipitated protein. The
The following procedure (19), which supernatant is then vortex mixed with 3
provides a relatively clean extract, is recom- mL of 2.7 M HCl. The lower aqueous acid
mended. layer is used for HPLC analysis.
Plasma porphyrins may also be extracted
❖ Procedure 1. Extraction of Porphyrins by vortex mixing 100 µL of sample with
from Feces 200 µL of acetonitrile:dimethyl sulfoxide
1. Weigh about 50 mg of feces into a 15- (DMSO) (4:1, vol/vol). The supernatant
mL graduated centrifuge tube. after centrifugation is used for HPLC sepa-
2. Add 1 mL of concentrated HCl and ration. This method, also suitable for the
vortex mix for 1 minute or until the extraction of red cell porphyrins, is recom-
particles disintegrate. mended for rapid porphyrin profile analy-
3. Add 3 mL of diethyl ether (peroxide- sis by HPLC.
free) and vortex mix for 1 minute.
3.1.4. Extraction of Porphyrins from
4. Add 3 mL of water and vortex mix for Tissues
1 minute.
5. Centrifuge at 2000× g for 10 minutes Porphyrins in tissues can be effectively
to give an upper ether layer, a pad of extracted by homogenizing the sample in
97
C.K. Lim

acetonitrile-DMSO (4:1, vol/vol), using 3.2.1. Uroporphyrin I, II, III, and IV


1 mL of homogenizing medium per 100 Isomers
mg of tissues. Repeated extraction may
be necessary for complete recovery. The Uroporphyrin I and III isomers can be
supernatant after centrifugation is thor- rapidly and effectively separated by isocrat-
oughly mixed with 2 volumes of water ic reversed-phase (RP)-HPLC on octade-
or HPLC aqueous phase buffer before cylsilyl (ODS) C18 columns with 13%
separation. Injection of the organic (vol/vol) acetonitrile in 1 M ammonium
extract without suitable adjustment of acetate buffer, pH 5.16 (adjusted with
the aqueous content resulted in peak acetic acid), as eluent (Figure 2). Optimiza-
distortion. tion studies have shown that the molar
concentration and pH of ammonium
3.2. Separation of Porphyrin Isomers acetate buffer significantly affected the
retention and resolution of uroporphyrin
The separation of isomers, particularly isomers (18). The optimum buffer concen-
the type I and type III isomers, is impor- tration was 1 M, and the best pH range for
tant for the differential diagnosis of certain chromatography on a conventional ODS
porphyrias. For example, the copropor- column was between 5.10 and 5.20.
phyrin excreted in the urine and feces of For separation on a base-deactivated
patients with congenital erythropoietic (BDS) C18 column, the optimum pH was
porphyria (CEP) is type I, while in heredi- 5.55 (Figure 2), although 5.16 was also suit-
tary coproporphyria it is type III. able. BDS C18 columns are columns with

Figure 2. Separation of uroporphyrin I and III isomers. (a) On Hypersil-BDS C18 with acetonitrile:1 M ammonium acetate, pH
5.16 (9:91, vol/vol), as eluent; (b) on Hypersil-ODS with acetontrile:1 M ammonium acetate, pH 5.16 (13:87, vol/vol), as eluent;
and (c) on Hypersil-BDS C18 with acetonitrile:1 M ammonium acetate, pH 5.55 (9:91, vol/vol), as eluent. Flow-rate, 1 mL/minute.

98
Analysis of Heme and Its Precursors

fewer residual silanol groups through boxylic acid porphyrin could not be com-
exhaustive end-capping or made by different pletely separated by RP-HPLC, although
bonding procedures to those normally used the type I isomer easily resolved from the
for conventional ODS columns. Residual type III isomers either with 15% (vol/vol)
silanol groups on silica-based C18 columns acetonitrile in 1 M ammonium acetate
interact adversely with basic compounds, buffer, pH 5.16, as eluent on a convention-
causing peak tailing or broadening. In gen- al C18 column or with 28% acetonitrile:
eral, BDS C18 columns give better resolu- methanol (10:90) in 1 M ammonium
tion and faster separation for porphyrins acetate buffer, pH 5.55, as eluent on a
than conventional C18 columns. BDS C18 column. The four type III iso-
Methanol should not be used as the mers were resolved into three peaks in the
organic modifier for the separation of uro- elution order of 7c, 7d, and 7a + 7b (7),
porphyrins, especially when isocratic elution with 28% acetonitrile:methanol (10:90) in
is used. It causes severe peak tailing and 1 M ammonium acetate buffer, pH 5.16,
excessive retention with loss of resolution. as eluent. The letters a, b, c, and d are used
Methanol is a hydrogen-bonding organic throughout this chapter to denote the posi-
modifier. A layer of methanol adsorbed onto tions of methyl groups on rings A, B, C,
the hydrophobic hydrocarbonaceous C18 and D, respectively.
stationary phase surface can form extensive
hydrogen bonds with the 8 carboxylic acid 3.2.3. Type I and Type III
groups of uroporphyrin. The result is long Hexacarboxylic Acid Porphyrins
retention and peak tailing. This phenome-
non is not observed for porphyrins with one There are two type I and six type III
or more methyl groups, since the interaction hexacarboxylic acid porphyrin isomers.
is dominated by hydrophobic interaction The two type I isomers (6Iab and 6Iac)
between the hydrophobic methyl group(s) have been separated from the most abun-
and the stationary phase surface. dant type III isomer (6IIIad) by isocratic
A small proportion (e.g., 10%) of ace- RP-HPLC with 16% (vol/vol) acetonitrile
tonitrile can be added to methanol, and the in 1 M ammonium acetate buffer, pH
mixture (10% acetonitrile and 90% 5.16, as eluent on a Hypersil-ODS column
methanol) can be used as the organic mod- (ThermoQuest, Bellafonte, PA, USA). The
ifier. The more hydrophobic acetonitrile, complete separation of all 8 isomers has
which is also a nonhydrogen bonding not been achieved. Using the above system,
organic modifier, will be preferentially 6IIIac coeluted with 6IIIbd, and 6IIIab
adsorbed onto the stationary phase surface, coeluted with 6IIIbc (7).
thus preventing hydrogen bond formation.
The complete separation of uroporphyrin 3.2.4. Type I and Type III
I, II, III, and IV isomers has not been Pentacarboxylic Acid Porphyrins
achieved. They were resolved into three
peaks in the elution order of I, III + IV, and There are four type III and one type I
II (7,18). The resolution was not improved pentacarboxylic acid porphyrin isomers.
by employing a BDS C18 column. These 5 isomers have been separated by RP-
HPLC on a Hypersil-ODS column with
3.2.2. Type I and Type III 45% (vol/vol) acetonitrile:methanol (10:90)
Heptacarboxylic Acid Porphyrins in 1 M ammonium acetate buffer, pH 5.16,
as eluent (Figure 3a). The elution order was
The four type III isomers of heptacar- 5I, 5bcd, 5abc, 5acd, and 5abd. A reversal
99
C.K. Lim

of elution order between 5I and 5abd was 3.2.6. Protoporphyrin, Heme, and
observed when 19% acetonitrile in 1 M Related Compounds
ammonium acetate, pH 5.16, was used as
the mobile phase (Figure 3b). The presence Dicarboxylic acid porphyrins, heme, and
of methanol in the mobile phase resulted in related compounds are much more hydro-
an overall improvement in resolution. phobic than the other porphyrins described
above. They require a much higher propor-
3.2.5. Coproporphyrin I, II, III, and IV tion of organic modifier for elution. Since
Isomers acetonitrile is immiscible with 1 M ammo-
nium acetate above the proportion of about
Coproporphyrin isomers are easily sepa- 35%, it cannot be used as the sole organic
rated by RP-HPLC (7). The separations of modifier for the separation of this group of
the 4 isomers on a Hypersil-ODS and a compounds. Either a mixture of acetoni-
Hypersil-BDS C18 column are shown in trile:methanol (10:90) or methanol alone
Figure 4 (a and b). Better resolution with can be used instead. Methanol is completely
faster elution times was achieved on the miscible with 1 M ammonium acetate.
Hypersil-BDS C18 column. The Hypersil- The separation of dicarboxylic por-
BDS C18 column also required less ace- phyrins and metalloporphyrins by RP-
tonitrile (23%) for elution than the Hyper- HPLC has been described (10). A typical
sil-ODS column (30%), which is an separation of protoporphyrin and heme-
obvious advantage. related compounds on a Hypersil-ODS

Figure 3. Separation of type I and type III pentacarboxylic acid porphyrin isomers. Column, Hypersil-ODS; eluent (a), 45%
acetonitrile:methanol (10:90, vol/vol) in 1 M ammonium acetate, pH 5.16; eluent (b), 19% (vol/vol) acetonitrile in 1 M ammo-
nium acetate, pH 5.16. The letters a, b, c, and d denote the positions of methyl groups on rings A, B, C, and D, respectively.

100
Analysis of Heme and Its Precursors

column with 86% (vol/vol) methanol in 1 ammonium acetate, pH 5.16, has been
M ammonium acetate buffer, pH 5.16, as described for the separation of type I and
eluent is shown in Figure 5. type III isomers of 8-, 7-, 6-, 5-, and 4-car-
boxylated porphyrins (7). The system is
3.2.7. Separation of Porphyrin Mixtures applicable to analysis where the separation
from Uroporphyrin to of dicarboxylated porphyrins is not
Protoporphyrin required, e.g., urinary porphyrins. The elu-
tion of dicarboxylated porphyrins requires
From uroporphyrin to protoporphyrin, an acetonitrile content higher than its mis-
the compounds differ widely in hydropho- cibility with 1 M ammonium acetate. It
bicity. Gradient elution is therefore essen- should be emphasized that using this gra-
tial for the simultaneous separation of these dient system the acetonitrile content
porphyrins, including their isomers. should not be allowed to exceed 35%, and
A 15-minute linear gradient elution the column must not be washed with ace-
from 13% to 30% acetonitrile in 1 M tonitrile at the end of the separation due to

Figure 4. Separation of copropor-


phyrin I, II, III, and IV isomers.
(a) On Hypersil-ODS with 30%
(vol/vol) acetonitrile in 1 M
ammonium acetate, pH 5.16, as
eluent; (b) on Hypersil-BDS C18
with 23% (vol/vol) acetonitrile in
1 M ammonium acetate, pH 5.16,
as eluent. Flow-rate, 1 mL/minute.

101
C.K. Lim

the immiscibility problem. It is also impor- um acetate buffer, pH 5.16; solvent B, 10%
tant to remember that whenever acetoni- (vol/vol) acetonitrile in methanol. Various
trile and 1 M ammonium acetate is used elution programs can be used, depending
for elution, as in the separation of the indi- on the applications, with these two solvent
vidual group of porphyrin isomers by iso- mixtures for porphyrin separation (15). The
cratic elution, the column should not be pH of the buffer, 5.16, is optimal for the
washed with acetonitrile before removal of separation of porphyrin mixtures.
ammonium acetate with a solvent in which Figure 7 shows the separation of por-
it is completely miscible. The column may phyrins in the feces and urine of a patient
be washed with 90% methanol or acetoni- with porphyria cutanea tarda (PCT) on a
trile:methanol in water. C18-bonded RP column (Hypersil-ODS).
Porphyrin mixtures including protopor- It clearly demonstrates the flexibility and
phyrin are best separated by gradient elu- applicability of the system.
tion RP-HPLC with (1,7,17) or without
ion-pairing agents (7). Columns of silica
gel chemically bonded with different
hydrocarbon chain lengths, from C1 to
C18, have all been successfully used for the
RP-HPLC separation of porphyrin mix-
tures in biological materials (7,8). With the
increasing use of on-line HPLC-MS in
analysis, including the tetrapyrroles, gradi-
ent elution solvent mixtures that are fully
compatible with MS are necessary. This
rules out systems that use involatile inor-
ganic phosphate in separation.
A simple RP-HPLC system with 0.1%
trifluroroacetic acid (solvent A) and ace-
tonitrile (solvent B) as the gradient elution
solvent mixture has been used for the sepa-
ration of porphyrins. The system, fully
compatible with MS, is able to resolve the
type I and III isomers of 6-, 5-, and 4-car-
boxylated porphyrins. Separation of uro-
and heptacarboxylic acid porphyrin iso-
mers, however, was not achieved (Figure
6). The system is best used for the separa-
tion of porphyrins with fewer numbers of
carboxylic acid groups, including the dicar-
boxylic acid porphyrins.
The ammonium acetate buffer system
that is fully compatible with MS and pro-
vides high efficiency separation of por-
phyrins is the buffer of choice. It is recom- Figure 5. Separation of protoporphyrin and metallopor-
mended that the following gradient phyrins. Column, Hypersil-ODS; eluent, methanol:1 M
ammonium acetate, pH 5.16 (86:14, vol/vol); flow rate, 1
mixtures are used for elution: solvent A, mL/minute. Peaks: 1 = Zn-deuteroporphyrin; 2 = heme; 3 =
10% (vol/vol) acetonitrile in 1 M ammoni- Zn-protoporphyrin; 4 = protoporphyrin.

102
Analysis of Heme and Its Precursors

4. RETENTION MECHANISM OF (6COOH, 2CH3), pentacarboxylic acid


PORPHYRINS AND METALLO- porphyrin (5COOH, 3CH3), copropor-
PORPHYRINS IN RP-HPLC phyrin (4COOH, 4CH3), mesoporphyrin
(2COOH, 4CH3, 2CH2CH3), and proto-
Understanding the retention behavior porphyrin (2COOH, 4CH3, 2CH=CH2)
and mechanism is useful in the prediction was observed.
and elucidation of the possible nature of The hydrophobic interaction mecha-
substituent groups present in unknown nism also explains the elution order of por-
porphyrins. phyrin isomers. The elution in the order of
The most dominant mechanism of I, III, IV, and II, for coproporphyrin iso-
retention in RP-HPLC is hydrophobic mers is an example. Coproporphyrin II has
interaction. In porphyrins, this is between two pairs of adjacent CH3 groups at posi-
the side-chain substituents and the tions 1 and 8 and 4 and 5, respectively,
hydrophobic hydrocarbonaceous (ODS) using Fischer’s numbering system. These
stationary phase surface. The hydrophobic- provide the largest hydrophobic surface
ity of the porphyrin side-chain substituents areas available for interaction. It is therefore
increases in the order: the longest retaining isomer. The symmetri-
cal coproporphyrin I has no adjacent CH3
CH2COOH<CH2CH2COOH<CH3<CH2CH3<CH=CH2
groups and, thus, has the smallest hydro-
The number of the most hydrophobic phobic surface areas available for interac-
substituents available for interaction, there- tion. It is, therefore, the fastest eluting iso-
fore, determines the relative retention of mer. Coproporphyrin III and IV isomers
the porphyrin. This is dominated by the each have a pair of adjacent CH3 groups, at
number of alkyl (especially methyl) groups. positions 1 and 8 and 2 and 3, respectively.
Thus, the elution of porphyrins in the In this situation, the relative distance
order of uroporphyrin (8COOH), hep- between the adjacent CH3 pair and the
tacarboxylic acid porphyrin (7COOH, remaining two nonadjacent CH3 groups
1CH3), hexacarboxylic acid porphyrin becomes an important factor in determin-

Figure 6. Separation of a
standard mixture of por-
phyrins. Column, Hyper-
sil-ODS (25 cm × 4.6 mm
i.d., 5 µm particle size); sol-
vents, 0.1% trifluoroacetic
acid (solvent A) and ace-
tonitrile (solvent B); elu-
tion, linear gradient from
25% solvent B (75% sol-
vent A) to 50% solvent B in
30 minutes; flow rate, 1
mL/minute; detection, ab-
sorbance 404 nm. Peaks: 4,
5, 6, 7, and 8 refer, respec-
tively, to tetra(copro)-,
penta-, hexa-, hepta-, and
octa(uro)carboxylic acid
porphyrin; I and III denote
type I and type III isomers.

103
C.K. Lim

ing the relative hydrophobicity. In copro- The type III heptacarboxylic acid por-
porphyrin III, these are 5 (from position 1 phyrin isomers each have only a single
to 3) and 6 (from position 8 to 5) bond CH3 group that dominated the hydropho-
lengths apart, respectively. In copropor- bic interaction. These isomers are, there-
phyrin IV, each of the adjacent CH3 groups fore, very similar in hydrophobicity and
is 5 bond lengths away (from positions 2 to difficult to separate.
8 and 3 to 5) from their nearest nonadja- In metalloporphyrins, hydrophobic
cent CH3 group. The slightly shorter dis- interaction between the side-chain sub-
tance (one bond length) between one of the stituents and stationary phase surface is
adjacent pair CH3 groups and its nearest also an important retention mechanism.
nonadjacent CH3 group (5 instead of 6 However, the insertion of a metal ion,
bonds apart) is sufficient to make the type which completely occupies the center of
IV isomer slightly more hydrophobic and, the porphyrin hole, significantly altered the
therefore, be retained longer than the type electronic environment around the central
III isomers. Similar arguments apply to the N atoms. The retention of metallopor-
separation of the penta- and hexacarboxylic phyrins is also dependent on the ability,
acid porphyrin isomers. and therefore the species, of the inserted

Figure 7. Separation of porphyrins


in (a) the fecal extract and (b) the
urine of a patient with PCT. Col-
umn, Hypersil-ODS (250 × 4.6
mm, 5 µm particle size); solvent A,
10% acetonitrile in 1 M ammoni-
um acetate buffer (pH 5.16); sol-
vent B, 10% acetonitrile in
methanol; elution, linear gradient
at 1 mL/minute from 10% to 90%
solvent B in 30 minutes, followed
by isocratic elution at 90% solvent
B for a further 10 minutes; detec-
tion, 404 nm. Peaks; 4, 5, 6, 7, and
8 refer, respectively, to tetra(copro),
penta-, hexa-, hepta-, and octa
(uro)carboxylic acid porphyrin; 4-
Iso = isocoproporphyrin; pp = pro-
toporphyrin.

104
Analysis of Heme and Its Precursors

metal ion to accept axial ligands from the phyrin methyl esters on an ODS column
mobile phase. Addition of polar axial lig- by gradient elution from 70% (vol/vol)
ands leads to a decrease in the overall acetonitrile in water to 100% acetonitrile
hydrophobicity of a molecule and, conse- in 30 minutes is shown in Figure 8. The
quently, its retention (10). system is also fully compatible with MS.

5. SEPARATION OF PORPHYRIN 6. SEPARATION OF


METHYL ESTERS PORPHYRINOGENS

In some applications, it is more conve- The porphyrinogens are the true inter-
nient to separate porphyrins as their methyl mediates in the biosynthesis of heme. Por-
ester derivatives. The majority of methods phyrinogens are unstable to oxidation by
reported for the separation of porphyrin air, especially under acidic conditions and
methyl esters are by normal phase (adsorp- in the presence of light, to the correspond-
tion) chromatography on silica gel columns ing porphyrins, and are therefore rarely
(7). RP-HPLC, however, provides better analyzed. However, studies have shown
resolution and, although unable to separate that for isomer separation, the porphyrino-
the type I and III isomers of uro- and hep- gens are usually better resolved than the
tacarboxylic acid porphyrin methyl esters, is porphyrins (3–6). Isomerically pure por-
able to simultaneously separate the type I phyrinogens are needed as substrates for
and III isomers of hexacarboxylic acid, pen- enzymic experiments, and their separation
tacarboxylic acid, and coproporphyrin may also have application in situations
methyl esters in a single gradient elution where the complete resolution of isomers is
run. Furthermore, the polar hydroxylated essential. For example, investigation of the
porphyrins, e.g., hydroxyuroporphyrin, preferred order of uroporphyrinogen decar-
which are difficult to elute from a silica gel boxylation requires the complete separa-
column, are easily eluted from a RP column. tion of all four type III heptacarboxylic
The separation of a mixture of por- porphyrinogen isomers (14).

Figure 8. RP-HPLC of porphyrin


methyl esters. Column, Hypersil-
ODS (250 × 4.6 mm, 5 µm parti-
cle size); eluent, linear gradient
elution from 70% acetonitrile in
water to 100% acetonitrile in 30
minutes; flow-rate, 1 mL/minute.
Peaks; 1, 2, and 3 = hydroxylated
porphyrins; 4, 5, 6, 7, and 8 refer,
respectively, to tetra(copro)-,
penta-, hexa-, hepta-, and octa-
(uro)carboxyl porphyrin; I and III
denote type I and type III isomers;
mp = mesoporphyrin; pp = proto-
porphyrin.

105
C.K. Lim

Porphyrinogens are easily prepared by re- ammonium acetate buffer, pH 5.16 (8:12:
duction of porphyrins, usually with 3% (wt/ 80, by vol) as mobile phase (4,7). The
wt) sodium amalgam, as described below. superior resolution of porphyrinogen iso-
mers is again demonstrated.
❖ Procedure 2. Preparation of The complete separation of the five type
Porphyrinogens by Reduction I and III pentacarboxylic acid porphyrino-
of Porphyrins gen isomers was achieved on a Hypersil-
ODS column with methanol:1 M ammo-
1. Dissolve the porphyrin in 0.01 M nium acetate buffer, pH 5.16 (40:60,
KOH in a tube. Cover the tube with vol/vol), as eluent (3,7).
foil to prevent exposure to light. The type I, II, III, and IV isomers of
2. Flush the tube and solution with nitro- coproporphyrinogen could be rapidly sepa-
gen and add excess 3% sodium amal- rated on an ODS column with acetoni-
gam. trile:1 M ammonium acetate, pH 5.16
3. Shake or vortex mix the stoppered tube (25:75, vol/vol) as mobile phase. With this
vigorously until no red fluorescence is system, the four coproporphyrinogen iso-
detected under UV light. mers could also be simultaneously separated
4. Transfer the porphyrinogen solution from the four coproporphyrin isomers (5).
immediately into a clean tube contain- Protoporphyrinogen was easily separat-
ing a small volume of 0.1 M EDTA, ed from protoporphyrin by RP-HPLC
flush with nitrogen, and keep the stop- with methanol:1 M ammonium acetate
pered tube on ice in the dark until buffer, pH 5.16 (90:10, vol/vol), as eluent.
analysis. The porphyrinogens are stable The separation of these two compounds
for at least 1 hour under these condi- may be useful for studying their intercon-
tions. vertion either chemically or enzymatically.
Uroporphyrinogen I and III isomers
have been separated on an ODS column
with 6% (vol/vol) acetonitrile in 1 M 7. RETENTION BEHAVIOR OF
ammonium acetate buffer, pH 5.16, as elu- PORPHYRINOGENS IN RP-HPLC
ent. The elution order was III before I,
which is the reversed of that observed for Although hydrophobic interaction is
the corresponding porphyrins. The por- undoubtedly still the mechanism govern-
phyrinogens are also more hydrophilic ing the retention of porphyrinogens in RP-
than the porphyrins, requiring less organic HPLC, the reversal in the elution order
modifier for elution. observed for many of the porphyrinogen
The four type III heptacarboxylic acid isomers in comparison to porphyrins
porphyrin isomers, which could not be requires explanation. For example, while
separated by RP-HPLC, were completely coproporphyrin isomers were eluted in the
resolved as the porphyrinogens with ace- order I, III, IV, and II based on the
tonitrile:methanol:1 M ammonium acetate hydrophobic interaction hypothesis, the
buffer, pH 5.16 (7:3:90, by vol), as eluent corresponding coproporphyrinogen iso-
(Figure 9). The elution order was 7a, 7c, mers were eluted in the order of I, II, III,
7b, and 7d, which is also different from and IV, an apparent contradiction to the
that of the porphyrins (6,14). hydrophobic interaction hypothesis. How-
The six type III hexacarboxylic acid por- ever, reduction of the methine bridges of
phyrinogen isomers have been separated by the rigid porphyrin macrocycle resulted in
RP-HPLC with acetonitrile:methanol:1 M a relatively flexible porphyrinogen mole-
106
Analysis of Heme and Its Precursors

cule, which is able to adopt various confor- to 415 nm and 600 to 620 nm, respective-
mations. In the flexible coproporphyrino- ly, provides a highly sensitive and specific
gen molecules, the small CH3 group in method of detection. Heme is a nonfluo-
each isomer may be subjected to varying rescence compound and cannot be detect-
degrees of steric hindrance or shielding by ed fluorimetrically.
the larger propionic acid groups, depend- The porphyrinogens do not fluoresce
ing on the adopted conformation. This and have only relatively weak UV absorp-
alters the expected hydrophobic surface tion at the 220 to 240 nm region. They
areas available for interaction, which results can be detected with a UV detector set at
in a change in elution order. Since the con- 220 nm, but are best detected electrochem-
formations of the various groups of por- ically with an amperometric or coulomet-
phyrinogens under RP-HPLC conditions ric detector because of their ease of oxida-
are unknown, it makes prediction of the tion. Porphyrins and metalloporphyrins are
elution order difficult. also electro-active and can be detected with
an electrochemical detector.
In terms of sensitivity, specificity, and
8. HPLC DETECTORS FOR ability to positively identify and characterize
PORPHYRINS, METALLO- compounds, the best “detector” currently
PORPHYRINS, AND available is the mass spectrometer. On-line
PORPHYRINOGENS HPLC-ESIMS and tandem mass spectrom-
etry (MS/MS) provide unsurpassed speci-
Porphyrins and metalloporphyrins have ficity for the analysis of tetrapyrroles.
an intense absorption band at the 400 nm The methyl esters of porphyrins are usu-
region (Soret band). Detection at the Soret ally used in MS analysis because they ionize
band region with an UV-visible detector better in the ion source than free acid por-
set at 400 to 405 nm allows the simultane- phyrins. Porphyrins with a higher number
ous detection of all porphyrins and metal- of carboxylic acid groups, e.g., uro- and hep-
loporphyrins. tacarboxylic acids porphyrins, are more dif-
Porphyrins also have intense red fluores- ficult to ionize than those with a lesser num-
cence, and a fluorescence detector set at ber of carboxylic acid groups, such as copro-
excitation and emission wavelengths of 400 and protoporphyrins. With the recent devel-

Figure 9. Separation of heptacar-


boxyl porphyrinogen isomers.
Column, Hypersil-ODS (250 ×
4.6 mm, 5 µm particle size); elu-
ent, acetonitrile:methanol:1 M
ammonium acetate, pH 5.16
(7:3:90, by vol); flow-rate, 1
mL/minute; detection, ampero-
metric at +0.70 V.

107
C.K. Lim

opment and introduction of high sensitivity propane sulphonic acid (CAPS) at pH


high resolution hybrid electrospray ioniza- 11.0 and the running voltage and tempera-
tion orthogonal quadrupole-time of flight ture were 25 kV and 45°C, respectively.
mass spectrometer (e.g., Q-Tof II; Micro- Isomers of porphyrins are generally diffi-
mass, Altrincham, Cheshire, England, UK), cult to separate by CE, and this procedure
however, all free acid porphyrins can now be has not achieved the same resolution as
analyzed with great sensitivity without the HPLC.
need for derivatization. Detection in CE is also less sensitive
than in HPLC. Although CE is highly sen-
sitive in terms of mass detection, samples
9. SEPARATION OF PORPHYRINS BY at low concentration, e.g., urinary por-
CAPILLARY ELECTROPHORESIS phyrins, can be difficult to analyze because
of the small sample volumes (approximate-
CE has been used for the separation of ly 10 nL) injected. The requirement for
porphyrins and the majority of the meth- surfactants (e.g., SDS) in the running
ods used the micellar electrokinetic capil- buffer for porphyrin separation makes it
lary chromatographic (MECC) mode in incompatible with MS analysis.
conjunction with visible absorbance or flu-
orescence detection (2,20). The separation
of porphyrins in the urine of a patient with 10. FUTURE DEVELOPMENTS
CEP is shown in Figure 10. The running
buffer was 100 mM sodium dodecyl sulfate Further development in MS can be
(SDS) and 20 mM 3-(cyclohexylamino)-1- expected with the introduction of user-

Figure 10. Separation of por-


phyrins in the urine of a patient
with CEP by CE. Running buffer,
100 mM SDS, 20 mM CAPS (pH
11.0); voltage, 25 kV; temperature,
450°C; detection, absorbance 404
nm. Peaks: 4, 5, 6, 7, and 8 refer,
respectively, to tetra(copro)-, penta,
hexa-, hepta-, and octa(uro)car-
boxylic acid porphyrin.

108
Analysis of Heme and Its Precursors

friendly instruments of very high sensitivi- and coproporphyrinogen isomers with amperometric
ty and resolution. This will allow the por- detection. Biochem. J. 234:629-633.
6.Lim, C.K., F. Li, and T.J. Peters. 1987. High-perfor-
phyrins to be analyzed with even greater mance liquid chromatography of type-III heptacar-
sensitivity and specificity, especially in boxylic porphyrinogen isomers. Biochem. J. 247:229-
HPLC-MS/MS analysis. The use of micro- 232.
7.Lim, C.K., F. Li, and T.J. Peters. 1988. High-perfor-
column HPLC-nanospray MS will further mance liquid chromatography of porphyrins. A review.
enhance the sensitivity of detection and J. Chromatogr. Biomed. Appl. 429:123-153.
should be explored for possible application 8.Lim, C.K. and T.J. Peters. 1984. Urine and faecal por-
phyrin profiles by reversed-phase high-performance liq-
in the analysis of the biosynthetic interme- uid chromatography in the porphyrias. Clin. Chim.
diates from ALA to heme. Acta 139:55-63.
9.Lim, C.K., M.A. Razzaque, J. Luo, and P.B. Farmer.
2000. Isolation and characterization of protoporphyrin
glycoconjugates from rat Harderian gland by HPLC,
ABBREVIATIONS capillary electrophoresis and HPLC/electrospray ioniza-
tion MS. Biochem. J. 347:757-761.
10.Lim, C.K., J.M. Rideout, and T.J. Peters. 1984. High-
ALA, 5-aminolevulinic acid; CAPS, 3- performance liquid chromatography of dicarboxylic
(cyclohexylamino)-1-propane sulfonic acid; porphyrins and metalloporphyrins: retention behaviour
and biomedical applications. J. Chromatogr. 317:333-
CE, capillary electrophoresis; CEP, con- 341.
genital erythropoietic porphyria; DMSO, 11.Lim, C.K., J.M. Rideout, and D.M. Samson. 1979.
Determination of 5-aminolaevulinic acid and porpho-
dimethyl sulfoxide; ESIMS, electrospray bilinogen by high-performance liquid chromatography.
ionization mass spectrometry; HPLC, high- J. Chromatogr. 185:605-611.
performance liquid chromatography; LC, 12.Lord, G.A., J.L. Luo, and C.K. Lim. 1999. Capillary
zone electrophoresis/mass spectrometry of 5-aminolae-
liquid chromatography; MECC, micellar vulinic acid and porphobilinogen. Rapid. Comm. Mass
electrokinetic capillary chromatography; Spec. 14:314-316.
MRM, multiple reaction monitoring; MS, 13.Luo, J.L., J. Deka, and C.K. Lim. 1996. Determina-
tion of 5-aminolaevulinic acid dehydratase activity in
mass spectrometry; MS/MS, tandem mass erythrocytes and porphobilinogen in urine by micellar
spectrometry; ODS, octadecylsilyl; PBG, electrokinetic capillary chromatography. J. Chromatogr.
porphobilinogen; RP, reversed-phase; SDS, 722:353-357.
14.Luo, J. and C.K. Lim. 1993. Order of uropor-
sodium dodecylsulfate; SIR, selected ion phyrinogen III decarboxylation on incubation of por-
recording. phobilinogen and uroporphyrinogen III with ery-
throcyte uroporphyrinogen decarboxylase. Biochem.
J. 289:529-532.
15.Luo, J. and C.K. Lim. 1995. Isolation and characteri-
REFERENCES zation of new porphyrin metabolites in human porphyria
cutanea tarda and in rats treated with hexachloroben-
1.Bonnett, R., A.A. Charalambides, K. Jones, I.A. Mag- zene by HPTLC, HPLC and liquid secondary ion mass
nus, and R.J. Ridge. 1978. The direct determination of spectrometry. Biomed. Chromatogr. 9:113-122.
porphyrin carboxylic acids. High-pressure liquid chro- 16.Mauzerall, D. and S. Granick. 1956. The occurrence
matography with solvent systems containing phase- and determination of δ-aminolevulinic acid and por-
transfer agents. Biochem. J. 173:693-695. phobilinogen in urine. J. Biol. Chem. 219:435-446.
2.Chiang, S.C.C. and S.F.Y. Li. 1997. Separation of por- 17.Meyer, H.D., W. Vogt, and K. Jacob. 1984. Improved
phyrins by capillary electrophoresis in fused-silica and eth- separation and detection of free porphyrins by high-
ylene vinyl acetate copolymer capillaries with visible performance liquid chromatography. J. Chromatogr.
absorbance detection. Biomed. Chromatogr. 11:366-370. 290:207-213.
3.Li, F., C.K. Lim, and T.J. Peters. 1987. Separation and 18.Rideout, J.M., D.J. Wright, and C.K. Lim. 1983. High
characterization of pentacarboxylic porphyrinogen iso- performance liquid chromatography of uroporphyrin
mers by high-performance liquid chromatography with isomers. J. Liq. Chromatogr. 6:383-394.
electrochemical detection. Biochem. J. 243:421-423. 19.Rossi, E. and D.H. Curnow. 1986. Porphyrins, Ch.
4.Li, F., C.K. Lim, and T.J. Peters. 1989. Preparation, 10, p. 261-303. In C.K. Lim (Ed.), HPLC of Small
high-performance liquid chromatographic separation Molecules. IRL Press, Oxford.
and characterization of hexacarboxylic porphyrinogens. 20.Weinberger, R., E. Sapp, and S. Moring. 1990. Capil-
J. Chromatogr. 461:353-359. lary electrophoresis of urinary porphyrins with
5.Lim, C.K., F. Li, and T.J. Peters. 1986. High-perfor- absorbance and fluorescence detection. J. Chromatogr.
mance liquid chromatography of uroporphyrinogen 516:217-285.

109
Analysis of Intermediates and End
6 Products of the Chlorophyll
Biosynthetic Pathway

Constantin A. Rebeiz
University of Illinois, Urbana, IL, USA

1. INTRODUCTION of a Chl-protein biosynthesis center, where


the assembly of photosystem I, photosys-
1.1. Multibranched Chlorophyll tem II, and light harvesting Chl-protein
Biosynthetic Pathway complexes (LHC) takes place (44). The
multiple Chl biosynthetic pathways are
Since the 1963 seminal review of Smith considered, individually or in groups of
and French (62), our understanding of the two or three adjacent pathways, to consti-
chlorophyll (chl) biosynthetic pathway has tute Chl-apoprotein biosynthesis subcen-
changed dramatically. Several factors have ters earmarked for the coordinated assem-
contributed to this phenomenon, among bly of individual pigment–protein
which are: (i) development of systems complexes. Apoproteins destined for some
capable of Chl and thylakoid membrane of the biosynthesis subcenters may possess
biosynthesis in organello and in vitro signals for specific Chl biosynthetic
(17,28,42,43,46); (ii) development of enzymes peculiar to that subcenter, such as
powerful analytical techniques that allowed 4-vinyl reductases, formyl synthetases, or
the qualitative and quantitative determina- Chl a and Chl b synthetases. Once an
tion of various intermediates of the path- apoprotein formed in the cytosol or in the
way (this chapter); and (iii) recognition of plastid reaches its biosynthesis subcenter
the probability that the structural and destination and its signal is removed, it
functional complexity of thylakoid includes may bind nascent Chl formed via one or
a multibranched heterogeneous Chl bio- more biosynthetic pathways. During Chl
synthetic pathway (44). binding, it may fold properly and act, at
Chlorophyll biosynthetic heterogeneity that location, as a template for other thy-
(41,47,49) refers to the biosynthesis of Chl lakoid apoproteins having appropriate sig-
via multiple biosynthetic routes. The nals (44).
multibranched Chl a/b biosynthetic path- Figure 1 depicts various biosynthetic
way depicted in Figure 1 may be a template intermediates and end products of the 15

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

111
C.A. Rebeiz

biosynthetic pathways that constitute the nm, a bandwidth of 40 nm, and a photon
multibranched Chl a/b biosynthetic path- density of about 0.01 µmol/m2s.
way. This chapter will describe various ana-
lytical techniques that permit the qualitative ❖ Procedure 1. Preparation of an
and quantitative analysis of the intermedi- Ammoniacal Extract
ates of this complex pathway. Chapter 10 in
this volume describes in more general terms 1. Since homogenization of plant tissues
the isolation, quantification, and character- disrupts the plant vacuoles and releases
ization of total Chl a/b. organic acids, NH4OH is added to the
acetone to neutralize the medium and
prevent loss of the acid-labile central
2. MATERIALS AND METHODS Mg atom from the Mg-tetrapyrroles
(19). For many of the compounds
All tetrapyrrole intermediates and end which are labile, such as protopor-
products of the Chl biosynthetic pathway phyrin IX (Proto), the extraction
are loosely or tightly bound to plastid mem- should be carried out under subdued
brane lipoproteins (4,31,32,38). Before lighting and at ice bucket temperatures.
qualitative and quantitative analysis, these 2. Cut the plant tissue into small pieces
tetrapyrroles need to be extracted from their about 1 cm in length.
native environment with organic solvents.
On the basis of their polarity, plant 3. Homogenized in acetone: 0.1 N
tetrapyrroles fall into two broad categories: NH4OH (9:1 vol/vol) at 0° to 4°C for
(i) very apolar fully esterified tetrapyrroles 60 seconds, using a homogenizer (PT
which are soluble in apolar solvents such as 10/35 probe; Brinkman Instruments,
petroleum ethers and hexane; and (ii) less Westbury, NY, USA), at a ratio of 7 mL
apolar tetrapyrroles soluble in more polar of solvent per gram of tissue.
solvents such as acetone. Once tetrapyrroles 4. Centrifugation at 39 000× g for 12 min-
have been partitioned between polar and utes at 1°C.
apolar solvents, they can be subjected to a 5. Decant the ammoniacal acetone extract
variety of purification and analytical tech- and store at -80°C until required.
niques. Before discussing details of purifica- Isolated plastids and subplastidic prepa-
tion and analysis, tetrapyrrole extraction rations can be used, for example, to study
and partitioning into apolar and polar frac- precursor product relationships in organel-
tions will be described. lo or in vitro. Plastids or subplastidic
preparations (see Chapter 12), which are
2.1. Tetrapyrrole Extraction usually suspended in buffers with a pH
range of about 7.0 to 8.0, are extracted
Tetrapyrrole extraction from green tis-
with acetone: 0.1 N NH4OH (9:1 vol/vol)
sues can be carried out under laboratory
at a rate of 10 mL per 1 mL of preparation
light (about 5 µmol/m2s). However, pig-
and then treated as above.
ment extraction from etiolated tissues
should be performed under a green safe- 2.2. Tetrapyrrole Partitioning Between
light that does not photoconvert pro- Apolar and Polar Solvents
tochlorophyllide a (Pchlide a) and Pchlide
a ester (Pchlide a E) to chlorophyllide a Once an ammoniacle extract has been
(Chlide a) and Chl a (66). In our laborato- made, partitioning tetrapyrroles between
ry, we routinely use a low irradiance green apolar and polar solvents is an easy way of
light with a transmission maximum at 503 achieving partial purification prior to
112
Analysis of Intermediate and End Products of Chlorophyll
113

Figure 1. Integrated Chl a/b biosynthetic pathway adapted from Reference 44. To facilitate understanding of the text, various biosynthetic pathways are designated by the numbers 1 to 14.
C.A. Rebeiz

quantitative and qualitative analysis. (excitation light) is absorbed by a fluores-


cent compound, the absorbed light is
❖ Procedure 2. Preparation of a reemitted at a longer wavelength. The
Hexane-Extracted Acetone Residue reemitted light is called fluorescence. In
contrast to absorbance spectroscopy, fluo-
1. Prepare an ammoniacal acetone extract rescence is a 2-window technique as it
as above. allows the monitoring of emission or exci-
2. Using a separation funnel, transfer tation spectra. For example, by excitation
chlorophylls and other fully esterified at an appropriate fixed wavelength, the
tetrapyrroles to hexane by extraction emitted light can be scanned, thus generat-
with an equal volume of hexane. ing an emission spectrum. On the other
3. Using conical centrifuge tubes, carry hand, by monitoring the emitted light at a
out a second extraction with one-third fixed emission wavelength, the absorbed
volume of hexane. Emulsions are light can be scanned, thus generating an
resolved using a benchtop centrifuge at excitation spectrum.
room temperature. In addition to its high sensitivity (subpi-
comole quantities of tetrapyrroles are readi-
4. After centrifugation, remove the lower
ly detectable), fluorescence measurements
phase (hypophase) that consists of
are highly selective. For example, let us con-
hexane-extracted acetone (HEAR)
sider a mixture of several fluorescent com-
using a Pasteur pipet.
pounds with approximately an equal
5. The HEAR hypophase contains mono- propensity to fluoresce, i.e., having approx-
carboxylic tetrapyrroles such as Pch- imately similar relative quantum yields, and
lide, Chlide, and Mg-protoporphyrin different absorption and emission proper-
monomethyl ester (Mpe), and dicar- ties. It is possible to elicit the fluorescence
boxylic tetrapyrroles such as Proto and of any compound in the mixture by selec-
Mg-Proto. tive excitation at a specific wavelength that
6. The HEAR epiphase contains fully minimizes the detection of the other fluo-
esterified tetrapyrroles such as chloro- rescent species. Of course, the other com-
phylls and protochlorophyllide esters. pounds in the mixture may also exhibit
The epiphase is usually contaminated some fluorescence that needs to be correct-
by trace amounts of hypophase pig- ed for during quantitative measurements.
ments and in some situations it is desir- Conversely, the excitation spectrum of any
able to purify further the epiphase pig- one of the compounds in the mixture may
ments prior to quantitative analysis. also be observed by recording an excitation
spectrum (also referred to as an action spec-
2.3. Spectrofluorometry trum) at a selected emission wavelength
that minimizes the detection of the other
The use of fluorescence emission and compounds in the mixture. Furthermore, if
excitation spectrofluorometry has been the fluorescence measurements are made at
instrumental in the detection and spectral 77 K, significant narrowing and sharpening
analysis of various tetrapyrroles. It is there- of the emission and excitation bands are
fore, appropriate to precede the description observed. This in turn results in consider-
of the detection and quantitative determi- able improvement in band resolution.
nation of various tetrapyrroles by a brief The correct use of spectrofluorometry is
introduction to spectrofluorometry. slightly more complicated than the use of
When light of a particular wavelength absorbance spectroscopy. The excitation
114
Analysis of Intermediate and End Products of Chlorophyll

source, which in most cases is a Xenon arc, mixtures. In the following, a brief descrip-
has a limited life span. As the arc lamp tion of common chromatographic proce-
ages, so does its output. As a consequence, dures used in tetrapyrrole separation and
the sensitivity of the instrument decreases purification are described.
as a function of the age of the excitation
lamp. This can be corrected for by adjust- 2.4.1. Paper Chromatography
ing the sensitivity of the instrument on a
daily basis against a stable fluorescent stan- Paper chromatography is one of the old-
dard, such as an acrylic block of rhodamine est techniques used for the separation of
b, by adjustment of the photomultiplier various tetrapyrroles including Chls.
voltage. The stability of the excitation Although more powerful techniques have
source also varies continuously as the volt- since been developed, it is still a viable and
age in the laboratory fluctuates as a func- cheap alternative for the separation of
tion of power demands. This caveat can Proto by ascending chromatography, from
simply be corrected for by recording spec- more polar tetrapyrroles such as uropor-
tra in the ratio mode. In that mode, the phyrins (Uro) and coproporphyrins
excitation beam is split into two beams. (Copro) (37).
One beam passes through the sample (sam-
ple beam), and the other beam, the refer- 2.4.2. Thin Layer Chromatography
ence beam, passes through a quantum
counter such as a cuvette filled with a con- For the past 35 years, thin layer chro-
centrated rhodamine b solution (about 10 matography (TLC) has been the workhorse
mg/mL polyethylene glycol 600). Since of tetrapyrrole purification. However, a
rhodamine b exhibits a constant quantum comprehensive treatment of TLC is
yield between 380 and 600 nm by moni- beyond the scope of this chapter. In our
toring the ratio of the sample and reference laboratory, we prepare our own TLC plates
beams instead of monitoring only the sam- using a variety of adsorbents including cel-
ple beam, the effect of any fluctuation in lulose, silica gel H, and polyethylene.
the electrical current on signal intensity is Silica gel H plates are prepared by mix-
eliminated. The recording of fluorescence ing 42 g of silica gel H (EM Science, Gibb-
spectra in the ratio mode is routine in most stown, NJ, USA) and 135 mL of water.
commercial scanning spectrofluorometers. Cellulose (Cellulosepulver, MN 300;
Finally, the photomultiplier response of Macherey-Nagel, Duren, Germany) plates
the instrument as a function of wavelength are prepared by mixing 22 g of cellulose
is not constant and usually decreases dra- and 135 mL of water. Polyethylene plates
matically in the red region of the spectrum. are prepared by mixing 40 g of chromato-
In computer-interfaced instruments, this is graphic grade polyethylene powder (Poly-
corrected for on-line by multiplying the sciences, Warrington, PA, USA) and 120
signal ratios at every wavelength by a cor- mL of pure or 90% aqueous acetone. Mix-
rection factor. This feature is standard on ing is carried out for 60 seconds in a War-
most sophisticated scanning spectrofluo- ing blender. The plates (0.25 or 0.5 mm in
rometers. thickness) are prepared by using a Desaga
spreader. After air-drying for about 30
2.4. Purification of Tetrapyrroles minutes, the plates are activated by heating
at 105°C overnight. The activated plates
Prior to some analyses, it is desirable to are stored dry in a dessicator until further
purify individual tetrapyrroles from crude use. Unless otherwise indicated, we favor
115
C.A. Rebeiz

development at 4°C in darkness. More trum of the eluting band is recorded


detail about specific TLC systems will be between 580 and 700 nm.
described as needed.

2.4.3. High-Pressure Liquid 3. ANALYSIS OF DIVINYL


Chromatography PROTOPORPHYRIN IX

In some cases, high-pressure liquid chro- Proto is a divinyl (DV) dicarboxylic


matography (HPLC), also referred to as tetrapyrrole with vinyl groups at position 2
high-performance liquid chromatography, and 4 of the macrocycle (Figure 2). In
is very useful for qualitative and quantita- extracting Proto from plant tissues or iso-
tive tetrapyrrole analysis. Prior to quantita- lated organelles, it is essential to eliminate
tive analysis, it is advisable to check the loss of Mg from Mg-Proto and Mpe, since
purity of the segregating tetrapyrrole peaks Mg loss would generate Proto and Proto
by high-resolution spectroscopic tech- monomethyl ester that would contaminate
niques such as spectrofluorometry. It has the endogenous Proto pool. Elimination of
been our experience that separated HPLC Mg loss is achieved by extraction with
tetrapyrrole peaks are often contaminated ammoniacal acetone as described in section
by very small amounts of other tetra- 2.1.
pyrroles.
Unless otherwise indicated, we currently 3.1. Quantitative Determination of
use an Applied Biosystems HPLC system Proto by Room Temperature
(Foster City, CA, USA) that consists of a Spectrofluorometry
quaternary LC-620 pump, variable wave-
length absorption (LC-95) and fluores- Upon tetrapyrrole extraction from plant
cence (LC-240) detectors, and a cooled tissues and partitioning of the pigments
autosampler (ISS 200). Ten- to fifty-micro- between hexane and acetone, Proto passes
liter aliquots of the acetone:ammonium into the HEAR phase (section 2.2) along
hydroxide extract are injected. Separations with other dicarboxylic tetrapyrroles such
are performed on a an Applied Biosystems as DV and monovinyl (MV) Mg-Proto
Pecosphere 3 × 3C, C-18 reversed phase, 4 (Figure 3) and monocarboxylic tetra-
× 0.5 cm column or other columns as indi- pyrroles such as DV and MV Mpe (Figure
cated for specific analyses. A PEEK-C18 3), DV and MV Pchlide a (Figure 4), DV
guard column (Applied Biosystems) is used and MV Chlide a and b (Figure 5), and
to protect the separation column, and elu- pheophorbide (Pheobide) a and b (i.e.,
tion is usually achieved with isocratic or demetalated Chlide a and b). The fluores-
gradient solvent systems. The elutants are cence emission properties of Proto are such
monitored by on-line spectrofluorometry that it is possible to determine the amount
where fluorescence can be stimulated by of Proto in the presence of other di- and
excitation at 400, 420, or 440 nm, and monocarboxylic tetrapyrroles without fur-
signal is monitored with a total emission ther purification. For example, in HEAR at
accessory. The amounts of various room temperature, Proto exhibits respec-
tetrapyrroles are determined from a calibra- tive excitation and emission maxim at 404
tion curve using Proto, Mg-Proto, Pchlide, and 633 nm, while Mg-Proto and Mpe
Chl, and pheophytin standards. When exhibit respective excitation and emission
needed, the HPLC pump is stopped at maxima at 418 and 595 nm, Pchlides a at
each emission peak, and an emission spec- 438 and 638 nm, Chlide b at 461 and 660
116
Analysis of Intermediate and End Products of Chlorophyll

nm, Chlide a at 432 and 675 nm, pheo- Chlide a and b emissions.
phytin and Pheobide a [Pheo(bide) a] at F (F622 E400) = undeconvoluted total
413 and 674 nm and Pheo(bide) b at 439 fluorescence emission amplitude observed
and 660 nm. By excitation of the HEAR at 622 nm upon excitation of the tetrapyr-
fraction at 400 nm, i.e., close to the Soret role mixture at 400 nm. This factor cor-
excitation maximum of Proto, and by rects for Copro fluorescence emission.
recording an emission spectrum between F (F638 E440) = undeconvoluted total
580 and 700 nm, it is possible to maximize fluorescence emission amplitude observed
the fluorescence emission of Proto at 633 at 638 nm upon excitation of the
nm. The fluorescence emission of Mg- tetrapyrrole mixture at 440 nm. This fac-
Proto and Mpe are far removed from the tor corrects for Pchlide a fluorescence
emission maximum of Proto and do not emission.
interfere with its determination. Only fluo- F (F675 E440) = undeconvoluted total
rescence emissions from Copro at 622 nm, fluorescence emission amplitude observed
from Pchlide a at 638 nm and to a minor at 675 nm upon excitation of the tetrapyr-
extent from Chlide a and b, are likely to role mixture at 440 nm. This factor corrects
interfere. The possible contribution of for Chlide a and b fluorescence emission.
these fluorescences to Proto fluorescence is See Table 2 for further equations.
eliminated by spectral deconvolution. This The various fluorescence amplitudes to
is achieved by exciting the tetrapyrrole be substituted into Equation 1 are read
mixture at 440 nm, i.e., close to the Soret from the recorded emission spectra. Next,
excitation maximum of Pchlide a and the net fluorescence emission amplitude of
Chlide a and b, and by recording a second Proto, as calculated from Equation 1, is
emission spectrum of the mixture between converted to Proto concentration by refer-
580 to 700 nm. The net fluorescence ence to a standard calibration curve. The
amplitude due only to Proto emission is latter is prepared from standard Proto solu-
deconvoluted from the Copro, Pchlide a, tions of known concentrations and from
and Chlide a and b emissions with the use their fluorescence emission amplitudes.
of Equation 1 (45). The latter are recorded under the same
instrumental conditions by excitation at
Equation 1. Deconvolution of 400 nm. In this manner, Proto can be
Fluorescence Data determined with a precision of about 4%
(45). Minimum detection levels are about
Proto (F633 E400) = 1.055 F (F633 E400) 0.2 pmol/mL. Equation 1 can be used with
- 0.1190 F (F622 E400) - 0.442 F (F638 any spectrofluorometer capable of record-
E440) - 0.0141 F (F675 E440) ing corrected fluorescence emission and
Where: excitation spectra. The user has to con-
Proto (F633 E400) = deconvoluted net struct, however, the required calibration
fluorescence emission amplitude of Proto curve that relates net fluorescence emission
at 633 nm upon excitation of the tetrapyr- amplitudes to Proto concentrations.
role mixture at 400 nm. In the Laboratory of Plant Biochemistry
F (F633 E400) = undeconvoluted total and Photobiology, Proto calculations as
fluorescence emission amplitude observed well as all other spectrofluorometric calcu-
at 633 nm upon excitation of the tetrapyr- lations are automatically carried out by the
role mixture at 400 nm. This fluorescence PC interfaced with the spectrofluorometer,
amplitude is usually contaminated by con- using Porphyrin Analytical Tools® (PAT)
tributions from Copro, Pchlide a, and software (48).
117
C.A. Rebeiz

The washed diethyl ether extract can be ❖ Procedure 3. Extraction of


used for further manipulations. If the orig- Monocarboxylic and Dicarboxylic
inal HEAR fraction contains both Proto Tetrapyrroles with Diethyl Ether
and Mg-Proto, and one wishes to separate
these tetrapyrroles, additional chromato- 1. Prepare a HEAR fraction hypophase as
graphic steps are required (section 3.3). above and transfer to a conical cen-
trifuge tube.
3.2. Extraction of Proto from HEAR 2. Transfer monocarboxylic tetrapyrroles
to diethyl ether by adding:
Prior to structural analysis, it is often 1/5 volume of diethyl ether,
necessary to extract Proto from the HEAR 1/17 volume of saturated NaCl,
fraction into less polar solvents such as 1/70 volume of 0.37 MKH2PO4
diethyl ether. Since Proto is a dicarboxylic at pH 7.7.
tetrapyrrole (Figure 3), it is quite soluble in
3. Mix thoroughly and resolve the phases
the aqueous HEAR fraction. As a conse-
by centrifugation at 1500× g for 30 sec-
quence, its extraction into less polar sol-
onds at room temperature.
vents requires a procedure adapted for
dicarboxylic tetrapyrroles (26). Essentially, 4. Remove the ether phase using a Pasteur
the procedure involves first the transfer of pipet.
the less polar monocarboxylic tetrapyrroles 5. Re-extract the HEAR fraction 4 to 5
to diethyl ether. This is followed by extrac- times with small volumes of diethyl
tion of dicarboxylic tetrapyrroles such as ether using centrifugation to resolve
Proto and Mg-Proto into diethyl ether the emulsion.
after adjustment of the HEAR pH. 6. Combine the ether extracts for further

Figure 2. Structure of protoporphyrin


IX, which is the last common inter-
mediate of chlorophyll and heme.

118
Analysis of Intermediate and End Products of Chlorophyll

manipulation of monocarboxylic tetra- KH2PO4 adjusted to pH 4.8. This


pyrroles. washing step is extremely important for
7. For extraction of dicarboxylic tetra- the recording of sharp 77 K spectra in
pyrroles from the remaining ether- diethyl ether. It is best achieved by
extracted HEAR fraction, adjust the removing the diethyl ether extract with
pH to 4.0 to protonate the free car- a Pasteur pipet and slowly releasing it at
boxylic groups. the bottom of a 15-mL test tube filled
8. Extract 5 times with small volumes of with the phosphate buffer. As the
diethyl ether using centrifugation to diethyl ether bubbles rise to the top, the
breakdown the emulsion. aqueous contaminants pass into the
9. Wash the combined ether extracts con- buffer. The washed diethyl ether extract
taining dicarboxylic tetrapyrroles by can then be removed using a Pasteur
passing through a 0.5 M solution of pipet.

Figure 3. Mg-protoporphryin IX and its derivatives.


Compound Abbreviation R1 R2 R3
Divinyl-Mg-protoporphyrin IX DV Mg-Proto vinyl H H
Monovinyl-Mg-protoporphyrin IX MV Mg-Proto ethyl H H
Divinyl-Mg-protoporphyrin IX monomethylester DV Mpe vinyl methyl H
Monovinyl-Mg-protoporphyrin IX monomethylester MV Mpe ethyl methyl H
Divinyl-Mg-protoporphyrin IX diester DV Mpde vinyl methyl LCFA
Monovinyl-Mg-protoporphyrin IX diester MV Mpde ethyl methyl LCFA

119
C.A. Rebeiz

3.3. Chromatographic Separation of extract, is spotted at the origin of a 5 × 15


Proto from other Tetrapyrroles cm piece of No. 1 or No. 3 chromatogra-
phy paper (Whatman, Clifton, NJ, USA),
3.3.1. Separation by Paper allowed to dry, and then the paper is
Chromatography inserted into a cut 1000-mL glass cylinder
containing about 10 mL of 2,4- or 2,6-
Although more powerful techniques lutidine: 0.05 N NH4OH (5:3.5 vol/vol).
have since been developed, it is still a viable The cylinder is capped with a piece of alu-
and cheap alternative for the separation of minum foil. Development is carried out
Proto from more polar tetrapyrroles such as under subdued light at room temperature
Uro and Copro by ascending chromatogra- in a ventilated hood. When the solvent
phy (37). Essentially, the tetrapyrrole mix- front has migrated about 10 cm, the chro-
ture, as in an aliquot of HEAR or acetone matogram is viewed under a 360 nm UV

Figure 4. Protochlorophyllide group. Tetrapyrroles which incorporate the fifth isocyclic ring, ring E. LCFA = long-chain fatty acid.
Compound Abbreviation R1 R2 R3
Divinyl-protochlorophyllide a DV Pchlide a methyl vinyl H
Monovinyl-protochlorophyllide a MV Pchlide a methyl ethyl H
Divinyl-protochlorophyllide a ester DV Pchlide a E methyl vinyl LCFA
Monovinyl-protochlorophyllide a ester MV Pchlide a E methyl ethyl LCFA
Divinyl-protochlorophyllide b DV Pchlide b formyl vinyl H
Monovinyl-protochlorophyllide b MV Pchlide b formyl ethyl H
Divinyl-protochlorophyllide b ester DV Pchlide b E formyl vinyl LCFA
Monovinyl-protochlorophyllide b ester MV Pchlide b E formyl ethyl LCFA

120
Analysis of Intermediate and End Products of Chlorophyll

light. The separated tetrapyrroles are can be efficiently used for the quantitative
detected by their red fluorescence. In this separation of Proto from other
system, Proto migrates with a retention tetrapyrroles. Using large columns, it can
factor (Rf) of about 0.8, while Copro and also be conveniently used for preparative
Uro migrate with Rfs of about 0.6 and 0.3, purposes.
respectively (37). Although paper chro- We have successfully used the solvent
matography is useful for separating Proto system of Ho (23) for separating Proto
form other tetrapyrroles, it is not conve- from Uro, Copro, and other multi-
nient for preparative purposes because of caboxylic tetrapyrroles in HEAR fractions
the difficulty of eluting the separated prepared from cancer cell cultures (50).
tetrapyrroles. The same system can be used with HEAR
preparation of plant sources. With this sol-
3.3.2. Chromatographic Separation and vent system, there is no need to convert
Determination by HPLC tetrapyrroles to their methyl esters prior to
chromatography. If feasible, it is always
If HPLC instrumentation is available, it advantageous to avoid derivatization prior

Figure 5. Chlorin group. Tetrapyrroles reduced to the oxidation state of chlorophylls, i.e., a single bond between carbons 7 and 8.
Compound Abbreviation R1 R2 R3

Divinyl-chlorophyllide a DV Chlide a methyl vinyl H


Monovinyl-chlorophyllide a MV Chlide a methyl ethyl H
Divinyl-chlorophyllide b DV Chlide b formyl vinyl H
Monovinyl-chlorophyllide b MV Chlide b formyl ethyl H
Divinyl-chlorophyll a DV Chl a methyl vinyl phytol
Monovinyl-chlorophyll a MV Chl a methyl ethyl phytol
Other divinyl-chlorophyll a Other DV Chl a methyl vinyl LCFA

121
C.A. Rebeiz

to chromatography, since derivatization elicited by excitation close to its Soret


may generate artifacts and often results in absorbance maximum at around 400 to
lower yields that necessitate correction for 405 nm. In each case, a calibration curve
percent recoveries during quantitative using known amounts of Proto should be
determinations. constructed in order to convert fluores-
Separation of Proto from tri-, tetra-, cence amplitudes to Proto concentrations.
penta-, hexa-, and octacarboxylic tetra- At each concentration, an emission spec-
pyrroles can be achieved on a Bondapak trum elicited by excitation at the chosen
reverse phase C18-bonded column (Waters, Soret excitation wavelength should be
Milford, MA, USA). The column is equili- recorded, and a calibration curve relating
brated with the first binary mobile phase fluorescence emission amplitudes of pure
(0.1 M sodium phosphate in acetonitrile, Proto solutions to Proto concentration is
29:16 vol/vol, pH 3.0) for 10 minutes. constructed.
After sample injection, the mobile phase is
changed to 0.1 M sodium phosphate in ace- 3.4.2. Quantitative Determination by
tonitrile (18:129 vol/vol, pH 3.0) for 15 Spectrophotometry
minutes. The flow rate is usually about 1.10
mL/minute. The eluant is monitored by The concentration of purified Proto
spectrofluorometry. Fluorescence is elicited solutions can be determined in a variety of
by excitation at 405 nm and is monitored at organic solvents by absorbance spec-
an emission wavelength of 630 nm. The troscopy. The procedure is only limited by
amounts of various tetrapyrroles can be the availability of molar extinction coeffi-
determined from a calibration curve using cient values for the solvent under consider-
tetrapyrrole chromatographic standards ation and is usually used for the prepara-
(Porphyrin Products). In this system, Proto tion of Proto stock solutions of known
exhibits a retention time of about 12.68 concentrations.
minutes. Most HPLC manuals describe the At room temperature, Proto exhibits a
use of internal and external standards for stepladder (etio) absorption spectrum in
quantitative determinations. The proce- various organic solvents with a very strong
dures usually vary slightly from manufac- Soret absorption band between 340 and
turer to manufacturer depending on the 440 nm and 4 absorption bands of decreas-
used software. The method can be scaled up ing intensity between 480 and 650 nm
for preparative purposes. In that case, the (19). The most suitable wavelength for the
recovered fraction can be treated as quantitative determination of Proto is at its
described in section 3.2 to transfer the puri- Soret absorbance maximum, which varies
fied Proto to diethyl ether. from 400 to 406 nm depending on the sol-
vent. At the Soret absorbance maximum,
3.4. Quantitative Determination of Proto has a large molar extinction coeffi-
Purified Proto cient that allows the quantitative determi-
nations of dilute solutions. Since many
3.4.1. Quantitative Determination by other pigments, such as carotenoids, Chls,
Spectrofluorometry and Pchlides, absorb light in this spectral
region, it is important to use highly puri-
Purified Proto can be determined in fied Proto. It is also possible to use Proto
HEAR as described in section 3.1 for absorbance in the red region of the spec-
tetrapyrrole mixtures. It can also be deter- trum, at 622 nm, for quantitative determi-
mined in any solvent from emission spectra nations. At this wavelength, the Proto
122
Analysis of Intermediate and End Products of Chlorophyll

molar extinction coefficient is very low, In the following sections, each group of
and such determinations require the use of compounds are considered in turn, and
more concentrated solutions. similarities and differences in analytical
The molar extinction coefficient is relat- procedures are highlighted as appropriate.
ed to Proto absorbance and concentration
by Equation 2 (15).
5. ANALYSIS OF MG-
Equation 2. Beer's Law PROTOPORPHYRIN IX

A = εcl The Mg-Proto pool is a mixed DV-MV


Where: A = Absorbance. dicarboxylic tetrapyrrole pool (Figure 3).
ε = Molar absorption coeffi- Under most conditions, the proportion of
cient. DV Mg-Proto is much larger than that of
c = Concentration of Proto MV Mg-Proto.
in moles per liter.
l = Optical length in cm of 5.1. Quantitative Determination of Mg-
the used cuvette, which in Proto in Crude Extracts by
most cases is 1 cm. Spectrofluorometry
Upon tetrapyrrole extraction from plant
4. ANALYSIS OF OTHER tissues and partitioning of the pigments
TETRAPYRROLES between hexane and acetone (sections 2.1
and 2.2), the mixed DV and MV Mg-Proto
The quantitative determination of the pool passes into the HEAR phase along
other intermediates of the Chl biosynthetic with other dicarboxylic and monocarboxylic
pathway (Figure 1) can be carried out using tetrapyrroles. Since in HEAR, DV and MV
essentially the same procedure as described Mg-Proto exhibit similar emission maxima
for Proto. Table 1 reports the absorption at 594 to 596 nm at room temperature, it is
coefficients for the compounds in various possible to determine the concentration of
solvents, while Table 2 depicts the equations the mixed DM-MV Mg-Proto pool in the
used to deconvolute the spectra for quanti- presence of other di- and monocarboxylic
tative determinations. For each compound, tetrapyrroles without further purification.
the analysis involves the following steps: Since Mg-Proto and Mpe exhibit identical
1. Extraction in ammoniacal acetone spectrophotometric and fluorescence prop-
from tissues or organelles. erties, it is necessary to extract the mono-
carboxylic Mpe pool from the HEAR frac-
2. Quantitative determination by spectro-
tion prior to Mg-Proto determination.
fluorometry in crude HEAR extracts.
Extraction of monocarboxylic tetra-
3. Transfer of the tetrapyrrole from ace- pyrroles from the HEAR fraction into
tone to hexane and then from HEAR diethyl ether is achieved exactly as
to diethyl ether. described in section 3.2. Once this is done,
4. Chromatographic purification on thin the ether-extracted HEAR fraction is excit-
layer plates or HPLC. ed at 420 nm, i.e., close to the Soret excita-
5. Quantitative determination of ether- tion maximum of the mixed Mg-Proto
extracted tetrapyrroles or purified pool, and an emission spectrum is recorded
tetrapyrroles by spectrofluorometry between 580 and 700 nm. The Mg-Proto
and/or spectrophotometry. pool exhibits a well-defined emission band
123
124

C.A. Rebeiz
Table 1. Molar Extinction Coefficients (Ext Coefficient) of Metabolic Intermediates of the Chl a Biosynthetic Pathway in Different Solvents at
Room Temperature
Tetrapyrrole Solvent Abs ( nm) Ext Coefficient Reference

DV Proto DME Diethyl ether 404 158000 19


DV Proto DME Dioxan 406 164000 19
DV Proto DME CHCl3 407 171000 19
DV Proto DME Pyridine 409 163000 19
Disodium DV Proto 80% Acetone 402 108244 This work
Disodium DV Proto 80% Acetone 629 3883 This work
DV Mg-Proto DME Diethyl ether 419 308000 19
DV Mg-Proto DME Diethyl ether 551 18200 19
DV Mg-Proto DME Diethyl ether 589 18200 19
Disodium DV Mg-Proto 80% Acetone 417 165900 This work
Disodium DV Mg-Proto 80% Acetone 550 13817 This work
Disodium DV Mg-Proto 80% Acetone 589 12537 This work
MV Pchl(ide) a Diethyl ether 432 187517 30
MV Pchl(ide) a Diethyl ether 623 22620 30
MV Pchl(ide) a Acetone 432 165817 30
MV Pchl(ide) a Acetone 623 21394 30
MV Pchl(ide) a Methanol 434 108685 30
MV Pchl(ide) a Methanol 629 16674 30
DV Pchl(ide) a Diethyl ether 437 205000 23
DV Pchl(ide) a Diethyl ether 622 22100 23
MV Pchlide b phytyl ester Diethyl ether 442 111519 56
MV Pchlide b phytyl ester Diethyl ether 630 24357 56
MV Pchlide b phytyl ester 80% Acetone 446 88585 56
MV Pchlide b phytyl ester Diethyl ether 632 18171 56
MV Chl a Diethyl ether 430 104090 34
MV Chl a Diethyl ether 660 83450 34
Table 1, continued.

Tetrapyrrole Solvent Abs ( nm) Ext Coefficient Reference


MV Chl a 80% Acetone 430 81110 34
MV Chl a 80% Acetone 663 73300 34
DV Chl a Diethyl ether 435 110820 57
DV Chl a Diethyl ether 660 78340 57
DV Chl a 80% Acetone 438 95120 57
DV Chl a 80% Acetone 664 69290 57
MV Chl b Diethyl ether 455 129090 34
MV Chl b Diethyl ether 643 48630 34

Analysis of Intermediate and End Products of Chlorophyll


MV Chl b 80% Acetone 460 118210 34
MV Chl b 80% Acetone 645 41370 34
DV Chl b Diethyl ether 462 118900 57
DV Chl b Diethyl ether 643 43910 57
DV Chl b 80% Acetone 468 87830 57
DV Chl b 80% Acetone 651 35780 57
MV Pheophytin a Diethyl ether 665 50530 70
MV Pheophytin a Diethyl ether 410 109770 70
MV Pheophytin a 80% Acetone 666 49310 66
MV Pheophytin a 80% Acetone 409 114040 66
DV Pheophytin a Diethyl ether 667 45680 57
DV Pheophytin a Diethyl ether 417 107900 57
DV Pheophytin a 80% Acetone 665 16520 57
DV Pheophytin a 80% Acetone 419 41300 57
MV Pheophytin b Diethyl ether 654 32750 70
MV Pheophytin b Diethyl ether 433 173500 70
MV Pheophytin b 80% Acetone 655 31600 66
MV Pheophytin b 80% Acetone 436 160220 66
DV Pheophytin b Diethyl ether 657 29580 57
DV Pheophytin b Diethyl ether 440 117300 57
DV Pheophytin b 80% Acetone 657 21060 57
DV Pheophytin b 80% Acetone 443 70960 57

Molar extinction coefficients of Pchlide a were calculated from the specific absorption coefficients reported in Reference 30 and a molecular weight of
613 for Pchlide a. In Reference 30 the cited specific absorption coefficients were mistakenly assigned to Pchlide a E by the authors.
125
C.A. Rebeiz

with a maximum at 594 to 596 nm. In this tudes vary with the freezing pattern of the
spectral region, interference by other nat- sample. For a given spectrum, however, the
ural tetrapyrroles is not encountered, and ratio of fluorescence amplitudes at any two
correction for fluorescence band overlap is or more wavelengths is independent of the
unnecessary. The fluorescence amplitude at freezing pattern.
595 to 596 nm or the integrated area As a consequence, determination of the
under the fluorescence emission band is amounts of DV and MV Mg-Proto is a 2-
next converted to Mg-Proto concentration step process. First, the total amount of DV
by reference to calibration curves prepared plus MV Mg-Proto in the ether-extracted
under the same instrumental and analytical HEAR fraction is determined at room
conditions. The calibration curves should temperature, exactly as described in section
relate authentic Mg-Proto concentrations 3.2. Next, the DV/MV ratio of the Mg-
to fluorescence emission amplitude at 595 Proto pool is determined from 77 K spec-
to 596 nm or to integrated fluorescence tra recorded in diethyl ether. The amount
areas under the Mg-Proto emission band. of DV and MV Mg-Proto is then calculat-
In this manner, Mg-Proto can be deter- ed from the total amount of Mg-Proto, as
mined with a precision of about 7% to determined from the ether-extracted
8%. Minimum detection levels are about HEAR fraction at room temperature and
0.2 pmol/mL. from the DV/MV Mg-Proto ratio as deter-
mined at 77 K in diethyl ether (63).
5.2. Quantitative Determination of DV To calculate the DV/MV Mg-Proto
and MV Mg-Proto in Crude ratio, two sharp 77 K excitation spectra in
Extracts by Spectrofluorometry diethyl ether need to be recorded. For the
recording of sharp DV and MV Mp-Proto
Before structural studies or determina- and all other 77 K DV and MV Mg-
tion of DV and MV Mg-Proto, it is neces- tetrapyrroles spectra, it is essential to thor-
sary to extract Mg-Proto from the Mpe- oughly wash the diethyl ether fraction with
free ether-extracted HEAR fraction into a 0.5 M solution of KH2PO4 adjusted to
less polar solvents such as diethyl ether. pH 4.8 (or to pH 7.0 for monocarboxylic
Extraction of DV and MV Mg-Proto from Mg-tetrapyrroles) as described in section
the ether-extracted Mpe-free HEAR frac- 3.2. At a pH lower than 4.8, Mg loss
tion is achieved exactly as described in sec- becomes a problem, and at a pH higher
tion 3.2 for Proto. than 4.8, the dicarboxylic Mg-Proto pool
The use of low temperature (77 K) is will be lost by passing into the aqueous
essential to differentiate between DV and phase.
MV Mg-Proto and all other DV and MV The first 77 K excitation spectrum is
Mg-tetrapyrroles. At 77 K, due to consid- recorded from 380 to 500 nm by position-
erable fluorescence excitation and emission ing the emission monochromator at the
band narrowing, the DV and MV Mg- emission maximum of DV Mg-Proto at
Proto pools exhibit respective excitation 591 nm. In this spectrum, DV Mg-Proto
and emission maxima at 424 and 591 nm will exhibit a sharp and narrow excitation
(DV Mg-Proto) and 417 and 589 nm (MV band with a maximum at 424 nm (63).
Mg-Proto) (12). It is not possible, however, The second 77 K excitation spectrum is
to directly determine the amounts of DV recorded from 380 to 500 nm by position-
and MV Mg-Proto from 77 K spectra, ing the emission monochromator at 587
since for a given amount of DV and MV nm, just below the 589 nm emission max-
Mg-Proto, the overall fluorescence ampli- imum of MV Mg-Proto. In this spectrum,
126
Analysis of Intermediate and End Products of Chlorophyll

MV Mg-Proto will exhibit a sharp and nar- Essentially, the tetrapyrrole mixture, as in
row excitation band with a maximum at an aliquot of HEAR or acetone extract, is
417 nm (63). In most cases, if both DV spotted at the origin of a 5 × 20 cm TLC
and MV Mg-Proto are present, two sharp plate of silica gel H. While the plate is still
maxima are observed, one at 424 nm for wet, it is inserted into a cut 1000-mL glass
DV Mg-Proto and one at 417 nm for MV cylinder containing about 10 mL of
Mg-Proto. The net fluorescence excitation toluene:ethyl acetate:ethanol (8:2:2 vol/
amplitudes of DV and MV Mg-Proto are vol/vol). The cylinder is capped with a piece
then deconvoluted and calculated with of aluminum foil. Development is carried
Equations 3 and 4 (Table 2) (63). out at 4°C in darkness. After the solvent
The various undeconvoluted fluores- front has migrated about 10 cm, the plate
cence excitation amplitudes to be substitut- is viewed under 366 nm UV. The separated
ed into Equation 3 and 4 are read from the tetrapyrroles are detected by their red fluo-
recorded excitation spectra. Next, the ratio rescence. In this system, Proto remains at
of the deconvoluted DV Mg-Proto excita- the origin, and Mg-Proto moves with an Rf
tion amplitude to the deconvoluted MV of about 0.09. Monocarboxylic Mg-por-
Mg-Proto excitation amplitude is calculat- phyrins such as Mpe, Pchlides, and Chlides
ed. This ratio is referred to as the apparent move toward the center of the plate. Mg-
DV/MV Mg-Proto excitation ratio. That Proto is eluted in methanol:acetone (4:1
ratio is converted to an authentic DV/MV vol/vol). Elution is achieved by scraping the
Mg-Proto concentration ratio by reference wet silica gel H band into a small beaker
to a calibration curve. The latter relates containing a few milliliters of the metha-
apparent ratios of DV/MV Mg-Proto exci- nol:acetone mixture. This is followed by
tation amplitudes to known ratios of transfer of the slurry to a conical centrifuge
DV/MV Mg-Proto concentrations. The tube with a Pasteur pipet and centrifuga-
calibration curve is constructed as follows: tion at 1800× g for 1 minute on a table top
first, known concentrations of DV and centrifuge. The supernatant containing
MV Mg-Proto dissolved in diethyl ether Mg-Proto, with or without Proto, is
are mixed in different proportions. Second, removed using a Pasteur pipet. The propor-
fluorescence excitation spectra are recorded tions of DV/MV Mg-Proto components
on every mixture at 77 K as described can be determined by spectrofluorometry
above. Third, the apparent fluorescence in diethyl ether at 77 K as described in sec-
excitation ratios at 424/417 nm are plotted tion 5.2. The Mg-Proto pool is transferred
against the authentic concentration ratio to ether, as described in section 3.2, after
for every mixture. In this manner, DV and dilution with a small volume of HEAR.
MV Mg-Proto can be determined with a The ether extract should be washed with a
precision of about 5% to 6% (63). 0.5 M solution of KH2PO4 adjusted to pH
4.8 prior to 77 K spectroscopy.
5.3. Chromatographic Separation of
Mg-Proto 5.3.2. Chromatographic Separation of
DV Mg-Proto from MV Mg-Proto
5.3.1. Chromatographic Separation on on Thin Layers of Polyethylene
Thin Layers of Silica Gel H
Separation of the purified Mg-Proto
The mixed DV-MV Mg-Proto pool can pool into DV and MV components can be
be separated from monocarboxylic Mg-por- achieved on thin layers of polyethylene
phyrins on thin layers of silica gel H (12). developed in 90% aqueous acetone (12).
127
128

C.A. Rebeiz
Table 2. Spectrofluorometric and Spectrophotometric Equations Used to Deconvolute Spectral Band Overlaps

Compound Solvent DL Eq Equation

DV Mg-Proto (E424 F591) Diethyl ether 0.1 3 1.014 F (E424 F591) - 0.106. F (E417 F587)
MV Mg-Proto (E417 F587) Diethyl ether 0.1 4 1.013 F (E417 F587) - 0.131. F (E424 F591)
Pchlide a (F638 E440) HEAR 0.1 5 1.0080 F (F638 E440) - 0.0141 F (F675 E440)
- 0.0197 F (F633 E400) + 0.0028 F (F622 E440)

DV Pchlide a (E453 F625) Diethyl ether 0.1 6 1.061 F (E451 F625) - 0.068 F (E437 F625)
1.3307

MV Pchlide a (E437 F625) Diethyl ether 0.1 7 1.060 F (E437 F625) - 0.964 F (E451 F625)
0.8056

Pchlide a (623) Diethyl ether – 8 41.10 (Abs 623) - 4.93 (Abs 663) - 4.93 (Abs 644)
Pchlide a (626) 80% Acetone – 9 33.20 (Abs 626) - 4.48 (Abs 663) - 7.58 (Abs 645)
MV Pchlide b (E463 F643) Diethyl ether 0.1 10 1.0022 F (E463 F643) - 0.0507 F (E455 F660)
MV Chlide b (E455 F660) Diethyl ether 0.1 11 1.0022 F (E455 F660) - 0.0438 F (E463 F643)
MV Pchlide b (F643 E443) Diethyl ether 0.1 12 1.04 F (F643 E463) - 0.54 F (F635 E440)
Pchlide a (F635 E440) Diethyl ether 0.1 13 1.04 F (F635 E440) - 0.08 F (F643 E463)
MV Chlide a (E433 F674) HEAR 0.2 14 [1.2600 F (E433 F674) - 0.1661 F (E412 F674)
- 0.0894 F (E460 F660) - 0.2796 F (E438 F 660)]
- 0.1 Chlide a (E433 F674)

DV Chlide a (E458 F674) Diethyl ether 0.1 15 1.0205 F (E458 F674) - 0.0557 F (E447 F674)
MV Chlide a (E447 F674) Diethyl ether 0.1 16 1.0205 F (E447F674) - 0.3749 F (E458 F674)
Chlide a (663) Diethyl ether – 17 12.162 (Abs 663) - 1.08 (Abs 644) - 0.32
(Abs 624)

Chlide a (663) 80% Acetone – 18 14.1803 (Abs 663) - 2.9099 (Abs 645) - 0.2238
(Abs 626)
MV Chlide b (E460 F660 ) HEAR 0.2 19 [1.0520 F (E460 F660) - 0.1450 F (E438 F660)
- 0.0551 F (E433 F674) - 0.0030 F (E412 F674)]
+ 0.1810 Chlide b (E460 F660)
Table 2, continued.

Compound Solvent DL Eq Equation

DV Chlide b (E498 F666) Diethyl ether 0.1 20 1.0184 F (E498 F666) - 0.0425 F (E475 F660)
MV Chlide b (E475 F660) Diethyl ether 0.1 21 1.0185 F (E475 F660) - 0.4421 F (E498F666)
Chlide b (644) Diethyl ether – 22 20.96 (Abs 644) - 3.31 (Abs 663) - 0.59 (Abs 624)
Chlide b (645) 80% Acetone – 23 26.0064 (Abs 645) - 4.6613 (Abs 6663) - 0.3636
(Abs 626)

MV Pheo(bide) a (E412 F674) HEAR 0.2 24 [1.132 F (E412 F674) - 0.9713 F (E433 F674)

Analysis of Intermediate and End Products of Chlorophyll


- 0.0682 F (E460 F660) + 0.06496 F(E438 F 660)]
+ 0.0682 Pheo(bide) a (E412 F674 )

MV Pheo(bide) b (E438 F660) HEAR 0.2 25 [1.179 F (E438 F660) - 0.4033 F (E460 F660)
- 0.541 F (E433 F674) + 0.04873 F (E412 F 674)]
+ 0.095 Pheo(bide) b (E438 F660)

E = excitation wavelength; F = emission wavelength; Abs = absorbance wavelength. The (E…F…) terminology is used to refer to equations that use exci-
tation spectra, the (F….E…) terminology is used to refer to equations that use emission spectra, while the Abs terminology is used in equations that use
absorbance spectra. For example: (E424 F591) = Soret excitation amplitude at 424 nm recorded at an emission wavelength of 591 nm; (F638E440) =
fluorescence emission amplitude at 638 nm elicited by excitation at 440 nm; Abs 663 = absorbance at 663 nm. DL = detection limit (pmol/mL). Eq =
equation number. See section 3.1, Equation 1 for a fully described example using figures for protoporphyrin IX.
129
C.A. Rebeiz

In this solvent, DV and MV Mg-Proto coefficient that allows the quantitative


migrate with approximate Rfs of 0.58 and determinations of dilute solutions. Since
0.66, respectively. The separated bands can many other pigments such as carotenoids,
be eluted by scraping into a small beaker Chls, and Pchlides absorb light in this
containing diethyl ether and centrifugation spectral region, it is important to use a
of the slurry to recover the diethyl ether highly purified DV Mg-Proto fraction. It is
supernatant. The diethyl ether fractions also possible to use DV Mg-Proto
should be washed with a 0.5 M solution of absorbance in the red region of the spec-
KH2PO4 adjusted to pH 4.8 prior to 77 K trum, at about 550 or 589 nm, for quanti-
spectroscopy as in section 3.2. tative determinations. At these wave-
lengths, the molar extinction coefficient is
5.4. Quantitative Determination of very low, and quantitative determinations
Purified DV and MV Mg-Proto require more concentrated solutions. The
molar extinction coefficients are related to
Purified DV and MV Mg-Proto can be DV Mg-Proto absorbance and concentra-
determined in any solvent from room tem- tion by Beer's law (Equation 2). Table 1
perature emission spectra elicited by excita- reports absorption maxima and molar
tion close to their Soret absorbance maxi- extinction coefficient values for DV Mg-
ma between 417 and 424 nm. As described Proto in various solvents.
in section 3.4.1 for purified Proto, a cali- At room temperature, MV-Mg Proto
bration curve using known amounts of DV exhibits a 3-banded absorption spectrum
or MV Mg-Proto should be constructed in similar to that of DV Mg-Proto. The
order to convert fluorescence data to Mg- absorption maxima are blue-shifted however
Proto concentrations. in comparison to DV Mg-Proto. In diethyl
The concentration of purified DV Mg- ether at room temperature, the Soret
Proto solutions can be determined in a absorption maximum is observed at 412
variety of organic solvents by room tem- nm, and the α and β absorption bands
perature absorbance spectroscopy. The pro- exhibit maxima at 546 and 586 nm (12). To
cedure is only limited by the availability of our knowledge, precise molar extinction
molar extinction coefficients for the sol- coefficients for MV Mg-Proto have not
vent under consideration. Such determina- been determined. As an approximation, the
tions are usually used for the preparation of molar extinction coefficients of DV Mpe
DV Mg-Proto stock solutions of known may be used at the MV Mg-Proto maxima.
concentrations.
At room temperature, DV-Mg Proto
exhibits a 3-banded absorption spectrum 6. ANALYSIS OF MG-
in various organic solvents with a very PROTOPORPHYRIN IX
strong Soret absorption band between 360 MONOMETHYL ESTER
and 440 nm and 2 absorption bands (the
α and β bands) of about equal intensity The Mg-Proto monomethyl ester (Mpe)
between 530 and 610 nm (22). The most pool is a mixed DV-MV monocarboxylic
suitable wavelength for the quantitative tetrapyrrole pool (Figure 4). The same
determination of DV Mg-Proto is at its molar extinction coefficients (Table 1) and
Soret absorbance maximum, which varies Equations (Table 2) used for Mg-Proto are
from 417 to 420 nm depending on the sol- also used for Mpe determination. What
vent. At the Soret absorbance maximum, differs is the extraction of the two pools,
DV Mg-Proto has a large molar extinction since Mpe is a less polar monocarboxylic
130
Analysis of Intermediate and End Products of Chlorophyll

tetrapyrrole, while Mg-Proto is a more Separation of the crude (i.e., prior to sil-
polar dicarboxylic tetrapyrrole. ica gel H purification) or silica gel H-puri-
fied Mpe pool into DV and MV compo-
6.1. Quantitative Determination of Mpe nents can be achieved on thin layers of
in Crude Extracts by polyethylene developed in 90% aqueous
Spectrofluorometry acetone in darkness as described in section
5.3.2. In this solvent, DV and MV Mpe
Upon tetrapyrrole extraction from plant migrate with approximate Rfs of 0.54 and
tissues and partitioning of the pigments 0.68, respectively.
between hexane and acetone (sections 2.1
and 2.2), the mixed DV-MV Mpe pool 6.3. Quantitative Determination of
passes into the HEAR along with other Purified DV and MV Mpe
dicarboxylic and monocarboxylic tetra-
pyrroles. Since Mg-Proto and Mpe exhibit As in the case of DV and MV Mg-
similar spectrophotometric and fluores- Proto, the concentration of purified DV
cence properties, it is necessary to correct and MV Mpe can be determined in a vari-
for the presence of Mg-Proto. In a first ety of organic solvents either by fluores-
step, the total Mg-Proto plus Mpe content cence or absorbance spectroscopy, using
is determined on an aliquot of HEAR as the same Equations and molar extinction
described in section 5.1 for Mg-Proto. coefficients.
Next, the Mpe pool is extracted into
diethyl ether as described in section 3.2 for
monocarboxylic tetrapyrroles. The Mg- 7. ANALYSIS OF MG-
Proto content is then determined on the PROTOPRPHYRIN IX DIESTER
ether-extracted HEAR fraction as
described in section 5.2. The amount of The Mg-Protoporphyrin IX diester
Mpe is calculated by subtracting the (Mpde) pool (35) is a mixed DV-MV fully
amount of Mg-Proto from the total esterified tetrapyrrole pool (Figure 3). The
amount of Mg-Proto plus Mpe. proportion of DV and MV Mpde appears
Similarly, DV and MV Mpe are deter- to depend upon the plant species (12). The
mined, as for Mg-Proto (section 5.2), after same equations used for the determination
transfer of the Mpe pool to diethyl ether. of Mg-Proto and Mpe are also used for
Mpde determination. What differs are the
6.2. Chromatographic Separation of extraction procedures, since Mpde is an
Mpe from other Tetrapyrroles apolar fully esterified tetrapyrrole.

The mixed DV-MV Mpe pool can be 7.1. Quantitative Determination of


separated from other tetrapyrroles as Mpde in Crude Extracts by
described section 5.3.1 for Mg-Proto. Mpe Spectrofluorometry
moves with an Rf of about 0.31, just ahead
of Pchlide a and Chlide (4), while Mg-Proto Upon tetrapyrrole extraction from plant
moves with an Rf of about 0.09. Mpe is tissues and partitioning of the pigments
eluted in methanol:acetone (4:1 vol/vol) as between hexane and acetone, the mixed
described for Mg-Proto. The proportions of DV-MV Mpde pool passes into the apolar
DV/MV Mpe components are determined hexane phase along with other fully esteri-
by 77 K spectrofluorometry in washed fied tetrapyrroles. It is possible to deter-
diethyl ether as described in section 5.2. mine the concentration of the mixed DM-

131
C.A. Rebeiz

MV Mpde pool in the presence of other macrocycle. Gas chromatographic mass


fully esterified tetrapyrroles. This is best spectroscopic analysis of these long chain
achieved in etiolated tissues devoid of Chl fatty alcohols did not result in an elucida-
or in greening tissues containing only small tion of their chemical structure (35). It is
amounts of Chl. If large amounts of Chl possible, however, to separate the various
are present, prior separation of the Mpde Mpde components by HPLC. Successful
pool from Chl is required (see below). separation was achieved on a 20-cm
To determine the amount of Mpde, an Spherisorb ODS (Thermo Separation
aliquot of the hexane extract is dried under Products, San Jose, CA, USA) (5 µm) col-
a stream of N2 gas at ice bucket tempera- umn eluted isocratically with water:
ture. The residue is dissolved in 80% ace- methanol:acetone (5:85:10 vol/vol/vol) at
tone and monitored by room temperature a flow rate of 1 mL/minute. In this system,
emission spectrofluorometry as described standard Mg-Proto dimethyl ester exhibits
for Mg-Proto in section 5.1. Alternatively, a retention time of about 3 minutes, while
if large amounts of Mpde are present, a the Mpde pool separates into 3 major com-
very small aliquot of the hexane extract can ponents with retention times of about 9.6,
be diluted with 80% acetone prior to spec- 10.9, and 12.6 minutes (35).
trofluorometric determination.
Similarly, the amounts of DV and MV 7.4. Chromatographic Separation of
Mpde are determined from the total DV Mpde from MV Mpde
amount of Mpde measured at room tem-
perature and the DV/MV Mpde ratio Complete separation of the DV and
determined at 77 K (section 5.2). MV Mpde components has so far met with
limited success (Rebeiz, unpublished).
7.2. Separation of Mpde on Thin Layers
of Silica Gel H
8. ANALYSIS OF
The mixed DV-MV Mpde pool can be PROTOCHLOROPHYLLIDE a
separated from other fully esterified Mg-
porphyrins on thin layers of silica gel H The Pchlide a pool is a mixed highly
(35) as described in section 5.3.1 for Mg- dynamic DV-MV tetrapyrrole pool (Figure
Proto. The mixed DV-MV Mpde and Pch- 4). The DV and/or MV content of this
lide a E pools move with respective Rfs of pool vary widely depending on the green-
about 0.75 and 0.82. If present, Chl moves ing group affiliation of the plant species
in between Mpde and Pchlide a E. In this (24) and the phase of the photoperiod
system, dicarboxylic tetrapyrroles stay at (1,16). Prior to 1979, the possible exis-
the origin while monocarboxylic tence of a DV Pchlide a component in the
tetrapyrroles, such as Mpe, move with an Pchlide a pool of algae and higher plants
Rf of about 0.28 to 0.31 (35). Mpde can was not recognized. As a consequence, pre-
be eluted with diethyl ether. 1980 studies of the Pchlide a pool may
have overlooked the presence of substantial
7.3. Separation of Various Mpdes by amounts of DV Pchlide a.
HPLC Two different pools of MV Pchlide a,
formed via different Chl a biosynthetic
The Mg-Proto diester pool appears to be pathways, coexist in etiolated and green
esterified by a number of long chain fatty plants (44). One pool is formed from DV
alcohols probably at position 7 of the Pchlide a (Figure 1, pathway 2) by reduc-
132
Analysis of Intermediate and End Products of Chlorophyll

tion of the vinyl group at position 4 to mixed DV-MV Pchlide a in the presence of
ethyl, a reaction catalyzed by [4-vinyl]- other di- and monocarboxylic tetrapyrroles
Pchlide a reductase (65). The other is can be determined without further purifi-
formed by conversion of MV Mpe to MV cation, after correction for spectral band
Pchlide a (64), a reaction catalyzed by a overlap.
putative MV cyclopentanone ring syn- This is achieved by exciting the
thetase (Figure 1, pathway 9). MV Pchlide tetrapyrroles in the HEAR fraction at 440
a formation from DV Pchlide a via path- nm, i.e., close to the Soret excitation maxi-
way 2 takes place in both dark divinyl mum of Pchlide a and Chlides a, and by
(DDV)-light-dark divinyl (LDDV), i.e., recording an emission spectrum between
DDV-LDDV, plant species such as cucum- 580 to 700 nm. A second emission spec-
ber and in dark monovinyl (DMV)-light- trum is elicited by excitation at 400 nm,
dark monovinyl (LDMV), i.e., DMV- i.e., close to the Soret excitation maxima of
LDMV, plant species such as corn wheat Proto and Copro. The net fluorescence
and barley. Conversely, MV Pchlide a for- amplitude due only to Pchlide a emission
mation from MV Mpe via pathway 9 is deconvoluted from the Copro, Proto,
appears to predominate in DMV-LDMV and Chlide a emissions with the use of
plant species (1). Equation 5 (Table 2) (45). The Pchlide a
DV Pchlide a occupies a central position net fluorescence amplitude is converted to
in the DV carboxylic Chl a biosynthetic Pchlide a concentration by reference to a
pathway as a precursor of MV Pchlide a standard calibration curve. The latter is
and DV Chlide a (Figure 1, pathways 1 prepared as described in section 3.1 from
and 2). In contrast to MV Pchlide a, only standard Pchlide a solutions of known con-
one pool of DV Pchlide a, which is part of centrations and from their fluorescence
biosynthetic pathway 1, exists in etiolated emission amplitudes. Pchlide a can be
and green plants (44). determined with a precision of about 6%
(45). Minimum detection levels are about
8.1. Quantitative Determination of 0.1 pmol/mL.
Pchlide a in Crude Extracts by
Spectrofluorometry 8.2. Quantitative Determination of MV
and DV Pchlide a in Crude Extracts
Upon tetrapyrrole extraction from plant by Spectrofluorometry
tissues and partitioning of the pigments
between hexane and acetone (sections 2.1 It is necessary to extract the Pchlide a
and 2.2), the mixed DV-MV Pchlide a pool from the HEAR fraction into less
pool passes into the HEAR fraction along polar solvents such as diethyl ether prior to
with other dicarboxylic and monocar- structural studies or determination of MV
boxylic tetrapyrroles. In HEAR at room and DV Pchlide a (section 3.2).
temperature, DV and MV Pchlide a exhib- As in the case of other MV and DV Mg-
it similar emission maxima at 637 to 639 tetrapyrroles, the use of low temperature
nm. In this region of the spectrum, the (77 K) is essential to differentiate between
main interfering tetrapyrrole is DV Proto, MV and DV Pchlide a. At this tempera-
which emits at 632 to 633 nm. Lesser ture, due to considerable fluorescence exci-
interference by the long wavelength emis- tation and emission band narrowing, MV
sion tail of Copro, and the short wave- Pchlide a exhibits a split Soret excitation
length emission tail of Chlide a may also band with maxima at 437 and 443 nm and
be encountered. The concentration of the an emission maximum at 625 nm. Like-
133
C.A. Rebeiz

wise, DV Pchlide a exhibits split Soret exci- next calculated. That ratio is converted to
tation maxima at 443 and 451 nm and an an authentic DV/MV Pchlide a concentra-
emission maximum at 625 nm (63). In this tion ratio by reference to a calibration
case too, determination of the amounts of curve (see section 5.2). The latter relates
DV and MV Pchlide a is a 2-step process. apparent ratios of DV/MV Pchlide a exci-
First, the total amount of DV plus MV tation amplitudes to known ratios of
Pchlide a in the HEAR fraction is deter- DV/MV Pchlide a concentrations. In this
mined at room temperature exactly as manner, DV and MV Pchlide a can be
described in section 8.4. Next, the determined with a precision of 4% to 7%
DV/MV ratio of the Pchlide a pool is (63).
determined at 77 K in diethyl ether. The
amount of DV and MV Pchlide a is then 8.3. Chromatographic Separation of
calculated from the total amount of Pch- Pchlide a
lide a and from the DV/MV Pchlide a
ratio (63). 8.3.1. Chromatographic Separation on
To calculate the DV/MV Pchlide a Thin Layers of Silica Gel H
ratio, one sharp excitation spectrum needs
to be recorded at 77 K in diethyl ether (see The mixed DV-MV Pchlide a pool can
section 5.2). The 77 K excitation spectrum be separated from other monocarboxylic
is recorded from 380 to 500 nm by posi- Mg-porphyrins on thin layers of silica gel
tioning the emission monochromator at H as described in section 5.3.1 (12). Pch-
the emission maximum of MV and DV lide a moves with an Rf of about 0.2 just
Pchlide a at 625 nm. In this spectrum, the behind Mpe (Rf of 0.31) (4). Pchlide a is
short wavelength MV Pchlide a Soret exci- eluted in methanol:acetone (4:1 vol/vol) as
tation maximum at 437 nm will appear as described for Mg-Proto. The proportions
a sharp distinct peak. Likewise, the long of DV/MV Pchlide a components are
wavelength DV Pchlide a Soret excitation determined by spectrofluorometry in
maximum at 451 nm will also appear as a washed diethyl ether extracts at 77 K as
sharp distinct peak (63). The MV Pchlide described in section 8.4.
a long wavelength excitation maximum
and the DV Pchlide a short wavelength 8.3.2. Chromatographic Separation of
excitation maximum will overlap com- DV Pchlide a from MV Pchlide a
pletely and appear as a single excitation
maximum at 443 nm. For calculation of Separation of the of the crude (i.e., prior
the DV/MV Pchlide a ratio, only the Soret to silica gel H purification) or silica gel H-
excitation maxima at 437 and 451 nm are purified Pchlide a pool into DV and MV
used. Because of complete overlap of the components can be achieved on thin layers
DV and MV Soret excitation maxima at of polyethylene developed in 90% aqueous
443 nm, this wavelength cannot be used in acetone in darkness as described in section
the calculations. The net fluorescence exci- 5.3.2 (11). In this solvent, DV and MV
tation amplitudes of DV and MV Pchlide Pchlide a migrate with approximate Rfs of
a are deconvoluted and calculated with the 0.55 and 0.75, respectively. The separated
use of Equations 6 and 7 (63). The ratio of bands are eluted in diethyl ether.
the deconvoluted DV Pchlide a excitation Partial separation of DV and MV Pch-
amplitude to the deconvoluted MV Pch- lide a by HPLC on a polyvinyl alcohol
lide a excitation amplitude, referred to as polymer column has been described by
the apparent excitation amplitude ratio, is Shioi and coworkers (61). The separation
134
Analysis of Intermediate and End Products of Chlorophyll

was performed on a 250 × 4.6 mm ODP- exhibits a 5-banded absorption spectrum


50 Asahipak column (Showa Denko, in various organic solvents. The wave-
Tokyo, Japan) packed with octadecyl silica lengths of absorption maxima depend
polyvinyl alcohol polymer (ODP). Spheri- upon the solvent. A strong Soret
cal particle size was 5 µm, pore diameter absorbance band with an absorption maxi-
250 Å, and carbon loading 17%. After mum at 432 to 434 nm and a less intense
injection of a 20-µL aliquot, elution was at red absorbance band with a maximum at
27°C with a linear mobile-phase gradient 622 to 629 nm are observed (Table 1). The
from methanol:1 M ammonium acetate Soret and red absorbance maxima can both
(8.2:2 vol/vol) to acetonitrile:acetone (7:3 be used for quantitative determinations. If
vol/vol). The duration of the gradient was the Soret absorbance is used, a highly puri-
28 minutes, after which the final composi- fied sample free of carotenoids and other
tion of the mobile phase was maintained tetrapyrroles is required. Contamination
isocratic for an additional 5 to 10 minutes. by carotenoids does not interfere however
Resolution of MV from DV Pchlide a is, with quantitative measurements when the
however, only partial with considerable red absorbance maximum is used. The
overlap of the two Pchlides a. molar extinction coefficients are related to
DV Pchlide a absorbance and concentra-
8.4. Quantitative Determination of tion by Beer's law (Equation 2). Table 1
Purified DV and MV Pchlide a reports absorption maxima and molar
extinction coefficient values for MV Pch-
Purified DV and MV Pchlide a can be lide a in various solvents.
determined in organic solvents from room At room temperature, DV Pchlide a
temperature emission spectra elicited by exhibits an absorbance spectrum similar to
excitation close to their Soret absorbance that of MV Pchlide a. Although the red
maxima between 430 and 440 nm. In each absorbance maximum of MV and DV
case, a calibration curve using known Pchlide a are identical, the Soret ab-
amounts of DV or MV Pchlide a should be sorbance maximum of DV Pchlide a is red-
constructed in order to convert fluores- shifted by about 5 nm with respect to that
cence data to Pchlide a concentrations. At of MV Pchlide a. Quantitative determina-
each concentration, an emission spectrum tion of DV Pchlide a by absorbance spec-
elicited by excitation at the chosen Soret troscopy are performed as described for
excitation wavelength should be recorded. MV Pchlide a. DV Pchlide a molar extinc-
A calibration curve relating DV or MV tion coefficient values are reported in Table
Pchlide a concentration to fluorescence 1.
emission amplitudes of the pure DV and
MV Pchlide a solutions at their emission 8.5. Quantitative Determination of
maxima should be constructed as described Pchlide a in the Presence of
in section 3.4.1. for purified Proto. Chlorophyll(ides) by
Alternatively, the concentration of puri- Spectrophotometry
fied MV Pchlide a solutions can be deter-
mined in a variety of organic solvents by Under certain conditions, as during the
room temperature absorbance spec- early phases of greening of etiolated tissues,
troscopy. Such determinations are usually it is possible to determine the amount of
used for the preparation of MV Pchlide a the mixed MV/DV Pchlide a pool in the
stock solutions of known concentrations. presence of Chl a and b and/or Chl(ide) a
At room temperature, MV Pchlide a and b by absorbance spectroscopy. The
135
C.A. Rebeiz

technique uses simultaneous equations to 9. ANALYSIS OF MV PCHLIDE b


correct for Pchlide a and Chl(ide) a and b
absorbance band overlap, although these The Pchlide b pool is a MV tetrapyrrole
cannot be used when the ratio of Chl(ide) pool (Figure 4) (56). In green cucumber
a/Pchlide a exceeds 6.0. cotyledons, its concentration varies from
In diethyl ether, the amount of Pchlide 400 to 800 pmol per 100 mg of tissue pro-
a in the presence of Chl(ide) a and b can tein. MV Pchlide b is not formed and does
be determined from Equation 8 (Table 2). not accumulate in etiolated tissues (25). It
has been proposed that of the 8 possible
Equation 8. biosynthetic pathways capable of MV Chl
b formation only 2 pathways (biosynthetic
Pchlide a (623) = 41.10 (Abs623) - 4.93 pathways 3 and 10) proceed via MV Pch-
(Abs663) - 4.93 (Abs644). lide b (44). The photoconversion of MV
Where: Pchlide a (623) = Amount of Pchlide b to Chlide b has been recently
Pchlide a in nmoles/mL determined from demonstrated in vitro in barley etioplast
the absorbance of the pigment mixture at membranes (54). In reconstituted model
623 to 624nm. systems, Pchlide oxidoreductase A (POR
Abs663 = Absorbance of the pigment A) appears to bind preferentially MV Pch-
mixture at 663 nm. This factor corrects for lide b (51). This may indicate that in
the Chl(ide) a overlap. DDV-LDDV green tissues capable of Pch-
Abs644 = Absorbance of the pigment lide b biosynthesis, POR A may be
mixture at 644 nm. This factor corrects for involved in the conversion of Pchlide b to
the Chl(ide) b overlap. Chlide b.
Equation 8 was adapted from that
reported by Koski and Smith (29). In
deriving Equation 8, a molar extinction 9.1. Quantitative Determination of
coefficient of 24 457.104 for Pchlide a in Pchlide b in Crude Extracts by
diethyl ether, at 623 nm, was used. This Spectrofluorometry
molar extinction coefficient was calculated
from the molecular weight of MV Pchlide Upon tetrapyrrole extraction from plant
a (i.e., 613) and the specific absorption tissues and partitioning of the pigments
coefficient of MV Pchlide a (39.9), which between hexane and acetone (sections 2.1
was erroneously reported as that of MV and 2.2), the MV Pchlide b pool passes
Pchlide a phytyl ester by Koski and Smith into the HEAR fraction along with Pchlide
(30). In calculating the Pchlide a molar a and other dicarboxylic and monocar-
extinction coefficient, the following rela- boxylic tetrapyrroles. In 80% acetone or in
tionship was used: HEAR at room temperature, MV Pchlide
b exhibits a broad emission maximum at
Molar extinction coefficient (ε) = around 641 nm and a Soret excitation
Specific absorption coefficient (α) maximum at 447 nm (56). The emission
(molecular weight) maximum, at 641 nm, overlaps with the
In 80% acetone, the amount of Pchlide 77 K emission maximum of hexacoordi-
a in the presence of Chl(ide) a and b can nated Pchlide a (640 nm) (13) and should
be determined from Equation 9 (Table 2) not be confused with it. At 77 K in diethyl
in a manner similar to that described for ether, it exhibits a Soret excitation maxi-
Equation 8, if the ratio of Chl a/Pchlide a mum at 463 nm. Upon excitation at 463
is less than 6. nm, it exhibits a pronounced red emission
136
Analysis of Intermediate and End Products of Chlorophyll

maximum at 643 nm. The detailed spectral by positioning the emission monochroma-
properties of MV Pchlide b have been pre- tor at 660 nm, the emission maximum of
viously described (56). MV Chlide b in diethyl ether at 77 K. In
this spectrum, the MV Chlide b Soret exci-
9.1.1. Determination of Pchlide b in Crude tation maximum at 475 nm will also
Extracts Containing Chl(ide) a and b appear as a distinct peak. For deconvolu-
tion and calculation purposes, the best dis-
Under natural conditions, MV Pchlide crimination between MV Pchlide b and
b can only be observed in green tissues or MV Chlide b is accomplished by deconvo-
during the advanced stages of greening of luting the net Soret excitation fluorescence
etiolated tissues. Spectrofluorometric amplitudes of the diethyl ether extract at
determination of MV Pchlide b in the 455 and 463 nm, respectively. The net flu-
presence of Pchlide a and Chlide a and b is orescence excitation amplitudes of MV
best achieved in diethyl ether at 77 K. Pchlide b and MV Chlide b at 463 and
However, as mentioned for other MV and 455 nm are deconvoluted and calculated
DV Mg-porphyrins, 77 K spectroscopic with the use of Equations 10 and 11 (25).
determinations cannot be used alone for The apparent MV Pchlide b/MV Chlide b
quantitative measurements. A 2-step tech- excitation ratio is converted to an authentic
nique using room temperature and 77 K MV Pchlide b/MV Chlide b concentration
spectrofluorometry has therefore been ratio by reference to a calibration curve
developed. In a first step, the amount of that relates apparent ratios of MV Pchlide
MV Chlide b in HEAR is determined by b/MV Chlide b excitation amplitudes to
room temperature spectrofluorometry as known ratios of MV Pchlide b/MV Chlide
described in section 13.1. In a second step, b concentrations (see section 5.2). Using
the MV Chlide b/MV Pchlide b ratio is this method MV Pchlide b can be deter-
determined at 77 K in diethyl ether. The mined with a precision of about 10% (25).
amount of MV Pchlide b is then calculated
from the total amount of Chlide b as deter- 9.1.2. Determination of Pchlide b in
mined at room temperature and from the Crude Extracts Containing Pchlide a
MV Chlide b/MV Pchlide b ratio as deter-
mined at 77 K in diethyl ether (25). Experimental conditions may arise
To calculate the MV Chlide b/MV Pch- where MV Pchlide b needs to be deter-
lide ratio, two sharp 77 K excitation spec- mined in the presence of MV Pchlide a
tra in diethyl ether need to be recorded. and in the absence of MV Chlide b. This
First, the HEAR fraction is extracted with situation may be encountered in model
diethyl ether (section 3.2). To optimize the systems prepared from etiolated tissues and
detection of MV Pchlide b, the first 77 K synthetic exogenous MV Pchlide b
excitation spectrum is recorded from 380 (51,54). Under such conditions, quantita-
to 500 nm in the ether extract by position- tive determination of MV Pchlide by spec-
ing the emission monochromator at 643 trofluorometry can be achieved on an
nm, the emission maximum of MV Pch- aliquot of HEAR and on a diethyl ether
lide b in diethyl ether at 77 K. In this spec- extract of HEAR. In this manner, losses
trum, the MV Pchlide b Soret excitation incurred during purification of small
maximum at 463 nm will appear as a sharp amounts of MV Pchlide b are avoided.
distinct peak. To optimize the detection of Again, a 2-step technique using room tem-
MV Chlide b, a second 77 K excitation perature and 77 K spectrofluorometric
spectrum is recorded from 380 to 500 nm determinations are used. In a first step, the
137
C.A. Rebeiz

amount of Pchlide a in the HEAR fraction known ratios of Pchlide a/MV Pchlide b
is determined by room temperature spec- concentrations (see section 5.2). In this
trofluorometry as described in section 8.4. manner, MV Pchlide b can be determined
In a second, step the Pchlide a/MV Pchlide with a precision of about 7% (25).
b ratio is determined at 77 K in diethyl
ether. The amount of MV Pchlide b is then 9.2. Chromatographic Separation of
calculated from the total amount of Pch- Pchlide b from other Tetrapyrroles
lide a as determined at room temperature
and from the Pchlide a/MV Pchlide b ratio A promising HPLC technique has been
as determined at 77 K in diethyl ether described by Scheumann et al. for synthet-
(25). ic MV Pchlide b (54). Separation of MV
To calculate the Pchlide a/MV Pchlide Pchlide b from MV Pchlide a is achieved
b ratio, two sharp 77 K emission spectra in on a C-18 reverse phase silica gel column
diethyl ether need to be recorded. First, the (Hypersil ODS, 5 µm particle size; Shan-
Pchlide a and b pools are extracted from don Lipshaw, Pittsburgh, PA, USA). Elu-
the HEAR fraction with diethyl ether as tion is at a flow rate of 1.0 mL/minute,
described in section 3.2 for monocar- with a step gradient starting with 34% 25
boxylic tetrapyrroles. To optimize the mM aqueous NH4OAC, 15% acetone,
detection of MV Pchlide b, the first 77 K and 51% methanol, and increasing to 16%
emission spectrum is recorded from 580 to water, 60% acetone, and 24% methanol
700 nm by excitation at the Soret excita- within 20 minutes. Finally, the gradient is
tion maximum of MV Pchlide b at 463 held at 100% acetone for 14 minutes. In
nm. In this spectrum, MV Pchlide b emis- this system, MV Pchlide b and a are eluted
sion would appear as a sharp distinct peak with respective retention times of about 9
at 643 nm. To optimize the detection of and 15 minutes.
MV Pchlide a, a second 77 K emission
spectrum is recorded from 580 to 700 nm 9.3. Quantitative Determination of
by excitation at 440 nm. In this spectrum, Purified Pchlide b
MV Pchlide a would exhibit a sharp emis-
sion maximum at 625 nm and a broader Purified MV Pchlide b can be deter-
emission band between 632 and 650 nm. mined in any appropriate solvent from
The best discrimination between Pchlide a room temperature emission spectra elicited
and MV Pchlide b is accomplished by by excitation close to its Soret absorbance
deconvoluting the net emission fluores- maximum. In each case, a calibration curve
cence amplitudes of the crude diethyl ether using known amounts of MV Pchlide b
extract at 635 and 643 nm (25). should be constructed in order to relate
The net fluorescence emission ampli- MV Pchlide b concentration to fluores-
tudes of MV Pchlide b and Pchlide a at cence emission amplitudes of the pure MV
643 and 625 nm, respectively, are decon- Pchlide b solutions at its emission maxi-
voluted and calculated with the use of mum. A similar procedure using Soret
Equations 12 and 13 (25). Next, the calcu- excitation spectra instead of emission spec-
lated apparent Pchlide a/MV Pchlide b tra can also be used.
emission ratio is converted to an authentic Alternatively, absorbance spectroscopy
Pchlide a/MV Pchlide b concentration may be used. At room temperature, MV
ratio by reference to a calibration curve Pchlide b exhibits a 6-banded absorption
that relates apparent ratios of Pchlide spectrum in various organic solvents. The
a/MV Pchlide b emission amplitudes to wavelengths of absorption maxima depend
138
Analysis of Intermediate and End Products of Chlorophyll

on the solvent. In diethyl ether, a strong has been demonstrated, however, that the
Soret absorbance band with an absorption bulk of the Pchlide a E pool of cucumber
maximum at 442 nm and a less intense red cotyledons, which consists mainly of MV
absorbance band with a maximum at 630 Pchlide a E, is not formed from MV Pch-
nm are observed (Table 1). Table 1 reports lide a by esterification. Instead, MV Pchlide
absorption maxima and molar extinction a and MV Pchlide a E appear to be formed
coefficient values for MV Pchlide b phytyl in parallel from a common precursor at the
ester in various solvents. MV Pchlide b and level of Proto and/or Mg-Proto or Mpe
its esterified analog exhibit identical spec- (36). The MV Pchlide a GG, dihydroGG,
tral and molar extinction properties. The and tetrahydroGG may be considered
Soret and red absorption maxima can both intermediates on the way to the formation
be used for quantitative determinations. If of MV Pchlide a P.
the Soret absorbance is used, a highly puri- The DV Pchlide a pool is considered to
fied sample free of carotenoids and other originate from DV Mpde (Figure 1, path-
tetrapyrroles is required. Carotenoids do way 15) via a series of reactions similar to
not interfere with quantitative determina- those of MV Pchlide a E. Because of exper-
tions using the red absorbance maximum. imental difficulties, a precursor product
The molar extinction coefficients are relat- relationship between DV Mpde and DV
ed to MV Pchlide b absorbance and con- Pchlide a ester has not yet been demon-
centration by Beer's law (Equation 2). strated. The DV Pchlide a GG, DHGG,
and THGG may be considered intermedi-
ates on the way to the formation of DV
10. ANALYSIS OF PCHLIDE ESTER a Pchlide a E.
The protochlorophyllide ester (Pch-
lide E) a pool is a highly heterogeneous 10.1. Quantitative Determination of
fully esterified tetrapyrrole pool. It is pre- Pchlide a E in Crude Extracts by
sent in all etiolated and green tissues so Spectrofluorometry
far surveyed (Rebeiz, unpublished). It
consists of MV and DV Pchlide a E (Fig- Upon tetrapyrrole extraction from plant
ure 4) (10). tissues and partitioning of the pigments
The long chain fatty alcohols esterified between hexane and acetone (sections 2.1
at position 7 of the macrocycle are variable and 2.2), the mixed Pchlide a E pool pass-
and have been reported to consist of ger- es into hexane along with other fully ester-
anylgeraniol (GG), dihydroGG (DHGG), ified tetrapyrroles. Pchlide a and its ester
tetrahydroGG (THGG), and hexahy- exhibit identical spectrofluorometric and
droGG (i.e., phytol) in Scenedesmus obliqu- spectrophotometric properties, so quanti-
us (27), in etiolated and greening cucum- tative determination of Pchlide a E is
ber cotyledons (58), and in etiolated leaves exactly as described in section 8.1. In most
of kidney bean (59). cases, Pchlide a E in the hexane extract will
In the integrated Chl a/b pathway, the be contaminated by trace amounts of Pch-
MV Pchlide a E pool is considered to orig- lide a carried over from acetone to hexane.
inate from MV Mpde via biosynthetic This minor Pchlide a contamination can
pathway 13 (Figure 1). Because of experi- be eliminated by washing the hexane with
mental difficulties, a precursor product rela- ammoniacal acetone or by purification of
tionship between MV Mpde and MV Pch- Pchlide a E on thin layers of silica gel H as
lide a E has not yet been demonstrated. It described below.
139
C.A. Rebeiz

10.2. Chromatographic Separation of were separated on a 4 × 200 mm Nucleosil


Pchlide a E from other Mg- C18-reversed phase column, 5 µm particle
Porphyrins size (Macherey and Nagel). Elution was
with a linear gradient of 20% to 80% ethyl
The mixed DV-MV Pchlide a E pool acetate in 80% aqueous methanol for 30
can be separated from other fully esterified minutes at a flow rate of 1 mL/minute.
Mg-porphyrins on thin layers of silica gel Sharp separation of Pchlide a GG from
H as described in section 5.3.1 (10). The Pchlide a DHGG, THGG, and phytol was
Pchlide a E, Chl a and b, and Pchlide a achieved (27,60).
pools move with respective Rfs of about Most etiolated tissue of higher plants
0.9, 0.8, and 0.5. Pchlide a E is eluted with contains mainly MV and/or DV Pchlide
diethyl ether. a, lesser amounts of MV Pchlide a E, and
After separation on thin layers of silica only trace amounts of DV Pchlide a E. In
gel H, the Pchlide a E can be purified fur- most cases, it is possible to separate the
ther on polyethylene plates. The ether various Pchlide a esters with minimal
extract is washed with 0.5 M KH2PO4 manipulation, using an isocratic HPLC
buffer, pH 7.0, to remove traces of chro- solvent system. Fifty microliters of the
matographic solvents and dried under a crude acetone:ammonium hydroxide
stream of N2 gas. The dried residue is dis- extract (see section 2.1) of etiolated tissues
solved in diethyl ether and applied to thin is injected onto a Pecosphere 3 × 3C, C-
layers of polyethylene. The plates are 18 reverse phase, 4 × 0.5 cm column, and
allowed to dry completely by holding at eluted with an isocratic solvent system
room temperature for 10 minutes under that consists of water:acetone:methanol
subdued light before developing in the dark, (5:10:75 vol/vol/vol), at a rate of 1 mL/
at room temperature, in 2-propanol:acetone minute. The elutants are monitored by
(1:1 vol/vol) (10). MV and DV Pchlide a E on-line spectrofluorometry. The amounts
move with respective Rfs of 0.5 and 0.3. of eluting Pchlide a and various Pchlide a
The bands fluoresce weak red under UV E can be determined from a calibration
light of 366 nm and are eluted in diethyl curve using Pchlide a as a standard and
ether. The eluted bands are dried under a appropriate commercial software. With
stream of N2 gas before redissolving in this system, 4 Pchlide a E with retention
diethyl ether for spectroscopic analysis. times of about 8.2, 9.9, 12.2, and 15.1
Because of the low concentration of Pchlide minutes are quantitatively resolved (Rebeiz,
a E in most plant tissues, and because of unpublished).
low recoveries, the red fluorescence of the
separated MV and DV bands may be barely 10.3. Quantitative Determination of
visible. Nevertheless, the fluorescence of the Purified DV and MV Pchlide a E
eluted bands is readily detected by high-res-
olution spectrofluorometry. As was described for purified DV and
After separation of MV from DV Pch- MV Pchlide a in section 8.4, the concen-
lide a E, HPLC is the method of choice for tration of purified MV and/or DV Pchlide
separating various MV or DV Pchlide a a E can be determined either by spectroflu-
esters. For example, after separation of DV orometry or absorbance spectroscopy.
from MV Pchlide a E on Fractogel TSK The red absorbance maxima of MV and
DEAE-650 (Merk, Darmstadt, Germany), DV Pchlide a E are identical, but the Soret
the different MV and DV Pchlide a E of absorbance maximum of DV Pchlide a E is
dark-grown wheat roots and S. obliquus red-shifted by about 5 nm with respect to
140
Analysis of Intermediate and End Products of Chlorophyll

that of MV Pchlide a E. Quantitative 12. ANALYSIS OF


determination of purified DV Pchlide a E CHLOROPHYLLIDE a
by absorbance spectroscopy are performed
as described for MV Pchlide a E. Molar The Chlide a pool is a mixed highly
extinction coefficient for DV Pchlide a E dynamic DV-MV tetrapyrrole pool (Figure
are reported in Table 1. 5). The DV and/or MV content of this
pool vary widely depending on the green-
10.4. Determination of Pchlide a E in ing group affiliation of the plant species
the Presence of Chlorophyll(ides) (24), the phase of the photoperiod (9), and
a and b light pretreatment of the plant tissue (18).
DV Chlide a (Figure 5) occurs as a single
The same absorbance equations used for pool that occupies a central position in the
the determination of Pchlide a in the pres- integrated Chl a/b pathway as a precursor
ence of Chl(ide) a and b in diethyl ether of MV Chlide a (Figure 1, pathway 4), of
and 80% acetone (section 8.5) can be used DV Chlide b (Figure 1, pathway 7), and
for Pchlide a E determination in the pres- DV Chl a (Figure 1 pathway 1).
ence of Chl(ide) a and b. In contrast, 3 different pools of MV
Chlide a, formed via different Chl a biosyn-
thetic pathways, coexist in etiolated and
11. ANALYSIS OF MV PCHLIDE b green plants (44). One pool is formed by
ESTER photoreduction of MV Pchlide a via path-
way 2 (Figure 1). The second is formed
The Pchlide b E pool is a MV tetrapyr- from DV Chlide a via pathway 4 by conver-
role pool (Figure 4). Its occurrence in green sion of the vinyl group at position 4 to
plants was first reported by Shedbalkar et ethyl, a reaction catalyzed by (4-vinyl)-
al., but is less ubiquitous than that of MV Chlide a reductase (4VCR) (Figure 1) (13).
Pchlide b (57). The third is formed by conversion of MV
Upon tetrapyrrole extraction from plant Pchlide a via pathways 9 and 12 (Figure 1).
tissues and partitioning of the pigments
between hexane and acetone, MV Pchlide 12.1. Spectrofluorometric Determination
b E passes into hexane along with Pchlide of Chlide a in Crude Extracts
a E and Chl a and b. Since Pchlide b E Containing Mainly MV Chlide a
exhibits identical electronic spectroscopic
properties as MV Pchlide b, the same spec- Upon tetrapyrrole extraction from plant
trofluorometric equations used for MV tissues and partitioning of the pigments
Pchlide b determination can be used for between hexane and acetone (sections 2.1
Pchlide b E analysis (section 9.1). and 2.2), the mixed DV-MV Chlide a pool
Pchlide b E can be separated from other passes into HEAR along with other dicar-
tetrapyrroles by chromatography on thin boxylic and monocarboxylic tetrapyrroles.
layers of silica gel H in darkness at 4°C in In most higher and lower plants, except in
toluene:ethyl acetate:ethanol (8:2:2 prochlorophyte picoplankton and the Nec1
vol/vol/vol). It migrates with a Rf of about corn mutant where the DV forms abound,
0.56. It is eluted in diethyl ether and the Chlide a pool consists of MV Chlide a
rechromatographed in darkness at room (673 nm emission) and trace amounts of
temperature on thin layers of cellulose DV Chlide a (also 673 nm emission). It is
developed in ligroin (60–90):n-propanol usually accompanied by MV Chlide b (657
(99:1 vol/vol) (57). nm emission), MV pheophorbide a, i.e.,
141
C.A. Rebeiz

demetalated, de-esterified Chl a (673 nm mainly of DV Chlide a with or without


emission) and smaller amounts of MV smaller amounts of MV Chlide a. This situ-
Pheophorbide b (660 nm emission). It is ation prevails in the Nec1 corn mutant (6),
also accompanied by Pchlide a (638 nm in the primitive prochlorophyte picoplank-
emission), and in some cases by small ton of the subtropical waters of the North
amounts of Mg-Proto and Mpe (595 nm Atlantic as well as in the picoplankton of
emission). the euphonic zone of the world tropical and
Under these circumstances, it is possible temperate oceans, and the Mediterranean
to determine the amounts of MV Chlide a Sea, and in DV-LDDV plant species sub-
without prior purification by room temper- jected to alternating light/dark treatments
ature excitation spectrofluorometry. Since (18). Under these circumstances, total
the emission maxima of MV Chlide a and b Chlide a can be evaluated using Equation
and MV pheophorbide a and b fall in the 14 to calculate the net Soret fluorescence
red region of the spectrum, far away from excitation amplitude at 433 nm, i.e., 5 nm
the emission of other Mg-tetrapyrroles, it is below the DV Chlide a excitation maxi-
possible to record Soret excitation spectra mum at 438 nm. Next, the net fluorescence
with minimum Soret excitation overlap excitation amplitude of the DV Chlide a
from other Mg-tetrapyrroles. The concen- pool, at 433 nm, is converted to DV Chlide
tration of Chlide a can then be determined a concentration by reference to a standard
from the excitation spectra after correction calibration curve. The latter is prepared
for excitation band overlap. This is achieved from standard DV Chlide a solutions of
by recording 2 Soret excitation spectra in known concentrations and from their fluo-
HEAR at room temperature. One spectrum rescence excitation amplitudes at 433 nm.
is recorded at an emission wavelength near The latter are recorded at an emission max-
the emission maximum of MV Chlide a, at imum of 674 nm.
674 nm. The other spectrum is recorded at
an emission wavelength near the emission 12.3. Spectrofluorometric Determination
maximum of MV Chl b at 660 nm. The net of MV and DV Chlide a in Crude
Soret fluorescence excitation amplitudes Extracts
due only to MV Chlide a is deconvoluted
with the use of Equation 14 (7). The net Prior to structural studies or determina-
fluorescence excitation amplitude of MV tion of MV and DV Chlide a, it is neces-
Chlide a, as calculated from Equation 14, sary to extract the Chlide a pool from the
are converted to MV Chlide a concentra- HEAR fraction into diethyl ether as
tion by reference to a standard calibration described in section 3.2 for monocar-
curve relating MV Chlide a concentrations boxylic tetrapyrroles.
to fluorescence excitation amplitudes. In As in the case of other MV and DV Mg-
this manner, MV Chlide a can be deter- tetrapyrroles, the use of low temperature
mined with a precision of about 1% for (77 K) is essential to differentiate between
pure MV Chlide a to 24% for extracts con- MV and DV Chlide a. At 77 K, MV
taining only 6% Chlide a (7). Minimum Chlide a exhibits a Soret excitation band
detection levels are about 0.2 pmol/mL. with a maximum at 447 and an emission
maximum at 674 nm. DV Chlide a
12.2. Spectrofluorometric Determination exhibits a red-shifted Soret excitation max-
of Chlide a in Crude Extracts imum at 458 nm and an emission maxi-
Containing Mainly DV Chlide a mum at 674 nm (8). In this case too, deter-
In some cases, the Chlide a pool consists mination of the amounts of DV and MV
142
Analysis of Intermediate and End Products of Chlorophyll

Chlide a is a 2-step process. First, the total Pchlide a move with an Rf of about 0.2 just
amount of DV plus MV Chlide a in behind Mpe (8). The mixed Chlide a-Pch-
HEAR is determined at room temperature lide a band is eluted in methanol:acetone
exactly as described in section 12.1. Next, (4:1 vol/vol). Further separation of Chlide a
the DV/MV ratio of the Chlide a pool is from Pchlide a can be achieved after methy-
determined at 77 K in diethyl ether. The lation with freshly prepared diazomethane.
amount of DV and MV Chlide a is then The methylated pigments are chro-
calculated from the total amount of Chlide matographed on thin layers of silica gel H in
a as determined at room temperature and toluene:ethyl acetate:ethanol exactly as
from the DV/MV Chlide a ratio as deter- described above. In this solvent, Pchlide a E
mined at 77 K in diethyl ether (68). and Chl run with respective Rf values of
To calculate the DV/MV Chlide a ratio, about 0.90 and 0.86. Methyl Chlide a and
one sharp 77 K excitation spectrum in methyl Pchlide a run with respective values
diethyl ether need to be recorded (see sec- of about 0.68 and 0.75. The methylated
tion 5.2). The 77 K excitation spectrum is Chlide a is eluted in diethyl ether. It is usu-
recorded from 380 to 500 nm by position- ally contaminated with small amounts of
ing the emission monochromator at the methylated Pchlide a. The recovery of a
emission maximum of MV and DV Chlide highly pure methylated Chlide a prepara-
a at 674 nm. In this spectrum, the DV tion requires a second purification on silica
Chlide a Soret excitation maximum at 458 gel H as described above (8).
nm will appear as a sharp distinct peak.
The MV Chlide a Soret excitation maxi- 12.5. Separation and Analysis of DV and
mum at 447 nm will also appear as a sharp MV Chlide a
distinct peak (67). The net fluorescence
excitation amplitudes of DV and MV Separation of silica gel H-purified
Chlide a are deconvoluted and calculated methyl Chlide a into DV and MV compo-
with the use of Equations 15 and 16 (Table nents can be achieved on thin layers of
2) (68). The ratio of the deconvoluted DV polyethylene developed in 2-propanol:ace-
Chlide a excitation amplitude to the tone (1:1 vol/vol) as described for DV and
deconvoluted MV Chlide a excitation MV Pchlide a E (section 10.2) (10).
amplitude is converted to an authentic An efficient separation of cucumber DV
DV/MV Chlide a concentration ratio by and MV Chlide a from DV and MV Pch-
reference to a calibration curve that relates lide a has been described (21). Separation is
the apparent ratios of DV/MV Chlide a achieved on a 201 TP 250 × 4.6 mm I.D.,
excitation amplitudes to known ratios of polymeric octadecylsilica, 5 µm particle
DV/MV Chlide a concentrations (see sec- size, and 300 Å pore size column
tion 5.2). In this manner, DV and MV (Vydac/The Separation Group, Hesperia,
Chlide a can be determined with a preci- CA, USA). Elution at a rate of 1.2
sion of 2% to 6% (68). mL/minute is with a linear acetone gradient
from 30% aqueous acetone to 55% aque-
12.4. Chromatographic Separation of ous acetone in 8 minutes, then to 100%
Chlide a from other Tetrapyrroles acetone in 4 minutes. The mobile phase is
kept at 100% acetone for an additional 4
The mixed DV-MV Chlide a pool can be minutes. In this system, MV and DV
separated from other monocarboxylic Mg- Chlide a eluted separately between 3 and 4
porphyrins on thin layers of silica gel H as minutes, while MV and DV Chlide a elut-
described in section 5.3.1 (8). Chlide a and ed separately between 6 and 8 minutes.
143
C.A. Rebeiz

12.6. Quantitative Determination of early phases of greening of etiolated tissues,


Purified DV and MV Chlide a it is possible to determine the amount of
the Chlide a pool in the presence of Chlide
Purified DV and MV Chlide a can be b and Pchlide a by absorbance spec-
determined in any solvent from room tem- troscopy. These simultaneous equations
perature emission spectra elicited by excita- correct for Pchlide a and Chlide a and b
tion close to their Soret absorbance maxi- absorbance band overlap but do not cor-
ma between 430 and 440 nm. In each case, rect for the slight discrepancy between the
a calibration curve relating DV or MV molar extinction coefficients of the MV
Chlide a concentration to fluorescence and DV components.
emission amplitudes of the pure DV and In diethyl ether, the amount of Chlide a
MV Chlide a solutions, at their emission in the presence of Chlide b and Pchlide a
maxima, should be constructed. can be determined from Equation 17
The concentration of purified MV (Table 2), which was adapted from that
Chlide a solutions can be determined in a reported by Koski and Smith (30), using a
variety of organic solvents by room temper- molar extinction coefficient of 83 450 for
ature absorbance spectroscopy. At room Chl(ide) a in diethyl ether at 660 nm. This
temperature, MV Chlide a exhibits an 8- molar extinction coefficient was calculated
banded absorption spectrum. A strong Soret from the molecular weight of MV Chl a
absorbance band with an absorption maxi- (i.e., 893.5) and the specific absorption
mum at 430 nm and a less intense red coefficient of MV Chl a (93.4) in diethyl
absorbance band with a maximum at 660 to ether (34). Similarly, in 80% acetone, the
663 nm are observed (Table 1). The Soret amount of Chlide a in the presence of
and red absorption maxima can both be Chlide b and Pchlide a can be determined
used for quantitative determinations. If the from Equation 18.
Soret absorbance is used, a highly purified
sample free of carotenoids and other
tetrapyrroles is essential, but this is not nec- 13. ANALYSIS OF CHLIDE b
essary when the red absorbance maximum is
used. The molar extinction coefficients are In higher plants, the Chlide b pool con-
related to DV Chlide a absorbance and con- sists exclusively of MV Chlide b. In the
centration by Beer's law (Equation 2). Table corn Nec1 mutant (6), it consists of DV
1 reports absorption maxima and molar Chlide b (Figure 5) (Rebeiz, unpublished).
extinction coefficient values for MV Chlide In the primitive prochlorophyte picoplank-
a in diethyl ether and 80% aqueous acetone. ton of the subtropical waters of the North
At room temperature, DV Chlide a Atlantic as well as in the picoplankton of
exhibits an absorbance spectrum similar to the euphotic zone of the world tropical and
that of MV Chlide a, although the Soret temperate oceans and the Mediterranean
absorbance maximum of DV Chlide a is sea, the Chlide b pool is most probably a
red shifted by about 5 nm with respect to mixed DV-MV tetrapyrrole pool. Figure 1
that of MV Chlide a. shows the 5 different subpools of MV
Chlide b formed via different pathways.
12.7. Quantitative Determination of
Chlide a in the Presence of Chlide b 13.1. Spectrofluorometric Determination
and Pchlide a by Spectrophotometry of Chlide b in Crude Extracts
Under certain conditions, as during the Upon extraction from plant tissues and
144
Analysis of Intermediate and End Products of Chlorophyll

partitioning between hexane and acetone below the DV Chlide b excitation maxi-
(sections 2.1 and 2.2), the MV Chlide b mum. The net fluorescence excitation
pool passes into HEAR along with other amplitude of the Chlide b pool at 460 nm
dicarboxylic and monocarboxylic tetra- is next converted to DV Chlide b concen-
pyrroles such as MV Chlide a (673–674 tration by reference to a standard calibra-
nm emission), MV pheophorbide a (673 tion curve that relates standard DV Chlide
nm emission), and smaller amounts of MV b excitation amplitudes at 460 nm to con-
Pheophorbide b (660 nm emission). It is centrations.
also accompanied by Pchlide a (638 nm
emission) and in some cases by small 13.2. Quantitative Determination of MV
amounts of Mg-Proto and Mpe (595 nm and DV Chlide b in Crude Extracts
emission). by Spectrofluorometry
It is possible to determine the amounts
of MV Chlide b without prior purification Before structural studies or determina-
by room temperature excitation spectroflu- tion of MV and DV Chlide b, it is neces-
orometry, since the emission maxima of sary to extract Chlide b from HEAR into
MV Chlide a and b and MV pheophorbide diethyl ether (section 3.2).
a and b fall in the red region of the spec- As in the case of other MV and DV Mg-
trum, far away from the emission of other tetrapyrroles, the use of low temperature
Mg-tetrapyrroles. The concentration of (77 K) is essential to differentiate between
Chlide b can be determined from the exci- MV and DV Chlide b. At 77 K, MV
tation spectra after correction for excitation Chlide b exhibits a Soret excitation band
band overlap. This is achieved by recording with an excitation maximum at 475 nm,
2 Soret excitation spectra in HEAR at an excitation shoulder at 485 nm, and an
room temperature. One spectrum is emission maximum at 659 to 660 nm. DV
recorded close to the emission maximum Chlide b exhibits a red-shifted Soret excita-
of MV Chlide a at 674 nm. The other tion maximum at 490 nm, a Soret excita-
spectrum is recorded close to the emission tion shoulder at 498 nm, and an emission
maximum of MV Chlide b at 660 nm. The maximum at 666 nm (67). In this case too,
net fluorescence excitation amplitude of determination of the amounts of DV and
MV Chlide b, as calculated from Equation MV Chlide b is a 2-step process. First, the
19 (7), is converted to MV Chlide b con- total amount of DV plus MV Chlide b in
centration by reference to a standard cali- HEAR is determined at room temperature
bration curve that relates standard MV (section 13.1). Next, the DV/MV ratio of
Chlide b solutions of known concentra- the Chlide b pool is determined at 77 K in
tions to their fluorescence excitation ampli- diethyl ether. The amount of DV and MV
tudes. In this manner, MV Chlide b can be Chlide b is then calculated from the total
determined with a precision of about 1% amount of Chlide b as determined from
for pure MV Chlide b to 39% for extracts the HEAR fraction at room temperature
containing only 6% MV Chlide b. and from the DV/MV Chlide b ratio as
In those cases where the Chlide b pool determined at 77 K in diethyl ether (67).
consists mainly or exclusively of DV To calculate the DV/MV Chlide b ratio,
Chlide b with or without smaller amounts 2 sharp 77 K excitation spectra in diethyl
of DV Chlide a (as described above), total ether need to be recorded. The first 77 K
Chlide b can be evaluated using Equation excitation spectrum is recorded from 380
19 to calculate the net Soret fluorescence to 500 nm by positioning the emission
excitation amplitude at 460 nm, i.e., 8 nm monochromator at the emission maximum
145
C.A. Rebeiz

of MV Chlide b at 660 nm. In this spec- relating known amounts of standard DV or


trum, the MV Chlide b Soret excitation MV Chlide b or their methyl esters to their
maximum at 475 nm will appear as a dis- fluorescence emission amplitudes should
tinct broad peak. The second 77 K excita- be constructed.
tion spectrum is recorded from 380 to 500 Alternatively, absorbance spectroscopy
nm by positioning the emission mono- may be used. At room temperature, MV
chromator at the emission maximum of Chlide b exhibits a 7-banded absorption
DV Chlide b at 666 nm (69). In this spec- spectrum with a strong Soret absorbance
trum, the DV Chlide b Soret excitation band, a solvent-dependent absorption
maximum at 490 nm will appear as a maximum at 455 to 460 nm, and a less
broad distinct peak. The net fluorescence intense red absorbance band with a maxi-
excitation amplitudes of DV and MV mum at 643 to 645 nm (Table 1). If the
Chlide b are deconvoluted and calculated Soret absorbance is used for quantitation, a
with the use of Equations 20 and 21 (Table highly purified sample free of carotenoids
2) (67). Next, the ratio of the deconvolut- and other tetrapyrroles is required, but this
ed DV Chlide b excitation amplitude to is not necessary when the red absorbance
the deconvoluted MV Chlide b excitation maximum is used. The molar extinction
amplitude is converted to an authentic coefficients are related to DV Chlide b
DV/MV Chlide b concentration ratio by absorbance and concentration by Beer's
reference to a calibration curve that relates law (Equation 2).
the apparent ratios of DV/MV Chlide b Quantitative determination of DV
excitation amplitudes to known ratios of Chlide b by absorbance spectroscopy can
DV/MV Chlide b concentrations (see sec- be performed as described for MV Chlide
tion 5.2). In this manner, DV and MV a, using the values given in Table 1.
Chlide b can be determined with a preci-
sion of 1% to 6% (68). 13.5. Quantitative Determination of
Chlide b in Crude Extracts in the
13.3. Chromatographic Separation of Presence of Pchlide a
Chlide b from other Tetrapyrroles
Under certain conditions, as during the
In green tissues, the Chlide b pool is early phases of greening of etiolated tissues,
accompanied by Chlide a. To our knowl- it is possible to determine the amount of
edge, purification of MV and DV Chlide b Chlide b in the presence of Chlide a and
has not been thoroughly investigated, but Pchlide a by absorbance spectroscopy. As
it might be possible to use the method of for Chlide a, Equations 22 and 23 (Table
separation of methylated derivatives, as 2) are used to correct for Pchlide a and
described for Chl a and b in section 14. Chlide a absorbance band overlap, but do
not correct for the slight discrepancy
13.4. Quantitative Determination of between the molar extinction coefficients
Purified DV and MV Chlide b of MV and DV components.

Purified DV and MV Chlide b and their


methyl esters can be determined in organic 14. ANALYSIS OF CHLOROPHYLL a
solvents from room temperature emission
spectra elicited by excitation close to their The Chl a pool is a mixed highly
Soret absorbance maxima between 455 and dynamic DV-MV tetrapyrrole pool (Figure
468 nm. In each case, a calibration curve 5). The DV and/or MV content of this
146
Analysis of Intermediate and End Products of Chlorophyll

pool vary widely depending on the green- formed in etiolated plants via pathway 13
ing group affiliation of the plant species by photoconversion of MV Pchlide a E (9).
(24), the phase of the photoperiod (9), and The third subpool appears to be formed via
light pretreatment of the plant tissue (2). pathway 14 by dark reduction of MV Pch-
The chemical structure of MV Chl a phy- lide a E during seed germination in total
tol (Chl a P) (Figure 5) was determined by darkness (Rebeiz, unpublished).
Fischer and Stern (20). In the integrated In the integrated Chl a/b pathway, DV
Chl a/b biosynthetic pathway, the MV Chl Chl a is depicted as a precursor of one of
a P pool is considered to consist of 4 dif- the MV Chl a subpools (Figure 1, path-
ferent subpools of MV Chl a P formed via ways 1 and 8). Its specific formation from
different Chl a biosynthetic pathways (44). DV Chlide a and its conversion to MV
Two subpools are formed from MV Pch- Chl a were recently documented (2). The
lide a and MV Chlide a via pathways 2 and nature of the fatty alcohol(s) at position 7
9 (Figure 1). The third subpool is formed has not been investigated. A second DV
from DV Chlide a and MV Chlide a via Chl a pool, with a long chain esterifying
pathways 4 and 5. The fourth subpool is fatty alcohol at position 7 of the macrocy-
formed from DV Chl a by direct conver- cle other than phytol, may also occur in
sion of the vinyl group at position 4 to some plant tissues enriched in DV Pchlide
ethyl via pathway 8 (2). It is conjectured a E such as cucurbit seed coats. Such a pool
that the 4 MV Chl a P subpools are part of may be formed by (photo)conversion of
different Chl-protein complexes having DV Pchlide a E via pathway 8 (Figure 1).
different roles in photosynthesis (44).
During the early phases of greening of 14.1. Quantitative Determination of Chl
etiolated tissues, in addition to MV Chl a a in Crude Extracts
P, each MV Chl a subpool (Figure 5) may
also contain Chl a GG, Chl a DHGG, and Upon tetrapyrrole extraction from
Chl a THGG. These Chls may be consid- green(ing) plant tissues and partitioning of
ered as transient intermediates on the way the pigments between hexane and acetone
to the formation of Chl a P (52,53). (sections 2.1 and 2.2), Chl a passes into
In the integrated Chl a/b biosynthetic hexane along with Pchlide a E. In the
pathway, 3 additional subpools of MV Chl process, Chl a may become contaminated
a, with long chain fatty alcohols at position by very small amounts of Chlide a, which
7 of the macrocycle other than phytol, are may be removed by washing the hexane
depicted. These Chls occur in trace extract with an equal volume of acetone:
amounts and may be either intermediates, water:0.1 N NH4OH (8:1:1 vol/vol/vol).
on the way to the formation of MV Chl a P, In most green higher and lower plants,
or end products of the Chl biosynthetic the Chl a pool in the hexane fraction con-
pathway. Because of experimental difficul- sists mainly of MV Chl a (673 nm emis-
ties, the esterifying long chain fatty alco- sion). During the early stages of greening
hol(s) at position 7 of the macrocycle have of DDV-LDDV plant species such as
not yet been characterized. The 3 subpools cucumber, the Chl a pool may also contain
have been detected in DDV-LDMV plant small amounts of DV Chl a (673 nm emis-
species such as corn and barley during etio- sion). In green tissues, the Chl a pool is
lation and/or during the early phases of usually accompanied by MV Chl b (657
greening. One subpool is formed from MV nm emission), MV pheophytin a, i.e.,
Chlide a via pathway 12 during the early demetalated Chl a (673 nm emission), and
phases of greening. The second subpool is smaller amounts of MV Pheophytin b (660
147
C.A. Rebeiz

nm emission). It is also accompanied by 14.3. Chromatographic Separation of Chl


small amounts of Pchlide a E (638 nm a from other Tetrapyrroles by Thin
emission). Layer Chromatography
Since Chl a and Chlide a exhibit iden-
tical spectral properties, it is possible to To facilitate purification of the Chl a
determine the amounts of Chl a from and b pools, it is possible to partially sepa-
room temperature excitation spectra with- rate Chl a from Chl b by solvent–solvent
out prior purification by using Equation extraction. This can be achieved by addi-
14 (section 12.1). For green tissue, a very tion to the hexane extract enough 84%
small aliquot of the hexane extract may be aqueous methanol at ice bucket tempera-
diluted in 80% acetone prior to spectro- tures to form two phases: a deep green
fluorometric determinations. For etiolat- epiphase enriched in Chl a and β-carotene
ed tissues subjected to a brief light treat- and a pale green hypophase enriched in
ment, Chl a is concentrated by drying an Chl b and xanthophylls.
aliquot of the hexane extract under a The mixed DV-MV Chl a pool can be
stream of N2 gas at ice bucket tempera- separated from other dicarboxylic and
ture. The residue is dissolved in a small monocarboxylic Mg-porphyrins on thin
volume of 80% acetone prior to spectro- layers of silica gel H as described in section
fluorometric analysis. 5.3.1. Chl a and b move together just
In some cases, such as in prochlorophyte behind Pchlide a E and away from other
picoplanktons and the Nec1 corn mutant, Mg-porphyrins with an Rf of about 0.8
the Chl a pool consists mainly of DV Chl (10). The Chl a plus b pools are eluted in
a, with or without smaller amounts of MV diethyl ether.
Chl a. Under these circumstances, total Once the Chl a plus b pools have been
Chl a can be evaluated as for Chlide a (sec- separated from other Mg-porphyrins and
xanthophylls, DV and MV Chl a can be
tion 12.2).
readily separated from DV and MV Chl b
by chromatography on thin layers of cellu-
14.2. Quantitative Determination of MV lose. Essentially an aliquot of the diethyl
and DV Chl a in Crude Extracts by ether eluate containing Chl a plus b is spot-
Spectrofluorometry ted at the origin of a 5 × 20 cm thin layer
plate of cellulose. Once the Chl sample has
Before structural studies or determina- been spotted, the lower edge of the plate is
tion of the amounts of MV and DV Chl a dipped into a beaker containing acetone. As
in a mixture of the 2 tetrapyrroles, it is nec- the acetone moves up the plate through the
essary to transfer the Chl a pool from hexa- cellulose, it concentrates the diffuse Chl
ne to diethyl ether. This is achieved either spot into a sharp thin line at the chromato-
by dilution of a very small aliquot of the graphic origin. This in turn improves con-
washed hexane extract in diethyl ether or siderably the resolution of the separated
by drying a small aliquot of the washed bands. The plate is next inserted into a cut
hexane extract under a stream of N2 gas 1000-mL glass cylinder containing about
and dissolving the residue in diethyl ether. 10 mL of ligroin (b.p: 60°–80°C):n-
The amounts of DV and MV Chl a are propanol (99:1 vol/vol). The cylinder is
determined from room temperature and capped with a piece of aluminum foil.
77 K excitation spectra by using Equations Development is carried out at room tem-
15 and 16 (Table 2) exactly as described for perature in darkness. After the solvent front
DV and MV Chlide a. has migrated about 10 to 15 cm, the plate is
148
Analysis of Intermediate and End Products of Chlorophyll

viewed under 366 nm UV light. The sepa- Hypersil MOS2, end-capped, C-8
rated Chls are detected by their red fluores- (6.2%–6.8% carbon), 120 Å pore size, 100
cence. MV Chl a and b migrate with × 4.6-mm column (Shandon Lipshaw) at
respective Rfs of about 0.5 and 0.21, while 30°C. Pigments are separated at a flow rate
DV Chl a and b migrate with respective Rfs of 1 mL/minute by a linear gradient
of about 0.56 and 0.16 (68). The Chl expressed by minute; % solvent A; % sol-
bands are eluted in diethyl ether. vent B and programmed as follows: (0; 75;
25), (1; 50; 50), (20; 30; 70); (25; 0; 100),
14.4. Chromatographic Separation of Chl and (32; 0; 100). The column is recondi-
a by HPLC tioned to original conditions over a 7-minute
period. In this system, solvent A consists of
HPLC has become a very popular ana- methanol:1 M ammonium acetate (70:30
lytical tool for the quantitative separation vol/vol). Solvent B consists of 100%
of complex plant extracts containing Chl a methanol. DV and MV Chl a, elute much
and b. A few HPLC applications will be slower (26–27 min) than DV and MV Chl
described below. b (20–21 min).

14.4.1. Separation of Chl a from Chl b 14.4.3. Separation and Determination of


and Pheophytins Chl a Esterified with Alcohols
other than Phytol
In addition to Chl a and b, the extracts
of green plants often contain demetallated Chl a pools esterified with alcohols
Chl known as pheophytins. These tetra- other than phytol have been observed dur-
pyrroles can be separated and their con- ing dark germination and during the very
centration can be determined by HPLC. early stages of greening of etiolated tissues.
Reversed phase C-18 bonded columns are In dark-grown barley and corn seed-
used. Five- to 50-µL aliquots of the lings, we have consistently observed the
ammoniacal acetone extract are injected. presence of trace amounts of Chl a esteri-
Elution at room temperature and at flow fied with long chain fatty alcohols (LCFA)
rate of 1 mL/minute is achieved with an other than phytol (Figure 1, pathway 14).
isocratic solvent system consisting of Experimental difficulties caused by the low
water:methanol:acetone (5:75:20 vol/vol/ concentration of these tetrapyrroles have so
vol). On an ODS C-18 Spherisorb col- far hindered determination of the nature of
umn (Thermo Separation Products) Chl the esterifying LCFA at position 7 of the
b, Chl a, pheophytin b, and pheophytin a macrocycle. Because of interference by the
elute with the following retention times: sizable Pchlide a E pool, the dark-formed
2.04, 2.88, 6.83, and 7.99 minutes, Chlide a E pool constituents can only be
respectively (39). observed after separation of the fully ester-
ified tetrapyrroles by HPLC. In the HPLC
14.4.2. Separation of DV Chl a from MV system described in section 10.2, the dark-
Chl a formed Chlide a Es exhibit retention times
of about 10.4 to 11.1 minutes (Rebeiz,
Several HPLC protocols have been unpublished).
developed for the separation of DV and The HPLC system described in section
MV Chl a from other plant pigments 10.2 can also be used to monitor the for-
(5,21,33,61). For example, the protocol mation of Chlide a Es in etiolated tissues
developed by Barlow et al. uses a 3-µm during the first 500 milliseconds of green-
149
C.A. Rebeiz

ing. As evidenced by on-line emission spec- scribed for Chlide a (sections 12.6 and
tra of eluting peaks, the formation of 4 dif- 12.7).
ferent Chlide a E can be detected during
the first 500 milliseconds, following a 2.5-
millisecond light treatment of etiolated 15. ANALYSIS OF CHL b
barley and corn (Figure 1, pathway 13).
The various Chlide a Es exhibit retention In most higher plants, the Chl b pool
times of 4.5, 5.2, 6.3, and 7.7 minutes. consists exclusively of MV Chl b (Figure
The chemical nature of the esterifying 5). In the Nec1 corn mutant, it consists
LCFAs at position 7 of the macrocycle is exclusively of DV Chl b (Figure 5) (6,68).
still undetermined. It is not clear either In primitive prochlorophyte picoplank-
whether the various Chlide a E are formed tons, the Chlide b pool is a mixed DV-MV
by photoconversion of Pchlide a Es or by tetrapyrrole pool.
rapid esterification of newly formed Chlide In the integrated Chl a/b pathway, the
a (Rebeiz, unpublished). MV Chl b pool consists of 8 different sub-
pools formed via different Chl b biosyn-
14.4.4. Determination of Chl a Esterified thetic pathways (Figure 1). Four subpools
with Geranylgeraniol Derivatives are formed from DV Pchlide a via path-
ways 2, 5, 6, and 8. Two subpools are
During the early stages of greening of formed from MV Pchlide b via pathways 3
etiolated tissues, newly formed Chlide a is and 10. The last 2 subpools are formed
converted to Chl a P via Chl a GG, Chl a from MV Chlide a via pathways 9 and 11.
DHGG, and Chl a THGG (53). Separa- MV Chl b formation via pathways 5 and 8
tion of these various Chl a has been is confined to DDV-LDDV plant species
achieved after pheophytinization (i.e., such as cucumber, while its formation via
demetalation) on a LiChrosorb RP-8, 5 to pathways 9, 10, and 11 is confined to
10 µm particle size column (Knauer, DDV-LDMV plant species such as corn
Oberusel, Germany). Isocratic elution is at barley and wheat and to DMV-LDMV
a flow rate of 1.5 mL/minute, in plant species such as Johnson grass (Figure
methanol:water (95:5 vol/vol) at room 1). Biosynthetic pathways 2, 3, and 6 occur
temperature (55). After extraction with in all three greening groups.
ethyl acetate, pheophytinization is achieved As far as we know, the esterifying alco-
by treatment with HCL (14). In this sys- hol at position 7 of the MV Chl b macro-
tem, the pheophytinized GG Chl a deriva- cycle is phytol. Because of the extreme
tives exhibit the following retention times: biosynthetic heterogeneity of the Chl b
pheophytin a GG (Pheo a GG) = 10 to 11 pool, it is possible that a more thorough
minutes, Pheo a DHGG = 11.8 to 12 min- investigation of the esterifying alcohol at
utes, Pheo a THGG = 14 to 14.2 minutes, position 7 of the Chl b macrocycle may
and Pheo a P = 16 to 16.4 minutes (55). reveal the presence of minor amounts of
LCFAs other than phytol.
14.5. Quantitative Determination of In the integrated Chl a/b pathway, the
Purified DV and MV Chl a DV Chl b pool consists of 2 different sub-
pools formed via 2 different Chl b biosyn-
Since DV and MV Chl a exhibit identi- thetic pathways (Figure 1). One subpool is
cal absorbance properties to DV and MV formed from DV Chlide b via pathway 7.
Chlide a, the concentration of purified Chl The second subpool is formed from DV
a solutions is determined exactly as de- Chl a via pathway 1. DV Chl b formation
150
Analysis of Intermediate and End Products of Chlorophyll

from DV Chlide a via ancestral pathways 1 plants, except in prochlorophyte pico-


and 7, takes place only in DDV-LDDV planktons and the Nec1 corn mutant, the
plant species. The accumulation of massive pheophytin and pheophorbide a pools are
amounts of DV Chl b is observed in the MV in nature.
Nec1 corn mutant and in primitive pro- It is possible to determine the amounts
chlorophyte picoplanktons. of pheophytin a and pheophorbide a with-
The isolation and purification of Chl b out prior purification by room temperature
has been described in sections 14.3 and excitation spectrofluorometry. Since the
14.4. Similarly, because the spectral prop- emission maximum of MV Pheo(bide) a
erties of Chl b are identical to Chlide b, the falls in the red region of the spectrum far
former may be quantified using the proce- away from the emission of other tetra-
dures described in section 13. pyrroles, it is possible to record Soret excita-
tion spectra with minimum Soret excitation
overlap. The concentration of Pheo(bide) a
16. ANALYSIS OF PHEOPHYTINS can then be determined from the excitation
AND PHEOPHORBIDES a spectra after correction for excitation band
overlap. This is achieved by recording 2
The first degradation products of Chl a Soret excitation spectra in HEAR or 80%
are pheophytin a and/or pheophorbides a acetone at room temperature. One spec-
[Pheo(bides) a] (40). Pheophytins are trum is recorded close to the emission max-
demetalated Chls while pheophorbides are ima of MV Chl(ide) a and Pheo(bide) a at
demetalated Chlides. During senescence or 674 nm. The other spectrum is recorded
after treatment with certain chemicals, close to the emission maxima of MV
green plants tend to accumulate substantial Chl(ide) b and Pheo(bide) b at 660 nm.
amounts of pheophytins and pheophor- The net Soret excitation amplitude due
bides (3). Spectrofluorometric techniques only to MV Pheo(bide) a is deconvoluted
have been developed to determine the con- with the use of Equation 24 (7). The net
centration of pheophorbides and pheo- fluorescence excitation amplitude of MV
phytins in crude extracts containing Pheo(bide) a, as calculated from Equation
Chlides and Chls. Pheophorbides are 24, is converted to MV Pheo(bide) a con-
determined on aliquots of HEAR, while centration by reference to a standard cali-
pheophytins are determined on aliquots of bration curve that relates known concentra-
the hexane extract after drying in a stream tions of standard Pheo(bide) a to their
of N2 gas at ice bucket temperature and fluorescence excitation amplitudes. In this
dissolution in 80% aqueous acetone. manner, MV Pheo(bide) a can be deter-
mined with a percent error of about 1% for
16.1. Analysis of Pheo(bide) a in Crude pure MV Pheo(bide) a to 13% for extracts
Extracts by Spectrofluorometry containing only 5% MV Pheo(bide) a (7).
Minimum detection levels are about 0.2
Upon tetrapyrrole extraction from plant pmol/mL.
tissues and partitioning of the pigments
between hexane and acetone (sections 2.1 16.2. Separation of Pheophytin a from
and 2.2), pheophorbide a passes into the other Tetrapyrroles
HEAR fraction along with other dicar-
boxylic and monocarboxylic tetrapyrroles, On silica gel H, pheophytin a moves
while pheophytin a passes into hexane with Chl a and is eluted in diethyl ether.
along with Chls. In most higher and lower Pheophytin a can then be readily separated
151
C.A. Rebeiz

from Chl a and b by chromatography on nations when the red absorbance maximum
thin layers of cellulose. In ligroin (b.p: is used. The molar extinction coefficients are
60°–80°C):acetone:n-propanol (99:10:0.45 related to MV and DV Pheobide
vol/vol/vol), pheophytin a migrates close to absorbance and concentration by Beer's law
the solvent front with an Rf of about 0.9, (Equation 2). Table 1 reports absorption
while Chl a and b move with respective Rfs maxima and molar extinction coefficient
of about 0.46 and 0.23 (7). The pheophytin values for MV and DV Pheo(bide) a in
a band is eluted in diethyl ether. diethyl ether and 80% aqueous acetone.
Separation of pheophytin a and its
derivatives by HPLC has been described in
section 14.4. 17. ANALYSIS OF PHEO(BIDE) b

16.3. Quantitative Determination of Upon tetrapyrrole extraction from plant


Purified Pheo(bide) a tissues and partitioning of the pigments
between hexane and acetone (sections 2.1
Purified MV Pheo(bide) a can be deter- and 2.2), pheophorbide b passes into
mined in any appropriate solvent from HEAR along with other dicarboxylic and
room temperature emission spectra elicited monocarboxylic tetrapyrroles, while pheo-
by excitation close to its Soret absorbance phytin b passes into hexane along with
maximum between 410 and 414 nm. In Chls. In most higher and lower plants,
each case, a calibration curve using known except in prochlorophyte picoplanktons and
amounts of MV Pheo(bide) a should be the Nec1 corn mutant, the pheophytin and
constructed in order to convert fluores- pheophorbide b pools are MV in nature.
cence data to Pheobide a concentrations. Quantification of Pheo(bide) b in crude
The concentration of purified MV and extracts is carried out as described in sec-
DV Pheo(bide) a solutions can be deter- tion 16.1, but using Equation 25 (7). The
mined in a variety of organic solvents by net fluorescence excitation amplitude of
room temperature spectrophotometry. At MV Pheo(bide) b, as calculated from
room temperature, between 380 and 700 Equation 25, is converted to MV
nm, MV and DV Pheobide a exhibit a 7- Pheo(bide) b concentration by reference to
banded absorption spectrum in various a standard calibration curve prepared from
organic solvents. The wavelengths of standard MV Pheo(bide) b solutions of
absorption maxima depend upon the sol- known concentrations and from their fluo-
vent. A strong Soret absorbance band with rescence excitation amplitudes. In this
an absorption maximum at 409 to 410 nm manner, MV Pheo(bide) b can be deter-
[MV Pheo(bide) a] or 417 to 419 nm [DV mined with a percent error of about 1% for
Pheo(bide) a] and a less intense red pure MV Pheo(bide) b to 14% for extracts
absorbance band with a maximum at 665 to containing 14% MV Pheo(bide) a (7). At
666 nm [MV Pheo(bide) a] or 665 to 667 concentrations of 6% Pheobide b or less,
nm [DV Pheo(bide) a] are observed (Table the analytical error becomes substantial
1). The Soret and red absorbance maxima (133.5%). Minimum detection levels are
can both be used for quantitative determi- about 0.2 pmol/mL.
nations. If the Soret absorbance is used, a Similarly, the purification and quanti-
highly purified sample free of carotenoids tation of MV Pheo(bide) b is as described
and other tetrapyrroles should be used. for MV Pheo(bide) a, using the appropri-
Contamination by carotenoids does not ate molar extinction coefficients given in
interfere however with quantitative determi- Table 1.
152
Analysis of Intermediate and End Products of Chlorophyll

ABBREVIATIONS 4.Amindari, S.M., W.E. Splittstoesser, and C.A. Rebeiz.


1995. Photodynamic effects of several metabolic
tetrapyrroles on isolated chloroplasts, p. 217-246. In
Unless preceded by MV or DV, abbrevi- J.R. Heitz and K.R. Downum (Eds.), Light-Activated
ations for tetrapyrroles are used to desig- Pest Control. Am. Chem. Soc. Washington, DC.
5.Barlow, R.G., D.G. Cummings, and S.W. Gibb. 1997.
nate metabolic pools that may consist of Improved resolution of mono- and divinyl chlorophylls
MV and DV components. ALA, δ- a and b and zeaxanthin and lutein in phytoplankton
aminolevulinic acid; Chl(ide), Chlide extracts using reverse phase C-8 HPLC. Mar. Ecol.
Prog. Ser. 161:303-307.
and/or Chlide ester; Chl, chlorophyll; 6.Bazzaz, M.B. 1981. New chlorophyll chromophores
Chlide, chlorophyllide; Copro, copropor- isolated from a chlorophyll deficient mutant of maize.
phyrin; D, dark; DHGG, dihydrogeranyl- Photobiochem. Photobiophys. 2:199-207.
7.Bazzaz, M.B. and C.A. Rebeiz. 1979. Chloroplast cul-
geraniol; DME, dimethyl ester; DV, ture V. Spectrofluorometric determination of chloro-
divinyl; GG, geranylgeraniol; HEAR, phyll(ide) a and b and pheophytin (or pheophorbide) a
hexane-extracted acetone residue; HPLC, and b in unsegregated pigment mixtures. Photochem.
Photobiol. 30:709-721.
high-pressure liquid chromatography; L, 8.Belanger, F.C., J.X. Dugan, and C.A. Rebeiz. 1982.
light; LCFA, long chain fatty alcohol; Chloroplast biogenesis: identification of chlorophyllide
LHC, light harvesting Chl; Mpde, Mg- a (E458F674) as a divinyl chlorophyllide a. J. Biol.
Chem. 257:4849-4858.
Proto diester; Mpe, Mg-Proto monomethyl 9.Belanger, F.C. and C.A. Rebeiz. 1980. Chloroplast bio-
ester; MV, monovinyl; P, phytol; Pchl(ide), genesis 30. Chlorophyll(ide) (E459F675) and chloro-
Pchlide and/or Pchlide ester; Pchlide E, phyll(ide) (E449F675) the first detectable products of
divinyl and monovinyl protochlorophyll photoreduc-
Pchlide ester; Pchlide, protochlorophyllide; tion. Plant Sci. Lett. 18:343-350.
Pheo(bide), pheophorbide and/or pheophy- 10.Belanger, F.C. and C.A. Rebeiz. 1980. Chloroplast
tin; POR, Pchlide oxidoreductase; Proto, biogenesis. Detection of divinyl protochlorophyllide
ester in higher plants. Biochemistry 19:4875-4883.
protoporphyrin IX; THGG, tetrahydroger- 11.Belanger, F.C. and C.A. Rebeiz. 1980. Chloroplast
anylgeraniol; Uro, uroporphyrin. biogenesis. Detection of divinyl protochlorophyllide in
higher plants. J. Biol. Chem. 255:1266-1272.
12.Belanger, F.C. and C.A. Rebeiz. 1982. Chloroplast
biogenesis. Detection of monovinyl magnesium proto-
ACKNOWLEDGMENTS porphyrin monoester and other monovinyl magnesium
porphyrins in higher plants. J. Biol. Chem. 257:1360-
1371.
This work was supported by funds from 13.Belanger, F.C. and C.A. Rebeiz. 1984. Chloroplast
the Illinois Agricultural Experiment Sta- biogenesis 47: spectroscopic study of net spectral shifts
tion, by the John P. Trebellas Photo- induced by ligand coordination in metalated
tetrapyrroles. Spectrochim. Acta 40A:807-827.
biotechnology Research Endowment, and 14.Benz, J., C. Wolf, and W. Rudiger. 1980. Chlorophyll
by the C.A. and C.C. Rebeiz Endowment biosynthesis: hydrogenation of geranylgeraniol. Plant
for Fundamental Research. Sci. Lett. 19:225-230.
15.Bladon, P. and G. Eglinton. 1964. Ultraviolet, visible
and infrared spectroscopy, p. 22-34. In J.C.P. Schwarz
REFERENCES (Ed.), Physical Methods in Organic Chemistry. Oliver
& Boyd, London.
1.Adb El Mageed, H.A., K.F. El Sahhar, K.R. Robertson, 16.Carey, E.E. and C.A. Rebeiz. 1985. Chloroplast bio-
R. Parham, and C.A. Rebeiz. 1997. Chloroplast bio- genesis 49. Difference among angiosperms in the
genesis 77. Two novel monvinyl and divinyl light-dark biosynthesis and accumulation of monovinyl and
greening groups of plants and their relationship to the divinyl protochlorophyllide during photoperiodic
chlorophyll a biosynthetic heterogeneity of green greening. Plant Physiol. 79:1-6.
plants. Photochem. Photobiol. 66:89-96. 17.Daniell, H. and C.A. Rebeiz. 1982. Chloroplast cul-
2.Adra, A.N. and C.A. Rebeiz. 1998. Chloroplast biogen- ture IX. Chlorophyll(ide) a biosynthesis in vitro at rates
esis 81. Transient formation of divinyl chlorophyll a fol- higher than in vivo. Biochem. Biophys. Res. Commun.
lowing a 2.5 ms light flash treatment of etiolated cucum- 106:466-470.
ber cotyledons. Photochem. Photobiol. 68:852-856. 18.Duggan, J.X. and C.A. Rebeiz. 1982. Chloroplast bio-
3.Amindari, S. 1992. Structure-Function Photodynamic genesis 37: induction of chlorophyllide a (E459F675)
Herbicidal Studies of Phenanthroline, Dipyridyl and accumulation in higher plants. Plant Sci. Lett. 24:
Pyridine Analogs, p. 287. NRES, Urbana. 27-37.

153
C.A. Rebeiz

19.Falk, J.E. 1964. Porphyrins and Metalloporphyrins, p. phyll(ide) via the acidic and fully esterified biosynthet-
250-253. Elsevier, Amsterdam. ic branches in higher plants. Biochemistry 21:242-247.
20.Fischer, H. and A. Stern. 1940. Die Chimie des 37.Nicolas, R.E.H. and C. Rimington. 1949. Qualitative
Pyrroles, p. 321-324. Kademische, Verlagsgesellschaft. analysis of the porphyrins by partition chromatography.
21.Garrido, J.L. and M. Zapata. 1997. Reversed-phase Scand. J. Cli. Lab. Invest. 1:12-18.
high performance liquid chromatographic separation of 38.Parham, R. and C.A. Rebeiz. 1995. Chloroplast bio-
mono- and divinyl chlorophyll forms using pyridine- genesis 72: a (4-vinyl) chlorophyllide a reductase assay
containing mobile phases and a polymeric octadecylsil- using divinyl chlorophyllide a as an exogenous sub-
ica column. Chromatographia 44:43-49. strate. Anal. Biochem. 231:164-169.
22.Granick, S. 1948. Protoporphyrin 9 as a precursor of 39.Rebeiz, C.A. 1978. The separation of chlorophyll and
chlorophyll. J. Biol. Chem. 172:717-727. pheophytins by reversed phase HPLC. Chromatogra-
23.Houssier, C. and K. Sauer 1969. Optical properties of phy Rev. 4:8-9.
the protochlorophyll pigments II. Electronic absorp- 40.Rebeiz, C.A. and M.B. Bazzaz. 1979. Cell-free agri-
tion, fluorescence and circular dichroism spectra. culture: the concept and its initial implementation, p.
Biochim. Biophys. Acta 172:261-266. 453-471. In C.D. Scott (Ed.), Biotechnology in Energy
24.Ioannides, I.M., D.M. Fasoula, R.K. Robertson, and Production and Conservation. John Wiley & Sons,
C.A. Rebeiz. 1994. An evolutionary study of chloro- New York.
phyll biosynthetic heterogeneity in green plants. 41.Rebeiz, C.A., F.C. Belanger, S.A. McCarty, G.
Biochem. Sys. Ecol. 22:211-220. Freyssinet, J.X. Duggan, S.M. Wu, and J.R. Mattheis.
25.Ioannides, I.M., V.P. Shedbalkar, and C.A. Rebeiz. 1981. Biosynthesis and accumulation of novel chloro-
1997. Quantitative determination of 2-monovinyl pro- phyll a and b chromophoric species in green plants, p.
tochlorophyll(ide) b by spectrofluorometry. Anal. 197-212. In G. Akoyunoglou (Ed.), Photosynthesis V.
Biochem. 249:241-244. Chloroplast Development. Balaban International Ser-
26.Kim, J.S. and C.A. Rebeiz. 1996. Origin of the chloro- vices, Philadelphia.
phyll a biosynthetic heterogeneity in higher plants. J. 42.Rebeiz, C.A., and P. Castelfranco. 1971. Chlorophyll
Biochem. Mol. Biol. 29:327-334. biosynthesis in a cell-free system from higher plants.
27.Knaust, R. and H. Senger. 1994. Monovinyl and Plant Physiol. 47:33-37.
divinyl protochlorophyll in different stages of esterifica- 43.Rebeiz, C.A. and P. Castelfranco. 1971. Protochloro-
tion isolated from mutant C-2A' of the unicellular green phyll biosynthesis in a cell-free system from higher
alga Scenedesmu obliquus. Physiol. Plant. 90:490-496. plants. Plant Physiol. 47:24-32.
28.Kolossov, V., I.M. Ioannides, S. Kulur, and C.A. 44.Rebeiz, C.A., I.M. Ioannides, V. Kolossov, and K.J.
Rebeiz. 1999. Chloroplast biogenesis 82: development Kopetz. 1999. Chloroplast biogenesis 80. Proposal of a
of a cell-free system capable of the net synthesis of unified multibranched chlorophyll a/b biosynthetic
chlorophyll(ide) b. Photosynthetica 36:253-258. pathway. Photosynthetica 36:117-128.
29.Koski, V.M. 1950. Chlorophyll formation in seedlings 45.Rebeiz, C.A., J.R. Mattheis, B.B. Smith, C.C. Rebeiz,
of Zea mays L. Arch. Biochem. 29:339-343. and D.F. Dayton. 1975. Chloroplast biogenesis.
30.Koski, V.M. and J.H.C. Smith. 1948. The isolation Biosynthesis and accumulation of protochlorophyll by
and spectral absorption properties of protochlorophyll isolated etioplasts and developing chloroplasts. Arch.
from etiolated barley seedlings. J. Am. Chem. Soc. Biochem. Biophys. 171:549-567.
70:3558-3562. 46.Rebeiz, C.A., A. Montazer-Zouhoor, and H. Daniell.
31.Lee, H.J., M. Ball, and C.A. Rebeiz. 1991. Intraplas- 1984. Chloroplast culture X: thylakoid assembly in
tidic localization of the enzymes that convert δ- vitro. Isr. J. Bot. 33:225-235.
aminolevulinic acid to protoporphyrin IX in etiolated 47.Rebeiz, C.A., R. Parham, D.A. Fasoula, and I.M.
cucumber cotyledons. Plant Physiol. 96:910-915. Ioannides. 1994. Chlorophyll biosynthetic heterogene-
32.Lee, H.J., M.D. Ball, R. Parham, and C.A. Rebeiz. ity, p. 177-193. In D.J. Chadwick and K. Ackrill (Eds.),
1992. Chloroplast biogenesis 65: enzymic conversion of The Biosynthesis of the Tetrapyrrole Pigments. John
protoporphyrin IX to Mg-protoporphyrin IX in a sub- Wiley & Sons, New York.
plastidic membrane fraction of cucumber etiochloro- 48.Rebeiz, C.A. and D.G. Saab. 1995. Porphyrin Analyt-
plasts. Plant Physiol. 99:1134-1140. ical Tools. Software protected by copyright.
33.Lenning, K.V., J.L. Garrido, J. Aristegui, and M. Zap- 49.Rebeiz, C.A., S.M. Wu, M. Kuhadje, H. Daniell, and
ata. 1995. Temperature-programmed high performance E.J. Perkins. 1983. Chlorophyll a biosynthetic routes
liquid chromatography of mono- and divinyl chloro- and chlorophyll a chemical heterogeneity. Mol. Cell.
phyll forms from marine phytoplankton. Chro- Biochem. 58:97-125.
matographia 41:539-543. 50.Rebeiz, N., S. Arkins, K.W. Kelley, and C.A. Rebeiz.
34.Mackinney, G. 1941. Absorption of light by chloro- 1996. Enhancement of coproporphyrinogen III trans-
phyll solutions. J. Biol. Chem. 140:315-322. port into isolated leucocyte mitochondria by ATP.
35.McCarthy, S.A., F.C. Belanger, and C.A. Rebeiz. Arch. Biochem. Biophys. 333:475-481.
1981. Chloroplast biogenesis: detection of a magne- 51.Reinbothe, C., N. Lebedev, and S. Reinbothe. 1999. A
sium protoporphyrin diester pool in plants. Biochem- protochlorophyllide light-harvesting complex involved
istry 20:5080-5087. in de-etiolation of higher plants. Nature 397:80-84.
36.McCarthy, S.A., J.R. Mattheis, and C.A. Rebeiz. 1982. 52.Rudiger, W. 1993. Esterification of chlorophyllide and
Chloroplast biogenesis: biosynthesis of protochloro- its implication for thylakoids development, p. 219-240.

154
Analysis of Intermediate and End Products of Chlorophyll

In C. Sundqvist and M. Ryberg (Eds.), Pigment-Pro- 62.Smith, J.H.C. and C.S. French. 1963. The major
tein Complexes in Plastids: Synthesis and Assembly. accessory pigment in photosynthesis. Ann. Rev. Plant
Academic Press, New York. Physiol. 14:181-224.
53.Rudiger, W. and S. Schoch. 1991. The last steps of 63.Tripathy, B.C. and C.A. Rebeiz. 1985. Chloroplast
chlorophyll biosynthesis, p. 451-464. In H. Scheer biogenesis. Quantitative determination of monovinyl
(Ed.), Chlorophylls. Academic Press, New York. and divinyl Mg-protoporphyrins and protochloro-
54.Scheumann, V., H. Klement, M. Helfrish, U. Oster, phyll(ides) by spectrofluorometry. Anal. Biochem.
S. Schoch, and W. Rudiger. 1999. Protochlorophyl- 149:43-36.
lide b does not occur in barley etioplasts. FEBS Lett. 64.Tripathy, B.C. and C.A. Rebeiz. 1986. Chloroplast
445:445-448. biogenesis. Demonstration of the monovinyl and
55.Schoch, S. 1978. The esterification of chlorophyllide a divinyl monocarboxylic routes of chlorophyll biosyn-
in greening bean leaves. Z. Naturforsch 33c:712-714. thesis in higher plants. J. Biol. Chem. 261:13556-
56.Shedbalkar, V.P., I.M. Ioannides, and C.A. Rebeiz. 13564.
1991. Chloroplast biogenesis. Detection of monovinyl 65.Tripathy, B.C. and C.A. Rebeiz. 1988. Chloroplast
protochlorophyll(ide) b in plants. J. Biol. Chem. biogenesis 60. Conversion of divinyl protochlorophyl-
266:17151-17157. lide to monovinyl protochlorophyllide in green(ing)
57.Shedbalkar, V.P. and C.A. Rebeiz. 1992. Chloroplast barley, a dark monovinyl/light divinyl plant species.
biogenesis: determination of the molar extinction coef- Plant Physiol. 87:89-94.
ficients of divinyl chlorophyll a and b and their pheo- 66.Vernon, L.P. 1960. Spectrophotometric determination
phytins. Anal. Biochem. 207:261-266. of chlorophylls and pheophytins in plant extracts. Anal.
58.Shioi, Y. and T. Sasa. 1883. Formation and degradation Biochem. 32:1144-1150.
of protochlorophylls in etiolated and greening cotyle- 67.Withrow, R.B. and L. Price. 1957. A darkroom safe-
dons of cucumber. Plant Cell Physiol. 24:835-840. light for research in plant physiology. Plant Physiol.
59.Shioi, Y. and T. Sasa. 1983. Compositional hetero- 32:244-248.
geneity of protochlorophyllide ester in etiolated leaves 68.Wu, S.M., J.M. Mayasich, and C.A. Rebeiz. 1989.
of higher plants. Arch. Biochem. Biophys. 220:286- Chloroplast biogenesis: quantitative determination
292. of monovinyl and divinyl chlorophyll(ide) a and b
60.Shioi, Y. and T. Sasa. 1982. Separation of protochloro- by spectrofluorometry. Anal. Biochem. 178:294-
phylls esterified with different alcohols from inner seed 300.
coat of three cucurbitaceae. Plant Cell Physiol. 69.Wu, S.M. and C.A. Rebeiz 1985. Chloroplast biogene-
23:1315-1321. sis. Molecular structure of chlorophyll b (E489 F666).
61.Shioi, Y., K. Watanabe, K.-i. Takmiya, J.L. Garrido, J. Biol. Chem. 260:3632-3634.
and M. Zapata. 1995. Separation of mono-and divinyl 70.Zscheile, F.P. and C.P. Comar. 1941. Influence of
chlorophyll species by high-performance liquid chro- preparative procedure on the purity of chlorophyll
matography using an octadecyl polyvinyl alcohol poly- components as shown by absorption spectra. Bot.
mer column. Anal. Biochem. 231:225-229. Gazette 102:463-481.

155
7 Analysis of Heme and Hemoproteins

Angela Wilks
University of Maryland, Baltimore, MD, USA

1. INTRODUCTION to the elucidation of structure and func-


tion. It is, however, pertinent to keep in
Heme is perhaps the most ubiquitous mind that functional hemoproteins require
cofactor found in nature and the most the binding and coordination of heme,
functionally diverse. Hemoproteins are which must be synthesized and incorporat-
involved in cell respiration (cytochromes), ed into the protein during protein synthe-
oxygen-binding and transport (hemoglo- sis. In addition, the expression of a given
bin and myoglobin), oxidative biotransfor- hemoprotein is dependent on the nature of
mations (cytochrome P-450 and peroxidas- that particular protein. For example, the
es), and most recently, as sensors in membrane-bound cytochrome P-450 en-
2-component regulatory systems (guanyl- zymes have markedly different require-
ate cyclase, FixL, and CooA). The ability of ments than a soluble protein such as myo-
hemoproteins to carry out extremely globin. Therefore, the expression and
diverse reactions arises largely from the purification of the proteins will be dis-
protein environment in which the heme cussed in the context of both their function
molecule resides and specifically the nature and properties. In light of the excellent
of the heme–ligands. Other factors that reviews on the chemistry of heme and por-
contribute to the reactivity of the heme are phyrins in Chapters 1 and 2, this review
intrinsic to the heme itself, including the will focus solely on the expression, purifi-
substituents on the heme periphery and, in cation, and analysis of heme in the context
some cases, the covalent attachment of the of hemoproteins.
heme to the protein. The structures of the
most common heme–ligands and examples 2. HEMOPROTEIN EXPRESSION
of the hemoproteins in which they occur AND PURIFICATION
are found in Table 1.
The advent of modern cloning and mol- Recombinant expression systems for
ecular biological techniques has opened up both prokaryotic and eukaryotic hemopro-
the field of hemoprotein research in regard teins are now routinely utilized to obtain

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

157
A. Wilks

Table 1. Heme Ligand Structure and Function

158
Analysis of Heme and Hemoproteins

large quantities of protein for biochemical cells when grown on minimal media alone.
and biophysical characterization. The The individual globin chains of hemo-
advance in molecular biological techniques globin can be expressed as fusion proteins,
has not only furthered our understanding of in recombinant E. coli systems, or as is
hemoprotein function but also contributed more common, with the β-globin chain,
to the rapidly expanding role of hemopro- the apoprotein itself (27,41,72). It is also
teins in areas such as cell signaling and regu- possible in E. coli cells to co-express the α
lation of gene expression and function. and β-globins together and obtain active
holoprotein (41,42,44,47,100). In all of
2.1. Hemoprotein Expression Systems these systems, however, some heterogeneity
is observed. A functionally homogenous
2.1.1. Oxygen-Binding Proteins protein is best obtained by removal of the
heme, separation of the α and β-globin
The successful expression and purifica- chains, and subsequent reassembly in the
tion of recombinant myoglobins from a presence of cyanohemin to reform the
number of species have been reported active tetrameric protein (41,42,47).
(103,110,122,127). Initial studies on the More recently, a new class of ligand
expression in Escherichia coli of human binding hemoproteins that act as biological
myoglobin utilized a fusion protein con- sensors have been identified in a number of
sisting of the first 31 amino acids of the organisms. This class of proteins includes
phage lambda cII gene and the tetrapeptide the eukaryotic soluble guanylate cyclase,
Ile-Glu-Gly-Arg, followed by the myoglo- FixL of Rhizobia, and CooA of Rhodospiril-
bin gene sequence (122). The fusion prod- lum rubrum, which sense NO, O2, and
uct was then isolated, reconstituted with CO, respectively (91). While mammalian
heme, cleaved with trypsin, and purified to soluble guanylate cyclases have been
generate the active protein in gram quanti- intractable to expression in E. coli, some
ties. Subsequent studies focused on the progress has been made in insect cell lines
generation of synthetic genes for sperm (135). The oxygen sensor FixL is a modular
whale myoglobin, which allowed for opti- protein consisting of an N-terminal heme
mization of the preferred E. coli codon domain and a C-terminal kinase transmit-
usage and the incorporation of unique ter domain. Expression of both the full-
restriction sites throughout the gene length protein and the heme domain alone
(110,127). The introduction of multiple have been successfully carried out in E. coli
restriction sites enabled the design of cas- TG1 under control of the T7 promoter in
sette based primers for site-directed muta- a pUC8 derived vector (32). The CooA
genesis (110,127). The synthetic gene transcription factor has been subcloned
when cloned into pUC18 (Life Technolo- into pKK223-3 (Amersham Pharmacia
gies, Rockville, MD, USA) and expressed Biotech, Piscataway, NJ, USA) under con-
in E. coli DH5α (Life Technologies), trol of the T7 promoter and expressed in E.
produced active holoprotein indistinguish- coli JM109 cells (2,99).
able from that of the native protein. High
expression yields of myoglobin apoprotein 2.1.2. Oxidases
as inclusion bodies has been obtained in
the T7 promoter based vector pET17b It is only recently that the membrane-
(Novagen, Madison, WI, USA) (49). The bound eukaryotic cytochrome P-450
authors were able to refold and reconstitute enzymes have been routinely expressed in
the protein in quantities of 200 mg/L of recombinant E. coli systems. There are a
159
A. Wilks

number of factors critical for successful enzymes have also been expressed in yeast,
recombinant expression of this family of baculovirus, and mammalian expression
enzymes. Many of the specific require- systems (33). The yeast and mammalian
ments for the successful expression of the expression systems in general produce
cytochrome P-450 enzymes involve the much less total protein, but have been
ability of the cell to adjust their synthesis of extremely valuable in the area of metabo-
heme and lipids to a level that allows for lite and drug research.
correct folding and membrane association. The recent emergence of nitric oxide in
If the rate of expression exceeds the ability physiological functions, such as signal
of the cell to synthesize these factors, then transduction in the cardiovascular and ner-
the protein irreversibly accumulates as vous systems and cytostatic functions of
inclusion bodies. The requirements for the immune system, has led to an extensive
functional cytochrome P-450 expression in body of work on nitric oxide synthase
E. coli were determined to be factors con- (NOS) (66,77). A number of isoforms of
tained in the expression vector, i.e., the pro- nitric oxide synthase have been expressed
moter and lac repressor gene, the structure in baculovirus, including the human endo-
and/or sequence of the mRNA around the thelial, inducible and neuronal enzymes, as
initiation codon, as well as variables such as well as the rat neuronal nitric oxide syn-
the E. coli strain and culture conditions (3). thase (11,12,73,90). Large-scale expression
The powerful T7 phage promoter is capa- of nitric oxide synthase in baculovirus has
ble of synthesizing large amounts of the been achieved, but a critical factor in
recombinant protein, but the majority was obtaining soluble active protein was the
found to accumulate in inclusion bodies. addition of exogenous heme required for
The most successful vectors for the expres- correct folding and binding of the cofactor
sion of cytochrome P-450 are based on the tetrahydrobiopterin (62,64,98).
lac promoter and its derivatives. The two E. The successful expression of nitric oxide
coli expression vectors that have been suc- synthase isoforms in E. coli has developed
cessfully used for cytochrome P-450 more recently and was critical to the rapid
expression are pCW Ori+, a derivative of progress in elucidating many of the struc-
pHSe5 (28,71), and pSP19g10L, a deriva- tural and mechanistic features of NOS (66,
tive of pSPORT-1 (Life Technologies). 77). As with the other members of the cyto-
The fusion of a promoter and ribosomal chrome P-450 family, the NOS enzymes
binding site of a prokaryotic plasmid and were expressed successfully in the pCW
the nucleotide sequence from a eukaryotic Ori+ vectors (30,31,93,131). Factors
gene can have an inhibitory effect on trans- reported to be critical for maximizing the
lation due to the formation of mRNA sec- expression of the rat neuronal nitric oxide
ondary structure. In cytochrome P-450, synthase were the co-expression of groEL
17α-hydroxylase (CYP17) changes in the and groES chaperonins in the protease defi-
amino terminal codons promoted high cient BL21 (DE3) pLysS E. coli strain (93).
level expression, whereas the unmodified However, Gerber and Ortiz de Montellano
DNA failed to produce any detectable pro- have reported successful expression of the
tein (3). Utilization of the pCW Ori+ plas- rat neuronal nitric oxide synthase in the
mid vector together with the first 8 codons absence of chaperonins (31). The inducible
of the modified CYP17 has resulted in the NOS, unlike the neuronal isoform, requires
high-level expression of a number of calmodulin, and co-expression of calmod-
cytochrome P-450 enzymes (35,124). ulin with iNOS is essential for active holo-
The eukaryotic cytochrome P-450 protein (30,131). All of the isoforms
160
Analysis of Heme and Hemoproteins

require tetrahydrobiopterin, a cofactor that covalent attachment of heme in vivo.


is not synthesized in vivo by prokaryotes, Myeloperoxidase, which has significant
therefore, reconstitution with this cofactor homology to lactoperoxidase and a cova-
is required to obtain active protein. lently linked heme, may also have a similar
Yeast expression systems have not been mechanism for covalent attachment of the
as extensively used as recombinant E. coli heme to the protein.
in large part because the yield of active When discussing the expression and
holoprotein is in the 0.5 to 1.0 mg/L purification of the oxidases, it is important
range, compared with 5 to 10 mg/L in the to include heme oxygenase in this category,
recombinant E. coli systems. However, although it is not strictly speaking a hemo-
both the neuronal and macrophage NOS protein. Heme oxygenase catalyzes the rate-
enzymes have been successfully expressed limiting step in heme degradation, and the
in Saccharomyces cerevisiae (6,50,94). products biliverdin and carbon monoxide
The prototypical peroxidases, on which have been implicated in antioxidant activity
most of the mechanistic and structural and signal transduction pathways, respec-
studies of peroxidase chemistry have been tively (20,63,78). Heme oxygenase binds
determined, are the plant horseradish per- heme as a cofactor and substrate and, in the
oxidase (HRP) and the yeast cytochrome c absence of any reductant, has spectra simi-
peroxidase. The heterologous expression of lar to the oxygen-binding proteins hemo-
HRP has been primarily carried out in the globin and myoglobin (48,128,132,133).
baculovirus system, which yields active gly- The rat and human heme oxygenase have
cosylated holoprotein (39), and in E. coli, been expressed in recombinant E. coli sys-
where the protein is expressed as inclusion tems as both the full-length protein and a
bodies and is subsequently refolded in the soluble catalytically active form lacking the
presence of heme and calcium (104). hydrophobic C-terminal anchor domain
Cytochrome c peroxidase has been (48,126,128). The full-length rat heme
expressed primarily in yeast where active oxygenase (32 kDa) was expressed using the
holoprotein is obtained (84). Yeast cyto- lac promoter of pKK233-2 (Amersham
chrome c peroxidase has been expressed Pharmacia Biotech) in E. coli XL-1 blue.
heterologously in E. coli by replacing the fol The protein, although catalytically active,
gene in a vector used to overexpress dihy- was not homogenous as judged by a prote-
drofolate reductase with the cytochrome c olytic fragment at 28 kDa on sodium dode-
peroxidase gene (26). The expression levels cyl sulfate-polyacrylamide gel electrophore-
were 15 mg/L of cell culture, of which 10% sis (SDS-PAGE) (48). A more recent
was holoprotein and 90% apoprotein. approach, in which the protein was
Limited success has been achieved with expressed without the C-terminal hydro-
the mammalian lactoperoxidase (17), phobic domain as a soluble fully active pro-
myeloperoxidase (113), and prostaglandin tein, resulted in high-level expression of a
synthase (29), which have been expressed homogenous 30 kDa protein. The protein
exclusively in baculovirus Sf9 cells. In the was expressed under the control of the phoA
case of lactoperoxidase, the heme was only promoter at high level in E. coli DH5α cells
partially covalently bound (17). The auth- (128). The protein, although initially
ors reported that the noncovalently bound expressed in low phosphate media, was in
heme could be covalently attached to the later experiments expressed in LB media
protein on treatment with hydrogen perox- when it was observed that the expression
ide and proposed that such an autocatalyt- levels in either media were similar. The pro-
ic mechanism may be responsible for the tein was expressed in LB media for a num-
161
A. Wilks

ber of reasons, including the simpler proto- vectors (Novagen) to high levels utilizing
col and cost effectiveness. Recently, the sol- E. coli BL-21 (pLysS) strain (95). The
uble bacterial heme oxygenases of Syne- yields of protein from this vector ranged
chocystis sp. PCC 6803 and the pathogen from 100 mg/L in rich medium to 40
Corynebacterium diphtheriae have been mg/L in minimal media.
expressed in E. coli as catalytically active It is only recently that S. cerevisiae
proteins (14,129). The C. diphtheriae heme cytochrome c has been expressed heterolo-
oxygenase (HmuO) was expressed under gously in E. coli (81). The successful
the control of the T7 promoter with and expression of the holoprotein required the
without a 6-histidine tag at the C terminus, co-expression of cytochrome c heme–lyase
and no significant differences in protein for covalent attachment of heme a to the
yields or activity were noted (129). apoprotein. The successful expression of
the proteins was achieved by cloning the
2.1.3. Electron Transfer Proteins genes encoding the cytochrome (CYC1)
and the heme–lyase (CYC3) in parallel
As previously described, two of the crit- under the control of the Lac and Trc pro-
ical factors in the expression of many moters in the vector pUC18. The expres-
mammalian hemoproteins has been the sion system yielded 15 mg/L of active iso-
removal of a hydrophobic membrane 1-cytochrome c holoprotein.
anchor domain or redesigning the codons The expression systems described above
encoding the first 5 amino acids of the N encompass only a small fraction of hemo-
terminus. Sligar and coworkers (5) in early proteins, and while it is hard to generalize
studies completely synthesized the genes on the expression of a given class or a partic-
for both the complete rat hepatic ular hemoprotein, some simple generaliza-
cytochrome b5 with the membrane anchor tions can be made. First, hemoproteins with
and the protease-treated soluble form. In covalently attached hemes (cytochrome c)
addition, they incorporated an optimal and/or posttranslational modifications, such
ribosomal binding site and spacer region as glycosylation (myeloperoxidase, lactoper-
for expression of the proteins in E. coli (5). oxidase), have been more successful in
The soluble protein, when expressed in eukaryotic expression systems. Second, a
pUC, accounted for 8% of the total pro- critical factor in recombinant E. coli expres-
tein, with the membrane-bound protein sion of membrane bound hemoproteins,
being somewhat lower and fractionating such as the cytochrome P-450 enzymes, is
with the cell membrane. In later studies, the ability of the cells to synthesize heme
the use of the high expression T7 promoter and other cofactors required for correct fold-
was utilized in the expression of both the ing and activity of the holoprotein.
rat and human cytochrome b5. In these
later studies, polymerase chain reaction 2.2. Protein Purification Methods
(PCR) was used to engineer a sequence
encoding a 4-histidine tag at the N termi- 2.2.1. Oxygen-Binding Proteins
nus allowing rapid purification by nick-
el–chelate affinity chromatography (46). Purification of many hemoproteins has
Optimization of expression for structural been simplified with the development of
nuclear magnetic resonance (NMR) stud- recombinant expression systems and the
ies was carried out by Guiles and coworkers recent advances in affinity chromatogra-
in which they expressed the rat liver phy. Recombinant sperm whale myoglobin
cytochrome b5 in the T7 derived pET3C has been purified to homogeneity primari-
162
Analysis of Heme and Hemoproteins

ly utilizing ion exchange chromatography (5 × 30 s) prior to centrifugation


(110,127). Procedure 1 outlines how this (140 000× g for 30 min).
was achieved. 3. The resultant supernatant was stirred at
A significant amount of data on the 4°C, and ammonium sulfate was added
structure and function of hemoglobin has (final concentration 50%), and the
accumulated to date, however, there is less solution was stirred for a further 1
information available on the great number hour. The precipitates were collected
of naturally occurring hemoglobin mu- by centrifugation, and the supernatant
tants, of which many have significant clini- was taken to 95% saturation with
cal value. The problem of obtaining suffi- ammonium sulfate and stirred for 2
cient quantities of these naturally occurring hours. The precipitates were again col-
mutants has been circumvented by the lected by centrifugation, washed with
recent development of molecular biological and then resuspended in 50 mM sodi-
techniques. A similar approach to that um phosphate (pH 6.0).
taken with myoglobin, in which a synthet- 4. The protein was applied to a Bio-Gel
ic gene utilizing the preferred E. coli codon P100 column (2.5 × 100 cm; Bio-Rad
usage and the incorporation of a number of Laboratories, Hercules, CA, USA)
restriction sites for rapid and efficient cas- equilibrated with 50 mM Tris (pH
sette mutagenesis, has been described (41). 6.0). The fractions containing myoglo-
Initial studies utilized the co-expression of bin (as judged by the absorbance at 408
synthetic α and β-globins from the same nm) were pooled and concentrated.
operon downstream of the lac promoter
5. The protein was further purified by fast
(41). The two subunits combined intracel-
protein liquid chromatography (FPLC)
lularly with endogenously produced heme
on a Mono S HR-10 column (Am-
to give tetrameric hemoglobin in a yield of
ersham Pharmacia Biotech) using a lin-
5% to 10% of the total protein. This is
ear gradient of 50 mM sodium phos-
described in Procedure 2, which has been
phate (pH 6.0) to 75 mM sodium
adapted from Reference 42.
phosphate (pH 8.0) over 45 minutes.
The purified myoglobin was stored as
❖ Procedure 1. Purification of
is at -70°C until further use.
Recombinant Sperm Whale
Myoglobin
❖ Procedure 2. Purification of Human
1. Fresh overnight cultures of pMb221 Hemoglobin
(pUC18 containing the synthetically
constructed myoglobin gene) (5 mL) 1. Frozen cells (150 g) were resuspended
were used to inoculate 4 × 2 L of LB in 300 mL 15 mM Tris-HCl (pH 8.0),
media containing 100 µg/mL ampi- 0.1 mM EDTA, 1 mM dithiothreitol
cillin. The cells were grown for 18 (DTT), 1 mM MgCl2, 0.1 mM
hours and harvested by centrifugation. MnCl2, 40 mg RNase A, 5 mg RNase
2. The cells were resuspended in 50 mM B, 10 000 U of DNase. The cells were
Tris (pH 8.0), 1 mM ethylene diamine lysed by sonication and centrifuged (20
tetraacetic acid (EDTA), 1 mM min at 17 300× g). The supernatant
phenylmethylsulfonylfluoride (PMSF), was further clarified at 50 000 rpm for
50 U DNase I/mL, 5 U RNase/mL, 30 minutes.
and 50 µg/mL lysozyme and stirred for 2. The supernatant was clarified through a
1 hour at 4°C. The cells were sonicated coarse Sephadex G-25 column (4 × 70
163
A. Wilks

cm; Amersham Pharmacia Biotech) ❖ Procedure 3. Denaturation and


equilibrated with 50 mM Tris-HCl (pH Reassembly of Recombinant Human
8.0) containing 0.1 mM EDTA and 1 Hemoglobin
mM DTT (start buffer). The eluate was
loaded onto a DE52 column (4 × 8 cm; 1. Aliquots of purified 0.5 mM hemoglo-
Whatman, Clifton, NJ, USA) equili- bin (5 mL) were added to 200 mL of
brated with start buffer and washed with acid–acetone (2.5 mL HCl/L acetone)
a further 500 mL of the same buffer. at -20°C followed by centrifugation at
The hemoglobin fraction was eluted 7000 rpm in a Sorvall SS34 rotor
with a gradient of 500 mL each of start (Kendro Laboratory Products, New-
buffer and 15 mM Bis-Tris (pH 6.2) town, CT, USA) for 10 minutes at
containing 0.1 mM EDTA and 1 mM -20°C. The resultant hemoglobin pel-
DTT. All of the above buffers were equi- let was resuspended in 8 M urea, 5
librated with carbon monoxide. The mM sodium phosphate, and 50 mM
hemoglobin fraction was concentrated mercaptoethanol (pH 6.7) (urea
through PM30 Amicon membranes buffer). The sample was then dialyzed
(Millipore, Bedford, MA, USA). overnight in the same buffer. Following
dialysis any precipitation was removed
3. The concentrated hemoglobin fraction by centrifugation at 50 000 rpm in a
was passed through a coarse Sephadex 55.1 Ti rotor (Beckman Coulter,
G-25 column (as above) equilibrated Fullerton, CA, USA) for 30 minutes at
with 15 mM Tris-HCl (pH 8.0), and 15°C.
the eluate was high-performance liquid
chromatography (HPLC)-purified on a 2. The denatured protein was applied to a
DEAE 5PW TSK column (2 × 15 cm; CM Sepharose CL6B column (2 × 7
TosoHaas, Montgomeryville, PA, cm; Amersham Pharmacia Biotech)
USA) equilibrated with the same equilibrated in urea buffer. The column
buffer. The protein was eluted with a was washed with approximately 80 mL
gradient from 15 mM Tris-HCl (pH of the urea buffer until all of the non-
8.0) to 15 mM Bis-Tris-HCl (pH 7.0) binding protein was eluted. The β-glo-
at a flow rate of 5 mL over 85 minutes. bin was then eluted with a linear gradi-
ent of 180 mL each of urea buffer (pH
4. The hemoglobin fraction was then 6.7) and the same buffer containing 0.1
passed over a Sephadex G-25 column M NaCl. The α-globin was eluted with
equilibrated with 20 mM BisTris-HCl a second gradient of 0.1 to 0.2 M NaCl
(pH 7.0) and further purified by in the urea buffer. The eluted globin
HPLC on a SP 5PW TSK column (2 × fractions were concentrated over PM10
15 cm; TosoHaas) equilibrated with membranes (Millipore).
the same buffer. The protein was eluted
with a gradient of 20 mM BisTris-HCl 3. The starting concentrations in the urea
(pH 7.0) to 25 mM Tris-HCl (pH 8.0) buffer of the α and β-globins for
at 5mL/minute over 85 minutes. reconstitution were between 4 and 5
mg/mL, based on the Molar absorp-
The resulting protein showed significant tion coefficients of 1.0 × 104 and 1.54
heterogeneity within the subunit composi- × 104 for the α and β subunits, respec-
tion, which was only resolved by disassem- tively. Cyanohemin was added to a 1.2
bling the hemoglobin and reconstituting the molar excess of the combined globin
individual components in the presence of concentration, and the solution was
heme (42,47) as described in Procedure 3. diluted to a final concentration of 0.3
164
Analysis of Heme and Hemoproteins

mg/mL in cold deionized water equili- tion and solubilization of the membranes.
brated with carbon monoxide (CO) by The selection of a suitable detergent and its
bubbling with CO, and the pH was concentration in which to solubilize a
adjusted to 8.0 with a few crystals of given cytochrome P-450 enzyme is largely
Tris. The solution was left undisturbed a matter of trial and error. Solubilization of
overnight at 5°C and was then concen- the protein from bacterial cell pellets is
trated through PM30 membranes. The usually carried out in 100 mM Tris buffer
concentrated reassembled hemoglobin in the pH range 7.5 to 8.0, containing
was then reduced anaerobically with either 500 mM sucrose or 20% glycerol
sodium dithionite in the presence of (18,36). The outer membrane is then solu-
CO and passed through a Sephadex G- bilized in the presence of lysozyme and
25 column equilibrated with 15 mM protease inhibitors (1 mM PMSF, 2 µM
Tris-HCl (pH 8.4) to remove the leupeptin, 10 µM bestatin, and 0.04 U/mL
excess hemin and dithionite. aprotinin) with gentle stirring for 1 hour at
4. Separation of the excess α and β-globin 4°C. The spheroplasts are collected by cen-
chains was accomplished on a DE52 trifugation at 100 000× g for 1 hour. The
column (4 × 5 cm; Whatman) in 15 pellets can then be resuspended in the start
mM Tris-HCl (pH 8.4). The α-globin buffer for solubilization. Solubilization of
and intact hemoglobin were then elut- the cytochrome P-450 is carried out at 4°C
ed from the column with a gradient of for 60 minutes, and the solubilized enzyme
750 mL each of start buffer and 80 is centrifuged at 100 000× g for 60 min-
mM Tris-HCl (pH 8.0). The α-globin utes. In the purification of CYP4All, 1%
was eluted first followed by the hemo- Emulgen 911 appeared the most effective
globin as the major band with the in solubilizing the protein from bacterial
excess β-globin remaining bound to membranes (18). In the case of CYP1A1,
the column. CYP2C10, and CYP3A4, a combination
of 0.625% cholate and 0.62% Triton
2.2.2. Oxidases N101 was found to yield the greatest level
of soluble active holoprotein (36).
The purification of the cytochrome P- Although there is no overall scheme for
450 enzymes has been greatly simplified the purification of recombinant cytochrome
with the advent of recombinant E. coli P-450 enzymes, two general approaches
expression systems and the use of metal- have been taken. First, the purification of
chelate affinity chromatography tech- the native protein has largely been carried
niques. For the purpose of this review, we out by a series of ion exchange chromatogra-
will focus on the purification of cyto- phy steps on DEAE Sephacel (Amersham
chrome P-450 enzymes from bacterial sys- Pharmacia Biotech) and CM Sepharose
tems in light of the considerable advan- (36). More recently, a number of cyto-
tages in high level of expression, ease of chrome P-450 enzymes have been purified
manipulation, and the relatively low cost. utilizing nickel chelate chromatography
A number of laboratories have utilized the (18,69). In these proteins, a 6-histidine tail
pCW Ori+ vector (see Section 2.1.2) in the was engineered at the C terminus allowing
development of expression and purification binding to a nickel–nitriloacetic acid (Ni-
systems for many cytochrome P-450 en- NTA) agarose column. The protein can
zymes (30,31,33,35,38,65,131). then be eluted with imidazole in a 1-step
The critical step in the purification of purification. The nickel–chelate affinity
cytochrome P-450 enzymes is the prepara- method has also been extensively utilized in
165
A. Wilks

the purification of the nitric oxide synthase cytochrome c from E. coli cells has recently
enzymes (30,92,131). been reported (81). The preparation of the
soluble fraction is essentially the same as
2.2.3. Electron Transfer Proteins previously described for the other soluble
hemoproteins such as cytochrome b5 and
The purification of the engineered solu- myoglobin. Purification of the protein was
ble recombinant cytochrome b5 (5) has carried out by successive cation exchange
been the basis for a number of subsequent chromatography steps on CM Sepharose
purification schemes. CL6B and on Mono S HR 10/10 by FPLC.

❖ Procedure 4. Purification of 2.2.4. Affinity Chromatography of


Cytochrome b5 Heme-Binding Proteins
1. E. coli TB-1 cells were harvested and In recent years, the advances in affinity
lysed in 50 mM Tris-HCl (pH 8.0) chromatography have expanded dramati-
containing 0.1 mM DTT, 1 mM cally with the development of molecular
EDTA, RNase A (1 U/mL), DNase biological tools for the expression of fusion
(16 U/mL), and lysozyme (3 mg/mL). proteins, such as the polyhistidine-tag sys-
Cell debris was removed by centrifuga- tem (described above) and the cellulose
tion, and the supernatant was applied binding domain system (Novagen), which
to a DEAE Sephacel column (2.6 × 9.4 allow 1-step affinity purification of the tar-
cm) equilibrated in 50 mM Tris-HCl get protein. Historically however, affinity
(pH 8.0), 0.1 mM DTT, and 1 mM chromatography has utilized immobilized
EDTA. The protein was eluted with a coenzymes such as nucleotides or flavins.
linear gradient. One cofactor that has largely been over-
2. The fractions containing cytochrome looked in this aspect is heme, although
b5 were pooled, concentrated by ultra- hemin–agarose is commercially available
filitration, and applied to a Bio-Gel (Sigma, St. Louis, MO, USA) (120).
P-30 gel filtration column (1.9 × 73 Hemin–agarose has been used to copurify
cm; Bio-Rad) equilibrated in 50 mM hemopexin and HBP.93 from rabbit serum
Tris-HCl (pH 8.0). The fractions con- (121). However, one drawback in the use
taining cytochrome b5 were pooled of hemin–agarose is the stringent condi-
and reapplied to a DEAE Sephacel col- tions required for elution of the proteins.
umn as described above. The resulting In the case of the hemopexin and HBP.93
purified protein was then stored in liq- purification, the protein was eluted in 0.1
uid nitrogen. M acetic acid (121). One unusual charac-
Minor modifications of the above proce- teristic of the resin is the high affinity for
dure have been utilized in the purification heme itself, which eliminates the possibili-
of the soluble microsomal cytochrome b5, ty of hemin being used to specifically elute
in which the second ion exchange chro- the bound protein from the column. How-
matography step is replaced with a hydrox- ever, this property can be used to specifical-
yapatite column (95). The purification of ly remove heme or concentrate heme from
the outer mitochondrial membrane cyto- a large sample volume such as depro-
chrome b5 (89) has also been reported, and teinized body fluids.
the protocol is similar to that described One area of research that hemin–agarose
previously (5). has been particularly useful is in the identi-
The purification of S. cerevisiae iso-1- fication of heme-binding proteins from
166
Analysis of Heme and Hemoproteins

pathogenic bacteria. Pathogenic bacteria be carried out using the pyridine hemo-
require iron for their survival, and this chrome assay (24,25,114).
requirement is in part linked to their viru-
lence (8). The major source of iron within ❖ Procedure 5. Pyridine Hemochrome
the host is found in heme and hemopro- Method
teins, and many pathogens have developed
1. The isolated hemoprotein in a 4.0-mL
sophisticated uptake systems for the acqui-
sample volume is converted to a pyri-
sition of heme from their environment. A
dine hemochrome by the addition of
number of heme receptors and periplasmic
0.5 mL of pyridine and 0.5 mL 0.5 N
transport proteins have been isolated utiliz-
NaOH solution.
ing hemin–agarose, including outer mem-
brane lipoprotein heme receptors from 2. The reduced spectrum (+ dithionite)
Haemophilus influenzae (60) and Haemo- minus the oxidized spectrum (no
philus ducreyi (22), as well as a secreted dithionite) is calculated at 550 (proto-
hemoglobin–protease in pathogenic E.coli heme) or 557 nm (heme c). The
strain EB1 (80). millimolar extinction coefficients
(∆εox-red) at each of the wavelengths
are 30.0 and 19.1 at 557 and 550 nm,
3. DETECTION AND QUANTITA- respectively (87).
TION OF HEMOPROTEINS The method has also been applied to
more complicated samples such as mito-
A unique and useful property of hemo- chondria, yeast, and photosynthetic bacte-
proteins is the ability to determine the ria (1,4,54,56,87,101). In such cases, it
nature of the heme as well as quantitate was necessary to remove lipids and conta-
both the heme and protein based on the minating photosynthetic pigments by sol-
absorption characteristics of the chromo- vent extraction. Protoheme and heme c can
phore. In addition, the absorption proper- then be separated by differential extraction
ties provide valuable information as to the in acidic organic solvent (25). However,
redox and spin state of individual hemopro- the pretreatment of heme samples must be
teins in multicomponent systems. These approached with some caution. Degrada-
techniques are invaluable in the study of tion of hematins in alkali solution has been
complex systems where individual spectral observed, and the pyridine ferrohemo-
characteristics can be accurately determined. chrome of protoheme is especially labile.
Differential extraction of heme in HCl/
3.1. Detection of Heme in Microsomes acetone results in the partial extraction of
and Whole Cells heme c from tissue samples. Improved
A number of methods for heme detec- methods of detection and quantitation of
tion and quantitation in tissue extracts heme have been developed (as described
have been described. All of the methods above). These methods are based on the
rely on monitoring the distinctive Soret redox difference absorbance measured at 2
absorbance either directly or following wavelengths of the pyridine hemochrome.
extraction of the chromophore. These methods can be utilized without
prior separation of the hemes and organic
3.1.1. Pyridine Hemochrome solvent treatment (52). The method
described below has been successfully used
Assignment of heme type in isolated to quantitate the hemes in the photosyn-
hemoproteins as well as quantitation can thetic bacteria R. sphaeroides (52).
167
A. Wilks

❖ Procedure 6. Detection of Hemes in hemoproteins, such as hemoglobin, also


Photosynthetically Grown Cells of react with carbon monoxide and may
R. sphaeroides interfere with accurate determination of
cytochrome P-450 content. In tissue
1. Cells grown at 30°C are disrupted with extracts or microsomes contaminated with
a sonic oscillator. other membrane fractions, this can be a
2. Pyridine (0.5 mL) and 0.5 mL of 0.5 significant problem, which requires modi-
N NaOH solution are added to 4.0 mL fication of the procedure (23). The amount
of the disrupted cells. of cytochrome P-450 is calculated from the
3. The difference absorbance is measured difference spectra of the reduced CO sam-
between the oxidized minus reduced ple versus the reduced contents of the ref-
samples at 2 wavelength sets ∆(∆red - erence sample with any contribution from
ox)555.5-535 and ∆(∆red - ox)549.5-535. cytochrome b5 being cancelled out. The
4. The amount of protoheme and heme c change in absorbance between 450 nm rel-
in the samples can then be calculated ative to 490 nm can then be converted to a
from the millimolar absorption coeffi- concentration based on the millimolar dif-
cients 24.0 for ∆(∆red - ox)555.5-535 ference absorption coefficient (εmM = 91).
and 10.9 ∆(∆red - ox)549.5-535 for pro- In a separate sample at the same concentra-
toheme and 6.46 ∆(∆red - ox)555.5-535 tion (usually 1.5 mg/mL protein concen-
and 22.0 ∆(∆red - ox)549.5-535 for tration), the reduced minus oxidized spec-
heme c. trum can be measured to determine the
It is critical the measurements be carried cytochrome b5 content. By again applying
out within a few hours of cell disruption, the millimolar difference absorption coef-
and that the samples are maintained at ficient of 21 for the absorbance change at
4°C. The redox difference spectra should 556 minus 575 nm, or of 185 from the
be measured immediately following the change at 426 minus 409 nm, the
addition of dithionite, since the ferrohe- cytochrome b5 concentration can be calcu-
mochrome is labile. lated. One point of note is that NADH
should be used in preference to NADPH
3.1.2. Difference Spectrophotometric for the reduction of cytochrome b5, as
Analysis NADPH contributes to endogenous sub-
strate turnover by cytochrome P-450, and
The application of difference spec- therefore cytochrome b5 may only be
trophotometry to pigments, as developed reduced 75% to 85% in samples not com-
by Chance and coworkers, has become a pletely purged of oxygen.
powerful method to study electron-trans- The identification and quantitation of
port reactions in turbid solutions and to cytochromes by low temperature difference
monitor changes in oxidation state of spectral analysis in liver microsomes, yeast,
membrane bound respiratory hemopro- and bacteria have previously been
teins (10). In microsomes of liver, kidney, described (61,68,70,82). The major advan-
and the adrenal cortex, the main hemopro- tage of low temperature spectral analysis is
tein components are cytochromes b5 and the sharpening of bands which may other-
P-450, which can be measured by differ- wise be overlapping at room temperature.
ence spectrophotometry. The cytochrome In the yeast S. cerevisiae, the characteris-
P-450s are best characterized by the ab- tic α bands in the 550 nm region of the
sorbance band at 450 nm for the reduced reduced cytochromes b, b1, c, and c1 are
carbon monoxide adduct. However, other ideal for measuring transitions in cells
168
Analysis of Heme and Hemoproteins

undergoing adaptation from an anaerobic of microsomal mono-oxygenase systems


to an aerobic environment. In E. coli, low has been developed based on principal
temperature difference spectrum (α and β component analysis (PCA) or singular
regions) analysis of the succinate-reduced value decomposition (SVD) analysis (45).
minus oxidized cells has identified absorp- This method is widely used to interpret flu-
tion peaks at 528 and 556 nm representing orescence and absorbance spectral changes
the β and α bands, respectively, of b and c in complex biochemical systems (37,45,
type cytochromes. Also identified in the α 85,102). By deducing a set of standard
region, although at much lower intensity, absorbance spectra for the high-spin, low-
are bands due to cytochrome a1 (590 nm) spin, and P420 states of pure ferric micro-
and cytochrome d (628 nm) (82). These somal cytochrome P-450, and using PCA
bands revealed considerable heterogeneity analysis, the thermodynamic parameters of
and were further resolved by fourth-order substrate binding and spin transitions in
finite difference analysis into components yeast microsomes containing human cyto-
at 542, 545.5, 553.5, and 563 nm and chrome P-450 3A4 have been evaluated
peaks at 520.5, 528, 535, and 537 nm for (86).
the α and β bands, respectively.
3.1.4. Multicomponent Spectral Analysis
3.1.3. Substrate Binding and Spin of Hemoproteins in Biological
Transitions Materials
While the pyridine hemochrome meth- Spectral scans across a range of visible
od and difference spectrophotometric wavelengths (390–640 nm) contain a great
analysis are effective in analyzing the com- deal of information about the oxidation
position and quantitative aspects of heme state of individual hemoproteins. Informa-
content in tissues, measurement of hemo- tion can be obtained for tissue homo-
protein spin-transitions is somewhat more genates or subcellular fractions through
problematic. However, the measurement of comparison to reference spectra by multi-
spin-transitions in cytochrome P-450 sys- component analysis. A hemoprotein spec-
tems is particularly relevant given the direct tral analysis program (HSAP), a spread-
relationship of functional properties of the sheet program written with Microsoft
monoxygenase system with spin-state. A Excel 4.0, has been described (7). The
number of systems for cytochrome P-450 program measures absorbances at many
have been developed which rely on the dif- wavelengths in the range of 500 to 640 nm
ference in Soret absorption spectra between and obtains molar ratio values for the indi-
the high-spin (λmax 388–396 nm) and low- vidual hemoproteins. To correct for light-
spin (λmax 416–418 nm) (34,51,88,112). scattering errors in turbid samples, a tur-
However, microsomal cytochrome P-450s bidity calculation has been introduced to
are problematic in that they undergo spon- the HSAP (13). This program is effective
taneous inactivation to form the P-420 in measuring reduced hemoproteins, while
(inactive form) protein. The overlap of the the unambiguous identification of oxidized
450 and 420 wavelengths, together with hemoproteins was not so straightforward.
the turbidity of the sample and the overlap The assignment of oxidized hemoproteins
of the P-450 absorbance bands with other was accounted for by the addition of spec-
hemoproteins such as cytochrome b5, tral data in the strongly absorbing Soret
makes direct difference spectral analysis dif- region, where the peak maxima are suffi-
ficult. A functional model for the analysis ciently different to allow discrimination
169
A. Wilks

between the reduced and oxidized proteins. tain the heme-associated peroxidase activi-
Subsequent development of a difference ty. The gels (12 cm in length and 1.5 mm
spectral analysis program (DSAP) has fur- thickness) are run overnight at 4°C with a
ther aided the separation and identification constant current (8 mA/gel) (21).
of individual cytochromes of organelles The following procedure has been mod-
such as mitochondria (74). The combina- ified slightly by Dutta and Henry (21) for
tion of HSAP and DSAP provide a method use in electroblotting experiments on
for interpreting complex spectral informa- polyvinylene difluoride (PVDF) mem-
tion of biological samples and may prove brane. Following transfer of the protein to
useful with other measurements of tissue PVDF membranes, the gels are incubated
oxidation in assessing clinical problems in the TMBZ solution for 3 hours at 4°C
associated with free radical reactions. (in the dark). H2O2 is added to a final con-
centration of 30 mM, and the staining is
3.1.5. Detection of Hemoprotein visible after 1 hour (storage in the staining
Peroxidase Activity on SDS-PAGE solution for periods greater than 3 hours
results in irreversible background staining).
Electrophoresis of proteins is commonly The membranes can be quickly destained
used in determination of protein purity, by washing twice in 100% methanol, fol-
molecular weight, and in the case of iso- lowed by destaining in 70 mM sodium sul-
electrofocusing (IEF), the pI of a given fite for 5 minutes. The membranes can
protein. This technique can be useful in then be stained with Coomassie Brilliant
identifying hemoproteins of unknown Blue R-250, 50% methanol, and 10%
molecular weight in an impure protein acetic acid and destained in 90% methanol.
fraction or isolate by taking advantage of
the heme-associated peroxidase activity as a ❖ Procedure 7. TMBZ Staining of
visualization tool (96,116). In this proce- Polyacrylamide Gels
dure, a substrate specific for heme, 3,3′,5,
5′-tetramethylbenzidine (TMBZ) is incu- 1. A solution of 6.3 mM TMBZ in meth-
bated with the SDS-PAGE gel and visual- anol is prepared freshly prior to use.
ized by the addition of hydrogen peroxide Immediately before use, 3 volumes of
(H2O2). One major disadvantage of the the TMBZ methanol solution is mixed
use of this staining technique in SDS- with 7 parts of 0.25 M sodium acetate,
PAGE analysis is that the denaturing con- pH 5.0. The gels are then immersed in
ditions often lead to loss of noncovalently this solution for a period of 1 to 2
bound heme from the protein during the hours (in the dark) and mixed every 15
electrophoresis run. This problem has been minutes.
circumvented with the use of lithium 2. Following incubation, the gels are
dodecyl sulfate (LDS) in place of the SDS. stained with the addition of H2O2 to a
The gels are run as previously described final concentration of 30 mM. The
(59), except SDS is replaced by LDS in all staining should be visible within 3 to 5
buffers. The sample buffer is prepared with minutes with increasing intensity over
a lower concentration of DTT (5 mM), as a 30-minute period.
this can interfere with the TMBZ staining 3. The gels are then placed in isopropa-
of the gel following electrophoresis. The nol:0.25 M sodium acetate (pH 5.0)
samples are immediately loaded onto the (3:7). The solution is changed 3 times
gel following addition of LDS-PAGE sam- to remove any precipitated TMBZ and
ple buffer and are not boiled so as to main- clear the gel background.
170
Analysis of Heme and Hemoproteins

4. The gels can be scanned at 690 nm or ❖ Procedure 8. Detection of Heme and


photographed at this time. Stained gels Hemoproteins by Chemiluminescence
can be stored for a period of 2 months
in the dark at room temperature. 1. The detection of chemiluminescence is
based on the peroxidase-dependent
5. The gels can be destained in 70 mM breakdown of luminol, and the reac-
sodium sulfite and washed (3 × 20 tions were carried out by modification
min) with 30% isopropanol and of a standard procedure from the
stained in Coomassie brilliant blue ECL blotting system commercially
R-250 (21). available from Amersham Pharmacia
Biotech.
3.2. Detection and Quantitation of 2. Gel electrophoresis: SDS-PAGE gels,
Heme-Containing Proteins by run with or without reducing agents,
Chemiluminescence electroblots, or dotblots, are washed in
Dulbecco’s phosphate-buffered saline
The techniques described above for the (dPBS) and incubated for 1 minute in a
detection of peroxidase activity in hemo- 1:1 mixture of CL reagents 1 and 2
proteins, while useful, has a number of (ECL Blotting Kit). The gels or blots are
drawbacks including prolonged incuba- then placed between transparent acetate
tion periods, sensitivity to light, and car- sheets and exposed to X-Omat film
cinogenicity. The benzidine staining pro- (Eastman Kodak, Rochester, NY, USA)
cedure is also a useful marker for the for a period of time (10 s to 18 h).
presence of blood in clinical and forensic 3. Liquid scintillation methods: Measure-
samples. To this end, a number of com- ment of chemiluminescence was car-
mercially available products have been ried out by mixing 10 µL of each
developed (9,19) which provide both reagent (CL1 and CL2) in a microfuge
qualitative and semiquantitative evidence tube followed by 1 µL of sample. The
of blood in clinical samples and do not solution was mixed and immediately
utilize the use of TMBZ. placed in a plastic scintillation vial, and
The development of luminol-based the emission of light was measured for
chemiluminescence assays for detecting 60 seconds. The samples were mixed
HRP-labeled antibody have been reported and placed in the instrument individu-
to have increased sensitivity to the standard ally to keep constant the time between
chromogenic techniques (9). With the in- mixing and scintillation spectroscopy.
creased sensitivity of chemiluminescence, The values obtained were in counts per
the ability to detect peroxidase activity in minute (cpm).
other hemoproteins has been analyzed. The chemiluminescence assay with
Comparison of the luminescence assay with HRP and cytochrome c, when compared
the standard chromogenic techniques on to the heme staining procedure, shows in-
SDS-PAGE and electroblot for several creased sensitivity. While both methods
hemoproteins indicated increased sensitivi- could detect protein on SDS-PAGE and
ty for qualitative detection of hemoproteins electroblot to 10 and 100 ng, respectively,
(19). Utilizing a liquid scintillation proce- the chemiluminescence assay can detect 1
dure, linear and quantitative detection of ng of HRP and 10 ng of cytochrome c
peroxidase activity could be measured for when exposed for 18 hours (19).
biological samples containing heme in the Chemiluminescence methods developed
pmol range (see methods below). for scintillation detection assays has been
171
A. Wilks

utilized for the detection of blood in urine ture for the membrane-bound P-450 iso-
(hematuria). The detection of blood in zymes has led to the use of indirect meth-
urine showed a linear relationship over a ods for determining active-site structures,
wide range of concentrations with the detec- such as homology modeling (69,83),
tion level as low as 0.1 to 1 pg/µL (19). mechanism-based inactivation (57,58),
The use of chemiluminescence in the and photoaffinity labeling (67,75,134). Al-
detection of hemoproteins has been modi- though such probes have not yet been rou-
fied to specifically detect oxidative metabo- tinely used with hemoproteins, the future
lites of hemoglobin and myoglobin (125). development of photoaffinity probes with
Hemoglobin is a major target for reactive such a broad specificity opens up the possi-
metabolites of drugs as well as environmen- bility of determining topological informa-
tal toxins, and the relatively long half-life of tion on the active sites of hemoproteins
the red blood cell has led to its use as a whose structures are unknown.
marker of exposure to such toxic metabo-
lites (79). Hydrogen peroxide at low con- 4.1. Photoaffinity Labeling with
centrations reacts with the heme group of Synthesizing Trifluoromethyl-
myoglobin and hemoglobin to form a cova- diazirinylphenyl Diazenes
lent heme–protein adduct. The modified
hemoprotein is then capable of generating Recently, Tschirret-Guth and Ortiz de
hydrogen peroxide in an essentially autocat- Montellano (119) have advanced the
alytic process. Thus, the covalent modifica- active-site labeling methodology developed
tion of the protein is an important marker in their laboratory. In previous studies, it
for oxidative damage and, more specifically, was noted that reaction of arylhydrazines
in the case of myoglobin, a sensitive marker and aryldiazenes with hemoproteins gave
for ischemic reperfusion injury. an iron–aryl complex (76). Early work uti-
Utilizing the chemiluminescence proce- lized the fact that, upon oxidation, the aryl
dure outlined above, Vuletich and Osawa complex shifted to form a mixture of N-
have modified the reaction conditions to arylprotoporphyrin-IX regioisomers. The
specifically select for protein bound heme ratio of the 4 regioisomers represented a
adducts on SDS-PAGE gels (125). The use low-resolution topology of the active-site,
of DTT is largely excluded as a reducing but gave no information on the specific
agent as it degrades the heme and, hence, amino acid residues within the active-site.
reduces the peroxidase activity of the heme The recent synthesis of azidoaryldiazenes as
adduct. Replacement of DTT with tris(2- photoaffinity probes for the active-site of
carboxyethyl) phosphine (TCEP), which hemoproteins has addressed this question
does not degrade the heme, allows detec- (119). The formation of the iron–aryl
tion of the peroxidase activity of the heme complex with the heme and subsequent
adduct with no contamination from non- formation of the activated nitrene by oxi-
covalent heme-containing proteins, as the dation limits the interaction to amino acid
noncovalent heme is lost on denaturation residues within the active site of the hemo-
and electrophoresis. protein (Figure 1a). The reaction of azido-
phenyldiazene with myoglobin was specific
for the distal histidine (His-64), suggesting
4. HEMOPROTEIN ACTIVE-SITE the probe preferentially selected for nucle-
PROBES ophilic residues (117). To develop pho-
toaffinity probes that were capable of label-
The lack of any available crystal struc- ing multiple residues, the authors replaced
172
Analysis of Heme and Hemoproteins

the azido function with the more reactive electronic nature of the heme. In addition
trifluoromethyl group by synthesizing 3- to providing valuable information on the
and 4-(trifluoromethyldiazirinyl)phenyl ligation and coordination state of the
diazene (118) (Figure 1b). Irradiation of heme, these techniques have aided greatly
the sample following formation of the in elucidating and confirming the mecha-
iron–diazirinylphenyl complex at 350 nm nism of action of the enzyme.
generates a highly reactive carbene capable HO-1 catalyzes the rate limiting step in
of inserting into unactivated C-H bonds. the conversion of heme to biliverdin (Fig-
The reaction of sperm whale myoglobin ure 2), and although not a hemoprotein on
with the activated carbene and subsequent reconstitution with heme, the enzyme
analysis of the tryptic peptides by mass forms a 1:1 ferric heme:HO-1 (heme:HO-
spectrophotometric techniques identified 1) complex with electronic spectra similar
at least 4 residues within the heme-binding to that of myoglobin (128). At pH 6.0, the
site of the protein, Leu-29, Val-68, His-64, heme:HO-1 complex has a Soret at 404
and Ile-107. With the exception of His-64, nm and a charge transfer band at 632 nm,
which was previously identified in the ear- characteristic of a high spin species (Figure
lier azidophenyldiazene work (117), the 3 3b). On increasing the pH from 6.0 to
remaining residues were nonpolar and less 10.0, the heme Soret shifts from 404 to
than 3 to 4 Å from the heme. 409 nm, and the charge transfer band at
630 nm decreases with the appearance of
α/β bands at 539 and 574 nm, a process
5. SPECTROPHOTOMETRIC attributed to a pH-dependent high-spin to
ANALYSIS OF HEMOPROTEINS low-spin conversion of a water ligand to a
hydroxide (Figure 3b) (40,111). The
The unique chromophore of hemopro- UV/visible spectra of the reduced complex
teins allows the use of a number of spec- in the presence of CO has a Soret at 420
trophotometric techniques to yield detailed nm and α/β bands at 567 and 537 nm,
information on the nature of the heme lig- consistent with a histidine proximal ligand.
and. Although individual spectroscopy Although the UV/visible spectrum can
techniques can yield valuable information, provide some limited information on the
it is the combined use of techniques such proximal ligand of a hemoprotein of
as UV/visible, magnetic circular dichroism unknown structure, it is not conclusive.
(MCD), resonance Raman, and NMR that MCD is a difference technique that will
can provide powerful insight as to the have both positive and negative compo-
nature of the coordination, ligation, and nents yielding a more detailed spectrum
oxidation state of the heme. than the corresponding UV/visible spec-
It will not be the focus of this review to trum. The increased sensitivity of MCD
give a detailed account of each technique, has been shown to be an effective “finger-
but present a brief synopsis of the informa- printing tool” for the determination of the
tion that can be gained as it relates to ligand coordination, oxidation state, and
hemoprotein structure and function. On spin state of hemoproteins (15,123).
studies of the human heme oxygenase Assignment of the proximal ligand in a
(HO-1), we have previously used a combi- hemoprotein of unknown structure by
nation of spectrophotometric techniques MCD, like UV/visible spectroscopy and
such as UV/visible, resonance Raman, resonance Raman (see below), is dependent
MCD, and NMR spectrophotometry to on comparison to a protein whose proxi-
determine the ligation, coordination, and mal ligand is known, such as myoglobin
173
A. Wilks

(histidine), catalase (tyrosine), and cyto- dine ligation in the heme:HO-1 complex
chrome P-450 (cysteine). The sample to be (Figure 3a) (40). The derivative bands of
analyzed is prepared with a known sixth the heme:HO-1 complex are blue-shifted
ligand such as cyanide, which together when compared to myoglobin, but are still
with the 4 pyrroles can then be compared very similar in structure (Figure 3b) (40).
to the ferric cyano complex of hemopro- Raman spectroscopy is a vibrational
teins, whose proximal ligands are known. spectroscopy technique that can detect
The electronic absorption and MCD spec- transitions between different ligation states
tra of the cyanoferric heme:HO-1 complex of porphyrins as the spin state is changed
is very similar to that of cyanoferric myo- (107,109). The change in spin state alters
globin, thus confirming the proximal histi- the size of the iron and its displacement

Figure 1. (a) Labeling of


active-site residues of hemo-
proteins with photoac-
tivable probes. (b) Structure
of the photoactivatable
probes of trifluoromethyl-
diazirinylphenyldiazene as
synthesized by Tschirret-
Guth et al. (118).

174
Analysis of Heme and Hemoproteins

from the heme plane which directly effects corresponding to the ferric 6-coordinate
the vibrational frequencies of the por- low-spin species (6CLS) at 1503 (υ3),
phyrin ring bond stretches. This technique 1582 (υ2), and 1638 (υ10) increasing (Fig-
can be used to characterize the spin state ure 4). This again supports the ionization
and coordination state in both the ferric of a coordinating water molecule to a
and ferrous oxidation states of a given hydroxide above pH 8.0.
hemoprotein (108). As in the MCD exper- Resonance Raman studies of the heme:
iments, the resonance Raman spectra of HO-1 complex provided direct confirma-
the heme:HO-1 complex show a marked tion of histidine as the proximal ligand
change in the heme axial coordination and (111). The iron–imidazole or iron–thiolate
spin state on changing pH (111). At pH ligand is not generally detectable due to the
6.0, the spectrum shows the heme to be weak coupling of the π electronic system.
primarily 6-coordinate high-spin (6CHS) However, in the case of the iron–imidazole
similar to myoglobin, in which the heme is (Fe-His) ligation, the reduced ferrous 5-
coordinated through a proximal histidine coordinate species shows significant en-
with water bound as the sixth ligand. hancement due to the iron being out of the
Increasing the pH to 8.0 significantly alters plane of the porphyrin. This transition
the Raman spectrum, with the bands at provides a characteristic marker for the Fe-
1483 (υ3) and 1565 cm-1 (υ2) of the ferric His stretching mode in the low frequency
6CHS species decreasing, and the bands ferrous spectra. Interestingly, the position

Figure 2. Conversion of
heme to biliverdin by
HO-1. The abbreviations
are as follows; Me,
methyl; V, vinyl; Pr, pro-
pionic acid.

175
A. Wilks

of the band is also a strong indication of is weakly or not hydrogen-bonded (106).


the ionization state of the proximal histi- It is interesting that the structure of the
dine. The band at 216 cm-1 in the heme:HO-1 complex resembles the oxygen-
heme:HO-1 complex (111) is close to that binding proteins more than the oxygen-acti-
of myoglobin (221 cm-1) (53,55), where vating peroxidases. The presence in the
the proximal histidine is only weakly heme:HO-1 complex of a histidine proxi-
hydrogen-bonded (Figure 5). The presence mal ligand that is neither ionized nor hydro-
of strong hydrogen bonding to the proxi- gen-bonded has strong implications for the
mal histidine in cytochrome c peroxidase mechanism of action of HO-1. The ability
shifts the iron–imidazole stretch to 233 of the proximal ligand to destabilize the
cm-1, while complete ionization gives a dioxygen bond and assist in cleavage to form
value closer to 246 cm-1 (105,115). In the activated ferryl species is largely depen-
cytochrome c peroxidase, it has been dent on the electron density present on the
shown that removal of the hydrogen bond- proximal ligand. As the electron density
ing network by mutation of Asp-235 to an increases by partial or complete deprotona-
Ala shifts the Fe-His stretching mode from tion of the proximal histidine, the ability to
246 to 206 cm-1. The relatively low value carry out dioxygen bond cleavage increases.
for the heme:HO-1 complex can therefore In previous studies, we have generated a
be interpreted as a proximal histidine that ferryl species analogous to that of com-

Figure 3. (a) MCD and (b) electronic


absorption spectra of the cyanoferric
heme:HO-1 complex and of the cyano-
ferric sperm whale myoglobin. The
MCD spectra of the human HO-1 and
sperm whale myoglobin have previously
been reported in References 40 and 16,
respectively.

176
Analysis of Heme and Hemoproteins

pound II in peroxidase and shown that this trophilic mechanism is consistent with the
species is not an intermediate in the nor- presence of a proximal ligand that is nei-
mal physiological reaction of heme oxyge- ther ionized or hydrogen-bonded.
nase (128). Subsequent studies with ethyl- Isotopic labeling and 2-dimensional
hydroperoxide showed that the reaction NMR studies on the rat HO-1 have
proceeded by the normal pathway with the revealed an unusual heme electronic struc-
formation of α-meso-ethoxyheme, analo- ture (43). The proton contact shift patterns
gous to the formation of α-meso-hydroxy- of the heme reveal large differences in the
heme in the enzymatic conversion of heme delocalized spin density in which the spin
to biliverdin (130). The ethylhydroperox- density is highest at positions 3 (methyl)
ide reaction, therefore, favors an electro- and 2 (vinyl) and much smaller at posi-
philic mechanism with the addition of the tions 1 (methyl) and 4 (vinyl) (Figure 7).
terminal ethoxy or hydroxy (Fe-O-OH) The asymmetric spin density within a pyr-
moiety to the heme (Figure 6). The addi- role is unprecedented in any low-spin ferric
tion of the terminal oxygen of the ferric hemoprotein, where it is more common to
peroxo anion (Fe-O-O-) to the α-meso- find asymmetry between different pyrroles.
edge of the heme is ruled out as the termi- Based on studies with model hemes, the
nal oxygen of the ethylhydroperoxide distribution of the spin density suggests a
species is blocked. The proposed elec- direct electronic perturbation of the heme

Figure 4. Resonance Raman spec-


tra of the heme:HO-1 complex in
the spin and coordination state
frequency region as a function of
pH (413.1 nm excitation). The
resonance Raman data for the
heme:HO-1 complex has previ-
ously been reported (111).

177
A. Wilks

by the protein environment such that there 6. SUMMARY


is an increase in electron density at the α-
meso-edge of the heme. The increased elec- The area of hemoprotein research has
tron density at the α-meso-edge would advanced dramatically in recent years with
facilitate the electrophilic addition of the the advent of molecular cloning tech-
protonated terminal oxygen to the α- niques. The availability of high level ex-
meso-position (Figure 6). pression systems for hemoproteins, such as
The extensive characterization of the the cytochrome P-450 enzymes, and the
heme:HO-1 complex by spectroscopic ability to generate site-directed mutants has
techniques such as MCD, resonance advanced our knowledge of both mech-
Raman, and NMR have greatly aided the anism and structure. The rapidly ex-
biochemical and enzymatic studies in eluci- panding role of hemoproteins in cell signal-
dating the mechanism of action of HO-1. ing processes typified by the eukaryotic
It is also gratifying that the recent 3-dimen- nitric oxide synthases and soluble guanylate
sional structure of the heme:HO-1 complex cyclase, and in gene regulation with tran-
has in large part confirmed many aspects of scription factors such as CooA, has expand-
the previous spectroscopic data (97). ed the repertoire of hemoprotein motifs. It

Figure 5. Resonance Raman spec-


tra of the ferrous heme:HO-1
complex and the manganese (II)
protoporphyrin:HO-1 complex
in the low frequency region
(441.6 nm excitation). The reso-
nance Raman data for the
heme:HO-1 complex has previ-
ously been reported (111).

178
Analysis of Heme and Hemoproteins

is with great anticipation that we look for- tral analysis program; IEF, isoelectric focus-
ward to the discovery of new and unique ing; H2O2, hydrogen peroxide; LDS, lithi-
hemoproteins and the critical role they may um dodecyl sulfate; MCD, magnetic circu-
play in molecular and cellular processes. lar dichroism; NMR, nuclear magnetic
resonance; PAGE, polyacrylamide gel elec-
trophoresis; PCA, principal component
ABBREVIATIONS analysis; PMSF, phenylmethylsulfonyl flu-
oride; PVDF, polyvinylene difluoride
DTT, dithiothreitol; EDTA, ethylene membrane; SDS, sodium dodecyl sulfate;
diamine tetra acetic acid; HO-1, human SVD; singular value decomposition analy-
heme oxygenase isozyme 1; HRP, horserad- sis; TCEP, tris(2-carboxyethyl) phosphine;
ish peroxidase; HSAP, hemoprotein spec- TMBZ, 3,3′,5,5′-tetramethylbenzidine.

Figure 6. Electrophilic oxidation of the porphyrin macrocycle by the ferric peroxide complexes.

Figure 7. Schematic representa-


tion of the unpaired spin distri-
bution about the heme periphery
in HO-1 and myoglobin. The size
of the circle represents the popula-
tion of the spin distribution.
Adapted from Reference 33.

179
A. Wilks

ACKNOWLEDGMENTS 14.Cornejo, J., R.D. Willows, and S.I. Beale. 1998. Phy-
tobilin biosynthesis: cloning and expression of a gene
encoding soluble ferredoxin-dependent heme oxyge-
I would like to thank Paul Ortiz de nase from Synechocystis sp. PCC 6803. Plant J. 15:99-
Montellano for his continuous support and 107.
encouragement over the years. I also thank 15.Dawson, J.H. and D.M. Dooley. 1989. Iron Por-
phyrins. Part 3, p. 1-135. In A.P.B. Lever and H.B.
the National Institutes of Health for their Gray (Eds.). VCH publishers, New York.
financial support. 16.Dawson, J.H., S. Kadkhodayan, C. Zhuang, and M.
Sono. 1992. On the use of iron octa-alkylporphyrins
as models for protoporphyrin IX-containing heme sys-
tems in studies employing magnetic circular dichroism
REFERENCES spectroscopy. J. Inorg. Biochem. 45:179-192.
17.DePillis, G.D., S. Ozaki, J.M. Kuo, D.A. Maltby, and
1.Agalidis, I., E. Jauneau, and F. Reiss-Husson. 1974. P.R. Ortiz de Montellano. 1997. Autocatalytic pro-
Influence of iron deficiency on the cytochromes of cessing of heme by lactoperoxidase produces the native
Rhodopseudomonas spheroides. Eur. J. Biochem. 47:573- protein-bound prosthetic group. J. Biol. Chem.
580. 272:8857-8860.
2.Aono, S., H. Nakajima, K. Saito, and M. Okada. 18.Dierks, E.A., Z. Zhang, E.F. Johnson, and P.R. de
1996. A novel heme protein that acts as a carbon Montellano. 1998. The catalytic site of cytochrome
monoxide-dependent transcriptional activator in Rho- P4504A11 (CYP4A11) and its L131F mutant. J. Biol.
dospirillum rubrum. Biochem. Biophys. Res. Commun. Chem. 273:23055-23061.
228:752-756. 19.Dorward, D.W. 1993. Detection and quantitation of
3.Barnes, H.J., M.P. Arlotto, and M.R. Waterman. heme-containing proteins by chemiluminescence.
1991. Expression and enzymatic activity of recombi- Anal. Biochem. 209:219-223.
nant cytochrome P450 17 alpha-hydroxylase in 20.Durante, W., M.H. Kroll, N. Christodoulides, K.J.
Escherichia coli. Proc. Natl. Acad. Sci. USA 88:5597- Peyton, and A.I. Schafer. 1997. Nitric oxide induces
5601. heme oxygenase-1 gene expression and carbon monox-
4.Bartsch, R.G. 1971. Cytochromes: bacterial. Methods ide production in vascular smooth muscle cells. Circ.
Enzymol. 23:344-363. Res. 80:557-564.
5.Beck von Bodman, S., M.A. Schuler, D.R. Jollie, and 21.Dutta, C. and H.L. Henry. 1990. Detection of hemo-
S.G. Sligar. 1986. Synthesis, bacterial expression, and protein peroxidase activity on polyvinylidene difluo-
mutagenesis of the gene coding for mammalian cyto- ride membrane. Anal. Biochem. 184:96-99.
chrome b5. Proc. Natl. Acad. Sci. USA 83:9443-9447. 22.Elkins, C., C.J. Chen, and C.E. Thomas. 1995. Char-
6.Black, S.M. and P.R. Ortiz de Montellano. 1995. acterization of the hgbA locus encoding a hemoglobin
Characterization of rat neuronal nitric oxide synthase receptor from Haemophilus ducreyi. Infect. Immun.
expressed in Saccharomyces cerevisiae. DNA Cell Biol. 63:2194-2200.
14:789-794. 23.Estabrook, R.W., J. Peterson, J. Barn, and A. Hilde-
7.Boyle, R.C., A.L. Tappel, A.A. Tappel, H. Chen, and brant. 1972. The spectrophotometric measurement of
H.J. Andersen. 1994. Quantitation of heme proteins turbid suspensions of cytochromes associated with
from mixtures. J. Agric. Food Chem. 42:100-104. drug metabolism, p. 303-350. In C.F. Chignell (Ed.),
8.Braun, V., K. Hantke, and W. Koster. 1998. Bacterial Methods in Pharmacology. Vol. 2. Appleton-Century-
iron transport: mechanisms, genetics, and regulation. Crofts, New York.
Met. Ions Biol. Syst. 35:67-145. 24.Falk, J.E. 1963. Part A. Pyrrole pigments: chemistry
9.Champiat, D., A. Roux, O. Lhomme, and G. Nosen- and biochemistry of porphyrins and metallopor-
zo. 1994. Biochemiluminescence and biomedical phyrins, p. 3-33. In M. Florkin and E.H. Stotz (Eds.),
applications. Cell Biol. Toxicol. 10:345-351. Comprehensive Biochemistry, Vol. 9. Elsevier, Amster-
10.Chance, B. 1954. Spectrophotometry of intracellular dam.
respiratory pigments. Science 120:767. 25.Falk, J.E. 1964. Porphyrins and Metalloporphyrins.
11.Charles, I.G., A. Chubb, R. Gill, J. Clare, P.N. Lowe, Elsevier, New York.
L.S. Holmes, M. Page, J.G. Keeling, S. Moncada, and 26.Fishel, L.A., J.E. Villafranca, J.M. Mauro, and J.
V. Riveros-Moreno. 1993. Cloning and expression of a Kraut. 1987. Yeast cytochrome c peroxidase: mutagen-
rat neuronal nitric oxide synthase coding sequence in a esis and expression in Escherichia coli show tryptophan-
baculovirus/insect cell system. Biochem. Biophys. Res. 51 is not the radical site in compound I. Biochemistry
Commun. 196:1481-1489. 26:351-360.
12.Charles, I.G., C.A. Scorer, M.A. Moro, C. Fernandez, 27.Fronticelli, C., J.K. O’Donnell, and W.S. Brinigar.
A. Chubb, J. Dawson, N. Foxwell, R.G. Knowles, and 1991. Recombinant human hemoglobin: expression
S.A. Baylis. 1996. Expression of human nitric oxide and refolding of beta-globin from Escherichia coli. J.
synthase isozymes. Methods Enzymol. 268:449-460. Prot. Chem. 10:495-501.
13.Chen, H., A.L. Tappel, and R.C. Boyle. 1993. Oxida- 28.Gegner, J.A. and F.W. Dahlquist. 1991. Signal trans-
tion of heme proteins as a measure of oxidative damage duction in bacteria: CheW forms a reversible complex
to liver tissue slices. Free Radic. Biol. Med. 14:509- with the protein kinase CheA. Proc. Natl. Acad Sci.
517. USA 88:750-754.

180
Analysis of Heme and Hemoproteins

29.George, H.J., D.E. Van Dyk, R.A. Straney, J.M. Trza- 43.Hernandez, G., A. Wilks, R. Paolesse, K.M. Smith,
skos, and R.A. Copeland. 1996. Expression purifica- P.R. Ortiz de Montellano, and G.N. La Mar. 1994.
tion and characterization of recombinant human Proton NMR investigation of substrate-bound heme
inducible prostaglandin G/H synthase from bac- oxygenase: evidence for electronic and steric contribu-
ulovirus-infected insect cells. Protein Expr. Purif. 7:19- tions to stereoselective heme cleavage. Biochemistry
26. 33:6631-6641.
30.Gerber, N.C., C.R. Nishida, and P.R. Ortiz de Mon- 44.Hoffman, S.J., D.L. Looker, J.M. Roehrich, P.E.
tellano. 1997. Characterization of human liver indu- Cozart, S.L. Durfee, J.L. Tedesco, and G.L. Stetler.
cible nitric oxide synthase expressed in Escherichia coli. 1990. Expression of fully functional tetrameric human
Arch. Biochem. Biophys. 343:249-253. hemoglobin in Escherichia coli. Proc. Natl. Acad. Sci.
31.Gerber, N.C. and P.R. Ortiz de Montellano. 1995. USA 87:8521-8525.
Neuronal nitric oxide synthase. Expression in Escheri- 45.Hofrichter, J., J.H. Sommer, E.R. Herry, and W.A.
chia coli, irreversible inhibition by phenyldiazene, and Eaton. 1983. Nanosecond absorption spectroscopy of
active site topology. J. Biol. Chem. 270:17791-17796. hemoglobin. Proc. Natl. Acad. Sci. USA 80:2235-
32.Gilles-Gonzalez, M.A., G. Gonzalez, M.F. Perutz, L. 2239.
Kiger, M.C. Marden, and C. Poyart. 1994. Heme- 46.Holmans, P.L., M.S. Shet, C.A. Martin-Wixtrom,
based sensors, exemplified by the kinase FixL, are a C.W. Fisher, and R.W. Estabrook. 1994. The high-
new class of heme protein with distinctive ligand bind- level expression in Escherichia coli of the membrane-
ing and autoxidation. Biochemistry 33:8067-8073. bound form of human and rat cytochrome b5 and
33.Gonzalez, F.J. and K.R. Korzekwa. 1995. Cyto- studies on their mechanism of function. Arch. Bio-
chromes P450 expression systems. Annu. Rev. Phar- chem. Biophys. 312:554-565.
macol. Toxicol. 35:369-390. 47.Hui, H.L., J.S. Kavanaugh, M.L. Doyle, A. Wierz-
34.Guengerich, F.P. 1983. Oxidation-reduction properties ba, P.H. Rogers, A. Arnone, J.M. Holt, G.K. Ackers,
of rat liver cytochromes P-450 and NADPH-cyto- and R.W. Noble. 1999. Structural and functional
chrome p-450 reductase related to catalysis in reconsti- properties of human hemoglobins reassembled after
tuted systems. Biochemistry 22:2811-2820. synthesis in Escherichia coli. Biochemistry 38:1040-
35.Guengerich, F.P., N.A. Hosea, A. Parikh, L.C. Bell- 1049.
Parikh, W.W. Johnson, E.M. Gillam, and T. Shimada. 48.Ishikawa, K., M. Sato, and T. Yoshida. 1991. Expres-
1998. Twenty years of biochemistry of human P450s: sion of rat heme oxygenase in Escherichia coli as a cat-
purification, expression, mechanism, and relevance to alytically active, full-length form that binds to bacteri-
drugs. Drug Metab. Dispos. 26:1175-1178. al membranes. Eur. J. Biochem. 202:161-165.
36.Guengerich, F.P., M.V. Martin, Z. Guo, and Y.J. 49.Jennings, P.A., M.J. Stone, and P.E. Wright. 1995.
Chun. 1996. Purification of functional recombinant Overexpression of myoglobin and assignment of its
P450s from bacteria. Methods Enzymol. 272:35-44. amide, C alpha and C beta resonances. J. Biomol.
37.Halaka, F.G., G.T. Babcock, and J.L. Dye. 1985. The NMR. 6:271-276.
use of principal component analysis to resolve the spec- 50.Jones, S., M. Tasab, J.E. Ogden, D.J. Ballance, and
tra and kinetics of cytochrome c oxidase reduction by M.J. Powell. 1996. Expression of rat neuronal nitric
5, 10-dihydro-5-methyl phenazine. Biophys. J. oxide synthase in Saccharomyces cerevisiae. J. Biotech-
48:209-219. nol. 48:37-41.
38.Halpert, J.R., T.L. Domanski, O. Adali, C.P. Biagini, 51.Jung, C., O. Ristau, and H. Rein. 1991. The high-
J. Cosme, E.A. Dierks, E.F. Johnson, J.P. Jones et al. spin/low-spin equilibrium in cytochrome P-450—a
1998. Structure-function of cytochromes P450 and new method for determination of the high-spin con-
flavin-containing monooxygenases: implications for tent. Biochim. Biophys. Acta 1076:130-136.
drug metabolism. Drug Metab. Dispos. 26:1223-1231. 52.Kakuno, T., R.G. Bartsch, K. Nishikawa, and T.
39.Hartmann, C. and P.R. Ortiz de Montellano. 1992. Horio. 1981. Redox componenets associated with
Baculovirus expression and characterization of catalyt- chromatophores from Rhodospirillum rubrum. J.
ically active horseradish peroxidase. Arch. Biochem. Biochem. 70:79-94.
Biophys. 297:61-72. 53.Kincaid, J., P. Stein, and T.G. Spiro. 1979. Absence of
40.Hawkins, B.K., A. Wilks, L.S. Powers, P.R. Ortiz de heme-localized strain in T state hemoglobin: insensi-
Montellano, and J.H. Dawson. 1996. Ligation of the tivity of heme-imidazole resonance Raman frequencies
iron in the heme-heme oxygenase complex: X-ray to quaternary structure. Proc. Natl. Acad. Sci. USA
absorption, electronic absorption and magnetic circu- 76:549-552.
lar dichroism studies. Biochim. Biophys. Acta 54.King, M.T. and G. Drews. 1975. The respiratory elec-
1295:165-173. tron transport system of heterotrophically-grown Rho-
41.Hernan, R.A., H.L. Hui, M.E. Andracki, R.W. Noble, dopseudomonas palustris. Arch. Microbiol. 102:219-
S.G. Sligar, J.A. Walder, and R.Y. Walder. 1992. 231.
Human hemoglobin expression in Escherichia coli: 55.Kitagawa, T., K. Nagai, and M. Tsubaki. 1979.
importance of optimal codon usage. Biochemistry Assignment of the Fe-Nepsilon (His F8) stretching
31:8619-8628. band in the resonance Raman spectra of deoxy myo-
42.Hernan, R.A. and S.G. Sligar. 1995. Tetrameric hemo- globin. FEBS Lett. 104:376-378.
globin expressed in Escherichia coli. Evidence of hetero- 56.Knaff, D.B. and B.B. Buchanan. 1975. Cytochrome b
geneous subunit assembly. J. Biol. Chem. 270:26257- and photosynthetic sulfur bacteria. Biochim. Biophys.
26264. Acta. 376:549-560.

181
A. Wilks

57.Koenigs, L.L. and W.F. Trager. 1998. Mechanism- 73.Nakane, M., J.S. Pollock, V. Klinghofer, F. Basha, P.A.
based inactivation of cytochrome P450 2B1 by 8- Marsden, A. Hokari, T. Ogura, H. Esumi, and G.W.
methoxypsoralen and several other furanocoumarins. Carter. 1995. Functional expression of three isoforms
Biochemistry 37:13184-13193. of human nitric oxide synthase in baculovirus-infected
58.Koenigs, L.L. and W.F. Trager. 1998. Mechanism- insect cells. Biochem. Biophys. Res. Commun.
based inactivation of P450 2A6 by furanocoumarins. 206:511-517.
Biochemistry 37:10047-10061. 74.North, J.A., R. Dietrich, and A.L. Tappel. 1996. Mul-
59.Laemmli, U.K. 1970. Cleavage of structural proteins ticomponent analysis of heme protein spectra in bio-
during the assembly of the head of bacteriophage T4. logical materials. Anal. Biochem. 233:115-123.
Nature 227:680-685. 75.Ohnishi, T., S. Miura, and Y. Ichikawa. 1993. Pho-
60.Lee, B.C. 1992. Isolation of an outer membrane he- toaffinity labeling of cytochrome P-45011 beta with
min-binding protein of Haemophilus influenzae type b. methyltrienolone as a probe for the substrate binding
Infect. Immun. 60:810-816. region. Biochim. Biophys. Acta 1161:257-264.
61.Lindenmayer, A. and R.W. Estabrook. 1958. Low 76.Ortiz de Montellano, P.R. 1995. Arylhydrazines as
temperature spectral studies on the biosynthesis of probes of hemoprotein structure and function. Bio-
cytochromes in bakers yeast. Arch. Biochem. Biophy. chimie 77:581-593.
78:66-82. 77.Ortiz de Montellano, P.R., C. Nishida, I. Rodriguez-
62.List, B.M., P. Klatt, E.R. Werner, K. Schmidt, and B. Crespo, and N. Gerber. 1998. Nitric oxide synthase
Mayer. 1996. Overexpression of neuronal nitric oxide structure and electron transfer. Drug Metab. Dispos.
synthase in insect cells reveals requirement of haem for 26:1185-1189.
tetrahydrobiopterin binding. Biochem. J. 315:57-63. 78.Ortiz de Montellano, P.R. and A. Wilks. 2000. Struc-
63.Maines, M.D. 1997. The heme oxygenase system: a ture and mechanism of heme oxygenase. Adv. Inorg.
regulator of second messenger gases. Annu. Rev. Phar- Chem. 51:359-402.
macol. Toxicol. 35:517-554. 79.Osawa, Y., K. Nakatsuka, M.S. Williams, J.T. Kindt,
64.Mayer, B., P. Klatt, B.M. List, C. Harteneck, and K. and M. Nakatsuka. 1996. Reactions of reactive me-
Schmidt. 1996. Large-scale purification of rat brain tabolites with hemoproteins—toxicological implica-
nitric oxide synthase from baculovirus overexpression tions: covalent alteration of hemoproteins. Adv. Exp.
system. Methods Enzymol. 268:420-427. Med. Biol. 387:37-45.
65.McMillan, K., D.S. Bredt, D.J. Hirsch, S.H. Snyder, 80.Otto, B.R., S.J. van Dooren, J.H. Nuijens, J. Luirink,
J.E. Clark, and B.S. Masters. 1992. Cloned, expressed and B. Oudega. 1998. Characterization of a hemoglo-
rat cerebellar nitric oxide synthase contains stoichio- bin protease secreted by the pathogenic Escherichia coli
metric amounts of heme, which binds carbon monox- strain EB1. J. Exp. Med. 188:1091-1103.
ide. Proc. Natl. Acad. Sci. USA 89:11141-11145. 81.Pollock, W.B., F.I. Rosell, M.B. Twitchett, M.E.
66.McMillan, K., J.C. Salerno, and B.S. Masters. 1996. Dumont, and A.G. Mauk. 1998. Bacterial expression
Nitric oxide synthases: analogies to cytochrome P450 of a mitochondrial cytochrome c. Trimethylation of
monooxygenases and characterization of recombinant lys72 in yeast iso-1-cytochrome c and the alkaline con-
rat neuronal nitric oxide synthase hemoprotein. Meth- formational transition. Biochemistry 37:6124-6131.
ods Enzymol. 268:460-472. 82.Poole, R.K., R.I. Scott, and B. Chance. 1980. Low-
67.Miller, J.P. and R.E. White. 1994. Photoaffinity label- temperature spectral and kinetic properties of
ing of cytochrome P450 2B4: capture of active site cytochromes in Escherichia coli K-12 grown at lowered
heme ligands by a photocarbene. Biochemistry oxygen tension. Biochim. Biophys. Acta 591:471-482.
33:807-817. 83.Poulos, T.L. 1991. Modeling of mammalian P450s on
68.Miyake, Y. and N. Takayama. 1976. Spectral interme- basis of P450cam X-ray structure. Methods Enzymol.
diates during the reduction of hepatic microsomal 206:11-30.
cytochrome P-450. J. Biochem. (Tokyo) 79:1077- 84.Poulos, T.L. 1993. Peroxidases. Curr. Opin. Biotech-
1087. nol. 4:484-489.
69.Modi, S., M.J. Paine, M.J. Sutcliffe, L.Y. Lian, W.U. 85.Ramos, L.S., K.R. Beebe, W.P. Karey, M.E. Sanches,
Primrose, C.R. Wolf, and G.C. Roberts. 1996. A B.C. Erickson, B.E. Wilson, B.E. Wangen, and B.R.
model for human cytochrome P450 2D6 based on Kowalski. 1986. Chemometrics. Anal. Chem. 58:
homology modeling and NMR studies of substrate 294R-515R.
binding. Biochemistry 35:4540-4550. 86.Renaud, J.P., D.R. Davydov, K.P. Heirwegh, D. Man-
70.Mok, T.C., P.A. Rickard, and F.J. Moss. 1969. The suy, and G.H. Hui Bon Hoa. 1996. Thermodynamic
carbon monoxide-reactive haemoproteins of yeast. studies of substrate binding and spin transitions in
Biochim. Biophys. Acta 172:438-449. human cytochrome P-450 3A4 expressed in yeast
71.Muchmore, D.C., L.P. McIntosh, C.B. Russell, D.E. microsomes. Biochem. J. 319:675-681.
Anderson, and F.W. Dahlquist. 1989. Expression and 87.Rieske, J.S. 1967. The quantitative determination of
nitrogen-15 labeling of proteins for proton and nitro- mitochondrial hemoproteins. Methods Enzymol.
gen-15 nuclear magnetic resonance. Methods Enzy- 10:488-493.
mol. 177:44-73. 88.Ristau, O., H. Rein, S. Greschner, G.R. Janig, and K.
72.Nagai, K. and H.C. Thogersen. 1984. Generation of Ruckpaul. 1979. Quantitative analysis of the spin
beta-globin by sequence-specific proteolysis of a hybrid equilibrium of cytochrome P-450 LM2 fraction from
protein produced in Escherichia coli. Nature 309:810- rabbit liver microsomes. Acta Biol. Med. Ger. 38:177-
812. 185.

182
Analysis of Heme and Hemoproteins

89.Rivera, M., C. Barillas-Mury, K.A. Christensen, J.W. 103.Shu, F., V. Ramakrishnan, and B.P. Schoenborn.
Little, M.A. Wells, and F.A. Walker. 1992. Gene syn- 1996. High-level expression and deuteration of sperm
thesis, bacterial expression, and 1H NMR spectroscop- whale myoglobin. A study of its solvent structure by
ic studies of the rat outer mitochondrial membrane X-ray and neutron diffraction methods. Basic Life
cytochrome b5. Biochemistry 31:12233-12240. Sci. 64:309-323.
90.Riveros-Moreno, V., B. Heffernan, B. Torres, A. 104.Smith, A.T., N. Santama, S. Dacey, M. Edwards,
Chubb, I. Charles, and S. Moncada. 1995. Purifica- R.C. Bray, R.N. Thorneley, and J.F. Burke. 1990.
tion to homogeneity and characterisation of rat brain Expression of a synthetic gene for horseradish peroxi-
recombinant nitric oxide synthase. Eur. J. Biochem. dase C in Escherichia coli and folding and activation of
230:52-57. the recombinant enzyme with Ca2+ and heme. J. Biol.
91.Rodgers, K.R. 1999. Heme-based sensors in biological Chem. 265:13335-13343.
systems [In Process Citation]. Curr. Opin. Chem. Biol. 105.Smulevich, G., J.M. Mauro, L.A. Fishel, A.M. Eng-
3:158-167. lish, J. Kraut, and T.G. Spiro. 1988. Heme pocket
92.Rodriguez-Crespo, I., N.C. Gerber, and P.R. Ortiz de interactions in cytochrome c peroxidase studied by
Montellano. 1996. Endothelial nitric-oxide synthase. site-directed mutagenesis and resonance Raman spec-
Expression in Escherichia coli, spectroscopic characteri- troscopy. Biochemistry 27:5477-5485.
zation, and role of tetrahydrobiopterin in dimer for- 106.Smulevich, G., M.A. Miller, J. Kraut, and T.G.
mation. J. Biol. Chem. 271:11462-11467. Spiro. 1991. Conformational change and histidine
93.Roman, L.J., E.A. Sheta, P. Martasek, S.S. Gross, Q. control of heme chemistry in cytochrome c peroxidase:
Liu, and B.S. Masters. 1995. High-level expression of resonance Raman evidence from Leu-52 and Gly-181
functional rat neuronal nitric oxide synthase in Escherichia mutants of cytochrome c peroxidase. Biochemistry
coli. Proc. Natl. Acad. Sci. USA 92:8428-8432. 30:9546-9558.
94.Sari, M.A., S. Booker, M. Jaouen, S. Vadon, J.I. 107.Spiro, T.G. 1978. Resonance Raman spectra of
Boucher, D. Pompon, and D. Mansuy. 1996. Expres- hemoproteins. Methods Enzymol. 54:233-249.
sion in yeast and purification of functional macrophage 108.Spiro, T.G. 1985. Resonance Raman spectroscopy as
nitric oxide synthase. Evidence for cysteine-194 as iron a probe of heme protein structure and dynamics. Adv.
proximal ligand. Biochemistry 35:7204-7213. Prot. Chem. 37:111-159.
95.Sarma, S., R.J. DiGate, D.L. Banville, and R.D. 109.Spiro, T.G. and R.S. Czernuszewicz. 1995. Reso-
Guiles. 1996. 1H, 13C and 15N NMR assignments nance Raman spectroscopy of metalloproteins. Meth-
and secondary structure of the paramagnetic form of ods Enzymol. 246:416-460.
rat cytochrome b5. J. Biomol. NMR 8:171-183. 110.Springer, B.A. and S.G. Sligar. 1987. High-level
96.Schmidt, M.L. and J.Q. Trojanowski. 1986. Enzy- expression of sperm whale myoglobin in Escherichia
matic detection of native and derivatized horseradish coli. Proc. Natl. Acad. Sci. USA 84:8961-8965.
peroxidase in sodium dodecyl sulfate polyacrylamide 111.Sun, J., A. Wilks, P.R. Ortiz de Montellano, and
gels. Anal. Biochem. 155:371-375. T.M. Loehr. 1993. Resonance Raman and EPR spec-
97.Schuller, D.J., A. Wilks, P.R. Ortiz de Montellano, troscopic studies on heme-heme oxygenase complexes.
and T.L. Poulos. 1999. Crystal structure of human Biochemistry 32:14151-14157.
heme oxygenase-1. Nat. Struct. Biol. 6:860-867. 112.Tamburini, P.P., G.G. Gibson, W.L. Backes, S.G.
98.Seo, H.G., J. Fujii, H. Soejima, N. Niikawa, and N. Sligar, and J.B. Schenkman. 1984. Reduction kinetics
Taniguchi. 1995. Heme requirement for production of of purified rat liver cytochrome P-450. Evidence for a
active endothelial nitric oxide synthase in baculovirus- sequential reaction mechanism dependent on the
infected insect cells. Biochem. Biophys. Res. Com- hemoprotein spin state. Biochemistry 23:4526-4533.
mun. 208:10-18. 113.Taylor, K.L., D.J. Uhlinger, and J.M. Kinkade, Jr.
99.Shelver, D., M.V. Thorsteinsson, R.L. Kerby, S.Y. 1992. Expression of recombinant myeloperoxidase
Chung, G.P. Roberts, M.F. Reynolds, R.B. Parks, and using a baculovirus expression system. Biochem. Bio-
J.N. Burstyn. 1999. Identification of two important phys. Res. Commun. 187:1572-1578.
heme site residues (cysteine 75 and histidine 77) in 114.Teale, F.W.J. 1959. Cleavage of the haem-protein link
CooA, the CO-sensing transcription factor of Rho- by acid methyl ethyl ketone. Biochim. Biophy. Acta.
dospirillum rubrum. Biochemistry 38:2669-2678. 35:543.
100.Shen, T.J., N.T. Ho, V. Simplaceanu, M. Zou, B.N. 115.Teraoka, J. and T. Kitagawa. 1981. Structural impli-
Green, M.F. Tam, and C. Ho. 1993. Production of cation of the heme-linked ionization of horseradish
unmodified human adult hemoglobin in Escherichia peroxidase probed by the Fe-histidine stretching
coli. Proc. Natl. Acad. Sci. USA 90:8108-8112. Raman line. J. Biol. Chem. 256:3969-3977.
101.Shioi, Y., K. Takamiya, and M. Nishimura. 1972. 116.Thomas, P.E., D. Ryan, and W. Levin. 1976. An
Studies on energy and electron transfer systems in improved staining procedure for the detection of the
green photosynthetic bacterium Chloropseudomonas peroxidase activity of cytochrome P-450 on sodium
ethylica strain 2K. I. Isolation and characterization of dodecyl sulfate polyacrylamide gels. Anal. Biochem.
cytochromes from Chloropseudomonas ethylica strain 75:168-176.
2K. J. Biochem. (Tokyo) 71:285-294. 117.Tschirret-Guth, R.A., K.F. Medzihradszky, and P.R.
102.Shrager, R.I. and R.W. Hendler. 1982. Titration of Ortiz de Montellano. 1998. Specific azidophenyl-
individual components in a mixture with resolution diazene hemoprotein active site probes. Cross-linking
of difference spectra, pK’s and redox transitions. Anal. of the heme to His-64 in myoglobin. J. Am. Chem.
Chem. 54:1147-1152. Soc. 120:7404-7410.

183
A. Wilks

118.Tschirret-Guth, R.A., K.F. Medzihradszky, and 128.Wilks, A. and P.R. Ortiz de Montellano. 1993. Rat
P.R. Ortiz de Montellano. 1999. Trifluoromethyl- liver heme oxygenase. High level expression of a trun-
diazirinylphenyldiazenes: new hemeprotein active-site cated soluble form and nature of the meso-hydroxy-
probes. J. Amer. Chem. Soc. 121:4731-4737. lating species. J. Biol. Chem. 268:22357-22362.
119.Tschirret-Guth, R.A. and P.R. Ortiz de Motellano. 129.Wilks, A. and M.P. Schmitt. 1998. Expression and
1998. Synthesis of photoaffinity probes for heme- characterization of a heme oxygenase (Hmu O) from
containing proteins. J. Org. Chem. 63:9711-9715. Corynebacterium diphtheriae. Iron acquisition requires
120.Tsutsui, K. 1986. Affinity chromatography of heme- oxidative cleavage of the heme macrocycle. J. Biol.
binding proteins: synthesis of hemin-agarose. Meth- Chem. 273:837-841.
ods Enzymol. 123:331-338. 130.Wilks, A., J. Torpey, and P.R. Ortiz de Montellano.
121.Tsutsui, K. and G.C. Mueller. 1982. A protein with 1994. Heme oxygenase (HO-1). Evidence for elec-
multiple heme-binding sites from rabbit serum. J. trophilic oxygen addition to the porphyrin ring in the
Biol. Chem. 257:3925-3931. formation of alpha-meso-hydroxyheme. J. Biol.
122.Varadarajan, R., A. Szabo, and S.G. Boxer. 1985. Chem. 269:29553-29556.
Cloning, expression in Escherichia coli, and reconsti- 131.Wu, C., J. Zhang, H. Abu-Soud, D.K. Ghosh, and
tution of human myoglobin. Proc. Natl. Acad. Sci. D.J. Stuehr. 1996. High-level expression of mouse
USA 82:5681-5684. inducible nitric oxide synthase in Escherichia coli
123.Vickery, L.E. 1978. Spin states of heme proteins by requires coexpression with calmodulin. Biochem.
magnetic circular dichroism. Methods Enzymol. 54: Biophys. Res. Commun. 222:439-444.
284-302. 132.Yoshida, T., K. Ishikawa, and M. Sato. 1991.
124.von Wachenfeldt, C., T.H. Richardson, J. Cosme, Degradation of heme by a soluble peptide of heme
and E.F. Johnson. 1997. Microsomal P450 2C3 is oxygenase obtained from rat liver microsomes by mild
expressed as a soluble dimer in Escherichia coli follow- trypsinization. Eur. J. Biochem. 199:729-733.
ing modification of its N-terminus. Arch. Biochem. 133.Yoshida, T. and G. Kikuchi. 1978. Purification and
Biophys. 339:107-114. properties of heme oxygenase from pig spleen micro-
125.Vuletich, J.L. and Y. Osawa. 1998. Chemilumines- somes. J. Biol. Chem. 253:4224-4229.
cence assay for oxidatively modified myoglobin. Anal. 134.Yun, C.H., G.J. Hammons, G. Jones, M.V. Mar-
Biochem. 265:375-380. tin, N.E. Hopkins, W.L. Alworth, and F.P.
126.Wilks, A., S.M. Black, W.L. Miller, and P.R. Ortiz Guengerich. 1992. Modification of cytochrome
de Montellano. 1995. Expression and characterization P450 1A2 enzymes by the mechanism-based inacti-
of truncated human heme oxygenase (hHO- 1) and a vator 2-ethynylnaphthalene and the photoaffinity
fusion protein of hHO-1 with human cytochrome label 4-azidobiphenyl. Biochemistry 31:10556-
P450 reductase. Biochemistry 34:4421-4427. 10563.
127.Wilks, A. and P.R. Ortiz de Montellano. 1992. Intra- 135.Zabel, U., M. Weeger, M. La, and H.H. Schmidt.
molecular translocation of the protein radical formed 1998. Human soluble guanylate cyclase: functional
in the reaction of recombinant sperm whale myoglobin expression and revised isoenzyme family. Biochem. J.
with H2O2. J. Biol. Chem. 267:8827-8833. 335:51-57.

184
Hemoproteins Purification and
8 Characterization by Using Aqueous
Two-Phase Systems

Daniel Forciniti
Chemical Engineering Department, University of Missouri-Rolla,
Rolla, MO, USA

1. INTRODUCTION all the publications in this topic since 1956


can be found in (http://bama.ua.edu/
There are many techniques that can be ~rdrogers/aq2phase/). Detail recipes for
used to purify and characterize proteins, most of the protocols used in different
but one especially caught my attention applications of aqueous 2-phase systems
about 10 years ago. Is it possible to purify a can be found in Reference 22.
protein using just water? A smart trick that Aqueous 2-phase extraction is a tech-
does the job is aqueous 2-phase extraction. nique that is ideally suitable for the removal
In 1958, Albertsson (2) found that when 2 of cell debris and simultaneous concentra-
aqueous solutions of hydrophilic polymers tion of the target protein; furthermore, it
such as polyethyleneglycol (PEG) and dex- can be used as a very selective procedure by
tran (Dx) are mixed above critical concen- adding affinity ligands to one of the phase-
trations, a liquid–liquid phase separation forming polymers. Table 1 shows some
occurs. Moreover, when he added these 2 examples of the use of aqueous 2-phase sys-
polymers to a disrupted cell broth, he tems for the purification of various proteins.
found that proteins or enzymes and cell Worth mentioning is the isolation of Vitre-
debris tend to partition unequally between oscilla hemoglobin from Escherichia coli
the 2 phases or between the phases and the lysate by 3 extraction steps with an overall
interface, thus allowing for the extraction yield of 47% and a purity higher than 95%
of a particular protein. The high water con- (21). For either industrial or recombinant
tent of these systems, which gives the tech- proteins, the method offers notable advan-
nique its name, provides a gentle environ- tages (27,54). For industrial enzymes, the
ment for biologically active proteins, cells, method’s utility stems from the ease with
and cell organelles. Many proteins have which it can be adapted to continuous pro-
been partitioned in aqueous 2-phase and 3- duction and scaled up to meet industrial
phase systems, and the subject has been needs. For example, Genencor Intl. (Palo
described in several reviews (2,22,50,52, Alto, CA, USA) (27) currently uses a PEG/
56). A comprehensive database containing salt system coupled with ion exchange chro-

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

185
Table 1. Some Examples of the Use of Aqueous Two-Phase Systems for the Protein Purification
186

D. Forciniti
Concentration
Substance Source Yield Factor/Purity Observations Reference

Thaumatin Cell homogenate of 92% 65% Two extraction steps using PEG/phosphate 5
E. coli systems.
Lactate dehydrogenase Porcine muscle extract 54% 11.8-fold PEG/Dx followed by affinity precipitation. 19
Ecdysone (hormone) Spinach 88% ND UCON 50-hb-5111/Reppal PES-200a. 33
3-phosphoglycerate kinase Yeast homogenate 57% 5.2-fold UCON 50-HB-5100/Dx T-500. 47
NAD-kinase Lactobacillus 100% 3-fold PEG/salt. 23
cellobiosus
Leucine dehydrogenase Bacillus sphaericus 98% 9.5-fold PEG/crude Dx (unfractionated Dx of high, 23
>1 000 000 molecular weight).
Beta interferon Human fibroblasts 350-fold 31
Beta interferon Recombinant E. coli 76% ND PEG/salt/PEG phosphate ester followed by 18
back extraction.
Fractionation of proteins Human serum ND ND PEG/Dx and PEG/Aquaphase PPTb plus addition 7
of various affinity ligands. Separation done by
counter-current distribution.
Lactate dehydrogenases Recombinant B. 80% 8-fold Cu(II)poly-VI-VCL and Dx-70. 30
stearothermophilus
Chymosin Recombinant A. ND ND PEG-8000/NA2SO4 Industrial scale production 27
awamori (Genencor Intrl.).
Recombinant Protein Mammalian cell 90% 100-fold PEG/Pluronic F68/Ammonium sulfate. Industrial 4
culture scale production (MedImmune, Gaithersburg,
MD, USA)
Cutinase Recombinant ND 26-fold PEG-1500/sodium phosphate (plus Triton 9
Saccharomyces X-100, or Tween 20, or butyrate).
cerevisiae
Vitreoscilla hemoglobin E. coli lysate 47% 95% Sequential extraction: (i) PEG/Dx, 21
(ii) PEG/Na2SO4, (iii) PEG/MgSO4
aReppal PES-200 is a hydroxypropyl starch produced by CarbamyLab (Kristianstad, Sweden).
bAquaphase PPT is a modified starch product produced by Perstop (Sweden).
ND: not determined by the authors.
Hemoproteins Purification and Characterization

matography for the purification of chy- phase systems is presented in Table 2.


mosin. For recombinant proteins, its utility Recently, temperature-induced phase split-
is based on its attractiveness as a gentle first ting systems, made with copolymers of eth-
step or on its selectivity on a small scale by ylene oxide and propylene oxide, are gain-
using affinity ligands. For example, Mattias- ing popularity (20).
son and Galaev (30) purified lactate dehy- Aqueous 2-phase systems have a very low
drogenase (carrying a tag of 6 histidine interfacial tension (the reversible work to
residues) from recombinant Bacillus stearo- create unit area of surface at constant tem-
thermophilus with an 80% yield and con- perature, volume, and chemical potential)
centrated 8-fold using an aqueous 2-phase and densities very close to that of pure
system formed by Cu(II)poly-VI-VCL water. The interfacial tension, which ranges
(copolymers of N-vinylcaprolactam and 1- from 1 to 300 µN/m, of aqueous 2-phase
vinyl imidazole) and Dx-70. One applica- systems depends on polymer molecular
tion of the method is aqueous 2-phase affin- weight, polymer concentration, and tem-
ity partitioning chromatography, in which perature. We have found that the logarithm
the stationary (support) phase is coated with of the interfacial tension varies linearly with
the Dx-rich bottom phase of an aqueous 2- the logarithm of the difference in polymer
phase system, and the top phase is used as concentration between the top and bottom
the mobile phase. Aqueous 2-phase systems phases. If systems of the same tie-line
are routinely used, among other places, in length (TLL; the length of the line joining
the Institute of Enzyme-Technology (Julich, the composition of both phases and the
Germany), at the Departments of Biotech- total system composition in a concentra-
nology, Biochemistry, and Center for tion–concentration plane) are compared,
Chemistry and Chemical Engineering Lund increasing the molecular weight of one of
University, and at the Royal Free Hospital the two polymers increases the interfacial
School of Medicine, University of London. tension. A very complex temperature
In addition to its application as a separa- dependence of the interfacial tension was
tion process, aqueous 2-phase partitioning also found. An extensive collection of den-
often yields information on important sities and interfacial tensions is available
physical properties, replacing other more (15). The reader is referred to Table 3 for
cumbersome analytical techniques. For representative values. The very low density
example, aqueous 2-phase partitioning can difference between top and bottom phases
be used to determine the isoelectric point makes phase separation under gravity very
of proteins, to measure the hydrophobicity slow. So, centrifugation is often used to
and charge of a protein, to calculate disso- achieve phase separation.
ciation constants between enzymes and The preparation of aqueous 2-phase sys-
substrates, to fractionate cell populations, tems is straightforward. Stock solutions of
or to characterize cell surfaces. One of all the phase components are mixed in
these applications is cross-partitioning (see appropriate amounts (by weight), and then
section 3.5.1), which has been used to the emulsion is allowed to separate into 2
determine the isoelectric point of hemoglo- phases under gravity or in a centrifuge.
bins from different species (51). After complete phase separation is achieved,
The most common systems are created the phases are sampled. In the laboratory-
using either 2 incompatible polymers or scale experiments, aqueous 2-phase systems
one polymer and a salt. PEG/Dx and PEG/ with a final mass of 5 to 10 g are usually
phosphate are by far the most popular prepared. In some systems, one of the phase
ones. An extended list including some new polymers derivatized with charged groups
187
D. Forciniti

Table 2. Commonly Used Phase-Forming Species

Species 1 Species 2 References

PEG Dx 2,16
PEG Ficoll 2
Dx Ficoll 2
Methyl Cellulose Dx 2
PEG Benzoil Dextran 28
P(EO-PO) Water (NaCl) 47
P(EO-PO) Reppal (Hydroxypropyl starch) 20
PEG Sodium citrate/citric acid 48
PEG NaH2PO4/K2HPO4 2,43
PEG Ammonium Sulfate 2
Sodium Dextran Sulfate PEG (NaCl) 2
PEG NaCl 44
PEG SCNNa/NaCl 10
PEG GuHCl/NaCl 46
DX PVP 56
Dx PVA 56
PEG Polyvinyl methyl ether 56
Methyl Cellulose Hydroxypropyl Dextran 2
All systems contain a large amount of water (or buffer), up to 90% (wt/wt).

or affinity ligands is also included. The the top and bottom phases (VT/VB). The
derivatized species often constitute a minor working tie-line and the volume ratio
constituent (1%–5%) of the total system. between the top and the bottom phase can
The equilibrium data for aqueous 2- be chosen by using published equilibrium
phase systems are usually presented by plot- data (56). Since there are variations in the
ting the amount of one phase forming molecular weight of the polymers and,
species (A) versus the amount of the other sometimes, the experimental conditions
(B) in both top and bottom phases. The used to determine a binodal are not com-
locus of equilibrium compositions of top pletely specified, it may be appropriate to
and bottom phase in the A-B plane is called create specific a phase diagram if a particu-
the binodal (See Figure 1). Figure 1 shows lar system is going to be used routinely.
the phase diagram for a polyethylene The equilibrium curve or binodal is
propylene random copolymer P(EO- affected by:
PO)/salt system at 25°C. A few tie-lines are 1. Polymer molecular weight. The higher
also indicated. Simple geometrical argu- the molecular weight of the polymer
ments show that the length of the tie-line is used, the lower the polymer concentra-
equal to TLL = (∆A2 + ∆B2)1/2, where ∆A = tion (on a weight by weight basis)
[A]top - [A]bottom and ∆B = (B)bottom - needed to obtain phase separation. The
(B)top. The ratio between segments BO and binodal curve becomes more asymmet-
AO is equal to the volume ratio between rical as the molecular weight difference
188
Hemoproteins Purification and Characterization

Table 3. Densities and Interfacial Tensions of some Representative Systems


[PEG] [Dx] Interfacial Tension
System % wt/wt % wt/wt ρtop ρbottom µN/m

Dx-40/PEG-4000 7.64 10.29 1.023 1.092 59.10


Dx-40/PEG-20 000 5.05 8.50 1.017 1.068 87.60
Dx-500/PEG-4000 7.64 10.46 1.014 1.098 154.5
Dx-500/PEG-20 000 4.97 8.46 1.016 1.074 145.2

[PEG] and [Dx] are the overall concentration of PEG and Dx (before phase separation). The
densities (ρ in units of g/cm3) of each phase were measured with U-shaped oscillator
densitometer, and the interfacial tensions were measured with a spinning drop tensiometer
(Krüss Site 04, Hamburg, Germany). All systems are at 25°C.

between the polymers increases. Dx and Dx.


2. Polymer hydrophobicity. The polymers 3. Temperature. The effect of this variable
become less compatible when the hy- is small. In PEG/Dx mixtures, less
drophobicity of one of them is in- polymer is needed at low temperatures.
creased. This can be done by derivatiz- On the other hand, the phase transi-
ing the polymer. For example, two tion in PEG-salt systems is facilitated
phases are formed by hydroxypropyl by high temperatures.

Figure 1. Equilibrium curve for a P(EO-PO)/NaCl system at 25°C. Points to the left the equilibrium curve correspond to one
phase, and points to the right correspond to 2 liquid phases at equilibrium Point B representing the composition of the top phase,
point A the composition of the bottom phase, and points O and P two possible overall concentrations of P(EO-PO) and NaCl.
An overall concentration equal to O yields VT/VB = 1, and an overall concentration equal to P yields VT/VB = 1/3.

189
D. Forciniti

4. Polydispersity of the polymers. Com- ent pHs, we must consider any change in
mercial grade polymers have a wide the physical properties of the proteins due
distribution of molecular weights. to changes in the pH and how these
Therefore, phase separation occurs in a changes may affect the protein partition
small range of polymer concentrations coefficient. For example, whereas lysozyme
changing the binodal curve to a bin- does not change its conformation over the
odal zone. If the polymer molecular pH range 1.2 to 11.3 in dilute salt solu-
weight distribution is too wide, more tions at moderate temperatures, it polymer-
than two phases can be formed. izes reversibly at pH above 5 (45). This
5. Ionic Strength. The length of the tie- change in the molecular weight of the pro-
line increases when salts are added to tein will change the partition coefficient.
PEG-salt systems. In PEG/Dx/water The molecular weight and concentration
systems the ionic strength does not of the phase-forming species also affect the
have an appreciable influence on the partition coefficient strongly (13). For
position of the binodal. example, in a PEG/Dx system, low PEG
molecular weight favors the partitioning of
1.1. Distribution of Proteins in Aqueous proteins into the PEG-rich phase. The
Two-Phase Systems effect of polymer concentration on the par-
tition coefficient is also well known. If K p
The separation power of a given aque- is smaller than 1, an increase in either one
ous 2-phase system is given by the partition of the phase-forming species decreases Kp.
coefficient, Kp, of a protein. Kp is defined Similarly, if Kp is larger than 1, an increase
as the ratio of the protein concentration in in either of the phase-forming species
the top and bottom phases. This partition increases Kp. The amount of the phase-
coefficient depends on the difference in forming species also affects the volume ratio
chemical potential of the protein between between the phases. For example, in Figure
top and bottom phases, and therefore, it is 1, a system of total composition O has a
a function of the chemical nature of the top:bottom volume ratio of 1, whereas a
polymers, the protein, added electrolytes, system of total composition P has a
and temperature. It is usually manipulated top:bottom volume ratio of 1/3. Therefore,
by changing the pH (13,14) or the temper- the targeted protein can be not only puri-
ature, by adding salts or affinity ligands, or fied, but also concentrated in a single step.
by changing the molecular weight of the Generally, the partition coefficient in-
polymers or their concentration. creases with increasing temperature in Dx/
Manipulation of the pH is of primary PEG systems between 4° and 40°C (12).
importance in partitioning studies of pro- The partition coefficient of small and
teins. It is convenient to split the partition hydrophilic proteins is only slightly affect-
coefficient into 2 contributions, one that is ed by changes in temperature, whereas the
independent of pH, and the other that is partition coefficient of bigger and more
proportional to the net charge of the pro- hydrophobic proteins is strongly affected
tein. In so doing, electrostatic and nonelec- by temperature changes. High tempera-
trostatic contributions to the partition coef- tures (around 40°C) may be used to mini-
ficient can be conveniently (but artificially) mize protein association, whereas low tem-
separated. This approach predicts that the peratures may be desirable to maintain
logarithm of the partition coefficient is a protein stability.
linear function of the charge of the protein. Salts have unequal affinities for the top
In the analysis of partitioning data at differ- and bottom phases of aqueous 2-phase sys-
190
Hemoproteins Purification and Characterization

tems (25). This uneven partition of salts dent on protein’s charge, Z is the charge of
between the 2 phases affects the chemical the protein, and γ depends on the types
potential of the protein in each phase and and concentration of polymers, tempera-
thus its partition coefficient. The following ture, and types of electrolytes. Values of γ
mental exercise helps to understand this are plotted in Figure 2 for various salts (val-
phenomenon. Consider 2 phases at equi- ues are for Dx-500/PEG-8000 at 4°C).
librium in which a salt has been previously The net effect of a salt can be calculated by
partitioned. The different affinity of the γ = γcation - γanion. For example, the addi-
salt for the bottom and top phases creates 2 tion of tetrabutyl ammonium phosphate
distinct ionic atmospheres in both phases. yields a γ = -79, whereas the addition of
Picture a charged protein molecule at infi- potassium perchlorate yields a γ = 30. This
nite distance from the phases. Bring the figure can be used to select the appropriate
protein and try to insert it in each of the 2 electrolyte to move the desired protein to
phases. Since the protein is going to see dif- either the top or bottom phases. A pH
ferent ionic atmospheres in both phases, away from the isoelectric point of the pro-
the work needed to insert it in one or the tein must be selected, since the partition
other phase will be different. This differ- coefficient of proteins at their isoelectric
ence in the work of insertion of a charged point is quite insensitive to salt type and
protein in each of the 2 phases at equilibri- concentration (see Equation 1).
um is proportional to the electrochemical The partition coefficient of proteins can
potential difference of the protein between be manipulated by adding an affinity lig-
them. Consequently, the partition coeffi- and or by using liquid–liquid chromatog-
cient, which is a function of that potential raphy, counter–current extraction, or by a
difference, is strongly affected by the type combination of the first two. Affinity lig-
and concentration of salts and by the ands, such as PEG-palmitate (42) or PEG-
charge on the protein. red (24), have been routinely used to
In general, the partition coefficient of improve the selectivity of aqueous 2-phase
proteins away from their isoelectric point extraction. In affinity partitioning (26), it
depends on both the type and concentra- is customary to define ∆ln Kp = ln KL - ln
tion of cation and anion (25). For example, K0 (KL and K0 are the partition coeffi-
for positively charged proteins the partition cients of the protein with and without
coefficient in PEG/Dx systems is higher in added affinity ligand, respectively) to
potassium chloride than in potassium quantify the enhancement in partitioning
phosphate; the reverse is true for negatively as a result of the addition of the affinity lig-
charged proteins. The effect of the cation and. Liquid–liquid chromatography, with
on the partition coefficient of positively or without the addition of an affinity lig-
charged proteins is K approximately equal and, can be used to improve the selectivity
to Cs greater than Na greater than Li, of aqueous 2-phase systems. For example,
whereas the reverse order holds for nega- we have used liquid–liquid chromatogra-
tively charged proteins. It is possible to cor- phy with an immobilized Dx-rich phase to
relate the partition coefficient of a protein purify formate dehydrogenase (FDH) from
with its charge and with the type of salt. Candida boinidi with and without the
Johansson (25) found that, addition of PEG-red as an affinity ligand
(50). Another attractive alternative tech-
log Kp = log K0 + γZ [Eq. 1]
nique is the use of metal affinity aqueous
where, K0 depends on the particular pro- 2-phase extraction (1,40,55). In this tech-
tein and phase-system, but it is indepen- nique, a small portion (less than 1%) of the
191
D. Forciniti

total PEG is replaced by PEG-iminodi- forward. Two sets of 2-phase systems (usu-
acetic acid (PEG-IDA), which chelates di- ally Dx/PEG) covering a wide pH range
valent cations like copper or zinc. Histidine are prepared. One set contains NaCl,
groups on the protein surface recognize whereas the other set contains Na2SO4.
these metals, and the strength of the bind- The pH dependence of the partition coef-
ing is proportional to the number of histi- ficient of proteins in the presence of NaCl
dine groups on the protein surface. is usually different from that in the pres-
One of the most common analytical ence of Na2SO4. So, the partition coeffi-
uses of aqueous 2-phase systems is the cient versus pH curves cross each other at a
determination of isoelectric points by pH (cross-point) that usually agrees with
determining cross-partitioning points (3, the isoelectric point of the proteins. Dis-
41,51,53). The method uses the different crepancies between the isoelectric points
sensitivity of the partition coefficient on determined by cross-partitioning and by
the kind of salt used and determines the isoelectric focusing can be due to confor-
pH at which the partition coefficient is mational changes of the protein in the
independent of salt type. If the protein of phases system or by interactions between
interest does not interact with contaminat- the polymers and the proteins.
ing materials in the solution, its isoelectric The knowledge of the hydrophobicity of
point can be determined without prior a protein is useful for the design of reverse
protein purification (other than desalting). phase chromatography units, for the
The determination of isoelectric points understanding of protein–ligand interac-
of proteins by cross-partitioning is straight- tions, and for the understanding of protein

Figure 2. Effect of cation and anion on the partition coefficient of proteins. The values of γ obtained from this figure must be
used with Equation 1.

192
Hemoproteins Purification and Characterization

folding and refolding. The usual way of partition coefficient with the molecular size
measuring hydrophobicity of a solute is to of the solutes. For example, Petersen
measure the free energy of transfer of that (38,39) found that the partition coefficient
solute from water into an organic solvent. in a Dx/PEG system of cytochrome c oxi-
Because of the need to use an organic sol- dase was 20, whereas that of cytochrome c
vent, these methods are not well suited to was 0.275 and used these differences to
measure hydrophobicities of polar and flex- show that both oxidized and reduced
ible biological molecules. Aqueous 2-phase cytochrome c formed a 1:1 complex with
systems have been used to determine the the oxidase. In another study, Middaugh
relative hydrophobicity of various proteins and Lawson (32) determined the associa-
(56). The overall idea is to “calibrate” a tion constant of hemoglobin by using
series of aqueous 2-phase systems (Dx/ aqueous 2-phase systems. They found that
Ficoll and Dx/PEG have been used) by the partition coefficients of oxyhemoglobin
partitioning a series of small solutes (amino and methemoglobin produced a sigmoidal
acids) of increasing hydrophobicity. It has curve when they were plotted against pro-
been found that the partition coefficient of tein concentration. From these plots, they
amino acid i, correlates with the partition determined the dimer–tetramer association
coefficient of glycine by (56): constant for these proteins.
The potential uses of this technique are
1n Ki = 1n KGly + (HF)(RHi) [Eq. 2]
vividly pictured in the following examples.
where HF is known as the hydrophobicity PEG-coated liposomes are a current alter-
factor (a constant for a given 2-phase sys- native to increase the stability of liposomes.
tem) and RHi is the hydrophobicity of The behavior of the modified liposomes
solute i relative to glycine. For a protein, will depend on their surface properties.
the surface hydrophobicity of the i protein Because the partition behavior of a particle
relative to molecule j, HSFi is given by: is a signature of its surface properties, par-
titioning of PEG-coated liposomes in
HSFi = 1n Kij /HFj [Eq. 3]
aqueous 2-phase systems can be used to
The technique has been used to deter- anticipate their behavior in the blood
mine the hydrophobicity of several hemo- stream. For example, Moribe et al. (34)
proteins including hemoglobin, apomyo- used aqueous 2-phase systems to detect
globin, and cytochrome c (56). The change surface differences of PEG-coated lipo-
of surface properties caused by pH- somes. They partitioned the coated and
induced denaturation of some of these pro- uncoated liposomes (after exposing them
teins has been investigated by this tech- to plasma) in two kinds of systems, charge
nique (56). For example, it has been found sensitive (5% PEG-8000, 5% Dx T-500,
that the difference in hydrophobic index and 0.11 M NaPO4, pH 7.0) and charge
(expressed in equivalent CH2 groups) for insensitive (0.01 NaPO4, 5% PEG-8000,
denatured and native cytochrome c is 146, 5% Dx T-500, and 0.15 M NaCl). Here
which indicates the exposure, as the pro- charge sensitive or insensitive refers to sys-
tein denatures, of hydrophobic residues tems in which the charge of the substance
otherwise buried in the interior of the to be partitioned either affects its partition
polypeptide. behavior or not. They concluded that in
It also is possible to follow protein–pro- spite of PEG being a steric barrier for the
tein and protein–ligand association by 2- interaction between plasma proteins and
phase partitioning (29). The basic princi- the liposomes, a weak interaction remains
ple here is the strong dependence of the between the PEG-coated liposomes and
193
D. Forciniti

plasma proteins. Berggren et al. (6) used 2. MATERIALS


P(EO-PO)/Reppal 2-phase systems to
study the hydrophobicity of a series of pro- 2.1. Polymers
teins. They partitioned in these systems
several salts, Na2PO4, NaCl, and NaClO4, A variety of polymers have been used to
and several proteins of different hydropho- prepare 2-phase systems (see Table 2).
bicity, myoglobin, cytochrome c, lysozyme, PEG, a linear synthetic polymer of ethyl-
bovine serum albumin, and β-lactoglobu- ene oxide units, and Dx, poly(α-1,6-glu-
lin. They were able to correlate the parti- cose), are the most commonly used poly-
tion coefficients of these proteins with mers in the preparation of aqueous 2-phase
their tryptophan content. systems. Examples of other sugar polymers
used are Ficoll (polysucrose), pullulan, and
1.2. Batch and Continuous Partitioning maltodextrins. Derivatized carbohydrate
polymers have also been used; these
Most of the times, aqueous 2-phase par- include methylcellulose, hydroxyethylcel-
titioning is carried out in test tubes, so it is lulose (HEC), Reppal PES (hydroxypropyl
a batch operation. Attempts have been starch), benzoyl, dextran sulfate, and
made, however, to evolve the technique diethylaminoethyl (DEAE)/Dx. Examples
into a continuous operation, mostly to of synthetic polymers, besides PEG, are
improve the purification factor of a given polyvinyl alcohol (PVA), polyvinylpyrroli-
protein. One that we personally recom- done, pluronic, and random copolymers of
mend is liquid–liquid partitioning chro- ethylene oxide and propylene oxide:
matography. In liquid–liquid partitioning EO50PO50, EO20PO80 (UCON), etc.
chromatography, one phase (for example Several suppliers can be used worldwide:
the Dx-rich phase in a PEG/Dx system) is Sigma (St. Louis, MO, USA) (PEG, Dx,
immobilized on a convenient support, and Dx-SO4, Ficoll, Methylcellulose), Union
the other phase (in this case the PEG-rich Carbide (Bound Brook, NJ, USA) (PEG,
phase) is used as the mobile phase. UCON), Amersham Pharmacia Biotech
The column is made of agarose or silica (Piscataway, NJ, USA) (Dx and Ficoll),
diol beads whose surface is derivatized by Polysciences (Warrington, PA, USA)
growing a hydrophilic polymer (polyacryl- (PEG), Shearwater Polymers (Huntsville,
amide) on it. The Dx-phase is retained, AL, USA) (PEG and PEG derivatives), etc.
and the silica diol or agarose beads become For most applications, the polymers are
impermeable to the proteins as determined used as received. Multivalent ions in com-
in our laboratory. The PEG-rich phase is mercial Dx can be eliminated by dialysis,
used as the mobile phase. The elution ultrafiltration, or by a desalting step. Impu-
times suggest that the partitioning into rities in PEG (antioxidants, ethylene gly-
Dx-phase is significant. For a column used col, and diethylene glycol) can be eliminat-
by us, of 2.5 cm in diameter and 40 cm ed by ether or hexane precipitation of a
length (about 196.3-mL volume), the con- PEG/acetone solution (1,11). Often, the
tinuous phase (PEG) is about 10 mL, and molecular weight and the molecular weight
the Dx-phase is about 5 mL. If the Dx- distribution are given by the manufacturer.
phase is assumed to coat the beads uni- In the absence of accurate information, the
formly, then the ratio (radius of the bead molecular weight can be determined by a
with the coat)/(radius without the coat) is size exclusion chromatography-low angle
about 1.009. For a bead of radius 10 µm, light scattering tandem (no internal stan-
the film thickness is about 900 nm. dards are needed) or by size exclusion chro-
194
Hemoproteins Purification and Characterization

matography using the appropriate stan- Biotech, whereas narrow fractions of PEG
dards. Some companies, like Wyatt Tech- can be purchased from PolySciences.
nology (Santa Barbara, CA, USA), provide
determination of molecular weights for a 2.2. Buffers
fee. The molecular weights of PEG and Dx
can be determined by using a Superose A variety of buffers have been used to
12 column (Amersham Pharmacia regulate the pH in aqueous 2-phase sys-
Biotech) eluted with a 3% NaCl solution tems. The two most commonly used are
at room temperature (17). Figure 3 shows a phosphate and Tris buffers. The buffers
typical molecular weight distribution curve must be kept in a refrigerator and used
for Dx. Molecular weight standards for within 30 days.
PEG can be bought from Polysciences.
Since molecular weight standards for Dx 2.3. Additives
are difficult to obtain, narrow fractions of
pullulan (Polysciences) can be used. Poly- A series of additives are normally used in
disperse polymers of good quality can be aqueous 2-phase systems. Bacteriocides
purchased from speciality chemical compa- (either sodium azide or chloroacetamide) are
nies like Sigma. Polymer batches of narrow conveniently added to the polymer stock
molecular weight distributions can also be solutions or in solid form to the 2-phase sys-
purchased but at a much higher price. For tem. A series of salts (shown in Figure 2) are
example, less polydisperse Dx can be commonly used to drive the protein of
bought from Amersham Pharmacia interest into one or another phase.

Figure 3. Molecular weight distribution of Dx T-500. This was obtained by running a Dx solution through a Superose 12 col-
umn eluted with NaCl (3%). The column was calibrated with pullulan standards.

195
D. Forciniti

2.4. Polymer Derivatives lyzed first against sodium bicarbonate


and then against water.
Different PEG derivatives have been PEG-fatty acids can be also easily pro-
used in aqueous 2-phase partitioning duced in the laboratory by reacting PEG
experiments as affinity ligands. The three with the chloride or anhydride of the fatty
main kinds are PEG-dye, PEG-fatty acid, acid in toluene
and PEG-imidazole compounds. Several
dyes, including Cibacron Blue F3G-A
(Ciba-Geigy, Basel, Switzerland), Procion 3. METHODS
Red HE-3B, Procion Green H-4G, and
Procion Brown MX-5BR (I.C.I. Organic 3.1. Preparation of Stock Solutions
Division, Blackely, UK), and Remazol dyes
(Hoechst, Frankfurt, Germany) can be The concentration of each stock solu-
conveniently attached to PEG-amine (from tion is only limited by the solubility of the
Shearwater Polymers). Some PEG-dye polymers and by the increase in viscosity,
derivatives are available commercially (for which makes the solutions very cumber-
example, Sigma commercializes PEG-red, some to handle. For a polymer–polymer
MW 8000), but they can be easily prepared system, stock solutions of polymers and the
in the laboratory, as can other derivatives. appropriate buffer are prepared; the poly-
mer stocks can be prepared in water or
❖ Procedure 1. Preparation of PEG buffer. For a polymer–salt system, the stock
Derivatives polymer solution, and a stock salt solution
of the desired pH, are prepared. In the sys-
A. Preparation of PEG-Red tems for affinity partitioning, stocks of
derivatized polymers are also prepared.
1. Dissolve aminated PEG and Procion Sodium azide (1 mmol) or chloroac-
Red HE 3B (1:1.8 ratio) in water. etamide (up to 5g/L) should be added to
2. Adjust the pH to 11.0 (5 M NaOH) each stock solution as a bacteriocide. Stock
and incubate the mixture at 60°C for solutions of the polymers must be stored at
24 hours with constant stirring. 4°C and used within 30 days of being pre-
pared. Stock solutions of PEG must be
3. Remove the excess salts by dialysis. stored in the dark to prevent UV-induced
oxidation. Age and exposure to light
B. Preparation of Iminodiacetate PEG induces the formation of acidic groups in
(for Metal Affinity Two-Phase spite of the addition of antioxidants. A
Partitioning) decrease in pH and a yellowish color of a
PEG solution are clear indications that oxi-
1. The synthesis begins with PEG-chlo- dation has taken place. Preparation of
ride (either produced in the laboratory some representative stock solutions is de-
or obtained from Shearwater Poly- scribed below.
mers), to which iminodiacetic acid and PEG stock of 30% to 50% (wt/wt) is
potassium carbonate are added. prepared by accurately weighing the poly-
2. The solution is refluxed for 48 hours. mer and the water or buffer in a flask and
3. The reaction is stopped by adding sodi- stirring for an hour or more on a magnetic
um sulfate and allowed to separate into stirrer until a clear solution is obtained.
2 phases. Solid PEG, if properly stored, contains less
4. The PEG-rich phase is diluted and dia- than 0.5% water. Dx stock of 20% to 30%
196
Hemoproteins Purification and Characterization

(wt/wt) concentration is prepared by first (the specific optical rotation of Dx is +199°


making a paste of the powder with a small mL/gdm). This method is applicable for
amount of water and then adding the rest the determination of concentration of
of the water to reach the final mass. Be- other carbohydrate polymers as well.
cause of the presence of water (5%–10%) Freeze-drying can be used to measure
in commercial Dx, an amount of Dx in the concentration of polymers in the stock
excess to that needed may be weighted. solutions: (i) a known amount (from 5 to
Heating the Dx solution up to 95°C on a 20 g) of polymer stock solution is added to
hot plate is highly recommended to facili- a freeze-drying flask; (ii) the solution is
tate the dissolution of the polymer and to freeze-dried for about 8 hours; and (iii) the
reduce bacterial growth. dried polymer is dissolved in 2 mL of
The final polymer concentration in the water, then freeze-dried for another 8
stock solution can be easily determined by hours. The user should check for constant
refractive index measurements (either PEG weight at least the first time that this
or Dx), polarimetry (Dx), colorimetric technique is used. We have found that
essays (PEG), or freeze-drying (both poly- extensive freeze-drying followed by rehy-
mers). To measure the concentration of dration in a small amount of water and
PEG by refractive index measurements, it subsequent freeze-drying yields results that
is necessary to measure the refractive index are identical to those obtained with other
increment of the solution above the buffer techniques.
(the refractive index increment above water Two to four times concentrated stock
of a 1% PEG solution is 0.00139). The solutions of salts are prepared using reagent
density of the solution can be measured grade chemicals. The solutions may be
very precisely using a U-shaped oscillator adjusted to the required pH before making
densitometer (Anton PAAR USA, Asland, up the final mass of the solution. For
VA, USA) or estimated from ρPEG = example, in case of phosphate and citrate
[0.997 + 0.169 CPEG/100] and ρDx = solutions, the acid and the basic salts are
[0.0997 + (0.391 CDx/100)], with C in weighed in molar ratios determined by the
g/100 mL and the densities of PEG and desired pH. Salts can also be used directly
Dx in g/mL at 25°C. Alternatively, the in the solid form.
concentration of PEG can be obtained by a For cross-partitioning experiments, the
colorimetric assay: (i) 5 mL of 0.5 M per- following stocks are also required:
chloric acid are added to 1 mL of PEG a. Stock solutions of a series of 0.04 M
solution; (ii) after 15 minutes, the precipi- buffers (glycine or sodium phosphate)
tate (if any) is discarded; (iii) 1 mL of 5% spanning the pH range from 3.5 to
BaCl2 and 0.4 mL of 0.1 M iodine are 11.5.
added to 4 mL of a PEG solution; (iv) after b. Stock solutions of alkali (i.e., lithium,
15 minutes, the absorbance is measured at sodium, potassium) chlorides (0.33
525 nm against a blank of all the above M).
chemicals except PEG; and (v) a calibra- c. Stock solutions of alkali sulfates
tion curve is prepared in the concentration (0.167 M).
range of 0.1 to 0.6 g/mL of PEG. The con- Protein stock solution, 1 g/L. Proteins
centration of Dx in the stock solution can should be desalted before use. Dialysis
be measured using a polarimeter with a Na against a buffer made in nanopure water,
lamp at 589 nm and 25°C: (i) 5 g of the ultrafiltration operated in dialysis mode, or
solution is diluted to 25 mL with water; a desalting column can be used for this
and (ii) the optical rotation (a) is measured purpose. For example, to desalt a protein
197
D. Forciniti

Table 4. Some Convenient Aqueous Two-Phase Systems


Concentration of Species 1 Concentration of Species 2 Water (or Buffer)
in % (wt/wt) in % (wt/wt) in % (wt/wt) VT:VB

PEG-3400 6.5% Dx-500 6.50% 87 1:1


PEG-6000 6.74% Dx-70 10.82% 75.94 1:1
PEG-1500 13.66% K2PO4 (pH 7.0) 13.12% 73.22 1:1
PEG-1000 14.5% MgSO4 10% 75.5 1:1
The polymers molecular weights are nominal.

by ultrafiltration prepare 50 g of a 1 g/L phase systems for the first time should
protein solution and ultrafiltrate with 5 choose equal volumes of top and bottom
volumes of a 50 mmol Tris buffer, pH 7.0. phases to facilitate sampling and protein
Stock solutions of affinity ligands, e.g., partition coefficient determination. For ana-
PEG-bound ligand, can be prepared at a lytical purposes, 5- or 10-g systems are very
concentration of hundredfold or more, as convenient. Although any buffer can be
the final concentration of these species in used, for acidic and neutral pHs, phosphate
the 2-phase system is quite low. The cost of buffers are recommended, whereas for basic
the PEG derivatives limits the amount of pHs, Tris buffers can be used. Buffer con-
stock solution to be prepared. centration should be kept between 20 to 50
mM. It is less cumbersome to work at room
3.2. Selection and Preparation of temperature since the mixing, equilibration,
Aqueous Two-Phase Systems and sampling has to be done at the same
temperature. Because protein partitioning is
The selection of the appropriate phase only marginally affected by temperature
system depends on the final application. changes, low temperatures may be desirable
PEG/Dx is by far the most common pair of to maintain protein stability. The final
polymers used. Other incompatible poly- preparation of an aqueous 2-phase system is
mers were already mentioned and summa- quite straightforward, and a detailed recipe
rized in Table 2. High molecular weight Dx can be found in Reference 11. An example
(Dx-500 000) is highly recommended, since is given in Procedure 2.
it can be used with low molecular weight
PEGs reducing the viscosity of the phases. ❖ Procedure 2. Preparation of a
Also popular are PEG/K2HPO4-KH2PO4 Dx-500 000/PEG-4000 System
systems. After the phase forming species at Room Temperature
have been selected, the next step is to select a
particular tie-line. Good sources of tie-lines 1. Shake the stock solutions well so that
are the monograph by P.Å. Albertsson (2), there are no density gradients.
the book by Zaslavsky (56), which contains 2. Place a graduated centrifuge tube of 15
about 150 phase diagrams for Dx/PEG, Dx/ mL total volume on a weighing balance.
Polyvinylpyrrolidine (PVP), Dx/Polyvinyl 3. Weigh out the stock solutions into the
alcohol (PVA), Dx/Ficoll, PEG/Polyvinyl tube in order of their increasing densi-
methyl ether, and PEG-salt, and several arti- ties, and layer them carefully over each
cles (16,43,44). Some convenient 2-phase other. This facilitates the removal of
systems are shown in Table 4. As a rule of portions of one stock solution in case
thumb, those who are using aqueous 2- of error during weighing. Because of
198
Hemoproteins Purification and Characterization

the problem of accurately pipetting the the ratio between top and bottom phase
polymer stock solutions due to their volumes of both published and prepared
high viscosity, they are best measured systems may not be the same. The appear-
by weight and are easily transferred ance of only one phase after following a
using a Pasteur pipet with a broken tip. published recipe step-by-step is equally
4. Mix the contents of the test tube thor- frustrating. These apparent inconsistencies
oughly, first by hand, and then in a cause people to believe that the lack of
rotary shaker (20 min is enough) at the reproducibility is an inherent property of
equilibration temperature. aqueous 2-phase systems. Fortunately, this
is not true. The most common reasons for
5. Let the systems settle for a period of 30 these inconsistencies are:
minutes to 24 hours depending on the
system composition, or centrifuge • The selected tie-line is too close to the
them for 2 to 15 minutes at 1500× g. critical point. So, small differences in
Poor temperature control in centrifuges the molecular weight or molecular
makes it more convenient to sediment weight distribution of the polymers,
the systems in a water bath or in a presence of additives, or differences in
chromatographic chamber when work- temperature moved the system into
ing at a temperature other than ambi- the one phase region. Addition of
ent. In general, the time of phase sepa- small amounts of one of the two poly-
ration depends on the distance of the mers will move the system into the 2-
working tie-line from the critical point. phase region.
Close to the critical point, the phase • The selected tie-line is too far from the
separation time is long. At intermedi- critical point. When working at longer
ate tie-lines the phase separation time is tie-lines poor mixing is normally the
shorter. If the more viscous phase vol- cause for the lack of production of 2
ume is larger than the volume of the phases. Since the denser stock solution
less viscous phase, the phase separation is added to the centrifuge tube first, it
time increases. If the system is to be is quite difficult to mix the residue of
used in a liquid–liquid partition chro- stock solution that is trapped in the tip
matography system, one must chose a of the tube with the rest of the solu-
total concentration of polymers such tion. So, the 2-phase system is actually
that the PEG-rich phase constitutes prepared using a considerable smaller
most of the volume. This phase system amount of one of the two polymers.
must be allowed to settle for 24 to 48 To assure good mixing, mix the con-
hours before the PEG-rich phase is tent of the tube in a vortex mixer and
used as the mobile phase. inspect the tip and walls of the tubes
If instructions are followed carefully, the for stock solution residues. Continue
preparation of aqueous 2-phase systems mixing after no deposits are present,
should be routine laboratory work. Still, a and place the tube in a rotary shaker.
common source of frustration for those
using aqueous 2-phase systems for first 3.3. Preparation of Liquid–Liquid
time is their apparent lack of reproducibil- Partitioning Chromatography
ity. As indicated before, systems are nor- Systems
mally prepared according to some pub-
lished binodal. Often, the prepared system Specific guidelines for the use of this
differs from the one published. Specifically, method can be found in the various articles
199
D. Forciniti

by Muller (35–37). The main steps are attempting to run a liquid–liquid par-
outlined in Procedure 3. tition chromatography (LLPC) col-
umn. The partition coefficient of the
❖ Procedure 3. Preparation of target protein must be adjusted to be
Partitioning Chromatography Systems between 0.3 and 0.1 (the data shown
in Figure 2 can be used for this pur-
1. Measure the partition coefficient of the pose), and the salt concentration
raw materials in batch systems before should be high enough to shield elec-

Figure 4. Purification of hemoglobin from E. coli. Cell homogenate is mixed with Dx and PEG solutions. The Dx-rich phase is
discarded, and Na2SO4 is added to the PEG-rich phase. The Na2SO4-rich phase is discarded, and MgSO4 is added to the PEG-
rich phase. The protein is recovered from the salt-rich phase.

200
Hemoproteins Purification and Characterization

trostatic interactions between the pro- relate quite well with the partition coeffi-
teins and the gel (ionic strength of 0.05 cients of the proteins obtained in batch
or higher). experiments, so scale-up is straightforward;
2. After the optimum conditions to (iii) very low amounts of Dx are needed,
obtain an appropriate partition coeffi- which is of direct benefit as far as costs go.
cient have been identified, prepare In addition, if the losses are proportional to
enough top phase at the right pH and the total Dx, then the losses are expected to
at the right ionic strength to elute the be low as well; (iv) PEG precipitates large
column. The top (PEG-rich) phase is proteins (above 200 000 Da) at the station-
allowed to equilibrate for several days ary phase–mobile phase interface. This is
in the presence of small amount of bot- avoided at all costs, as the precipitate clogs
tom phase. the column; and (v) the eluant contains
significant amounts (around 10% wt/wt)
3. The column is packed according to
PEG. Depending on the final application,
Muller (36) and equilibrated until the
this can be removed as described in the fol-
UV noise of the effluent has dropped
lowing section.
below 0.005 OD units at 280 nm. This
ensures that all the Dx-rich phase that
is not bound to the beads has been ❖ Procedure 4. Determination of
washed out. Partition Coefficients
4. The sample is dissolved in the mobile
phase (it should not exceed 2%–3% of 1. Approximately 1 mL of the protein
the bed volume for analytical runs and solution to be purified is added to the
as twice as much for preparative runs) phase-forming species mixture replac-
and injected into the column. The elu- ing an equal amount of buffer. Mixing
tion is started immediately. and phase separation are done as
described above for systems that do not
As in any chromatographic separation, contain any protein.
sample preparation is quite important. If
the starting material is a cell homogenate, 2. Mixing has to be done carefully. It has
solids must be sedimented out by centrifu- to be vigorous enough to allow distrib-
gation for about 15 minutes at 2000× g. ution of the proteins between the 2
The clear supernatant is mixed with the phases but gentle enough to prevent
appropriate amount of PEG that is going protein denaturation (a rotary shaker is
to be used as a mobile phase. If aggregates highly recommended, whereas the use
are observed, they must be eliminated by of a vortex mixer is discouraged).
centrifugation. If no precipitation is 3. The phase systems are centrifuged at
observed, more PEG is added (to reach 1500× g for 20 minutes to speed phase
30%), the liquid is cooled in an ice bath settling.
for 10 minutes, and the protein precipitate 4. Sampling is done by pipetting carefully
is removed and resuspended in buffer. 1 mL of top phase and 1 mL of bottom
Some features of these type of systems phase from each partitioning tube (the
are: (i) the partition coefficients must be amount pipetted should be controlled
sufficiently different from 1 to make an by weighing for more precise sam-
impact on the retention times. That is, pling). Impurities may accumulate at
they must be in the right range to make a the liquid–liquid or liquid–air inter-
multicomponent chromatographic separa- faces. They do not constitute a prob-
tion possible; (ii) the elution volumes cor- lem unless they are pipetted during
201
D. Forciniti

sampling, so a positive pressure on the concentration is usually enough). If no


pipet as it enters the phases is always precipitate is present, poor sampling is
recommended. Blank phase systems are probably the source of error.
sampled in the same manner.
5. The samples are diluted with buffer. 3.4. Removal of PEG
The actual dilution depends on the
particular protein and on its partition Depending on the final application of
coefficient. Since the viscosity of the the protein purified by aqueous 2-phase
phases is very high, improper mixing of extraction, it may be desirable to eliminate
the sample and the dilution buffer may all or most of the polymer that contami-
result. Uncontrollable scattering from nates the protein of interest. Although the
regions of different densities within the overall yield of the separation may be
sample produces erroneous absorbance reduced, one of the easiest ways of elimi-
readings. As a general rule, mix the nating the polymer (usually PEG) from the
sample of the phase with the dilution protein solution is to repartition the PEG-
buffer and stir in a vortex mixer. rich phase against a salt (phosphate or sul-
6. Leave the solution resting and stir fate) rich phase. This is accomplished
again. Inspect the solution to detect rather easily by first separating the top
density differences along the axial (PEG-rich) phase from the bottom (Dx-
direction of the test tube. Continue rich) phase and by adding either solid sodi-
stirring until the solution is completely um phosphate or sulfate directly into the
transparent. Because of the relatively PEG-rich phase. By driving the protein
high absorbance of the blanks at 280 into the salt-rich phase most of the PEG is
nm, an absorbance reading of protein eliminated. If the size of the protein is suf-
containing samples between 0.5 and 1 ficiently different from the size of PEG,
is recommended. Hemoproteins are PEG-protein mixtures can be separated by
conveniently measured at 540 nm. ultrafiltration and by gel permeation chro-
Standard protein tests like Bradford’s matography. For example, we have used
test (8) can be also used. Ultra free-20 (Sigma) centrifuge tubes with
a nominal molecular weight cut-off of
7. The partition coefficient is calculated 10 000 to separate lysozyme from PEG-
from K = [Absorbancesample - Absor- 4000. The samples were centrifuged at
banceblank]top/[Absorbancesample - Ab- 12 000× g for 30 minutes. Up to 85% of
sorbanceblank]bottom. the PEG present is eliminated in this way.
8. For preparative applications a precise
mass balance is not necessary. For ana- 3.5. Methods for Characterization
lytical purposes, a protein mass balance Experiments
can easily be performed, since the vol-
umes of the phases are very easy to 3.5.1. Cross-Point Determination
measure, and the density of each phase
is well correlated with polymer concen- Prepare 2 sets of Dx/PEG systems (Set A
tration. If the mass balance is not close which contains alkali chloride and Set B
(within 5%), check for the formation which contains alkali sulfate) spanning a
of a precipitate at the liquid–liquid pH range from 3.5 to 11.5. Two runs are
interface. If a precipitate is present, one highly recommended for precision work.
should use more diluted protein solu- In the first run, 4 or 5 different pH values
tions (a decrease of 50% in protein are enough. In the second run, 5 or 6
202
Hemoproteins Purification and Characterization

points should be obtained in the neighbor- generally due to poor sampling, and a pro-
hood of the cross-point. tein mass balance should be done to assure
that sampling has been done correctly.
❖ Procedure 5. Cross-Point The sensitivity of cross-partitioning
Determination depends upon the angle at which the 2
lines intersect. If the lines are perpendicu-
1. Set A. Add to a 10-mL centrifuge tube lar, the sensitivity is at a maximum, while
(37): (i) 2.5 g of Dx stock solution; (ii) parallel or nearly parallel lines yield no
1.0 g of PEG stock solution; (iii) 3 g of cross-point or a “cross-point range”. The
sodium chloride; (iv) 2.5 g of buffer; slope of the lines depends upon the type of
and (v) 1 g of the protein stock solution. salt, the change in net charge of the protein
2. Set B. Add to a 10-mL centrifuge tube: with pH, the specific interactions between
(i) 2.5 g of Dx stock solution; (ii) 1.0 g the ions and the proteins, and the salt-
of PEG stock solution; (iii) 3 g of or induced changes in the interactions
sodium sulfate solution; (iv) 2.5 g of between polymer and protein. The sensi-
buffer; and (v) 1 g of the protein stock tivity can be manipulated by varying the
solution. molecular weight of the polymers, the tem-
3. The final phase system composition is perature, the concentration of the poly-
7.5% (wt/wt) Dx, 5.0 (wt/wt) PEG, mers, and the type of salt. So, cross-parti-
0.1 M alkali chloride or 0.05 M alkali tioning should be done using the lowest
sulfate, and 0.04 M glycine or sodium possible PEG molecular weight to mini-
phosphate buffer. mize problems associated with the high vis-
4. Prepare blanks of the phases without cosity of the phases. If the sensitivity is not
added protein. good enough, the experimentalist needs to
5. Mix, equilibrate, and sample the phases explore different conditions until a good
sensitivity is found.
as explained in Section 3.4.6.
The pH and the partition coefficient at
6. The pH in each phase is measured with the cross-point are only marginally depen-
a microelectrode directly on the undi- dent on the combination of salts used and
luted phases. Because of the high vis- on their concentration. For example, NaCl
cosity of the phases, the pH measure- can be replaced by potassium chloride
ments must be done over a relatively and/or sodium sulfate by lithium sulfate
long period of time. without affecting the results. Still, some
7. The partition coefficients of Sets A and small differences in pH values at the cross-
B are plotted versus the pH. The pH point with different salts have been ob-
and the partition coefficient values at served. These differences are similar to
which one Kp versus pH line (Set A) those encountered in the electrophoretic
crosses the other one (Set B) and are determination of isoelectric points, which
read from the axes. can also be slightly affected by the salt used.
The lines of Kp versus pH may not cross This independence of cross-partitioning
each other because of errors in pH or in the on the type and concentration of salt
values of the partition coefficients. One makes cross-partitioning a viable option for
must be sure that the pH has been mea- determining the isoelectric point of pro-
sured long enough to reach equilibrium and teins that are stable only at high salt con-
that the pH of both phases agrees within the centrations. In contrast, the type and con-
experimental uncertainty (approximately centration of salts have a strong influence
0.05 pH units). Erroneous values of Kp are on the shape of the ln Kp versus pH curves.
203
D. Forciniti

3.5.2. Surface Hydrophobicity phase system. Again, the hemoglobin pre-


ferred the PEG-rich phase. Solid magne-
Dx/Ficoll-400 systems are prepared by sium sulfate was added to the PEG-rich
weighting stock solutions of the polymers phase to form a PEG/MgSO4 2-phase sys-
to a final concentration of 12.5% Ficoll tem. In contrast to the PEG/Na2SO4 sys-
and 10.8% Dx-70. The systems are pre- tem, the hemoglobin preferred the salt-rich
pared in sodium phosphate buffer at pH phase. The use of Mg titration to separate
7.4 at a concentration of (56): the protein from PEG may be applicable to
Cbuffer = 0.11 - 0.67CNaCl [Eq. 4] other proteins too.
where the concentration of the buffer is var-
ied from 0.01 to 0.11 M and the concentra- 4.2. Partitioning of Hemoproteins in
tion of NaCl is varied from 0 to 0.15 M. PEG/Dx/Cu plus PEG Systems
The protein(s) of interest is partitioned in Small amounts of metal chelate PEG
this set of systems as indicated above. The (PEG-iminodiacetic acid loaded with
logarithm of the partition coefficient is plot- Cu++) added to conventional aqueous 2-
ted versus the ionic strength. The zero inter- phase systems have been used to extract
cept yields a parameter that represents the proteins that contain histidine (40,55). A
strength of all the interactions of the protein plot of ∆ln Kp is linear with the number of
with an aqueous environment relative to exposed histidine groups on the protein
that of a methylene group and the slope molecules. They were able to separate
yields a parameter that reflects the strength cytochrome c, myoglobin, and hemoglo-
of the hydration interactions of all the iono- bins. pH control is critical when using this
genic groups of the protein relative to that of technique, since at low pHs the free base of
the α-carboxyl group of DNP-amino acid. the imidazole form a noncoordinating imi-
dazolium side chain that does not bind
4. EXAMPLES Cu++. The enhancement in the partitioning
of the proteins into the PEG-rich phase
4.1. Isolation of Recombinant caused by the addition of PEG-IDA is
Hemoglobin from Cell Homogenates remarkable. For example, at pH 8.0, the
partition coefficient of human hemoglobin
The work by Hart and Bailey (21) is a is 0.38 (7% PEG-8000, 4.4% Dx T-500,
good example of the use of aqueous 2- 0.1 M NaCl, and 0.01 M sodium phos-
phase systems for the purification of a phate), whereas upon addition of PEG-IDE
recombinant protein. They isolated Vitre- the partition coefficient increases to 14.
oscilla hemoglobin from E. coli lysate. The
purification was done in three extraction 4.3. Cross-Partitioning of Hemoglobins
steps (see Figure 4) with an overall yield of
47% and a purity higher than 95%. In the Numerous hemoproteins have been
first partition step, a PEG/Dx system was studied by using cross-partitioning experi-
used. In this system, the contaminant pro- ments. The pH at the cross-point agrees
teins partitioned strongly into the Dx-rich very well with the isoelectric point pH
phase, whereas the hemoglobin preferred determined by using other techniques. For
the PEG-rich phase. Additional purifica- example, human A hemoglobin has a pH at
tion of the target protein is achieved by the cross-point of 7.0 (isoelectric point:
adding solid sodium sulfate to the PEG 7.0), and cytochrome c from horse yields a
phase, thus forming another PEG-salt 2- pH at the cross-point of 9.85 (isoelectric
204
Hemoproteins Purification and Characterization

point: 9.8). The cross-partitioning of he- The tune-up of the separation is quite
moproteins from cytochrome c (MW straight forward, and it includes optimiza-
12 000) to catalase (MW 240 000) in Dx/ tion of the partition coefficient of the target
PEG systems (51) yields partition coeffi- protein as compared to the contaminant
cients at the cross-point that do not show proteins, optimization of the amount of
the clear dependence on protein molecular affinity ligand, and inhibition of other
weight found with nonhemoproteins. Even enzymes that compete with the target pro-
though the molecular weights of human tein for the affinity ligand. For example, in
hemoglobin variants (A, F, S, and C) and the purification of FDH by using affinity
hemoglobins from different species are liquid–liquid partition chromatography, we
essentially the same, the partition coeffi- found that the optimum conditions were
cient at the cross-point of hemoglobins A achieved when the stationary phase was Dx-
and F and those of hemoglobins from dif- 500, the mobile phase PEG-20 000 (2.7%
ferent mammalian species show measurable PEG and 4.5 Dx), 75 mM potassium bro-
differences. Although the 4 human hemo- mide, 12 mM phosphate buffer at pH 7.5,
globin variants differ in charge, adult and a concentration of PEG-red in the
hemoglobins A, S, and C have the same the mobile phase of 5 × 10-5 M. The sample
partition coefficient, while the fetal hemo- was 500 µL of crude extract of C. boidinii
globin (F) has a lower partition coefficient. heated for 10 minutes at 55°C. We also
showed that the separation of the ligand
4.4. Liquid–Liquid Partitioning from the enzyme is quite straightforward.
Chromatography The eluate containing the FDH–ligand
complex was treated with potassium phos-
There are numerous examples of the use phate forming a PEG-salt system. Using a
of this technique for the purification of concentration of potassium phosphate of
nucleic acids and proteins. In a few of 10% (wt/wt) the K value of FDH was
them, the addition of affinity ligands to 0.003, and the volume ratio was 1:4.22 (top
improve the separation has been explored. to bottom). The yield of the enzyme in the
Because the elution volume is extremely lower phase was 99%. The column was sta-
sensitive to the partition coefficient of the ble for more than 1 year, and scale-up was
protein (if it is smaller than 3), even in the straightforward.
absence of affinity ligands considerable res-
olution can be obtained. For example,
Muller (37) separated a synthetic mixture 5. CONCLUDING REMARKS
of lysozyme (retention volume, V = 17
mL), peroxidase (V = 19 mL), cytochrome Aqueous 2-phase extraction is a very
c (V = 25 mL), myoglobin (V = 30 mL), β- well-established technique that has been
lactoglobulin (V = 39 mL), ovotransferrin used in biochemical laboratories for the last
(V = 50 mL), ovalbumin (V = 60), and 40 years. It is a versatile, easy to use, and low
human serum albumin (V = 100) (0.6–1 cost technique. Although the primary use of
mg each) in a 300 × 10 mm column the method is the purification of proteins
packed with Lipargel coated with a solu- and other biological materials, it can be also
tion of Dx-40 and eluted at a flow rate of used for protein characterization studies.
0.3 mL/minute with a PEG-6000 solution. Uncountable proteins have been purified
We have demonstrated that affinity lig- using this technique either from cell
ands can be used in LLPC to increase the homogenates or from previously fractionat-
purification factor of a given protein (49). ed mixtures. Although it has been used to
205
D. Forciniti

separate cell debris from proteins providing 4.Alred, A. 1999. Purification of a recombinant protein
a first cheap purification step, it has been from mammalian cell culture: an industrial applica-
tion. 11th International Conference on Partitioning in
also used to purify proteins in a single step Aqueous Two-Phase Systems, Gulf Shore, Alabama.
from cell debris by making use of a variety 5.Asenjo, J.A., R.E. Turner, S.L. Mistry, and A. Kaul.
of affinity ligands. The possibility of using 1994. Separation and purification of recombinant pro-
teins from Escherichia coli with aqueous two-phase sys-
the technique in continuous mode by tems. J. Chromatogr. A 668:129-138.
immobilizing 1 phase opens even more pos- 6.Bergreen, K., G. Johansson, and F. Tjerneld.
sibilities. One of its main advantages is its 1995. Effects of salts and the surface hydrophobici-
ty of proteins on partitioning in aqueous two-phase
low cost and easy use. The cost of chemicals systems containing thermoseparating ethylene
is minimum (except if affinity ligands has to oxide-propylene oxide copolymers. J. Chromatogr.
be used), and the hardware needed to A 718:67-79.
7.Birkenmeir, G., G. Kopperschlager, P.A. Albertsson,
implement it is available in any biochem- G. Johansson, F. Tjerneld, H.E. Akerlund, S. Berner,
istry laboratory. Members of the aqueous 2- and H. Wickstroem. 1987. Fractionation of proteins
from human serum by counter-current distribution. J.
phase systems community can be reached at Biotechnol. 5:115-129.
our Web page for helping those who wish to 8.Bradford, M.M. 1976. A rapid and sensitive method
use this technique. I hope that this chapter for the quantitation of microgram quantities of protein
utilizing the principle of protein dye binding. Anal.
will encourage researchers in the heme and Biochem. 72:248-254.
related compounds research field to consid- 9.Fernandez, S., G. Johansson, and R. Hatti-Kaul.
er aqueous 2-phase systems for their isola- 1999. Two-phase partitioning of recombinant Cuti-
nase. 11th International Conference on Partitioning in
tion procedures. Aqueous Two-Phase Systems, Gulf Shore, Alabama.
10.Forciniti, D. 1994. Protein refolding using aqueous
two-phase systems. J. Chromatogr. A 668:95-100.
ABBREVIATIONS 11.Forciniti, D. 1999. Preparation of aqueous two-phase
systems. In R. Hatti-Kaul (Ed.), Aqueous Two-Phase
Systems–Methods and Protocols. Humana Press, New
Dx, dextran; HF, hydrophobicity factor; Jersey.
12.Forciniti, D., C.K. Hall, and M.-R. Kula. 1991. Effect
HSFi, surface hydrophobicity of protein I; of temperature on protein partitioning. Bioseparations
Kp, partition coefficient (= Ct/Cb); K0, 2:115-128.
partition coefficient at zero charge; K0, 13.Forciniti, D., C.K. Hall, M.-R. Kula. 1991. Protein
partitioning at the isoelectric point: effect of polymer
partition coefficient in the absence of affin- concentration and polymer molecular weight. Biotech-
ity ligands; KL, partition coefficient in the nol. Bioeng. 38:986-994.
presence of affinity ligands; PEG, poly- 14.Forciniti, D., C.K. Hall, and M.-R. Kula. 1992. Pro-
tein partitioning. Effect of pH and polymer molecular
ethyleneglycol; P(EO-PO), polyethylene weight. Chem. Eng. Sci. 47:165-175.
propylene random copolymer (UCON); 15.Forciniti, D., M.-R. Kula, and C.K. Hall. 1990. Inter-
PVA, polyvinyl alcohol; PVP, polyvinyl- facial tension in aqueous two-phase systems. Influence
of temperature and polymer molecular weight. J.
pyrrolidone; RHi, hydrophobicity of solute Biotechnol. 16:279-290.
i relative to glycerin; TLL, tie-line length. 16.Forciniti, D., M.-R. Kula, and C.K. Hall. 1991. Influ-
ence of polymer molecular weight and temperature on
phase composition in aqueous two-phase systems.
Fluid Phase Equilib. 61:243-262.
REFERENCES 17.Forciniti, D., M.-R. Kula, and C.K. Hall. 1991. Mol-
ecular weight distribution and aqueous two-phase sys-
1.Aguinada-Diaz, P.A. and R.Z. Guzman. 1996. Affin- tems. Biotechnology 20:151-162.
ity partitioning of metal ions in aqueous polyethylene 18.Guan, Y., T.H. Lilley, T.E. Treffry, C.-L. Zhou, and
glycol/salt two-phase systems with PEG-modified P.B. Wilkinson. 1996. Use of aqueous two-phase sys-
chelators. Sep. Sci. Technol. 31:1483-1499. tems in the purification of human interferon-alpha1
2.Albertsson, P.Å. 1986. Partition of Cell Particles and from recombinant E. Coli. Enzyme Microb. Technol.
Macromolecules, 3rd ed. Wiley (Interscience), New 19:446-455.
York. 19.Guoqiang, D., R. Kaul, and B. Mattiasson. 1994.
3.Albertsson, P.Å., S. Sasakawa, and H. Walter. 1970. Integration of aqueous two-phase extraction and affin-
Cross partition and isoelectric points of proteins. ity precipitation for the purification of lactate dehy-
Nature 228:1329-1330. drogenase. J. Chromatogr. A 668:145-154.

206
Hemoproteins Purification and Characterization

20.Harris, P.A., G. Karlström, and F. Tjerneld. 1991. 36.Muller, W. 1994. Columns using aqueous two-phase
Enzyme purification using temperature- induced phase systems. In H. Walter and G. Johansson (Eds.), Meth-
formation. Bioseparation 2:237-246. ods in Enzymology. Academic Press, NY.
21.Hart, R.A. and J.E. Bailey. 1991. Purification and 37.Muller, W. 1994. Separation of proteins and nucleic
aqueous two-phase partitioning properties of recombi- acids. In H. Walter and G. Johansson (Eds.), Methods
nant Vitreoscilla hemoglobin. Enzyme Microb. Tech- in Enzymology. Academic Press, New York.
nol. 13:788-795. 38.Petersen, L.C. 1978. Cytochrome c-cytochrome aa3
22.Hatti-Kaul, R. (Ed.). 1999. Aqueous Two-Phase Sys- complex formation a low ionic strength studied by
tems–Methods and Protocols, Chs. 2 and 3. Humana aqueous two-phase partition. FEBS Lett. 94:105-108.
Press, New Jersey. 39.Petersen, L.C. 1978. Measurements of cytochrome c-
23.Hustedt, H., K.H. Kroner, and M.-R. Kula. 1984. cytochrome aa3 complex formation by aqueous two-
Applications of phase partitioning in biotechnology, p. phase partition. Biochem. Soc. Trans. 6:1274-1275.
529-589. In H. Walter, D.E. Brooks, and D. Fisher 40.Plunkett, S.D. and F.H. Arnold. 1990. Metal affinity
(Eds.), Partitioning in Aqueous Two-Phase Systems. extraction of human hemoglobin in an aqueous poly-
Academic Press, New York. ethylene glycol-sodium sulfate two-phase system.
24.Johansson, G. 1984. Partitioning of proteins, p. 161- Biotechnol. Tech. 4:45-48.
226. In H. Walter, D.E. Brooks, and D. Fisher, (Eds.), 41.Sasakawa, S. and H. Walter. 1972. Partition behavior
Partitioning in Aqueous Two-Phase Systems. Academ- of native proteins in aqueous Dx-poly(ethylene glycol)
ic Press, New York. phase systems. Biochemistry 11:2760-2765.
25.Johansson, G. 1994. Charge determination by parti- 42.Shanbhag, V.P. and P.E.H. Jensen. 1999. Affinity par-
tioning. In H. Walter and G. Johansson (Eds.), Meth- titioning using poly(ethylene glycol) with covalently
ods in Enzymology. Academic Press, New York. coupled hydrophobic groups. In R. Hatti-Kaul (Ed.),
26.Kopperschlager, G. 1994. Affinity extraction with dye Aqueous Two-Phase Systems—Methods and Protocols.
ligands. In H. Walter and G. Johansson (Eds.), Meth- Humana Press, New Jersey.
ods in Enzymology. Academic Press, New York. 43.Silva, L.H.M., J.S.R. Coimbra, and A.J.A. Meirelles.
27.Lorch, J. 1999. Two-phase aqueous extraction as a 1997. Equilibrium phase behavior of poly(ethylene
process development tool. 11th International Confer- glycol) + potassium phosphate + water two-phase sys-
ence on Partitioning in Aqueous Two-Phase Systems, tems at various pH and temperatures. J. Chem. Eng.
Gulf Shore, Alabama. Data 42:398-401.
28.Lu, M., F. Tjernald, G. Johansson, and P.Å. 44.Snyder, S.M., K.D. Cole, and C.C. Szlag. 1992. Phase
Albertsson. 1991. Preparation of benzoyl-Dx and its composition viscosities, and densities for aqueous two-
use in aqueous two-phase systems. Bioseparation 2: phase systems composed of polyethylene glycol and
247-255. various salts at 25 C. J. Chem. Eng. Data 37:268-274.
29.Lundeberg, S. and L. Backman. 1994. Protein-protein 45.Sophianopulos, A.J. and K.E. Van Holde. 1964. Phys-
and protein-ligand interactions, p. 241-254. In H. ical studies of muramidase (lysozyme). J. Biol. Chem.
Walter and G. Johansson (Eds.), Methods in Enzymol- 239:2516-2524.
ogy. Academic Press, New York. 46.Spears, T. and D. Forciniti. 1994. Protein refolding
30.Mattiasson, B. and I.Y. Galaev. 1999. Affinity parti- using chaotropic aqueous two-phase systems. In R.D.
tioning using aqueous two phase systems formed by Rogers (Ed.), Aqueous Two-Phase Systems: From
thermosensitive polymers. 11th International Confer- Metal Ions to Biomolecules. ACS Books, Washington.
ence on Partitioning in Aqueous Two-Phase Systems, 47.Tjerneld, F., P.A. Alred, R.F. Modlin, A. Kozlowski,
Gulf Shore, Alabama. and J.M. Harris. 1995. Purification of biomolecules
31.Menge, U., M. Morr, U. Mayr, and M.-R. Kula. using temperature-induced phase separation. In R.D.
1983. Purification of human fibroblast interferon by Rogers and M.A. Eiteman (Eds.), Aqueous Biphasic
extraction in aqueous two-phase systems. J. Appl. Separations: Biomolecules to Metal Ions. Plenum
Biochem. 5:75-90. Press, New York.
32.Middaugh, C.R. and E.Q. Lawson. 1980. Analysis of 48.Vernau, J. and M.-R. Kula. 1990. Extraction of pro-
protein association by partitioning in aqueous two- teins from biological raw materials using aqueous
phase polymer systems: applications to the tetramer- PEG/citrate phase systems. Biotechnol. Appl. Bio-
dimer association of hemoglobin. Anal. Biochem. chem. 12:397-404.
105:364-368. 49.Walsdorf, A., D. Forciniti, and M.-R. Kula. 1990.
33.Modlin, R.F., P.A. Alred, and F. Tjerneld. 1994. Uti- Investigation of affinity partition chromatography
lization of temperature-induced phase separation for using formate dehydrogenase as a model. J. Chro-
the purification of ecdysone and 20-hydroxyecdysone matogr. 523:103-117.
from spinach. J. Chromatogr. A 668:229-236. 50.Walter, H., D.E. Brooks, and D. Fisher (Eds.). 1984.
34.Moribe, K., K. Maruyama, and M. Iwatsuru. 1997. Partitioning in Aqueous Two-Phase Systems, p. 498-
Estimation of surface state of poly(ethylene glycol)- 528. Academic Press, New York.
coated liposomes using an aqueous two-phase parti- 51.Walter, H. and D. Forciniti. 1994. Cross-partition-
tioning technique. Chem. Pharm. Bull. 45:1683- ing: determination of isoelectric point by partitioning,
1687. p. 223-233. In H. Walter and G. Johansson (Eds.),
35.Muller, W. 1989. Aqueous two-phase polymer systems Methods in Enzymology. Academic Press, New York.
for liquid/liquid partition-chromatography of biopoly- 52.Walter, H. and G. Johansson (Eds.). 1994. Methods
mers. Ber. Bunsen-Ges. Phys. Chem. 93:956-961. in Enzymology, Vol. 228. Academic Press, New York.

207
D. Forciniti

53.Walter, H., S. Sasakawa, and P.Å Albertsson. 1972. tioning in Aqueous Two-Phase Systems, Gulf Shore,
Cross partition of proteins. Effect of ionic composition Alabama.
and concentration. Biochemistry 11:3880-3883. 55.Wuenschell, G.E., E. Naranjo, and F.H. Arnold.
54.Winter, C., D. Ansaldi, J. Clifford, P. Lester, and B. 1990. Aqueous two-phase metal affinity extraction of
Wolk. 1999. Initial isolation of recombinant proteins heme proteins. Bioprocess Eng. 5:199-202.
from whole fermentation lysates using aqueous two 56.Zaslavsky, B.Y. 1995. Aqueous Two-Phase Partition-
phase systems. 11th International Conference on Parti- ing. Marcel Dekker, New York.

208
Structural Study of Heme Proteins by
9 Electron Microscopy of 2-Dimensional
Crystals

Terrence G. Frey
San Diego State University San Diego, CA, USA

1. INTRODUCTION spectroscopy. Thus, electron microscopy of


2D crystals with the application of image
Characterization of membrane proteins processing techniques, sometimes called
has always lagged behind that of soluble electron crystallography because many of
proteins, because most techniques of pro- the computational tools and techniques are
tein purification and structural study derived from X-ray crystallography, was the
depend upon having a water-soluble prepa- first technique to reveal the structure of a
ration. The development of myriad classes membrane protein in three dimensions
of detergents over the past decades has (43). It continues to play a very significant
greatly facilitated the purification of inte- role in the structural study of membrane
gral membrane proteins and many of the proteins. In this paper, I will review the
techniques of structural characterization. techniques of electron crystallography
Direct measurement of the 3-dimensional applied to integral membrane heme-con-
(3D) structures of integral membrane pro- taining proteins describing in general
teins has proceeded more slowly, but signif- terms:
icant progress was made in the past 10 to • Techniques for growth of 2D crystals.
15 years, and the atomic structures of a • Specimen preparation for electron
number of membrane proteins are now microscopy and collection of micro-
known. Although X-ray crystallography graphs.
and 2-dimensional (2D) nuclear magnetic
resonance (NMR) spectroscopy have been • Data processing to produce a 3D
used to elucidate the structures of thou- model.
sands of soluble proteins, their utility in • Comparison of results from electron
the structural study of membrane proteins crystallography and X-ray crystallogra-
has been limited by the difficulty of grow- phy.
ing 3D crystals in the case of X-ray tech- The reader should recognize that in the
niques or by the large size of protein–deter- context of these methods, heme-containing
gent micelles in the case of NMR membrane proteins are not intrinsically

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

209
T.G. Frey

different from other membrane proteins, of 2D crystalline “purple membrane” with-


and the techniques described and refer- in the plasma membrane of Halobacterium
enced here are applicable to other classes of halobium. The purple membrane can be
membrane proteins (86). The methods readily purified and consists of large 2D
used are in many cases very technical, and a arrays of bacteriorhodopsin molecules
full description is well beyond the scope of arranged on a 2D lattice and belonging to
a single article or even a whole volume. the 2-sided space group p3. This was one
Thus, I will outline the methods in rela- of the specimens exploited by Henderson
tively general terms and give the reader ref- and Unwin to demonstrate the efficacy of
erences for greater depth of description of recording noisy electron micrographs at
the techniques involved. Amos et al. have low electron doses and then recovering a
written an excellent and comprehensive high resolution image of a single unit cell
review of 3D structure determination of by averaging thousands of unit cells within
2D crystals (4). a crystal (78). They subsequently also cal-
culated the first 3D image of a membrane
protein and at a resolution sufficient to
2. GROWTH OF 2D CRYSTALS demonstrate transmembrane α-helices
(43). A number of other membrane pro-
2D crystals can be defined as crystals in teins occur naturally in 2D crystals; these
which a motif, a protein, or assembly of include gap junctions and the photosyn-
proteins in the case of a protein crystal, is thetic membrane of the bacterium
repeated at the points of a 2D lattice. Thus, Rhodopseudomonas viridis (68,76). Others
a 2D crystal is one that is characterized by can be induced to form 2D crystals within
a unit cell that periodically repeats at pre- their native membranes by the addition of
cise locations in two dimensions but not in specific ligands (62,71), by removing lipid
the third. Note that a 2D crystal is not a enzymatically (59), or by other methods
2D object but is 3D. The nomenclature (61), and since membrane proteins are
used to describe space groups of 2D crys- inserted the same way into their native
tals was first described by Holser (45) and bilayers, these crystals are all of the type
revived by Fuller et al. (37). The first sym- shown in Figure 1a. It is also possible to
bol describes the type of lattice and the sec- form 2D crystals of a membrane protein by
ond symbol the class of symmetry perpen- expressing a recombinant form in high
dicular to the crystal. The following concentration in another type of cell, as
symmetry operations describe symmetry was the case with cardiac gap junction pro-
elements within the plane of the crystal. tein (76). The latter approach affords the
There are a number of excellent reviews of opportunity to modify the protein using
membrane protein crystallization, and recombinant DNA technology, either by
some of these are devoted to growth of 2D removing components that might inhibit
crystals (50,55,60). Thus, I will not go into crystallization or to identify the location of
great detail describing these techniques a specific sequence in the final structure.
here, but will offer my own perspective.
2.2. Growth by Detergent Extraction
2.1. Naturally Occurring Crystals
In some cases, it is possible to form 2D
One of the first membrane proteins crystals of a protein by extracting native
studied by electron crystallography, bacte- membranes in which it is present at high
riorhodopsin, occurs naturally in patches concentration using detergent to remove
210
Structural Study of Heme Proteins by Electron Microscopy

not only excess lipid but also other conta- gent used, two different crystal forms have
minating proteins. For example, cytochrome been obtained. Multiple extractions with
c oxidase constitutes approximately 10% of nonionic Triton® X-114 and X-100 pro-
the protein of the mitochondrial inner mem- duces a vesicular preparation of nearly pure
brane, and one can purify a membrane frac- cytochrome oxidase and 25% by weight of
tion of nearly pure cytochrome oxidase by residual phospholipid. The molecules of
treating beef heart mitochondria with appro- cytochrome oxidase are arranged as dimers
priate concentrations of detergents followed related by a crystallographic 2-fold axis.
by centrifugation to separate detergent solu- The crystal is formed when a large vesicle
bilized components from the membrane collapses causing molecules from two sides
residue (30,66,80). This requires 2 to 3 of the vesicle to interdigitate in the center
detergent treatments, and in each case, the of the vesicle, as shown in Figure 1b, form-
pellet becomes enriched in cytochrome ing a 2D crystal in the space group p22121
oxidase while other membrane proteins with unit cell dimensions of a = 100 Å, b =
and excess phospholipid are removed by 125 Å, and a thickness of 210 Å (42). In
decanting the supernatants. Depending this case, each unit cell of the crystal con-
upon the type and concentration of deter- tains one molecule from each layer of the

Figure 1. Molecular packing in


four classes of 2D crystal. (a) All
molecules oriented in the same
direction in a single bilayer. (b)
One crystal composed of two layers
of a collapsed vesicle. (c) Molecules
with alternating orientation in a
single bilayer. (d) A crystal of class
in panel a rolled into a cylinder
producing a structure with helical
symmetry.

211
T.G. Frey

collapsed vesicle, i.e., each unit cell con- with purified detergent solubilized protein
tains two dimers. The space group and detergent solubilized lipids, either
describes a primitive orthorhombic lattice “native” lipids purified from the same type
with a 2-fold axis of rotation perpendicular of membrane as the protein are derived or
to the membrane. The symmetry operators synthetic lipids using Procedure 1
21 indicate 2-fold screw axes (rotation by (55,60,82,84). This is described in more
360°/2 followed by translation by half of a detail in Chapter 11.
unit cell) parallel to the a and b crystal axes
in the plane of the crystal. ❖ Procedure 1. Growth of 2D Crystals
Treatment with sodium deoxycholate
removes a large proportion of phospholipid 1. Dissolve lipids in an organic solvent
and dissociates cytochrome oxidase dimers (e.g., chloroform) and dry a measured
into monomers. The resulting 2D crystal is amount on the surface of a suitable ves-
not vesicular but consists of sheets of sel under a stream of nitrogen.
cytochrome oxidase monomers arranged on 2. Suspend the dried lipid in an appropri-
a primitive 2D lattice in the space group ate volume of buffered detergent by
p121, denoting no symmetry perpendicular sonication, producing mixed deter-
to the membrane and a 21 screw axis of sym- gent–lipid micelles.
metry along one of the crystal axes (37).
These crystals are of the class shown in Figure 3. Mix the protein and lipid solutions
1c. The unit cell dimensions are a = 69 Å and approximately 1–10 mg/mL) to give a
b = 140 to 170 Å depending upon the prepa- relatively high protein to lipid ratio
ration; the thickness is approximately 110 Å (approximately 1:1 by weight).
(35,37). In both cases, the structures of the 4. Remove the detergent by dialysis or by
preparations are more complex than adsorption.
described here. The vesicular p22121 dimer As the detergent concentration decreas-
crystals are predominantly multilayered vesi- es, the protein–detergent micelles and the
cles with many small 3D crystals containing protein–lipid micelles merge and eventual-
layers of the 2D crystal motif stacked in reg- ly form bilayer membranes containing a
ister (42,51,80). Single 2D crystals are more high concentration of protein. Under the
rare but can readily be found. The p121 crys- proper conditions, the protein molecules
tal form contains single layer crystals as well within the bilayers arrange themselves onto
as stacks of the simple 2D crystal motif in a 2D lattice forming 2D crystals (see Fig-
which successive layers are offset from one ure 2) (50,55,81). There are two common
another, and electron micrographs show sev- methods to remove detergent. One is to
eral differently appearing projections (35). adsorb excess detergent by adding com-
Multilayered crystals of membrane proteins mercially available resin beads, e.g., Bio-
are commonly observed and often severely Beads® (Bio-Rad Laboratories, Hercules,
inhibit structural study because of the het- CA, USA) (81). A slower more controlled
erogeneity in the structures of different crys- method is to dialyze the detergent–pro-
tals (35,54,69). tein–lipid solution against detergent-free
buffer. The speed of this process depends
2.3. Reconstitution of Purified Protein on the characteristics of the detergent
with Purified Lipids employed, principally the critical micelle
concentration (cmc)(50,55). In aqueous
The most general method for growing solution, detergent molecules aggregate to
2D crystals of membrane proteins begins form micelles that are in equilibrium with
212
Structural Study of Heme Proteins by Electron Microscopy

individual detergent molecules; the cmc is ple which should be pure and monodis-
essentially the concentration of individual perse. The latter property can be difficult
molecules in the presence of micelles. Since to achieve in the case of membrane pro-
detergent micelles are too large to pass teins which, owing to their hydrophobic
through the pores of common dialysis tub- surfaces, can adopt multiple aggregation
ing, dialysis of detergent solutions proceeds states even in the presence of detergents.
by the movement of individual detergent Thus, the choice of detergent can be criti-
molecules through the dialysis tubing. cal and affects both the aggregation of the
Thus, the rate of dialysis depends on the protein and the methods employed in
cmc; the higher the cmc, the faster dialysis removing it during crystallization trials.
occurs. Of course dialysis also proceeds For example, Suarez et al. found that beef
more rapidly at higher temperature. Triton heart mitochondrial cytochrome c oxidase
detergents have low cmc’s, so dialysis even is polydisperse in many common deter-
at room temperature takes a relatively long gents, but became monodisperse when
period of time. For this reason, Weiss et al. transferred to dodecyl-maltoside (also
used the Bio-Bead adsorption method to known as lauryl maltoside), a detergent
crystallize Complex III (cytochrome c oxi- consisting of a maltose polar head and a 12
doreductase) purified in Triton X-100 from carbon saturated hydrocarbon tail that they
Neurospora mitochondria (81,84). Kim et synthesized for this purpose (now commer-
al. grew crystals of purified cytochrome c cially available) (70). Choice of detergent
oxidase essentially identical to the dimer also affects the crystallization process,
crystals described above by reconstitution since, as mentioned above, the cmc deter-
with purified phospholipids (53). mines the dialysis rate, and other properties
Many factors can affect the prospects of affect the efficacy of adsorption to beads.
success in crystallizing a membrane pro- Frequently, two detergents within the same
tein. As with most crystallization experi- generic class can have significantly different
ments, a critical factor is the protein sam- cmcs. For example, dodecyl-maltoside has

Figure 2. Reconstitution of purified phospholipids and purified membrane protein into a protein–lipid bilayer by mixing
detergent solubilized lipid (left) and detergent solubilized protein (center) and then removing the detergent molecules.

213
T.G. Frey

a cmc of 0.15 mM, while decyl-maltoside 3. SPECIMEN PREPARATION AND


has a cmc of 1.6 mM. In some cases it may ELECTRON MICROSCOPY
be desirable to slow down dialysis of a
detergent with a high cmc, and this can be Preparation of specimens for electron
accomplished by dialyzing the sample microscopy is a critical step that must take
against a buffer containing the detergent at into consideration the goals of the project
a concentration that is below its cmc and and the limitations imposed by the elec-
then reducing the detergent concentration tron microscope. There are many books
when the dialysate is changed. Useful describing general techniques and methods
information on detergent properties can be for biological electron microscopy, and the
found in Jap et al. (50) and in Crystalliza- reader should consult these for detailed
tion of Membrane Proteins edited by Michel protocols (7,22,40). Generally, the goal is
(60) as well as in a pamphlet published by to obtain the 2D or 3D structure of the
Calbiochem-Novabiochem (6). protein at the highest possible resolution.
Other factors to be considered and var- The limitations imposed by the electron
ied are: microscope are:
• pH. • The specimen should be thin, general-
• Ionic strength. ly less than 1000 Å. A related goal is to
obtain a preparation of 2D crystals
• Temperature. that are a single unit cell thick, rather
• Choice of lipid. than multiple layers forming small 3D
• Protein concentration (generally sever- crystals.
al mg/mL). • The specimen must be stable at very
high vacuum, approximately 1 x 10-7
• The presence of other solutes that
Torr or better. This usually requires
might influence protein conformation
removing most or all of the water,
(specific ligands, etc.) or solubility.
unless the specimen is maintained at
Given all of the conditions that might very low temperature, -160°C or
be varied, it is useful or essential to have a below. Removal of water will generally
convenient system for microdialysis of destabilize the specimen conforma-
many different samples. There are a num- tion, so one is then faced with finding
ber of commercial microdialysis cells suit- some mechanism to stabilize and sup-
able for this purpose. One can also con- port the specimen structure.
struct microdialysis cells from glass tubing • Biological specimens yield very low
similar to those originally described by contrast, as they are composed of light
Zeppenzauer for crystallization of soluble elements. Contrast is commonly
proteins (87). Convenient and inexpen- increased by adding some form of
sive microdialysis cells can also be con- heavy atom contrast agent (e.g., stain),
structed from a standard microcentrifuge but adequate contrast can be achieved
tube by cutting off the conical tube leav- on unstained specimens by adjusting
ing the tube cap and collar. In this case the focus of the objective lens (see
the compartment is formed by the tube below).
cap over which one lays a small piece of
dialysis tubing that is then sealed with the 3.1. Negative Staining
collar of the tube (Dr. Alok Mitra, per-
sonal communication). One of the most common techniques of

214
Structural Study of Heme Proteins by Electron Microscopy

specimen preparation is negative staining Freshly evaporated carbon films are gen-
first developed in the 1960s. Negative erally hydrophilic and readily adsorb pro-
staining works well with 2D membrane teins and lipid vesicles. As they are stored,
protein crystals, but the useful resolution however, they quickly become more
that can be achieved is limited to about 10 hydrophobic and then do not adsorb the
Å, and 20 Å is more common. The speci- specimen as well. This change is readily
men in an aqueous suspension of approxi- observed as one draws a liquid drop off of
mately 1 mg protein/mL is adsorbed onto the grid with a piece of filter paper. If the
a thin carbon film. Carbon films are gener- liquid draws off evenly leaving a thin film,
ated by evaporation of carbon onto a suit- the grid is hydrophilic, but if it is all
able surface, for example freshly cleaved rapidly drawn off leaving none behind,
mica, from which it can be floated off onto the grid is hydrophobic and will probably
a water surface and lowered onto the sur- not adsorb the specimen. One can over-
face of electron microscope grids (com- come this change to some extent by
monly 400 mesh copper grids) previously increasing the concentration of the speci-
placed on a support beneath the film. The men, but it is often necessary to render
carbon film can be lowered gently onto the the surface hydrophilic again by a process
grid surfaces by draining water out of the called glow discharge, in which the films
vessel. Alternatively, one can coat grids are exposed to the ions formed by an elec-
with a plastic film, generally Collodion, tric discharge in the residual gases in a
but Formvar is also used, after which a car- vacuum apparatus pumped down to sev-
bon film is evaporated onto the plastic eral tens of microns of pressure. A glow
film. These carbon–Collodion films can be discharge accessory is common in com-
used directly, but best results are usually mercial vacuum evaporators found in
obtained if the plastic is dissolved away electron microscopy laboratories, or an
with a solvent, since pure carbon films are inexpensive glow discharge apparatus can
thinner and more conductive giving less be constructed from a plastic vacuum
movement when irradiated with an elec- dessicator, a Tesla coil vacuum tester, and
tron beam. Ideally, one should use the an inexpensive rotary pump (3). Proce-
thinnest carbon film possible in order to dure 2 describes a typical method to pre-
minimize its contribution to the image, pare negatively stained specimens.
but very thin carbon films are fragile, and
400 mesh grids are too coarse to support ❖ Procedure 2. Preparation of
them adequately. In this case, one can first Negatively Stained Specimens
coat the grid with a holey film, a plastic
film containing numerous circular or 1. Allow a drop of the specimen to adsorb
ellipsoidal holes of varying sizes. There to a carbon-coated grid for 1 to 5 min-
are several methods by which holey plas- utes.
tic films can be formed (36,57,63), and 2. Wash the grid with several drops of
they are generally stabilized to electron water or buffer followed by several
irradiation by evaporation of carbon onto drops of a suitable negative stain, com-
them. Holey films make an excellent sup- monly 1% to 2% uranyl acetate, but
port for very thin carbon films and can uranyl formate and phosphotungstic
even be used to support thin layers of acid are also used. Uranyl stains gener-
stain over the holes without a carbon sup- ally provide higher resolution, but
port film. must be used at a pH of about 4.5,

215
T.G. Frey

which may disrupt the structures of 2. Rinse with water or volatile buffer to
some specimens. remove sample that is not adsorbed to
3. Draw off excess stain with a piece of fil- the surface.
ter paper leaving a very thin layer that 3. Freeze by plunging into a cryogen; liq-
dries down forming a glass-like elec- uid nitrogen is acceptable, but ethane
tron dense replica surrounding the or propane cooled in liquid nitrogen
specimen molecules supporting and freezes much more rapidly.
contrasting them. 4. Place the sample in a freeze frac-
In the electron microscope, the actual ture–etch instrument and remove ice by
protein structure appears light against a sublimation at approximately 70°C.
dark background formed by the stain, 5. Contrast the surface by evaporating a
hence the term “negative” stain. It is heavy atom, generally platinum evapo-
important to realize that negative staining rated with carbon, but tungsten–tanta-
contrasts the 3D surface of the molecule lum may produce smaller metal grains
with atoms that are approximately 7 Å in yielding higher resolution (12). This
diameter. Thus, the resolution in negative- creates a shadow effect that highlights
stained specimens is limited to approxi- the surface topography and can also be
mately 10 Å at best and more commonly used to measure the thickness of the
20 Å. Furthermore, the internal structure specimen.
of protein domains are not contrasted. This technique has been applied to con-
trast selectively the surface of vesicle crystals
3.2. Other Methods of cytochrome oxidase dimers indicating
that the enzyme protrudes 20 to 30 Å
Images of negatively stained specimens beyond the bilayer surface on the exterior
are a projection of the electron density of surface (corresponding to the matrix side of
the 3D stain replica onto the 2D image the inner mitochondrial membrane) (33)
plane. Ultimately, one would wish to cal- and to measure the thickness of a number
culate a 3D structure from multiple images of 2D crystals (37). Berriman, Leonard,
of tilted specimens (see below), but infor- and coworkers used shadowing to demon-
mation on the 3D configuration of the strate that crystals of the mitochondrial
molecule can often be quickly obtained by cytochrome bc1 complex thinned markedly
other common specimen preparation tech- during electron irradiation (5), and Smith
niques. Indeed, this information is often and Ivanov have published a procedure to
essential to confirm the molecular packing compute the surface relief structure from
of a new crystal form, information that is images of shadowed specimens (67). A
required in calculating the 3D structure. related technique, freeze fracture–replica-
One of the most useful techniques is heavy tion, can also be used to study the structure
metal shadowing of freeze-dried specimens of membrane protein crystals in order to
(Procedure 3). help determine the molecular packing (13).
Heuser has adapted the technique of rapid
❖ Procedure 3. Heavy Metal slam freezing and freeze fracture–etch to
Shadowing look at molecules adsorbed to a slurry of
small mica chips (44). Conventional plastic
1. Adsorb the specimen to a flat embedding and thin sectioning can also be
hydrophilic surface, such as a carbon used to evaluate the gross structure of a new
film or freshly cleaved mica. crystal preparation of a membrane protein

216
Structural Study of Heme Proteins by Electron Microscopy

and to help confirm the molecular packing since the resulting images would be of the
model (31,51,80). actual biological molecules rather than the
distribution of heavy atom stains around
3.3. Low Dose Electron Microscopy them. There are several problems in achiev-
ing this goal, however, beginning with the
The challenges of specimen preparation low contrast afforded by biological speci-
for electron microscopy are compounded mens and by their sensitivity to exposure to
by the sensitivity of biological specimens to high energy electrons. Through the use of
electron irradiation. The exposure required low dose techniques, one can record elec-
to record a single image at moderate reso- tron micrographs of unstained specimens
lution, approximately 1 nm, can be as high at electron doses low enough to minimize
as 100 to 300 electrons/Å2. However, mea- radiation damage. The disadvantage is that
surements of electron damage at much although these images may technically be
lower exposures paint a gloomy picture for high resolution, the signal-to-noise ratio
prospects of achieving even this modest
(S/N) is very low, often less than 1.0, and
resolution. An exposure of even 20 to 30
cannot be interpreted. The S/N can be
electrons/Å2 results in loss of 20% to 30%
increased by averaging many images of
of the mass of a typical biological specimen
identical structures such as the unit cells of
(20), exposure of less than 10 electrons/Å2
a crystal; statistically the S/N is increased
disrupts the higher order features of a pro-
tein crystal (38), and an electron dose of by a factor equal to the square root of the
approximately 0.5 electrons/Å2 is sufficient number of structures averaged. This can be
to inactivate enzymes (39). The primary a very powerful tool in the case of 2D crys-
function of stain is to increase the contrast tals, where a very small area might contain
of biological specimens so lower electron 100 unit cells giving an increase in the S/N
doses can be used to achieve useful images. of tenfold. A more typical situation would
Furthermore, heavy atom stains are more be a crystal containing several thousand
resistant to damage by electron irradiation. unit cells giving and an increase in S/N of
Nevertheless, studies in the 1970s demon- thirty- to fiftyfold. This was first demon-
strated the efficacy of recording electron strated by Unwin and Henderson with
micrographs using minimal electron expo- their images of unstained purple mem-
sure even for negatively stained specimens brane containing thousands of bacteri-
(77,83). Now, most electron microscopes orhodopsin molecules; these electron
allow one to focus and correct astigmatism micrographs appear featureless, but the
on an area of the specimen grid adjacent to averaged image was clearly interpreted at 7
the specimen and then record an image of Å resolution as resulting from the presence
the specimen, exposing it to only the elec- of transmembrane α-helices (78).
trons required to expose the photographic The remaining problem is how to pre-
film. This procedure is absolutely essential pare unstained specimens for the high vacu-
when recording images of unstained speci- um conditions in an electron microscope.
mens that are much more sensitive to elec- Unwin and Henderson dried their purple
tron irradiation than are stained specimens. membrane specimens in a thin layer of 1%
glucose in order to surround them with a
3.4. Unstained Specimens hydrophilic substance that could also sup-
port them structurally. This approach has
Ideally, one would like to record elec- worked very well with purple membrane
tron micrographs of unstained specimens, and with a number of other examples, but

217
T.G. Frey

has the disadvantage that glucose has a (Pleasanton, CA, USA) and by Oxford
density very similar to protein and thus Instruments (Concord, MA, USA) for the
actually reduces the already low contrast popular side entry electron microscopes.
rather than increasing it. This was accept- The techniques of cryoelectron microscopy
able in the case of bacteriorhodopsin mole- have been described in an excellent review
cules as nearly all of the protein lies within by Dubochet et al. (21), and I will only
the lipid bilayer surrounded by the lower summarize the important points here. The
density hydrocarbon tails of the lipid mol- key to this technique is to freeze the speci-
ecules. Cytochrome oxidase crystals, how- men very rapidly in a thin layer of water.
ever, project much of their structure With freezing velocities above approxi-
beyond the lipid bilayer surface, and these mately 10 000 degrees/second, the water is
portions of the structure are virtually invisi- transformed to vitreous ice, a noncrys-
ble above 10 Å resolution if the crystals are talline ice form that has a density and
embedded in glucose (17,18,42). Better structure similar to liquid water. It is very
results have been obtained with auroth- difficult to freeze a thick specimen this
ioglucose, a glucose derivative containing rapidly, but a thin layer of water clinging to
gold atoms, or with glucose mixed with an electron microscope grid can be readily
uranyl acetate (79). These mixtures provide frozen to vitreous ice by plunging it into a
low resolution contrast of the hydrophilic suitable cryogen such as liquid propane or
domains of membrane proteins while liquid ethane cooled by liquid nitrogen.
embedding the structure in a hydrophilic The specimen grid is held in a pair of fine
substance. Kuhlbrandt and others have tweezers clamped into a simple device for
obtained excellent results using tannic acid plunging, and Procedure 4 is followed (see
rather than glucose (54). Figure 43 in Reference 21).

3.5. Frozen Hydrated Specimens ❖ Procedure 4. Preparation of Frozen


Hydrated Specimens
The ideal method is to maintain an
aqueous environment around the crystal as 1. Apply 1 to 5 µL of the specimen sus-
is the case with 3D protein crystals studied pended in a suitable buffer at relatively
by X-ray diffraction. Although environ- high concentration, approximately 5 to
mental chambers have been constructed 20 mg/mL, to a grid with a hydrophilic
that maintain significant partial pressure of substrate, either continuous carbon
water around the specimen by differential film or holey film that has been recent-
pumping, these proved too unstable for ly glow discharged.
high resolution imaging. Taylor and
2. Blot the grid by pressing it firmly by
Glaeser demonstrated another approach,
hand between two layers of filter paper.
freezing the specimen in a thin layer of ice
and keeping it frozen at low temperature, 3. Plunge immediately into the cryogen;
-130°C or less, with a specially designed this step is facilitated if the plunge
cryoelectron microscope stage (72). The device has a foot pedal release.
early attempts, particularly by Dubochet’s 4. Transfer very quickly to liquid nitro-
group and Unwin’s group, provided very gen, quickly flicking off excess cryogen,
useful results but suffered from stage vibra- and store under liquid nitrogen until
tions that limited resolution. This spurred use.
efforts to construct more stable cryospeci- The result is a specimen embedded in a
men holders now marketed by Gatan material very similar to its native environ-
218
Structural Study of Heme Proteins by Electron Microscopy

ment at a very low temperature where from the image. This is analogous to
movement is inhibited. The specimen may absorption contrast in the light microscope
be adsorbed to a thin carbon film as for and contrasts relatively low resolution
negative staining, or it may be suspended details; and (ii) phase contrast is generated
over the holes of a holey film. The latter when the phases of electrons are retarded as
method has the advantage that the speci- they pass through the specimen. The phase
men is not in contact with a solid support of these electrons are further modified by
prior to freezing, but the holey film has a the objective lens, and the extent of this
significantly smaller area suitable to record phase shift depends upon the:
images. The specimen grid should be • Angle of diffraction.
frozen immediately after blotting, but the
thin film of water supporting the specimen • Spherical aberration of the objective
may still dry significantly if the relative lens.
humidity of the environment is low. To • Focus of the objective lens.
minimize drying, one can maintain higher Thus, it is possible to control the
humidity around the specimen by: amount of phase shift of the diffracted
• Blowing humidified air across it (21). electrons by changing the focus of the
objective lens, and with an appropriate
• Freeze in a cold room where humidity
level of underfocus, some of the diffracted
is high.
electrons can be further phase-shifted by
• Use a specially constructed freezing approximately 90°, generating appropriate
device that incorporates a humidity phase contrast when combined with undif-
chamber (64). fracted electrons at the image plane. But
Adrian et al. have adapted this proce- for each choice of underfocus, only elec-
dure to incorporate heavy atom salts in trons diffracted at particular angles are
the vitrified water layer in order to phase-shifted by 90°, generating proper
increase the contrast of the frozen speci- contrast. Electrons diffracted at other
mens (1). A very important benefit of cry- angles are phase-shifted by smaller amounts,
oelectron microscopy is the reduction of generating less contrast, or are phase-shift-
electron beam damage at low tempera- ed in the wrong direction, generating
ture. In measurements of loss of higher inverted contrast.
resolution information as a function of Diffraction angle correlates with resolu-
electron irradiation, specimens at -170°C tion, and electrons diffracted at higher
can be exposed to 5 to 10 times the num- angles contain higher resolution informa-
ber of electrons as those at room tempera- tion. The changes in the phase of electrons
ture (38). as a function of their diffraction angle is
The low contrast provided by the rela- described by the contrast transfer function
tively small density differences between vit- (CTF) (23,26). In order to visualize indi-
reous ice and protein can be enhanced by vidual macromolecules, one must adjust
appropriate choice of focus of the objective the objective lens to a relatively large
lens. In the brightfield mode of a transmis- underfocus, one micron or more. This gen-
sion electron microscope, contrast is gener- erates contrast of lower resolution, features
ated by two mechanisms: (i) amplitude making the molecules visible, but may
contrast is generated when electrons are introduce contrast reversals at high resolu-
scattered by the specimen at a wide enough tion. Figure 3c is an optical diffraction pat-
angle to cause them to be intercepted by tern (equivalent to a plot of Fourier trans-
the objective aperture subtracting them form intensities) of the cytochrome oxidase
219
T.G. Frey

crystal in Figure 3a, and the effects of the reversed contrast, are shown in the plot of
CTF can be seen in the concentric rings of the CTF displayed as an insert in Figure 3c
high background noise. The regions of the on the same scale as the as the diffraction
Fourier transform, where phases have been pattern. By definition, CTF values greater
shifted in the wrong direction generating than zero represent incorrect phase shifts,

Figure 3. (a) An electron micrograph of a frozen hydrated crystal of cytochrome oxidase dimers; one unit cell is outlined. (b) A
Fourier-filtered image with dramatically increased S/N calculated from 5 electron micrographs similar to panel a. One unit cell is
outlined with unit cell axes of a = 100 Å and b = 125 Å. (c) Optical diffraction pattern of the crystal in panel a; the optical dif-
fraction pattern is equivalent to a plot of the intensities of the Fourier transform. The reciprocal lattice vectors, a* and b*, are indi-
cated. The inset is a plot of the phase CTF, χ(α), for this defocus shown on the same scale, and the zeros in the CTF are indicat-
ed by horizontal lines showing regions of minimal contrast in the diffraction pattern.
220
Structural Study of Heme Proteins by Electron Microscopy

and the circled diffraction spots lie within stained specimens, one normally records
rings of the diffraction pattern that have several images at different tilt angles for
been phase-shifted in the wrong direction. each specimen, since these specimens are
In order to calculate a high resolution more resistant to radiation damage.
image of a biological specimen, one must Unstained specimens are much more sensi-
correct for the effects of the CTF. Since dif- tive to electron irradiation, and generally,
ferent values of underfocus optimally con- only one high resolution image is recorded
trast features at different levels of resolu- from each. Specimens are more stable to
tion (different diffraction angles), it is irradiation at low temperature (10,38), so
sometimes advantageous to record more it is possible to record more than one
than one image of the same specimen, the micrograph from a single specimen at low
first at lower defocus for high resolution temperature, particularly if the highest res-
information, and the second at greater olution is not required.
defocus for lower resolution information
(11,28,73). 3.7. Specific Labeling

3.6. Collecting Tilt Data Most studies of protein structure by elec-


tron crystallography do not yield reso-
Transmission electron micrographs are lution sufficient to construct an atomic
2D projections of the 3D electron density model, and methods to identify the loca-
of the specimen. While these projections tions of functionally important sites
reveal important information about the and/or components are needed in order to
structure of the specimen, much detail is exploit fully lower resolution structures.
lost when structural features are projected There are two general approaches to iden-
upon one another. If the specimen is tilted tify specific sites on low resolution struc-
and another micrograph recorded, a differ- tures: (i) specific labeling with molecules
ent set of features will be superimposed, visible by electron microscopy; and (ii)
providing new information about struc- comparison with structures lacking one or
ture. This is most readily seen if the two more components.
projections are viewed as a stereo pair. A Both of these approaches have been
full 3D reconstruction requires many applied with success to low resolution
images of the specimen tilted at different structures of heme proteins derived from
angles, greatly exceeding the number that electron microscopic data.
can be recorded from a single specimen The most common method of specific
without very significant radiation damage labeling in electron microscopy of biologi-
to unstained specimens (14,43). Thus, low cal structures is the use of specific antibody
dose images of many different but identi- molecules. Antibodies are most commonly
cal specimens must be recorded in order to used to determine the distribution proteins
sample the 3D Fourier transform (see sec- in cells and organelles, but can also be used
tion 3.6 below and Figure 4). All images to identify the position of antibody epi-
must be translated to bring them into topes on molecular structures. Frey et al.
alignment at a common origin, so it is usu- used subunit-specific antibodies to deter-
ally necessary to record images at a variety mine that the surface exposed in vesicle
of tilt angles and merge the data, beginning crystals of cytochrome oxidase dimers cor-
with images recorded with the smallest tilt responded to the matrix surface of the
and then adding images in order of increas- inner mitochondrial membrane, conclud-
ing tilt angle. In the case of negatively ing that the interior surface of the vesicles
221
T.G. Frey

corresponds to the surface exposed to the binding sites by electron microscopy in


intermembrane space (30). They subse- many other specimens (2,82).
quently prepared monovalent antibody In many cases, the protein being studied
fragments, Fabs, to label subunit IV on the binds another protein with sufficient affini-
surface of these crystals, concluding that ty and specificity that the protein ligand
this subunit lies 20 to 30 Å from the 2-fold can be used to label its binding site. This is
axis of the dimer and near the a crystal axis the case with cytochrome c, one of the
(32). Based upon the prediction of a trans- substrates for cytochrome c oxidase. Frey
membrane α-helix from residues 80 to 97, and Murray (35) incubated crystals of cyto-
the volume of the N-terminal domain of chrome oxidase monomers with cyto-
subunit IV could account for the 20 to 30 chrome c, which binds to cytochrome oxi-
Å structure projecting from the bilayer sur- dase with an affinity comparable to that of
face that was detected in freeze-dried and specific antibodies. After extensive image
shadowed specimens (34). Fab fragments processing, the site of cytochrome c binding
have also been used as bulky affinity labels to cytochrome oxidase monomers was
to identify their corresponding epitope deduced from difference images (Figure

Figure 4. Lattice lines of the 3D Fourier transform of a 2D crystal. The positions of the reflections in the diffraction pattern
of an untilted crystal are shown as black ellipses. In the 3D Fourier transform, these reflections extend perpendicular to the plane
of the crystal as shown by the lines of periodically varying intensity. The Fourier transform of an electron micrograph of a crystal
that has been tilted is a central section of the 3D Fourier transform that intercepts the lattice lines at the points shown by the X’s.

222
Structural Study of Heme Proteins by Electron Microscopy

5a), consistent with the cytochrome c bind- plex is to compare structures of the intact
ing site in the atomic structure, determined complex with structures of subcomplexes.
by X-ray diffraction and from biochemical This approach was used in the study of the
studies (see section 4.1 and Figure 5b). mitochondrial cytochrome bc1 complex.
Other labeling studies have taken a dif- Weiss, Leonard, and coworkers crystallized
ferent approach, using heavy atom cluster Neurospora mitochondrial cytochrome c
molecules that have been modified to react reductase (cytochrome bc1or Complex III)
selectively with certain functional groups by reconstituting it with purified lipids and
of proteins, generally reactive sulfhydryl adsorbing excess detergent with Bio-Beads.
groups of cysteine residues (29). A special This produced crystals of the type shown in
issue of The Journal of Structural Biology is Figure 1c, although these were generally
devoted to results from this approach using formed in the two layers of a collapsed vesi-
gold cluster compounds (1999, volume cle giving two overlapping crystalline layers
127, issue 2). Crum et al. used a mono- (56,84). Their low resolution 3D recon-
maleimide derivative of an undecagold struction of the intact complex is shown in
cluster compound to label specifically Cys- Figure 6b. Hovmoller et al. subsequently
115 of cytochrome oxidase subunit III in formed crystals of the purified subcomplex
crystals of cytochrome oxidase dimers. lacking two large “core” proteins, and com-
They then identified the binding site by parison of the 3D structure with that of the
low dose cryoelectron microscopy of speci- intact complex allowed them to identify the
mens embedded in glucose and uranyl functional components as shown in Figure
acetate (16). 6 (46,47). The core subunits, which proba-
A different approach to identify the vari- bly function in facilitating the assembly of
ous components of a macromolecular com- the complex, were later purified, and their

Figure 5. A comparison of the structure of cytochrome oxidase monomers determined by electron crystallography (a) and by
X-ray crystallography (b). (a) A low resolution structure in projection calculated from electron micrographs of frozen hydrated
crystals of cytochrome oxidase monomers. The dark peak outlined in white contour lines is the position of cytochrome c bind-
ing calculated from difference images. (b) A ribbon diagram produced from the atomic coordinants calculated from the high res-
olution X-ray structure and displayed by the program RasMol. The cytochrome c binding site is placed between Cys-115 of sub-
unit III and the acidic residues of subunit II as determined biochemically.

223
T.G. Frey

low resolution structure was determined lution information. The simplest method
from helical aggregates confirming the to screen micrographs for quality is optical
assignment in Figure 6 (48). The cyto- diffraction, since the diffraction pattern is
chrome b6f complex found in the thylakoid the Fourier transform of the object and is a
membrane of chloroplasts has a function in diagnostic of several important image char-
photosynthesis very similar to that of acteristics. Formation of an optical diffrac-
cytochrome bc1 in mitochondria, but lacks tion pattern is readily accomplished with
the core subunits. The low resolution pro- an optical diffractometer consisting of a
jection structure of the cytochrome b6f laser (generally a 1–5 mW He/Ne laser),
complex purified from Chlamydomonas beam expansion–spatial filter, and a single
reinhardtii was determined by electron crys- diffraction lens to collect the diverging
tallography of 2D crystals grown by recon- beam and focus it to a point at some dis-
stitution and found to be very similar to the tance (24,65). When an electron micro-
cytochrome bc1 subcomplex lacking the graph is placed in the optical path just after
core subunits (8). the diffraction lens, the focused diffraction
pattern of the illuminated portion of the
4. DATA PROCESSING micrograph can be observed at the focus of
the lens. The undiffracted beam appears as
4.1. Selecting Micrographs — Optical a bright spot in the center of the diffraction
Diffraction pattern and is surrounded by the diffrac-
tion pattern of the object image. In the
Although data collection by low dose case of crystalline objects, the diffraction
electron microscopy is a critical element in pattern consists of peaks of light at the
determining the structure of a membrane points of a 2D lattice whose spacings are
protein, data processing is equally impor- the reciprocal of the crystal lattice; thus,
tant. The first step is to identify which of the lattice in the diffraction image is called
the many micrographs recorded are suit- the “reciprocal lattice”. The structural
able for further processing; generally, only a information common to all unit cells with-
minority of micrographs contain high reso- in the illuminated area of the micrograph

Figure 6. A comparison of the structure of cytochrome c reductase (cytochrome bc1 complex) determined by electron crystal-
lography and by X-ray crystallography. (a) A drawing interpreting the positions of 5 subunits in the dimeric complex with respect
to the lipid bilayer in the center. Core subunits I and II face the matrix space. (b) Balsa wood models of the low resolution struc-
tures determined by electron microscopy–crystallography of: (i) a subcomplex lacking core subunits I and II on the left, and (ii) the
intact complex. (iii) A ribbon diagram based upon the atomic coordinants of all subunits determined by X-ray crystallography.

224
Structural Study of Heme Proteins by Electron Microscopy

is contained at the points of the diffraction circles in the diffraction pattern (Figure
pattern’s reciprocal lattice, while nonperi- 3c), and the radii of the circles can be
odic noise is distributed throughout the used to calculate accurately the focus of
diffraction pattern. Although one can the objective lens. If the objective lens
obtain equivalent information by digitizing has residual astigmatism, the focus is
the electron micrograph and calculating its different in orthogonal directions, and
diffraction pattern, an optical diffractome- the zeroes produce concentric ellipses
ter accomplishes this instantaneously, rather than circles.
allowing one to move the micrograph
around in the beam to select the best area 4.2. Digitizing
quickly. One generally looks for two crite-
ria revealed by the optical diffraction In order to process electron micrographs
pattern: by computer, they must first be converted
to digital form. Although it is now possible
1. Does the image contain high resolution to purchase sensitive high resolution digi-
information about the crystal structure? tal cameras for transmission electron
The diffraction pattern is the Fourier microscopes, film is still the best media on
transform of the object illuminated, which to record low dose high resolution
representing its structure in frequency images. The most accurate scanners have
space, with points furthest from the been mechanical, based upon wrapping the
center representing higher frequency micrograph around a rotating drum or on
components contributing higher reso- precise movement in two dimensions, and
lution information. One therefore looks these are generally quite expensive. More
for micrographs whose diffraction pat- recently, high resolution digital cameras
terns display diffraction spots on the based upon charge-coupled device (CCD)
reciprocal lattice that extend relatively technology have become available, and
far from the origin of the diffraction these represent a suitable lower cost alter-
pattern. Once the diffraction constant native to mechanical scanners for many
of a particular optical diffractometer is applications. Whatever the device used,
calibrated, usually with a diffraction there are several factors to consider in digi-
object or grating of known spacing, the tizing an image, and these have been cov-
resolution of the information from each ered in earlier publications (19,26). The
micrograph can be calculated based first is the resolution one requires or
upon the distance from the origin of expects in the digitized image. In digital
the furthest diffraction spot. sampling of a continuous function (analog
2. Is the micrograph properly focused signal), one must sample the function at an
with astigmatism corrected? Character- interval that is one half the interval or reso-
istics of the transfer of information lution one wants to obtain in the digitized
from the object to the image are image; this is called the Nyquist sampling
described by the CTF as described rate. Thus, if one requires 10 Å informa-
below. A plot of the CTF in diffraction tion in a digital image, it must be sampled
space shows that it periodically passes (digitized) at 5 Å or smaller intervals. In
through zero at points determined by order to obtain very high resolution infor-
the wavelength of the electron wave, mation from a low dose image, one must
the spherical aberration of the objective digitize the image of a large 2D crystal, at
lens, and by the focus of the objective very small intervals, producing a very large
lens. The zeroes appear as concentric data file. Electron micrographs of 2D crys-
225
T.G. Frey

tals are typically recorded at about electron microscopy and computational


40 000×, and 1 Å spacing in the specimen averaging improved, leading to improve-
corresponds to 4 µm on the film. Thus, in ments in image resolution, it became
order to record 10 Å information in the apparent that disorder in membrane pro-
digitized electron micrograph, one would tein crystals was limiting the resolution
have to sample it at 20-µm (5 Å) intervals, that could be achieved after averaging the
and for 4 Å resolution, the sampling would unit cells contained within an image. This
have to be at 8-µm intervals. In practice, problem was first recognized by Crowther
one actually samples more finely than and Sleytr who developed the first comput-
strictly required by the Nyquist limit, since er programs to attempt to correct for crys-
there is some fall off in the transfer of high talline disorder (15). Henderson et al. later
resolution information that depends on the developed a method and software to cor-
sampling aperture size. rect the lattice disorder in images of very
large 2D protein crystals in order to
4.3. Fourier Filtering improve the S/Ns of their images at higher
resolution, and their method is described
One of the principal goals of image pro- in Procedure 5 (41). Although this may
cessing applied to electron micrographs of seem like a laborious process, the improve-
2D crystals is to extract an image with high ments in data can be dramatic, and most
S/N from a micrograph with a very low steps of the procedure are automated.
S/N; this is accomplished by signal averag- A similar approach to this problem
ing. The Fourier transform of a perfect 2D developed out of efforts that began in
crystal of infinite extent would be nonzero Joachim Frank’s group to create software
only at the points of a 2D reciprocal lattice, tools to align electron microscope images
but as seen in Figure 3, the Fourier trans- of individual particles using correlation
form of an actual image of a 2D protein methods. The single particle correlation
crystal contains nonperiodic noise, and the methods can also be used to align indi-
peaks at the points of the reciprocal lattice vidual unit cells if the S/N is high
are spread out somewhat as the crystal is enough to permit accurate alignment, or
finite and not perfectly ordered. If the it can be applied to patches of unit cells
image is reconstructed using only the in the case of low S/N images of
information lying at the reciprocal lattice unstained specimens. Once the patches
points of the Fourier transform, the result of unit cells are aligned, they can be aver-
is the average of all of the unit cells within aged to increase dramatically the S/N of
the digitized image, and the S/N is the resulting images (27). The single par-
increased by the square root of the number ticle averaging software can also be used for
of unit cells averaged. This is shown in Fig- structural study of molecules that are not
ure 3b compared with the original image in crystalline and have yielded dramatic
Figure 3a. results when applied to images of individ-
ual ribosomes and ribosomal subunits (58).
4.4. Correlation Alignment
❖ Procedure 5. Resolution of the
The resolution of the averaged image Electron Micrograph
depends upon the inherent resolution of
the original electron micrograph (defined 1. Apply a mask to the Fourier transform
by the CTF) and upon the order of the that passes only the information that
crystal being averaged. As techniques of lies within a specified radius of each
226
Structural Study of Heme Proteins by Electron Microscopy

reciprocal lattice point. Å or greater). Correction of the CTF is not


2. Calculate the inverse Fourier transform absolutely required if all of the information
of the “masked” transform to produce a contained in a micrograph lies within the
“coarsely” filtered image in which each first zero of the CTF; this is the region
unit cell is averaged with its nearest around the origin of the Fourier transform
neighbors. and within the first ring of low noise where
3. Select a reference image from the coarse- the CTF goes through zero on the inset
ly filtered image and calculate the cross- graph. However, if one wishes to obtain an
correlation function of this reference accurate representation of the object, even
image with the entire filtered image. at low resolution, the amplitudes of the
Fourier transform must be increased by
Identify the positions of each unit cell varying amounts to compensate for the fact
by searching for the peaks in the crosscor- that the CTF is not -1.0 across this fre-
relation function. quency spectrum. The most significant
Reinterpolate the sampling of the origi- correction is for those bands of the fre-
nal image based upon the positions of all of quency spectrum of the Fourier transform
the unit cells identified in the crosscorrela- where the CTF has caused a phase shift of
tion function in order to remove crystal lat- 180°. In Figure 3c, the circled lattice points
tice disorder. contain information about the crystal
structure whose phases have been shifted
4.5. Correcting the CTF by 180°, and these will contribute incorrect
information to the image unless they are
The performance of the objective lens of corrected. Once the defocus and residual
an electron microscope is defined by the astigmatism of a particular micrograph has
CTF, which is the Fourier transform of the been defined, the CTF can be calculated,
point-spread-function that describes how a and the phase changes are easily corrected.
point on the object (specimen) appears in Correction of the amplitudes is more com-
the image (23,26). Figure 3c shows the plicated because: (i) the amount by which
effect of the CTF in modulating the inten- amplitudes must be increased can be diffi-
sities of the Fourier transform (displayed in cult to determine, since one must include
the optical diffraction pattern) of the image contributions from amplitude contrast;
in Figure 3a, and the inset graph shows the and (ii) regions of the Fourier transform
phase contrast component of the CTF for near the zeroes of the CTF require very
this defocus, demonstrating that it causes large corrections, which can greatly magni-
periodic phase reversals (phase shifts of fy the contribution of noise in the image.
180°) within concentric bands of spatial The proper methods for making this cor-
frequencies rection are beyond the scope of this article,
Note: When the CTF is plotted in this but more detailed information can be
manner, correct transfer of contrast is indi- found in the literature (73,89).
cated when sin c(a) = -1.
Not shown in the inset is the effect of 4.6. 3D Reconstruction
amplitude contrast generated when elec-
trons are scattered into the objective lens The ultimate goal is to calculate a 3D
aperture removing them from the image; structure of the protein under study. Elec-
amplitude contrast is important at low spa- tron micrographs are 2D projections of the
tial frequencies contributing contrast to 3D electron density. Most people are intu-
low resolution features (approximately 50 itively aware that one can gain a better
227
T.G. Frey

knowledge of a complex 3D object’s struc- where m equals the number of views, D is


ture by viewing it from several angles. the particle diameter, and d is the desired
This intuitive approach is quantitatively resolution (14).
achieved by a number 3D reconstruction Fourier transforms of 2D crystals have
algorithms that make use of 2D projec- a special property that renders the use of
tions along different directions. In the Fourier transforms computationally effi-
case of 2D crystals, the most common of cient; they are sampled on a 2D lattice
these algorithms make use of a property of parallel to the plane of the crystal and are
Fourier transforms described by the cen- continuous along “lattice lines” perpen-
tral section theorem; this states that the dicular to the crystal plane at the points
Fourier transform of a 2D projection of a of the 2D reciprocal lattice as shown in
3D object is a central section (a section Figure 4. For example, in the Fourier
that passes through the origin) of the 3D transform of the cytochrome oxidase
Fourier transform of the object. Thus, as crystal in Figure 3, the information about
one collects 2D projections along differ- the crystalline structure is contained at
ent angles, one can fill in the 3D Fourier the points of the reciprocal lattice defined
transform and estimate its value at a reso- by the lattice vectors a* and b*, while
lution limited by: nonperiodic noise is distributed over the
1. The number of 2D projections and the entire transform. If one could view the
angle between them. This is so because 3D Fourier transform, one would see that
higher resolution information is con- the information intersected by this cen-
tained further from the origin of the tral section varies continuously along
Fourier transform where the 2D cen- lines, lattice lines, parallel to one another,
tral sections are further apart; eventual- and perpendicular to the plane of the
ly they diverge enough that the values transform in Figure 3c as diagrammed in
of the 3D Fourier transform can no Figure 4. The Fourier transforms of
longer be estimated from the values on images of tilted crystals sample these lat-
the 2D sections. tice lines at the positions where the cen-
2. The size of the object. The reason for tral section intersects them as shown in
this limitation is more subtle and arises Figure 4, and collecting the information
from the fact that larger objects have required for a complete 3D reconstruc-
Fourier transforms that vary more tion of the crystal requires collecting
rapidly than smaller objects with the central sections at different tilt angles in
same level of detail. Thus, the 2D sec- order to sample these lattice lines finely
tions for larger objects must be more enough to estimate their value continu-
closely spaced to achieve comparable ously out to the desired resolution. The
resolution compared to smaller objects. higher the resolution and the larger the
Another way of viewing this is to rec- unit cell, the more projections required
ognize that defining the structure of a and the finer the angular intervals
larger object requires more data (e.g., between them. Once the lattice lines
more projections). have been measured from central sec-
The semiquantitative relationship between tions, they can be sampled at appropriate
resolution, object size, and number of tilts was regular intervals, and the 3D structure
expressed by Crowther et al. in the equation: calculated by an inverse 3D Fourier
m ≅ πD/d transformation (4).

228
Structural Study of Heme Proteins by Electron Microscopy

5. COMPARISON WITH Although in projection, the dimers


STRUCTURES FROM X-RAY observed in 2D crystals by cryoelectron
DIFFRACTION microscopy appear very similar to those
generated from the X-ray coordinants
5.1. Cytochrome c Oxidase (obtained from the Protein Data Bank and
displayed with the program RasMol; see
When the first atomic structure of a Figure 7) (75), comparison of their sizes
eukaryotic cytochrome oxidase determined indicates that they must be different struc-
by X-ray crystallography was published in tures. The maximum dimension parallel to
1995 (74), its structure had previously been the membrane of dimers in the 2D crystals
determined in 2D projection at approxi- is approximately 100 Å, the length of the a
mately 8 to 10 Å resolution (79) and in 3 crystal axis along which the molecules are
dimensions at approximately 15 Å resolu- aligned. The X-ray model, on the other
tion (35). This is too low a resolution to hand, has a maximum dimension of
discern subunit boundaries, let alone trace approximately 150 Å. Thus, it appears that
the polypeptide chains, but a number of the dimer in 2D crystals must be a more
structural features had been deduced by compact structure with the individual
specimen preparation to contrast different monomers more closely aligned than the
domains selectively, by various labeling crystallographic dimers in the 3D crystals
experiments, and by comparing the struc- used to determine the structure by X-ray
tures of both 2D crystal forms, the diffraction. This is also indicated by the
monomer form, and the dimer form. The fact that the dimer in 2D crystals has its
transmembrane α-helices predicted by highest concentration of mass around the
hydropathy plots based on amino acid 2-fold axis, while the dimer in the 3D crys-
sequences proved to be fairly accurate, and tals is less densely packed in this region.
according to the X-ray model, they separate The size and shape of a cytochrome oxidase
the molecule into two hydrophilic domains monomer observed in 2D crystals com-
that protrude 35 to 40 Å beyond the lipid pares more favorably with the X-ray struc-
bilayer into the intermembrane space and ture (Figure 5). At 95 x 53 Å, the structure
into the matrix space of mitochondria. This determined by electron microscopy (35) is
is in contrast to the marked asymmetric somewhat smaller than the 110 x 63 Å
distribution of protein mass, 60 Å into the structure measured from X-ray coordi-
intermembrane space and less than 10 Å nants, but this difference can be explained
into the matrix space, proposed from 3D by the ambiguity in determining the mole-
reconstructions by electron crystallography cular boundary in a 2D projection.
(18,42,79). It is difficult to reconcile these The sites identified by electron micro-
and accept the X-ray model as being more scopy following specific labeling with Fabs,
accurate. On should note, however, that cytochrome c, and a monomaleimide
Frey et al. correctly determined that the undecagold cluster are, for the most part,
matrix side domain projected 20 to 30 Å confirmed by the atomic structure deter-
based upon the lengths of shadows cast in mined by X-ray crystallography. As shown
specimens that had been freeze-dried and in Figure 5, the cytochrome c binding site
shadowed with platinum–carbon (32–34). in images of cytochrome oxidase monomers
This highlights the importance of using is in essentially the same position as the
different specimen preparation techniques binding site in the X-ray structure deduced
in studying complex biological structures from the positions of Cys-115 of subunit
by electron microscopy. III and the acidic residues of subunit II that
229
T.G. Frey

have been shown to bind to opposite sur- and the maleimide group bound to the
faces of cytochrome c in the active site sulfhydryl of Cys-115.
(9,25). Although the position of Cys-115
of subunit III in the X-ray structure 5.2. Cytochrome c Reductase
appears to be quite distant from the peak
identified for the undecagold cluster com- The low resolution structure of the
pound bound to Cys-115 in the low reso- Neurospora cytochrome c reductase (cyto-
lution structure determined by electron chrome bc1 complex) determined by elec-
crystallography (16), one must remember tron microscopy is very similar the atomic
that the dimer observed in 2D crystals by structure of the beef heart mitochondrial
electron microscopy is much more com- enzyme determined by X-ray diffraction
pact than the dimer found in 3D crystals (49,85,88). As shown in Figure 6, the
by X-ray diffraction. In order to compare structure calculated from electron micro-
these 2 structures, the monomers in the X- graphs of 2D crystals displays an asymmet-
ray structure must each be moved approxi- ric distribution of mass protruding 30 Å on
mately 25 Å towards one another placing one side of the bilayer and 70 Å on the
Cys-115 of subunit III within the 15 Å other with a 50 Å domain within the lipid
length of the link between the undecagold bilayer (56,81). The smaller domain pro-
cluster observed by electron microscopy truding 30 Å was identified as the

Figure 7. A comparison of the structure of a cytochrome oxidase dimer determined by (a) electron crystallography at 15 Å res-
olution and (b) a ribbon diagram based upon the atomic structure determined by X-ray crystallography. The position of sub-
unit IV in the electron microscopy structure was determined by labeling with anti-IV Fabs. The structures are on the same scale.

230
Structural Study of Heme Proteins by Electron Microscopy

hydrophilic subunits of cytochrome c1 and examples of heme proteins described here


the Rieske iron sulfur protein (subunits IV have yielded only low resolution structures,
and V in Figure 7), and the larger domain however, and the complete structures were
as the core subunits I and II based upon obtained by X-ray crystallography of 3D
the structure of a subcomplex lacking sub- crystals. The failure of electron crystallog-
units I and II (47,52). These assignments raphy to produce high resolution structures
are confirmed in the X-ray structure in of these enzymes may be explained in part
which the cytochrome c1 and Rieske iron by their large sizes. Cytochrome c oxidase
sulfur domains extend 30 Å beyond the has a molecular weight of 200 000
bilayer and the core subunits 70 Å beyond. (400 000 for the dimer form), and cyto-
The dimensions of the cytochrome c chrome c reductase has a molecular weight
reductase dimer are also similar: 120 x 75 of 250 000 (500 000 for the dimer), and
Å in the electron microscopy structure ver- both contain many different polypeptide
sus 143 x 102 Å in the X-ray structure. subunits. Nevertheless, low resolution
Here, the somewhat smaller structure models calculated from electron micro-
determined by electron microscopy can be graphs provided early insight into the
attributed to: (i) shrinkage when the 2D structures of these critically important
crystals are prepared for electron micro- enzymes. Identification of functional sites
scopy by negative staining; and/or (ii) the and domains by specific labeling and by
fact that the structure determined by elec- crystallization of subcomplexes also proved
tron microscopy is of the Neurospora valuable in elucidating the structures of
enzyme and that determined by X-ray dif- these enzymes and in explaining some
fraction is of the beef heart enzyme. The aspects of their function. Furthermore, the
structure of cytochrome b6f in projection technique of electron crystallography has
(8) is very similar to the structure of the been proven to be capable of determining
cytochrome bc1 subcomplex calculated the atomic structures of a number of pro-
from atomic coordinants, but a 3D struc- teins and will surely prove useful in eluci-
ture of the cytochrome b6f complex is not dating the structures of other heme-con-
yet available. taining membrane proteins.

ABBREVIATIONS REFERENCES

1.Adrian, M., J. Dubochet, S.D. Fuller, and J.R. Har-


cmc, critical micelle concentration; ris. 1998. Cryo-negative staining. Micron 29:145-160.
CCD, charge-coupled device; CTF, con- 2.Aebi, U., B. Heggeler, L. Onorato, J. Kistler, and
trast transfer function; S/N, signal to noise M.K. Showe. 1977. New method for localizing pro-
teins in periodic structures: Fab fragment labeling
ratio; 2D, two-dimensional; 3D, three- combined with image processing of electron micro-
dimensional. graphs. Proc. Natl. Acad. Sci. USA 74:5514-5518.
3.Aebi, U. and T.D. Pollard. 1987. A glow discharge
unit to render electron microscope grids and other
surfaces hydrophilic. J. Electron Microsc. Tech.
6. SUMMARY 7:29-33.
4.Amos, L.a., R. Henderson, and P.N.T. Unwin. 1982.
Three-dimensional structure determination by electron
Determination of the structures of microscopy of two-dimensional crystals. Prog. Bio-
membrane proteins by electron microscopy phys. Mol. Biol. 39:183-231.
of 2D crystals has proven to be a very pow- 5.Berriman, J., R.K. Bryan, R. Freeman, and K.R.
Leonard. 1984. Methods for specimen thickness deter-
erful method that, in the best cases, can mination in electron microscopy. Ultramicroscopy
yield atomic models of these proteins. The 13:351-364.

231
T.G. Frey

6.Bhairi, S.M. 1997. Detergents: A Guide to the Proper- 23.Erickson, H.P. and A. Klug. 1971. Measurement and
ties and Uses of Detergents in Biological Systems. Cal- compensation of defocusing and aberrations by Fouri-
biochem-Novabiochem Corporation, San Diego. er processing of electron micrographs. Phil. Trans. R.
7.Bozzola, J.J. and L.D. Russell. 1999. Electron Soc. Lond. B 261:105-118.
Microscopy. Principles and Techniques for Biologists. 24.Erickson, H.P., W.A. Voter, and K. Leonard. 1978.
Jones and Bartlett, Boston. Image reconstruction in electron microscopy: enhance-
8.Bron, P., J.J. Lacapere, C. Breyton, and G. Mosser. ment of periodic structure by optical filtering. Meth-
1999. The 9 Å projection structure of cytochrome b6f ods Enzymol. 49:39-63.
complex determined by electron crystallography. J. 25.Ferguson-Miller, S., D.L. Brautigan, and E. Margo-
Mol. Biol. 287:117-126. liash. 1978. Definition of cytochrome c binding
9.Capaldi, R.A., V. Darley-Usmar, S. Fuller, and F. Mil- domains by chemical modification. III. Kinetics of
lett. 1982. Structural and functional features of the reaction of carboxydinitrophenyl cytochromes c with
interaction of cytochrome c with complex III and cytochrome c oxidase. J. Biol. Chem. 253:149-159.
cytochrome c oxidase. FEBS Lett. 138:1-7. 26.Frank, J. 1973. Computer processing of electron
10.Chiu, W. and T.W. Jeng. 1982. Electron radiation sen- micrographs, p. 215-274. In J.K. Koehler (Ed.),
sitivity of protein crystals. Ultramicroscopy 10:63-69. Advanced Techniques in Biological Electron Microscopy.
11.Conway, J.F. and A.C. Steven. 1999. Methods for Springer-Verlag, Berlin.
reconstructing density maps of “single” particles from 27.Frank, J., W. Chiu, and L. Degn. 1988. The charac-
cryoelectron micrographs to subnanometer resolution. terization of structural variations within a crystal field.
J. Struct. Biol. 128:106-118. Ultramicroscopy 26:345-360.
12.Costello, M.J., J. Escaig, K. Matsushita, P.V. Viitanen, 28.Frank, J. and P. Penczek. 1995. On the correction of
D.R. Menick, and H.R. Kaback. 1987. Purified lac the contrast function in biological electron microscopy.
permease and cytochrome o oxidase are functional as Optik 98:125-129.
monomers. J. Biol. Chem. 262:17072-17082. 29.Frey, P.A. and T.G. Frey. 1999. Synthesis of
13.Costello, M.J. and T.G. Frey. 1982. Membranous undecagold labeling compounds and their applications
cytochrome c oxidase. A freeze-fracture electron micro- in electron microscopic analysis of multiprotein com-
scopic analysis. J. Mol. Biol. 162:131-156. plexes. J. Struct. Biol. 127:94-100.
14.Crowther, R.A., D.J. De Rosier, and A. Klug. 1970. 30.Frey, T.G., S.H. Chan, and G. Schatz. 1978. Structure
The reconstruction of a three-dimensional structure and orientation of cytochrome c oxidase in crystalline
from projections and its application to electron membranes. Studies by electron microscopy and by
microscopy. Proc. R. Soc. A 317:319. labeling with subunit-specific antibodies. J. Biol.
15.Crowther, R.A. and U.B. Sleytr. 1977. An analysis of Chem. 253:4389-4395.
the fine structure of the surface layers from two strains 31.Frey, T.G. and T. Chang. 1986. The structure of mem-
of Clostridia, including correction for distorted images. brane bound cytochrome c oxidase. Ann. NY Acad.
J. Ultrastruct. Res. 58:41-49. Sci. 483:120-130.
16.Crum, J., K.J. Gruys, and T.G. Frey. 1994. Electron 32.Frey, T.G., M.J. Costello, and S.H. Chan. 1984.
microscopy of cytochrome c oxidase crystals: labeling Selective contrast in electron microscopy of crystalline
of subunit III with a monomaleimide undecagold clus- cytochrome oxidase. Ultramicroscopy 13:85-91.
ter compound. Biochemistry 33:13719-13726. 33.Frey, T.G., M.J. Costello, B. Karlsson, J.C. Hasel-
17.Deatherage, J.F., R. Henderson, and R.A. Capaldi. grove, and J.S. Leigh. 1982. Structure of the
1982. Relationship between membrane and cytoplas- cytochrome c oxidase dimer. Electron microscopy of
mic domains in cytochrome c oxidase by electron two-dimensional crystals. J. Mol. Biol. 162:113-
microscopy in media of different density. J. Mol. Biol. 130.
158:501-514. 34.Frey, T.G., L.A. Kuhn, J.S. Leigh, Jr., M.J. Costello,
18.Deatherage, J.F., R. Henderson and R.A. Capaldi. and S.H. Chan. 1985. Cytochrome oxidase: structural
1982. Three-dimensional structures of cytochrome c insights from electron microscopy and from secondary
oxidase vesicle crystals in negative stain. J. Mol. Biol. structure prediction. J. Inorg. Biochem. 23:155-162.
158:487-499. 35.Frey, T.G. and J.M. Murray. 1994. Electron
19.De Rosier, D.J. and P.B. Moore. 1970. Reconstruction microscopy of cytochrome c oxidase crystals.
of three-dimensional images from electron micro- Monomer-dimer relationship and cytochrome c bind-
graphs of structures with helical symmetry. J. Mol. ing site. J. Mol. Biol. 237:275-297.
Biol. 52:355-369. 36.Fukami, A. and K. Adachi. 1965. A new method of
20.Dubochet, J. 1975. Carbon loss during irradiation of preparation of a self-perforated micro plastic grid and
T4 bacteriophages and E. coli bacteria in electron its application. J. Electron Microsc. 14:112-118.
microscopes. J. Ultrastruct. Res. 52:276-288. 37.Fuller, S.D., R.A. Capaldi, and R. Henderson. 1979.
21.Dubochet, J., M. Adrian, J.J. Chang, J.C. Homo, J. Structure of cytochrome c oxidase in deoxycholate-
Lepault, A.W. McDowall, and P. Schultz. 1988. Cryo- derived two-dimensional crystals. J. Mol. Biol.
electron microscopy of vitrified specimens. Q. Rev. 134:305-327.
Biophys. 21:129-228. 38.Glaeser, R.M. and K.A. Taylor. 1978. Radiation dam-
22.Dykstra, M.J. 1992. Biological Electron Microscopy. age relative to transmission electron microscopy of bio-
Theory, Techniques, and Troubleshooting. Plenum logical specimens at low temperature: a review. J.
Press, New York. Microsc. 112:127-138.

232
Structural Study of Heme Proteins by Electron Microscopy

39.Hahn, M., J. Seredynski, and W. Baumeister. 1976. 55.Kühlbrandt, W. 1992. Two-dimensional crystalliza-
Inactivation of catalase monolayers by irradiation with tion of membrane proteins. Q. Rev. Biophys. 25:1-49.
100 keV electrons. Proc. Natl. Acad. Sci. USA 73:823- 56.Leonard, K., P. Wingfield, T. Arad, and H. Weiss. 1981.
827. Three-dimensional structure of ubiquinol:cytochrome c
40.Hayat, M.A. 1986. Transmission Electron Microscopy. reductase from Neurospora mitochondria determined by
Academic Press, Orlando. electron microscopy of membrane crystals. J. Mol. Biol.
41.Henderson, R., J.M. Baldwin, K.H. Downing, J. Lep- 149:259-274.
ault, and F. Zemlin. 1986. Structure of purple mem- 57.Lunsdorf, H. and E. Spiess. 1986. A rapid method of
brane from halobacterium halobium: recording, mea- preparing perforated supporting foils for the thin car-
surement and evaluation of electron micrographs at 3.5 bon films used in high resolution transmission electron
A resolution. Ultramicroscopy 19:147-178. microscopy. J. Microsc. 144:211-213.
42.Henderson, R., R.A. Capaldi, and J.S. Leigh. 1977. 58.Malhotra, A., P. Penczek, R.K. Agrawal, I.S.
Arrangement of cytochrome oxidase molecules in two- Gabashvili, R.A. Grassucci, R. Junemann, N.
dimensional vesicle crystals. J. Mol. Biol. 112:631-648. Burkhardt, K.H. Nierhaus, and J. Frank. 1998.
43.Henderson, R. and P.N. Unwin. 1975. Three-dimen- Escherichia coli 70 S ribosome at 15 A resolution by
sional model of purple membrane obtained by electron cryo-electron microscopy: localization of fMet-
microscopy. Nature 257:28-32. tRNAfMet and fitting of L1 protein. J. Mol. Biol.
44.Heuser, J. 1989. Protocol for 3-D visualization of mol- 280:103-116.
ecules on mica via the quick- freeze, deep-etch tech- 59.Mannella, C.A. 1984. Phospholipase-induced crystal-
nique. J. Electron Microsc. Tech. 13:244-263. lization of channels in mitochondrial outer mem-
45.Holser, W.T.Z. 1958. Point groups and plane groups branes. Science 224:165-166.
in a two-sided plane and their subgroups. Z. Kristallo- 60.Michel, H. 1991. Crystallization of Membrane Pro-
gr. 110:266-281. teins. CRC Press, Boca Raton.
46.Hovmoller, S., K. Leonard, and H. Weiss. 1981. 61.Miyazawa, A., Y. Fujiyoshi, M. Stowell, and N.
Membrane crystals of a subunit complex of mitochon- Unwin. 1999. Nicotinic acetylcholine receptor at 4.6
drial cytochrome reductase containing the cyto- A resolution: transverse tunnels in the channel wall. J.
chromes b and c1. FEBS Lett. 123:118-122. Mol. Biol. 288:765-786.
47.Hovmoller, S., M. Slaughter, J. Berriman, B. Karls- 62.Mohraz, M. and P.R. Smith. 1984. Structure of
son, H. Weiss, and K. Leonard. 1983. Structural (Na+,K+)-ATPase as revealed by electron microscopy
studies of cytochrome reductase. Improved membrane and image processing. J. Cell Biol. 98:1836-1841.
crystals of the enzyme complex and crystallization of a 63.Murray, J.M. and R. Ward. 1987. Preparation of holey
subcomplex. J. Mol. Biol. 165:401-406. carbon films suitable for cryo-electron microscopy. J.
48.Hutchinson, E.G., W. Tichelaar, G. Hofhaus, H. Elec. Microsc. Tech. 5:285-290.
Weiss, and K.R. Leonard. 1989. Identification and 64.Murray, J.M. and R. Ward. 1987. Principles for the
electron microscopic analysis of a chaperonin oligomer construction and operation of a device for rapidly
from Neurospora crassa mitochondria. EMBO J. freezing suspensions for cryo-electron microscopy. J.
8:1485-1490. Electron Microsc. Tech. 5:279-284.
49.Iwata, S., J.W. Lee, K. Okada, J.K. Lee, M. Iwata, B. 65.Salmon, E.D. and D. DeRosier. 1981. A surveying
Rasmussen, T.A. Link, S. Ramaswamy, and B.K. Jap. optical diffractometer. J. Microsc. 123:239-247.
1998. Complete structure of the 11-subunit bovine 66.Seki, S., H. Hayashi, and T. Oda. 1970. Studies on
mitochondrial cytochrome bc1 complex (see com- cytochrome oxidase. I. Fine structure of cytochrome
ments]. Science 281:64-71. oxidase-rich submitochondrial membrane. Arch.
50.Jap, B.K., M. Zulauf, T. Scheybani, A. Hefti, W. Biochem. Biophys. 138:110-121.
Baumeister, and U. Aebi. 1992. 2D crystallization: 67.Smith, P.R. and I.E. Ivanov. 1980. Surface reliefs
from art to science. Ultramicroscopy 46:45-84. computed from micrographs of isolated heavy metal
51.Jayaraman, U., T. Chang, T.G. Frey, and J.K. Blasie. shadowed particles. J. Ultrastruct. Res. 71:25-36.
1987. Electron density profile of two-dimensionally 68.Sosinsky, G.E. and G.A. Perkins. 2000. Electron
crystalline membranous cytochrome c oxidase at low crystallographic methods for investigating gap junc-
resolution. Biophys. J. 51:475-486. tion structure [In Process Citation]. Methods 20:140-
52.Karlsson, B., S. Hovmoller, H. Weiss, and K. 155.
Leonard. 1983. Structural studies of cytochrome 69.Stokes, D.L. and N.M. Green. 1990. Three-dimen-
reductase. Subunit topography determined by electron sional crystals of CaATPase from sarcoplasmic reticu-
microscopy of membrane crystals of a subcomplex. J. lum. Symmetry and molecular packing. Biophys. J.
Mol. Biol. 165:287-302. 57:1-14.
53.Kim, C.H., M. Radermacher, M. Kessel, J. Frank, 70.Suarez, M.D., A. Revzin, R. Narlock, E.S. Kempner,
and T.E. King. 1985. Three-dimensional reconstruc- D.A. Thompson, and S. Ferguson-Miller. 1984. The
tion of cytochrome oxidase vesicle crystals prepared by functional and physical form of mammalian
cholate solubilization. J. Inorg. Biochem. 23:163-169. cytochrome c oxidase determined by gel filtration,
54.Kühlbrandt, W. 1988. Structure of light-harvesting radiation inactivation, and sedimentation equilibrium
chlorophyll a/b protein complex from plant photosyn- analysis. J. Biol. Chem. 259:13791-13799.
thetic membranes at 7 A resolution in projection. J. 71.Taylor, K.A., L. Dux, S. Varga, H.P. Ting-Beall, and
Mol. Biol. 202:849-864. A. Martonosi. 1988. Analysis of two-dimensional crys-

233
T.G. Frey

tals of Ca2+-ATPase in sarcoplasmic reticulum. Meth- cytochrome-c reductase from Neurospora mitochondria
ods Enzymol. 157:271-289. and structure analysis by electron microscopy. Methods
72.Taylor, K.A. and R.M. Glaeser. 1976. Electron Enzymol. 126:191-201.
microscopy of frozen hydrated biological specimens. J. 82.Wikoff, W.R., G. Wang, C.R. Parrish, R.H. Cheng,
Ultrastruct. Res. 55:448-456. M.L. Strassheim, T.S. Baker, and M.G. Rossmann.
73.Toyoshima, C. and N. Unwin. 1988. Contrast trans- 1994. The structure of a neutralized virus: canine par-
fer for frozen-hydrated specimens: determination from vovirus complexed with neutralizing antibody frag-
pairs of defocused images. Ultramicroscopy 25:279- ment. Structure 2:595-607.
291. 83.Williams, R.C. and H.W. Fisher. 1970. Electron
74.Tsukihara, T., H. Aoyama, E. Yamashita, T. Tomiza- microscopy of Tobacco Mosaic virus under condi-
ki, H. Yamaguchi, K. Shinzawa-Itoh, R. Nakashima, tions of minimal beam exposure. J. Mol. Biol. 52:
R. Yaono, and S. Yoshikawa. 1995. Structures of metal 121-123.
sites of oxidized bovine heart cytochrome c oxidase at 84.Wingfield, P., T. Arad, K. Leonard, and H. Weiss.
2.8 A [see comments]. Science 269:1069-1074. 1979. Membrane crystals of ubiquinone:cytochrome c
75.Tsukihara, T., H. Aoyama, E. Yamashita, T. Tomiza- reductase from Neurospora mitochondria. Nature
ki, H. Yamaguchi, K. Shinzawa-Itoh, R. Nakashima, 280:696-697.
R. Yaono, and S. Yoshikawa. 1996. The whole struc- 85.Xia, D., C.A. Yu, H. Kim, J.Z. Xia, A.M. Kachurin,
ture of the 13-subunit oxidized cytochrome c oxidase L. Zhang, L. Yu, and J. Deisenhofer. 1997. Crystal
at 2.8 A [see comments]. Science 272:1136-1144. structure of the cytochrome bc1 complex from bovine
76.Unger, V.M., N.M. Kumar, N.B. Gilula, and M. Yea- heart mitochondria [published erratum appears in Sci-
ger. 1999. Three-dimensional structure of a recombi- ence 1997 Dec 19;278(5346):2037]. Science 277:60-
nant gap junction membrane channel. Science 283: 66.
1176-1180. 86.Yeager, M., V.M. Unger, and A.K. Mitra. 1999. Three-
77.Unwin, P.N.T. 1974. Electron microscopy of the dimensional structure of membrane proteins deter-
stacked disk aggregate of Tobacco Mosaic Virus pro- mined by two-dimensional crystallization, electron cry-
tein. II. The influence of electron irradiation on the omicroscopy, and image analysis. Methods Enzymol.
stain distribution. J. Mol. Biol. 87:657-670. 294:135-180.
78.Unwin, P.N.T. and R. Henderson. 1975. Molecular 87.Zeppenzauer, M. 1971. Formation of large crystals, p.
structure determination by electron microscopy of 253. In W.B. Jacoby (Ed.), Methods of Enzymology.
unstained crystalline specimens. J. Mol. Biol. 94:425- Academic Press, New York.
440. 88.Zhang, Z., L. Huang, V.M. Shulmeister, Y.I. Chi,
79.Valpuesta, J.M., R. Henderson, and T.G. Frey. 1990. K.K. Kim, L.W. Hung, A.R. Crofts, E.A. Berry, and
Electron cryo-microscopic analysis of crystalline S.H. Kim. 1998. Electron transfer by domain move-
cytochrome oxidase. J. Mol. Biol. 214:237-251. ment in cytochrome bc1. Nature 392:677-684.
80.Vanderkooi, G., A.E. Senior, R.A. Capaldi, and H. 89.Zhu, J., P.A. Penczek, R. Schroder, and J. Frank.
Hayashi. 1972. Biological membrane structure. 3. The 1997. Three-dimensional reconstruction with contrast
lattice structure of membranous cytochrome oxidase. transfer function correction from energy-filtered cryo-
Biochim. Biophys. Acta 274:38-48. electron micrographs: procedure and application to the
81.Weiss, H., S. Hovmoller, and K. Leonard. 1986. 70S Escherichia coli ribosome. J. Struct. Biol. 118:197-
Preparation of membrane crystals of ubiquinol- 219.

234
Analysis and Reconstitution of
10 Chlorophyll–Proteins

Harald Paulsen and Volkmar H.R. Schmid


Institut für Allgemeine Botanik der Johannes-Gutenberg
Universität Mainz, Mainz, Germany

1. INTRODUCTION complexes is not always an easy task, as sev-


eral of these complexes are quite unstable
It is hard to believe that only some 30 and dissociate very easily. This may be
years ago, it was a matter of debate whether illustrated by analyses of the major LHC
chlorophyll (Chl) and other photosynthe- (LHCII) of photosystem II (PSII).
sis pigments are protein-bound or just dis- Although this is the most abundant Chl-
solved in plant membranes. Philip Thorn- containing complex and certainly one of
ber, who vividly described this debate in the more stable ones, its Chl-protein stoi-
his recollection of photosynthesis research chiometry reported by various laboratories
in the 1960s (90), was one of the expo- has fluctuated between 6 (44) and 15 (14).
nents who finally convinced their col- One of the major problems is the fact that,
leagues that most, if not all, Chl in plants is with few exceptions, Chl-binding polypep-
in fact organized in protein complexes. It tides are membrane proteins that need to
was his laboratory that devised quite a be solubilized by detergents for isolation
number of chromatographic and elec- and analysis. The quantification of proteins
trophoretic techniques for isolating (bacte- in detergent solution is often difficult,
rio)chlorophyll-containing complexes from which in turn tends to render Chl/protein
bacteria and plants. These isolation tech- ratios unreliable. Moreover, looking at the
niques later paved the way for structural crystal structure of LHCII monomeric
analyses of, e.g., photosynthetic reaction complexes (49) where most of the Chls are
centers of purple bacteria (24) as well as exposed at the surface of the protein com-
bacterial (53) and plant (49) light-harvest- plex, it may be not too surprising that
ing complexes (LHC). Many of these pro- some of these pigments are easily lost upon
tocols for isolating pigment–protein com- treatment of the complex by detergent.
plexes are still, in a more or less modified The major secret of solubilizing Chl-con-
version, being used in many laboratories taining protein complexes without produc-
today. ing large amounts of unbound pigments
Isolating and analyzing Chl-protein has been the choice of the right detergent.

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

235
H. Paulsen and V.H.R. Schmid

Unfortunately, however, the one optimal 2. ISOLATION AND ANALYSIS OF


detergent for all Chl-protein complexes CHLOROPHYLL–PROTEIN
does not seem to exist. Optimized isolation COMPLEXES
protocols with individual detergents or
detergent mixtures have been developed for As pointed out in the introduction to
just about every Chl-protein. Only a selec- this chapter, there is a tremendous number
tion of these procedures can be referred to of different isolation procedures described
in this chapter. in the literature that have been devised for
A new approach to studying Chl-pro- various Chl-containing complexes. It is
tein complexes was opened up when Plum- impossible to give a complete overview of
ley and Schmidt (72) showed that LHCII these techniques. Therefore, we will point
can be reconstituted in vitro upon denatur- to procedures that we believe are good
ing it in detergent solution. Under renatur- examples of different separation principles
ing conditions in detergent solution, the used for Chl-proteins and focus on those
protein refolds and comcomitantly assem- techniques that are also useful for isolating
bles its pigments. This astonishing capabil- reconstituted pigment–protein complexes
ity of self-organization in vitro seems to (see below, section 3.4). Likewise, we will
extend to all Chl a/b-containing complexes describe in some more detail only those
in plants and also to bacterial LHCs (see analytical procedures that have successfully
section 3.1). On the other hand, no suc- been used to characterize recombinant pig-
cessful reconstitution of the Chl a-contain- ment–protein complexes.
ing complexes of reaction centers and inner
antennas has been reported. Possibly this 2.1. Isolation of Chlorophyll–Protein
indicates some structural feature that is Complexes
common only among Chl-a/b proteins
which enables them to refold in the pres- 2.1.1. Solubilization
ence of detergents and pigments.
The approach of reconstitution is so Most Chl-protein complexes are local-
powerful, because it allows the generation ized in photosynthetic membranes; there-
of recombinant pigment–protein complex- fore, the first step in the isolation of these
es in vitro. Not only can the pigment and complexes notoriously includes the solubi-
lipid composition in these complexes be lization of the membranes by use of deter-
altered, in order to assess their contribution gents. The detergent employed for solubi-
to structure and function, but also bacteri- lization needs to be wisely chosen, as it will
ally expressed apoproteins carrying direct- influence the aggregational state and the
ed mutations can be used. This way, the integrity of the pigment–protein complex-
impact of individual protein domains, or es, and it will often limit the choice of
even single amino acids, on pigment bind- techniques accessible for further purifica-
ing or functioning of the complex can be tion. Ionic detergents for instance that are
addressed. Such a detailed analysis of struc- difficult to remove quantitatively from the
ture–function relationships is much more protein complexes, such as sodium dode-
difficult to achieve in an in vivo system. cylsulfate (SDS), usually preclude subse-
In this chapter, we will focus on the quent ion exchange chromatography.
analysis and reconstitution of Chl-a/b pro- Nonionic glycosidic detergents such as
teins. We hope the reader will also find the octylglucoside (OG) or dodecylmaltoside
selected references to the analysis and (LM) have proven useful to solubilize
reconstitution of other proteins helpful. many pigment–protein complexes under
236
Analysis and Reconstitution of Chlorophyll–Proteins

mild nondenaturing conditions (Table 1). native and recombinant Chl-protein com-
However, other nonionic detergents like plexes usually is the analysis of the pigment
Triton® X-100 or ionic detergents such as and protein components. Very often, the
lauryldimethylamine oxide (LDAO) and first step consists of an extraction of the
SDS (or lithiumdodecylsulfate, LDS, complexes by adding 80% acetone, which
where SDS would precipitate at low tem- denatures and precipitates the protein moi-
peratures) are also in use. SDS or LDS, in ety, whereas the noncovalently bound pig-
combination with nonionic detergents, ments end up in the supernatant and can
allows the control of the denaturing strin- subsequently be separated and analyzed. If
gency in partially denaturing polyacry- Chl-protein complexes have been separat-
lamide gels and, thus, is useful in separat- ed by polyacrylamide gel electrophoresis,
ing more stable complexes from less stable acetone does not extract pigments from gel
ones (70). slices. This can be done quantitatively by
using 2-butanol (52).
2.1.2. Purification The pigments in the 80% acetone
extract can be analyzed and quantified
The overview of separation procedures either spectrophotometrically or by high-
given in Table 2 is far from being complete, performance liquid chromatography
but rather gives a few examples of where (HPLC). For the quantification of mix-
the procedures have been used. tures of Chl a and Chl b in 80% acetone
The majority of the procedures used for (buffered to pH 7.8), the algorithm by
the isolation of native Chl-protein com- Porra et al. (73) is frequently used:
plexes includes ultracentrifugation steps in concentrationChl a (µg/mL) = 12.25 A663.6 - 2.55 A646.6
a density gradient or various chromato- concentrationChl b (µg/mL) = 20.31 A646.6 - 4.91 A663.6
graphic techniques or both; these tech- where A646.6 and A663.6 are the
niques are also useful to isolate recombi- absorbances at the given wavelengths at 1
nant Chl-protein complexes. Some of the cm pathlength, minus the absorbance at
gel electrophoretic techniques tend to have 750 nm. Some alternative algorithms are
a more strongly denaturing effect on pig- discussed in the same reference. For the
ment–protein complexes and, therefore, determination of bacteriochlorophyll a in
may lead to the loss of pigments. However, acetone:methanol (7:2, vol/vol), a molar
gel electrophoresis provides a rapid means absorption coefficient of ε770 = 76
to separate in vitro reconstituted Chl-pro- 000/Mcm (19) has been measured. For the
tein complexes from unbound pigments rough quantification of α- and β-
and, thus, is useful for analyzing reconsti- carotenoids and their derivatives, the spe-
tution products. Therefore, gel elec- cific absorption coefficient of Davies (22)
trophoresis along with sucrose gradient is very useful: ε440 = 240 L/gcm.
centrifugation and ion exchange tech- Numerous HPLC protocols for separat-
niques will be described in some more ing Chls and carotenoids have been devel-
detail in section 3.4. oped (29,34,65,93). One of the simplest
ones is a gradient from 80% to 100% ace-
2.2. Analysis of Chlorophyll–Protein tone. Usually, the conversion of peak areas
Complexes to pigment quantities is calibrated on the
basis of absorption coefficients such as
2.2.1. Biochemical Characterization those given above.
Quantification of the precipitated and
The first biochemical characterization of redissolved protein can be performed by
237
Table 1. Detergents Used for Solubilizing Chl-Proteins
238

H. Paulsen and V.H.R. Schmid


Detergent Used to Solubilize References

OG PSII core complexes 37


LH1 from Rhodospirillum rubrum 64
Chl-protein complexes of thylakoids 15,69
LM PSI for crystallization 30
PSII core complexes with antenna 7,37
complexes attached
PSII core and antenna subunits 20
CP43 and CP47 1
LHCII subunits 21
LHCI-730 86
PSI and PSII from red algae 89
Chl a,b,c complex of M. squamata 97
Heptylthioglucoside PSII for crystallization 74,75
CP22 (PsbS) 31
Triton® X-100 LHCII 13,48
PSI 56
Chl a,b,c complex of M. squamata 76
Lauryldimethyl-amine oxide (LDAO) LH1 and LH2 from Rhodospirillum molischianum 92
SDS or LDS Chl-protein complexes of thylakoids 4,25
Digitonin Chl a,c complexes from various algae 10,26,27,39
Zwittergent 14 PSI and PSII from Prochlorothrix hollandica 95
Zwittergent 16 LHCI 38,51,91
Table 2. Separation Procedures Used to Isolate Chl-Proteins
Used for References

Sucrose-gradient PSI 57
ultracentrifugation
PSII core and antenna subunits 20
PSII core complex and PSII-LHCII 37
super complex
LH1 and LH2 from R. molischianum 92
LHCI 63
LHCII 13,48

Analysis and Reconstitution of Chlorophyll–Proteins


Chl a,c complexes from various algae 11,26,27,39
Chl a,b,c complex of M. squamata 76
Sucrose gradient with subsequent PSI reaction center complex 80
anion-exchange chromatography
PSI 80
CP43 and CP47 1
D1-D2-cytochrome b559, CP43 5
and CP 47 and LHCII subunits
LHCII complexes 61
Recombinant CP24 62
Recombinant CP29 35
PSI and PSII from red algae 89
Chl a,b,c complex from M. squamata 97
PSI and PSII from P. hollandica 95
Anion exchange chromatography Water-soluble Chl-protein from cauliflower 43,60
and Brussels sprouts
Gel filtration chromatography PSII-LHCII super complex 7
LH1 from R. rubrum 64
Perfusion chromatography PSII 81
LHCI 91
Nickel chelating chromatography Recombinant His-tagged LHCII 28,79
239
H. Paulsen and V.H.R. Schmid

applying a number of generally


used protein assays such as the
bicinchoninic acid reaction (in
Reference 88, see Chapter 10), the
Lowry test (71), or the Bradford
test (9). Detergents are a problem
References

in many protein assays. If the pro-


tein is precipitated from a solution
68,72
77,87

69,84
2,45 containing SDS or LDS, co-pre-
36

46

54

47
31
21
5
3
cipitation of the dodecylsulfate
salt can be reduced by acidifying
the solution to pH 4.0–5.0 with
acetic acid. Note that acidification
is incompatible with subsequent
Chl analysis in the supernatant, as
Chl-protein complexes of thylakoids
His-tagged bacterial photosynthetic

Native and reconstituted Chl a,b,c

it will turn the Chls into their


Chloroplast protein complexes

pheophytins. If the protein is pre-


Chl-proteins from thylakoids
complex from M. squamata

cipitated from a very dilute solu-


tion, it may be necessary to collect
it by extended ultracentrifugation
Recombinant LHCII

in order to pellet it quantitatively.


Subunits of PSII
reaction center

LHCII subunits

It is often desirable to exchange


CP22 (PsbS)
PSI and PSII

PSII LHCs

or remove detergents from Chl-


Used For

protein complexes before their


LHCII

analysis or further handling.


Detergents having a critical micel-
lar concentration in the millimo-
Partially denaturing LDS (Laemmli) gel electrophoresis

lar range, such as OG, can most


easily be removed by dialysis. A
method to exchange detergents in
Blue native polyacrylamide gel electrophoresis

protein solutions by hydrophobic


Precipitation by mono- and divalent cations

interaction chromatography on
Preparative flat-bed isoelectric focussing

phenyl sepharose has been


described (78); however, deter-
Copper chelating chromatography

Fluid-phase isoelectric focussing

gents that stably interact with pro-


teins such as dodecylsulfate are
only inefficiently exchanged by
this technique. Removal of these
Modified Laemmli gel

detergents, SDS or LDS, is often


problematic, particularly with
recombinant Chl a/b-protein
Deriphat gel
Table 2, continued

complexes that are reconstituted


at high concentrations of LDS.
The bulk of LDS can be removed
by precipitating the dodecylsulfate
with 200 mM potassium salt at
0°C in the presence of a nonionic
240
Analysis and Reconstitution of Chlorophyll–Proteins

detergent. The remaining dodecylsulfate absence of any pigment–protein complex-


can be removed by binding the complexes es. As the detergent concentration is low-
to an ion exchange column and washing ered, Chl concentrations in the decreasing
them with buffer containing a nonionic number of detergent micelles rise until
detergent (41). energy transfer becomes possible. There-
fore, if energy transfer from Chl b to Chl a
2.2.2. Spectroscopic Characterization is detected even upon heat-denaturing the
Chl-protein complexes, it cannot be
The spectroscopy used to analyze isolat- indicative of intact complexes and most
ed Chl-protein complexes very much likely the detergent concentration is too
depends on which complex is to be ana- low.
lyzed and on its properties to be examined. CD spectroscopy is useful to character-
Techniques generally used for a first char- ize pigment–protein complexes, as both
acterization of pigment–protein complexes the protein and the pigment moieties give
are absorption, fluorescence, and circular rise to CD signals in the UV and visible
dichroism (CD) spectroscopy (96). regions, respectively (96). CD signals in
An immediate test for the intactness of the visible region in which Chls and
isolated Chl a/b LHCs is the measurement carotenoids absorb have been taken as a
of energy transfer from Chl b to Chl a. criterion to test the structural authenticity
This can easily be measured by using a of recombinant Chl a/b complexes (68,72).
steady-state fluorescence spectrophotome- Beyond this fingerprint comparison, CD
ter. The excitation is set to an absorption spectra provide information about the pig-
wavelength of Chl b (usually at around 470 ment organization in pigment–protein
nm, at the long-wavelength side of the complexes (94) and the state of oligomer-
Soret absorption band in order to mini- ization of such complexes (33,41). Signals
mize simultaneous excitation of Chl a). of CD in the UV domain have been used
Fluorescence emission is scanned over the to compare protein folding in a recombi-
Chl a and Chl b emission wavelength nant with that in wild-type LHCII (66).
region, between 650 and 750 nm. Emis-
sion from Chl a exclusively (maximum at 3. RECONSTITUTION OF
around 680 nm, no shoulder at 660 nm) PIGMENT–PROTEIN
indicates quantitative energy transfer from COMPLEXES
Chl b to Chl a. Alternatively, the emission
wavelength is set to the Chl a emission Reconstitution represents the controlled
wavelength around 680 nm, and excitation folding of an LHC-apoprotein in the pres-
is scanned in the Chl and carotenoid ence of detergents and pigments (and
absorption domain. Excitation signals at lipids), giving rise to complexes with very
455 nm and around 475 nm indicate ener- similar properties when compared to the
gy transfer from Chl b and carotenoids, authentic complexes isolated from leaf
respectively. material.
Care must be taken to avoid deceptive
energy transfer in dilute detergent solu- 3.1. Survey of Reconstituted
tions. For instance in 0.1% (wt/vol) LM Pigment–Protein Complexes
solution, as is often used in sucrose density
gradient centrifugations, efficient energy The first pigment–protein complexes
transfer from Chl b to Chl a is detected, which were reconstituted are the major
even with unbound pigments, in the LHCII of higher plants and the core LHC
241
H. Paulsen and V.H.R. Schmid

(LH1) of photosynthetic bacteria. In these their applications are summarized. Both


initial experiments, authentic proteins iso- procedures will be given in more detail in
lated from LH1 (64) and total thylakoid section 3.3.2.
membrane proteins (72) were employed. The result of reconstitutions of LHCI
New possibilities were opened up by the and LHCII by these two methods is
availability of cDNAs for several LHC- depicted in Figure 1. It is obvious that
apoproteins of different origins, which application of the detergent exchange
allowed the use of bacterially overexpressed method yields reconstituted LHCI and
LHC proteins in reconstitution experi- LHCII, whereas with the freeze-thaw
ments (17,68). Since bacterially expressed method, only reconstituted LHCII is
proteins can easily be mutated, the intro- obtained.
duction of various structural alterations Additionally, a reconstitution technique
into recombinant pigment–protein com- was developed, which is based on detergent
plexes was facilitated. Using C and N ter- mixing, which also prompts protein fold-
minally truncated apoproteins, the signifi- ing and pigment binding. This method
cance of these protein domains for the allows protein refolding to initiate very
formation of LHCII could be identified quickly and, therefore, facilitates time-
(18,67). Later, reconstitution of different resolved spectroscopic measurements (8).
LHCs of higher plants, CP29 (35), CP26
(82), and CP24 (62), as well as LHCI-730 3.3. Reconstitution Procedures
(86) and LHCI-680a (Schmid and
Paulsen, unpublished) were accomplished. Reconstitutions of plant LHCs usually
Additionally, an LHCI (LhcaR1) of the red start from isolated plant pigments and bac-
alga Porphyridium cruentum (32), a LHC terially expressed apoprotein.
of the green alga Chlorella fusca, and a Chl
a,b,c-containing complex of the prasino- 3.3.1. Pigment Isolation
phyte Mantoniella squamata were success-
fully reconstituted (54). Recently, the Reconstitution is performed either with
peripheral LHC (LH2) of a purple bacteri- a total thylakoid pigment extract or with (a
um has also been reconstituted (92). mixture of ) individual pigments. Some
Moreover, it was shown that it is not pigments are commercially available [e.g.,
only possible to reconstitute monomeric Chl a and b, lutein, α- and β-carotene
LHC but also the oligomeric form of the from Sigma (St. Louis, MO, USA); lutein
major LHCII complex and of LHCI-730 and zeaxanthin from Roth (Germany)] but
(41,79,86). others are not. Therefore, their isolation is
described in the following protocol. All iso-
3.2. Comparison of Reconstitution lation steps should be performed in dim
Procedures light. As a source for pigment isolation,
homogenized whole leaves or thylakoids
All the LHCII reconstitution experi- isolated as in, e.g., Reference16, can be
ments until 1992 were performed by the used. Thylakoids are suspended in a small
original freeze-thaw method (72). Later, a volume of dilute buffer as 10 mM Tricine-
new method, based on detergent exchange NaOH (pH 7.8) and extracted by addition
was developed (66), which proved to be of acetone to a final concentration of 80%.
very powerful as it has allowed reconstitu- Proteins are removed by a 10-minute cen-
tion of the more labile complexes in recent trifugation at 15 000× g. For isolation of
years. In Table 3, these two methods and total pigment extract or individual pig-
242
Analysis and Reconstitution of Chlorophyll–Proteins

ments, the supernatant is treated in differ- 6. The aliquots are dried in a nitrogen
ent ways. stream and can be stored for months to
years at -20°C under nitrogen or argon.
❖ Procedure 1. Total Pigment Extract For the isolation of individual Chls and
carotenoids, the following procedure is
1. The pigment solution in acetone is useful:
mixed with 0.25 volumes diethyl ether
in a separating funnel. ❖ Procedure 2. Isolation of Individual
2. To improve phase separation, solid Pigments
NaCl (e.g., 35 g to 600-mL solution) is
added and dissolved by gently moving 1. The acetonic pigment solution is
the funnel. If the lower acetone phase cooled to 0°C.
remains colored, the ether extraction 2. Dioxane is added to give a final con-
should be repeated, adding more NaCl centration of about 15% (vol/vol) (42).
if phase separation is poor. 3. To the homogenous solution 0.16 vol-
3. Combine and dry the ether phases, umes of water is added drop-wise
either by the addition of solid NaCl or under constant stirring, and the resul-
by placing the ether solution in a tant solution is kept on ice for 1 hour
-20°C freezer for at least 1 hour. Then without further stirring.
ice or NaCl can be removed by filtra- 4. Aggregated Chls (as well as pheophytin
tion through a sintered glass funnel and β-carotene) are collected by cen-
(precooled to -20°C if ice crystals are to trifugation (15 000× g, for 10 min),
be removed). and the pelleted pigments are used for
4. Evaporate the ether in a rotary evapora- column chromatographic separation of
tor or nitrogen stream. the Chls and β-carotene. If the super-
5. Pigments are dissolved in acetone, natant still contains a substantial
quantified on the basis of their Chl con- amount of the Chl originally present,
tent (see section 2.2.1), and aliquoted. more water may be added drop-wise,
and the additional precipitate collect-
ed. If the addition of water is too exces-
sive or too fast, xanthophylls will also
aggregate and contaminate the crude
Chl preparation. The supernatant is
kept for xanthophyll isolation.
5. For the separation of individual Chls
(and xanthophylls), a reversed phase
C18 material [e.g., 55–105 µ-Bonda-
pak (Waters, Milford, MA, USA)] with
acetone–water mixtures as the mobile
phase is suitable. Chromatography can
be done either at low pressure or by
Figure 1. Partially denaturing gel electrophoresis of recon- using an HPLC apparatus. A column
stitution mixtures with LHCI- (Lhca4; lanes a and b) or volume of at least 4 mL (low pressure
LHCII- (Lhcb1; lanes c and d) apoprotein. The mixtures
were subjected to either the freeze-thaw (lanes a and c) or the
development) or 0.8 mL (high pressure
detergent exchange method (lanes b and d). The resolution processing) is recommended for each
of bands with monomeric complexes (M) and free pigments milligram of raw pigments applied.
(FP) is visible.
243
H. Paulsen and V.H.R. Schmid

Table 3. Comparison of the Experimental Steps of the Two Most Commonly Used Reconstitution
Techniques

Detergent Exchange
Freeze-Thaw Method Method

References that the 17,18,54,55,68,72 32,35,40,41,62,66,82,86


method is used in:

Protein denaturation by + +
LDS and heating

Addition of OG - +

Addition of pigments (and lipids) + +

Freeze at -20°C and thaw at + -/(+)


22°C 3 times

Removal of LDS by KCl addition - +

Application to nondenaturing + +
polyacrylamide gel electrophoresis or
sucrose density gradient centrifugation

6. Preequilibrate the column with 86% 12. For isolation of individual xantho-
acetone. phylls, the pigments in the supernatant
7. Dissolve the Chl pellet in a small vol- of the dioxane precipitation (step 4) are
ume of 86% acetone. ether-extracted and dried as described
8. To remove any particulate material the for total pigment extracts (steps 1–4).
solution is centrifuged (15 000× g for 13. The residue is dissolved in ethanol
10 min). and made up to 8% KOH by addi-
9. The supernatant is loaded on the tion of 0.1 volumes from an 80%
column. (wt/vol) stock solution in water (22).
10. Elution of Chl b (green) and Chl a The mixture is overlaid with nitrogen
(blue-green) is done with acetone of and kept overnight in a tightly
that concentration. For the elution of capped bottle at 55°C in the dark.
pheophytin (brown) and β-carotene The saponification with KOH con-
(orange), the acetone concentration has verts residual Chl and lipids into
to be raised to 90% and 95%, respec- more hydrophilic products.
tively. To obtain pure pigments, only 14. Xanthophylls are extracted by ether as
the central part of the individual described for total pigment extract
pigment fractions should be collected. (steps 1–2).
11. The eluted pigments are transferred to 15. The ether fraction is washed twice with
ether, dried, quantitated, and stored as 3 volumes of water.
described for total pigment extracts 16. The xanthophylls are then obtained
(steps 1–6). from the ether solution as described
244
Analysis and Reconstitution of Chlorophyll–Proteins

above (total pigment extracts, steps described by Nagai and Thøgersen (58), is
3–4). They can be either used as “total described here in detail.
xanthophylls” (see section 2.2.1 for
absorption coefficient for carotenoids) ❖ Procedure 3. Isolation of
for reconstitution or, alternatively, sub- Recombinant LHC-Apoproteins
jected to a column chromatography
procedure as outlined for the Chls 1. Start with an 5 mL overnight culture
(step 5) to isolate the individual xan- [Luria-Bertani medium supplemented
thophylls. with 100 µg ampicillin/mL (LB-Amp)]
17. Proceed as in steps 6 to 9, but use of Eschericia coli that harbors the
74% acetone for preequilibration and respective expression plasmid.
as solvent. 2. Inoculate a 250-mL Erlenmeyer flask
18. Isocratic elution is started with 74% containing 100 mL LB-Amp with 1 mL
acetone. Following elution of neoxan- of the overnight culture and grow the
thin and violaxanthin the acetone con- cells on a rotary shaker at 170
centration is raised to 80% acetone for rounds/minute and 37°C to mid-log
the elution of lutein. phase (OD550 of around 0.5), which
takes about 2 hours.
The purity of the individual pigments is
3. Induce overexpression by addition of a
most conveniently tested by analytical
1 M isopropyl-β-D-thiogalactoside
HPLC or by thin-layer chromatography
solution to 1 mM final concentration.
(TLC) with, e.g., RP18-plates (Merck,
The cells are cultivated for another 4 to
Darmstadt, Germany) and methanol as
5 hours under the same conditions.
solvent. For the quantification of Chls see
section 2.2.1; carotenoids can be quanti- 4. Harvest the cells by centrifugation
fied by means of the absorption coefficients (5 min at 5 000× g). The cell pellets are
of Davies (23). The specific absorption either stored at -20°C or processed fur-
coefficients, for example for an ethanolic ther immediately.
solution, are ε439 = 224.3 L/gcm (neoxan- 5. Suspend the cell pellet in 500 µL lysis
thin), ε443 = 255 L/gcm (violaxanthin), buffer [50 mM Tris-HCl (pH 8.0),
ε445 = 255 L/gcm (lutein), and ε453 = 262 25% (wt/vol) sucrose, and 1 mM
L/gcm (β-carotene). EDTA) and bring it to 800 µL with
lysis buffer.
3.3.2. Protein Isolation 6. Add 200 µL lysozyme from a 1%
(wt/vol, in lysis buffer) solution which
Originally, authentic proteins isolated is freshly prepared each time. The solu-
from the membranes of the corresponding tion is mixed well and incubated at
species were extracted and used for recon- room temperature for 30 minutes.
stitution experiments (72). Meanwhile, 7. Add 10 µL DNase I solution [0.1%
however, cDNAs derived from the genes (wt/vol) solution in 20 mM Tris-HCl
are cloned into expression vectors, e.g., (pH 8.0), 50 mM NaCl, 1 mM dithio-
pDS-vectors (12), in order to obtain large threitol (DTT), and 50% (vol/vol)
quantities of the desired protein. All bacte- glycerol; this solution can be stored at
rially expressed LHC-apoproteins are accu- -20°C], together with 10 µL of 0.1 M
mulated in the form of insoluble inclusion MnCl2 and 1 M MgCl2. Incubation
bodies. Therefore, inclusion body isolation, for another 30 minutes at room tem-
which follows mainly the procedure perature follows.
245
H. Paulsen and V.H.R. Schmid

8. Add 2 mL of detergent solution A [1% USA)] or 7-fold (SW40 or SW41 rotors,


(wt/vol) deoxycholic acid (sodium salt), Beckman Coulter).
1% (vol/vol) Nonidet® P-40, 0.2 M
NaCl, 20 mM Tris-HCl (pH 7.5), Procedure 4. Freeze-Thaw Method
2 mM EDTA, 30 mM DTT). The
solution is mixed well and kept at room 1. Suspend 8 µg of LHCII or 25 µg of
temperature for 5 minutes. LHCI inclusion body protein in 16 µL
9. Centrifuge the solution for 10 minutes storage buffer (section 3.3.1).
at 10 000× g. 2. Mix the protein solution with an equivalent
10. Suspend the pellet in 2 mL detergent volume of 2× reconstitution buffer
solution B [0.5% (wt/wt) Triton® [200 mM Tris-HCl (pH 9), 4% (wt/vol)
X-100, 1 mM EDTA-NaOH (pH LDS, 100 mM DTT, 2 mM benzamidine,
7.8), and 20 mM DTT] and keep the 10 mM ε-aminocaproic acid, and — for
solution at room temperature for gel separation — 25% (wt/vol) sucrose].
5 minutes. 3. The protein solution is heated for
11. Collect inclusion bodies by centrifuga- 1 minute in a boiling water bath and
tion as in step 5. Sometimes the over- cooled on ice.
expressed protein does not form very 4. Dried Chls and total xanthophylls corre-
stable inclusion bodies. In this case, the sponding to 24 µg and 5 µg, respectively,
volume of the detergent solutions (LHCI: total pigment extract equivalent
should be reduced (e.g., by 50%, but to 30 µg Chl) are dissolved in 1.5 µL
the appropriate amount has to be ethanol (the final ethanol content must
determined empirically). not exceed 8% as otherwise protein pre-
12. Suspend the pellet in storage buffer cipitation may occur). Pigments must be
[50 mM Tris-HCl (pH 8.0), 1 mM EDTA, dissolved completely, which is best done
20 mM DTT]. If the protein pellet by vigorously vortex mixing followed by
appears slimy at this point and is not an incubation for 30 seconds in an ultra-
easily resuspended, it is advisable to sonic bath. When pigments sediment
repeat steps 4–7 once again. during subsequent centrifugation of the
pigment solution (1 min at 15 800× g),
13. Assess the protein content by, e.g., the this step has to be repeated.
Bradford protein assay (9), which is
compatible with DTT. Aliquots of the 5. Add the protein solution to the pig-
protein solution can be stored at ment solution under vortex mixing.
-20°C. 6. The resultant solution is placed in a
-20°C freezer for 2 hours and then thawed
3.3.3. Reconstitution Procedures at room temperature for 15 minutes.
7. Repeat step 6 twice.
In the following, we describe reconstitu- 8. The solution is ready to be analyzed on
tion of Chl-protein complexes by (i) freeze- a partially denaturing gel or in sucrose
thaw cycles, (ii) detergent exchange, and density gradients (see section 3.4).
(iii) detergent mixing. Quantities and vol-
umes given are for subsequent separation ❖ Procedure 5. Detergent Exchange
in analytical polyacrylamide slab gels. For Method
sucrose gradient ultracentrifugation, the
amounts should be scaled up 4-fold [SW60 1. Prepare a protein solution as in step 1
rotor, Beckman Coulter (Fullerton, CA, of Procedure 4.
246
Analysis and Reconstitution of Chlorophyll–Proteins

2. Reconstitution is continued by the means of a stopped-flow device which


addition of 3.7 µL of 10% (LHCII) or allows time resolved measurements
20% (LHCI) OG to the protein down to the millisecond range. The
solution. dilution brought about by the mixing
3. Boil the solution for 1 minute and cool of the 2 samples results in folding of
it down on ice. the protein (8).
4. Add 1.2 µL 1 M DTT to the sample. 6. The success of reconstitution can be
5. Prepare a pigment solution as in step 4 examined by gel electrophoretic analy-
of Procedure 4. sis or by following spectroscopic sig-
nals, e.g., the energy transfer from Chl
6. Transfer the protein solution to the
b to Chl a (section 2.2.2).
pigments during mixing.
Besides monomeric complexes, oligo-
7. Add 4.27 µL 2 M (LHCII) or 6.78 µL
meric complexes can also be reconstituted.
1 M (LHCI) KCl solution.
Very convenient is the generation of the
8. The resultant solution is mixed and heterodimeric LHCI-730, which requires
kept at 4°C for 20 minutes. equal amounts of the apoproteins (12.5 µg
9. Precipitated potassium dodecylsulfate is of both) in the starting protein solution
sedimented by centrifugation (15 800× g (86). Using a sophisticated, multistep pro-
for 5 min at 4°C). cedure, trimerization of LHCII was
10. The supernatant is loaded on a gel or achieved which allowed crystallization of
sucrose gradient (section 3.4). the reconstituted complex. This method is
described in detail in Hobe et al. (41).
Meanwhile, another reconstitution method
❖ Procedure 6. Detergent Mixing for trimeric LHCII has been developed,
Method where refolding of the protein occurs while
1. Twenty-three micrograms LHCII it is immobilized, via a His-tag, on a metal-
inclusion body protein is dissolved chelate column material. This method
in 116 µL of 0.2% (wt/vol) SDS, facilitates faster production and isolation of
100 mM lithium borate (pH 9.0), trimeric complexes (79).
12.5% (wt/vol) sucrose, and 5 mM
DTT. 3.4. Isolation of Reconstituted
2. Transfer the solution to a cuvette. Complexes
3. Twelve micrograms Chl and 3 µg total All the techniques described below
xanthophylls are dissolved in 3 µL should be performed at 0° to 4°C in dim
ethanol (step 4 of Procedure 4). light.
4. Dissolved pigments are mixed with
116 µL of a solution with 2% (wt/vol) 3.4.1. Partially Denaturing
OG, 0.075% (wt/vol) phosphatidyl- Polyacrylamide Gel Electrophoresis
glycerol, 100 mM lithium borate (pH
9.0), 12.5% (wt/vol) sucrose, and Most of the commonly used partially
5 mM DTT. denaturing gel systems go back to the
5. The pigment solution is added to the recipes given by either Laemmli (50) or
protein solution in the cuvette and Neville (59). They proved to be also suit-
mixed rapidly by stirring. Alternatively, able for isolation of reconstituted complex-
these 2 solutions are rapidly mixed by es. For reconstituted LHCII, we prefer the

247
H. Paulsen and V.H.R. Schmid

Laemmli system (68), and for LHCI, we For both gel types, the same running
prefer a modified Neville system (85). For buffer with 25 mM Tris, 196 mM glycine,
most applications, analytical slab minigels and 0.1% (wt/vol) LDS (only required in
are a good choice. The 30% (wt/vol) the cathode buffer) is used. The buffer is
acrylamide stock solution we use has an best prepared as a tenfold stock solution
acrylamide: N,N′-methylenebisacrylamide and diluted before use.
ratio of 30. Prior to electrophoresis, the gel and the
Laemmli Gel buffer should be cooled to 4°C. After
Prepare the required volume of the removal of the comb, the gel pockets are
resolving gel solution with 12% (wt/vol) rinsed with running buffer. Samples equiv-
acrylamide, 400 mM Tris-HCl (pH 8.8) alent to 10 µg Chl are applied to 4-mm-
and 10% (vol/vol) glycerol. While the solu- wide wells of a 1-mm-thick gel. Elec-
tion is stirred, ammonium persulfate (APS) trophoresis is either conducted with a
[10% (wt/vol) stock solution] and constant voltage of 60 V (Laemmli) or
N,N,N′,N′-tetramethylethylenediamine with a constant current of 0.1 mA/ mm2 of
(Temed) are added to final concentrations gel cross-section (Neville gel) for about 2.5
of 0.07% (wt/vol) and 0.05% (vol/vol), hours. Afterwards, the gel sandwich is dis-
respectively. The gel solution is poured assembled, the gel documented, and indi-
between the assembled glass plates up to vidual bands can be excised and the protein
about 7 mm below that point where the eluted for further characterization (see sec-
bottom of the comb will be located. The tion 2.2).
gel surface is overlaid with a thin layer of
water to obtain homogenous polymeriza- 3.4.2. Sucrose Density Gradients
tion and a plane gel surface. After 1 hour,
the gel should be polymerized, the water is Compared to nondenaturing gel elec-
poured off, and, if necessary, the gel surface trophoresis, ultracentrifugation through
is dabbed with filter paper. The stacking sucrose density gradients is a more gentle
gel solution with 4.5% (wt/vol) acry- method for isolating labile LHC. More-
lamide, 130 mM Tris-HCl (pH 6.8), 10% over, this method allows the isolation of
(vol/vol) glycerol, 0.05% (wt/vol) APS, sufficient material for further analyses and
and 0.05% (vol/vol) Temed is poured on has the advantage that the green band col-
top of the resolving gel, and the comb is lected from the centrifuge tube can be used
inserted by gently pushing it down starting immediately without the need to extract it
from one side. from a gel.
Neville Gel Sucrose gradients can be formed either
The resolving gel is composed of 12% by means of a gradient mixer in combina-
(wt/vol) acrylamide, 424 mM Tris-HCl tion with a peristaltic pump or, more con-
(pH 9.1), and 10% (wt/vol) sucrose. Poly- veniently, by the freeze-thaw method
merization is initiated by the addition of described by Bassi and Simpson (6). For
10% APS solution and Temed to final con- the latter method, a solution with 0.5 M
centrations of 0.03% (wt/vol) and 0.075% sucrose, 5 mM Tricine-NaOH (pH 7.8),
(vol/vol), respectively. The stacking gel is and 0.1% (wt/vol) LM is filled in the cen-
composed of 4% (wt/vol) acrylamide, 54 trifuge tubes. The tubes are placed in a
mM Tris-H2SO4 (pH 6.1), 10% (wt/vol) -20°C freezer. Three hours before sample
sucrose, and polymerization is initiated by application, the tubes are transferred to a
final concentrations of 0.06% (wt/vol) refrigerator and kept there until completely
APS and 0.075% (vol/vol) Temed. thawed. Subsequently, the upper tenth of
248
Analysis and Reconstitution of Chlorophyll–Proteins

the gradient solution is carefully removed, exchange chromatography is suitable for


which results in gradients with a sucrose the isolation of these proteins. This
concentration of about 0.1 to 1 M sucrose. method is a good choice if one intends
Depending on the rotor used, centrifuga- large-scale isolation. Furthermore, the
tion at 4°C is performed at 450 000× g for advantages described for sucrose density
16 hours (SW60, Beckman Coulter) or gradients also apply to this method.
260 000× g for 23 hours (SW40 or 41, Diethylaminoethyl (DEAE)-cellulose is
Beckman Coulter). Subjecting a reconsti- mostly used as stationary phase for column
tution mixture containing the 2 apopro- development by gravity, fast flow Q-
teins of LHCI-730 to density-gradient Sepharose® or Poros Q (both from Amer-
ultracentrifugation results in a separation sham Pharmacia Biotech, Piscataway, NJ,
as is shown in Figure 2. The resolution of USA) for pump-mediated column process-
zones with free pigments, monomeric ing (41,91,97). The mobile phase is usual-
complexes, and the heterodimeric LHCI- ly composed of a slightly alkaline buffer
730 is clearly visible. The bands of interest (e.g., phosphate buffer, Tris), and a deter-
are collected with a flat-tipped syringe nee- gent such as LM, both in low concentra-
dle from the top. tions [10 mM and 0.05% (wt/vol), respec-
tively]. Prior to sample application, the
3.4.3. Anion Exchange Chromatography column is washed first with 4 column vol-
umes of, e.g., 10 mM sodium phosphate
As a consequence of the low isoelectric buffer (pH 7.4) and then with 2 volumes
points of the higher plant pigment pro- of the buffer supplemented with the deter-
teins, ranging from 4 to 5 (21), anion gent, which is used for the solubilization of
the pigment–protein complexes, e.g.,
0.05% (wt/vol) LM. Then the sample,
adjusted to the same phosphate buffer (pH
7.4) and detergent concentration, is
applied. After the sample has completely
entered the column bed, the column is
washed with 2 column volumes of buffer
including detergent. Elution is achieved by
the buffer plus detergent supplemented
with NaCl. Mostly, a gradient of 0 to 400
mM NaCl works well. The steepness of the
NaCl gradient has to be determined indi-
vidually. Eluted bands can be characterized
with regard to apoprotein composition
(section 3.4.1), pigment composition (sec-
tion 2.2.1), and spectral properties (section
2.2.2).

4. CONCLUDING REMARKS

Figure 2. Sucrose gradient fractionation of reconstitution Several aspects have been mentioned in
mixtures containing the two apoproteins of LHCI-730. FP, the previous paragraphs of how reconstitu-
free pigments; m-LHCI, monomeric LHCI; d-LHCI, dimer-
ic LHCI (LHCI-730). tion of Chl a/b-protein complexes can be
249
H. Paulsen and V.H.R. Schmid

used as a fine and sophisticated surgical dodecylmaltoside; OG, octylglucoside; PS,


tool in functional analyses of these com- photosystem; SDS, sodium dodecylsulfate;
plexes. Single Chl-binding amino acids, for Temed, N,N,N′,N′-tetramethylethylenedi-
instance, may be exchanged simply by amine.
mutating the bacterially expressed apopro-
tein (83). If the corresponding Chl then is
lacking in the reconstituted complex, its ACKNOWLEDGMENTS
individual spectroscopic properties may be
deduced from spectral differences between We thank Martin Lohr (Universität
this and the native complex. Mainz, Germany) for carefully reading the
It should, however, be kept in mind that manuscript. Work in the authors’ laborato-
reconstitution itself is quite a striking ry was supported in part by grants from the
example of self-organization of a biological Deutsche Forschungsgemeinschaft (Pa
structure. A cue as simple as the mixing of 324/6-1, Schm 1203/2-1).
2 different detergents is sufficient to initi-
ate the correct folding of a medium-size REFERENCES
membrane protein and the binding of up
to 15 or so pigment molecules of several 1.Alfonso, M., G. Montoya, R. Cases, R. Rodriguez,
different kinds to their correct binding and R. Picorel. 1994. Core antenna complexes, CP43
and CP47, of higher plant photosystem II. Spectral
sites. The effort seems worthwhile to try properties, pigment stoichiometry, and amino acid
and understand this self-organization composition. Biochemistry 33:10494-10500.
process itself: what is the sequence of 2.Allen, K.D. and L.A. Staehelin. 1991. Resolution of
16 to 20 chlorophyll protein complexes using a low
events between the prereconstitution mix- ionic strength native green gel system. Anal. Biochem.
ture of components and the fully formed 194:214-222.
stable complex, what are the structural fea- 3.Allen, K.D. and L.A. Staehelin. 1992. Biochemical
characterization of photosystem-II antenna polypep-
tures involved, and which is the driving tides in grana and stroma membranes of spinach. Plant
force? Once we know the answer to these Physiol. 100:1517-1526.
questions, we may understand why Chl 4.Anderson, J.M., J.C. Waldron, and S.W. Thorne.
1978. Chlorophyll-protein complexes of spinach and
a/b-protein complexes appear to reconsti- barley thylakoids. FEBS Lett. 92:227-233.
tute more easily than other pigment–pro- 5.Bassi, R., B. Pineau, P. Dainese, and J. Marquardt.
1993. Carotenoid-binding proteins of photosystem II.
tein complexes. We may then learn to Eur. J. Biochem. 212:297-303.
design biomimetic structures that auto- 6.Bassi, R. and D. Simpson. 1987. Chlorophyll-protein
nomously form in vitro. complexes of barley photosystem I. Eur. J. Biochem.
163:221-230.
7.Boekema, E.J., H. van Roon, F. Calkoen, R. Bassi,
and J.P. Dekker. 1999. Multiple types of association of
ABBREVIATIONS photosystem II and its light-harvesting antenna in par-
tially solubilized photosystem II membranes. Biochem-
istry 38:2233-2239.
APS, ammonium persulfate; CD, circu- 8.Booth, P.J. and H. Paulsen. 1996. Assembly of light-
lar dichroism; Chl, chlorophyll; CP, harvesting chlorophyll a/b complex in vitro. Time-
resolved fluorescence measurements. Biochemistry
chlorophyll protein; DTT, dithiothreitol; 35:5103-5108.
HPLC, high-performance liquid chro- 9.Bradford, M.M. 1976. A rapid and sensitive method
matography; LB-Amp, Luria-Bertani for the quantitation of proteins utilizing the principle
of protein-dye binding. Anal. Biochem. 72:248-254.
medium supplemented with ampicillin; 10.Büchel, C. and G. Garab. 1997. Organization of the
LDAO, lauryldimethylamine oxide; LDS, pigment molecules in the chlorophyll a/c light-harvest-
lithiumdodecylsulfate; LHC, light-harvest- ing complex of Pleurochloris meiringensis (Xantho-
phyceae): characterization with circular dichroism and
ing complex; LH1 and LH2, light-harvest- absorbance spectroscopy. J. Photochem. Photobiol. B
ing complexes of purple bacteria; LM, Biol. 37:118-124.

250
Analysis and Reconstitution of Chlorophyll–Proteins

11.Büchel, C. and C. Wilhelm. 1993. Isolation and char- 25.Delepelaire, P. and N.H. Chua. 1981. Electrophoretic
acterization of a photosystem I-associated antenna purification of chlorophyll a/b protein complexes from
(LHC I) and a photosystem I-core complex from the Chlamydomonas reinhardtii and spinach and analysis of
chlorophyll c-containing alga Pleurochloris meiringensis their polypeptides. J. Biol. Chem. 256:9300-9307.
(Xanthophyceae). J. Photochem. Photobiol. B Biol. 26.Douady, D., B. Rousseau, and L. Caron. 1994. Fucox-
20:87-93. anthin chlorophyll a/c light-harvesting complexes of
12.Bujard, H., R. Gentz, M. Lanzer, D. Stueber, M. Laminaria saccharina — partial amino acid sequences
Mueller, I. Ibrahimi, M.T. Haeuptle, and B. Dob- and arrangement in thylakoid membranes. Biochem-
berstein. 1987. A T5-promoter-based transcription- istry 33:3165-3170.
translation system for the analysis of protein expres- 27.Durnford, D.G. and B.R. Green. 1994. Characteriza-
sion in vivo and in vitro. Methods Enzymol. tion of the light harvesting proteins of the chromo-
155:416-433. phytic alga, Olisthodiscus luteus (Heterosigma carterae).
13.Burke, J.J., C.L. Ditto, and C.J. Arntzen. 1978. Biochim. Biophys. Acta 1184:118-126.
Involvement of the light-harvesting complex in cation 28.Flachmann, R. and W. Kühlbrandt. 1996. Crystalliza-
regulation of excitation energy distribution in chloro- tion and identification of an assembly defect of recom-
plasts. Arch. Biochem. Biophys. 187:252-263. binant antenna complexes produced in transgenic
14.Butler, P.J.G. and W. Kühlbrandt. 1988. Determina- tobacco plants. Proc. Natl. Acad. Sci. USA 93:14966-
tion of the aggregate size in detergent solution of the 14971.
light-harvesting chlorophyll a/b-protein complex from 29.Francis, G.W., B. Huseby, and O.M. Andersen. 1993.
chloroplast membranes. Proc. Natl. Acad. Sci. USA An improved HPLC system for the analysis of photo-
85:3797-3801. synthetic pigments. Chromatographia 35:189-192.
15.Camm, E.L. and B.R. Green. 1980. Fractionation of 30.Fromme, P., H.T. Witt, W.D. Schubert, O. Klukas,
thylakoid membranes with the nonionic detergent W. Saenger, and N. Krauss. 1996. Structure of photo-
octyl-β-D-glucopyranoside. Plant Physiol. 66:428- system I at 4.5 angstrom resolution: a short review
432. including evolutionary aspects. Biochim. Biophys. Acta
16.Camm, E.L. and B.R. Green. 1982. The effects of 1275:76-83.
cations and trypsin on extraction of chlorophyll-pro- 31.Funk, C., W.P. Schröder, A. Napiwotzki, S.E. Tjus,
tein complexes by octyl glucoside. Arch. Biochem. Bio- G. Renger, and B. Andersson. 1995. The PSII-S pro-
phys. 214:563-572. tein of higher plants: a new type of pigment-binding
17.Cammarata, K.V., F. Plumley, and G.W. Schmidt. protein. Biochemistry 34:11133-11141.
1990. Reconstitution of light-harvesting complexes: a 32.Gantt, E., F.X. Cunningham, B. Grabowski, and S.
single apoprotein binds Chla, Chlb and xanthophylls, Tan. 1998. Relatedness of caroteno-chlorophyll anten-
p. 341-344. In M. Baltscheffsky (Ed.), Current na complexes in algae and plants, p. 239-247. In G.
Research in Photosynthesis, Vol. 2. Kluwer Academic Garab (Ed.), Photosynthesis: Mechanism and Effects,
Publishers, Dordrecht. Vol. I. Kluwer Academic Publishers, Dordrecht.
18.Cammarata, K.V. and G.W. Schmidt. 1992. In-vitro 33.Garab, G., J. Kieleczawa, J.C. Sutherland, C. Busta-
reconstitution of a light-harvesting gene product–dele- mante, and G. Hind. 1991. Organization of pig-
tion mutagenesis and analyses of pigment binding. ment–protein complexes into macrodomains in the
Biochemistry 31:2779-2789. thylakoid membranes of wild-type and chlorophyll b-
19.Clayton, R.K. and B.J. Clayton. 1981. B 850 pig- less mutant of barley as revealed by circular dichroism.
ment–protein complex of Rhodopseudomonas Photochem. Photobiol. 54:273-281.
sphaeroides. Extinction coefficients, circular dichroism, 34.Gilmore, A.M. and H.Y. Yamamoto. 1991. Resolution
and the reversible binding of bacteriochlorophyll. Proc. of lutein and zeaxanthin using a non-endcapped, lightly
Natl. Acad. Sci. USA 78:5583-5587. carbon-loaded C18 high-performance liquid chro-
20.Dainese, P. and R. Bassi. 1991. Subunit stoichiom- matographic column. J. Chromatogr. 543:137-145.
etry of the chloroplast photosystem II antenna 35.Giuffra, E., D. Cugini, R. Croce, and R. Bassi. 1996.
system and aggregation state of the component Reconstitution and pigment-binding properties of
chlorophyll a/b binding proteins. J. Biol. Chem. recombinant CP29. Eur. J. Biochem. 238:112-120.
266: 8136-8142. 36.Goldsmith, J.O. and S.G. Boxer. 1996. Rapid isola-
21.Dainese, P., G. Hoyer-Hansen, and R. Bassi. 1990. tion of bacterial photosynthetic reaction centers with
The resolution of chlorophyll a/b-binding proteins by a an engineered poly-histidine tag. Biochim. Biophys.
preparative method based on flat bed isoelectric focus- Acta 1276:171-175.
ing. Photochem. Photobiol. 51:693-703. 37.Hankamer, B., J. Nield, D. Zheleva, E. Boekema, S.
22.Davies, B.H. 1965. Analysis of carotenoid pigments, p. Jansson, and J. Barber. 1997. Isolation and biochemi-
489-532. In T.W. Goodwin (Ed.), Chemistry and Bio- cal characterisation of monomeric and dimeric photo-
chemistry of Plant Pigments. Academic Press, New York. system II complexes from spinach and their relevance
23.Davies, B.H. 1976. Carotenoids, p. 38-165. In T.W. to the organisation of photosystem II in vivo. Eur. J.
Goodwin (Ed.), Chemistry and Biochemistry of Plant Biochem. 243:422-429.
Pigments, Vol. 2. Academic Press, London. 38.Haworth, P., J.L. Watson, and C.J. Arntzen. 1983.
24.Deisenhofer, J., O. Epp, K. Miki, R. Huber, and H. The detection, isolation, and characterisation of a
Michel. 1985. Structure of the protein subunits in the light-harvesting complex which is specifically associat-
photosynthetic reaction center of Rhodopseudomonas ed with photosystem I. Biochim. Biophys. Acta
viridis at 3 Å resolution. Nature 318:618-624. 724:151-158.

251
H. Paulsen and V.H.R. Schmid

39.Hiller, R.G., C.D. Scaramuzzi, and J. Breton. 1992. reconstituted algal chlorophyll a/b-binding light-har-
The organisation of photosynthetic pigments in a Crypto- vesting complexes of Chlorella fusca with different pig-
phyte alga — a linear dichroism study. Biochim. Bio- ment compositions and pigment–protein stoichiome-
phys. Acta 1102:360-364. tries. Photosynth. Res. 49:71-81.
40.Hobe, S., R. Förster, J. Klingler, and H. Paulsen. 56.Mullet, J.E. and C.J. Arntzen. 1980. Simulation of
1995. N-proximal sequence motif in light-harvesting grana stacking in a model membrane system. Media-
chlorophyll a/b-binding protein is essential for the tion by a purified light-harvesting pigment–protein
trimerization of light-harvesting chlorophyll a/b com- complex from chloroplasts. Biochim. Biophys. Acta
plex. Biochemistry 34:10224-10228. 589:100-117.
41.Hobe, S., S. Prytulla, W. Kühlbrandt, and H. 57.Mullet, J.E., J.J. Burke, and C.J. Arntzen. 1980.
Paulsen. 1994. Trimerization and crystallization of Chlorophyll proteins of photosystem I. Plant Physiol.
reconstituted light-harvesting chlorophyll a/b complex. 65:814-822.
EMBO J. 13:3423-3429. 58.Nagai, K. and H.C. Thøgersen. 1987. Synthesis and
42.Iriyama, K., N. Ogura, and A. Takamiya. 1974. A sequence-specific proteolysis of hybrid proteins pro-
simple method for extraction and partial purification duced in E. coli. Methods Enzymol. 153:461-481.
of chlorophyll from plant material, using dioxane. J. 59.Neville, D.M. 1971. Molecular weight determination
Biochem. 76:901-904. of protein-dodecyl sulfate complexes by gel elec-
43.Kamimura, Y., T. Mori, T. Yamasaki, and S. Katoh. trophoresis in a discontinous buffer system. J. Biol.
1997. Isolation, properties and a possible function of a Chem. 246:6328-6334.
water soluble chlorophyll a/b protein from Brussels 60.Nishio, N. and H. Satoh. 1997. A water-soluble
sprouts. Plant Cell Physiol. 38:133-138. chlorophyll protein in cauliflower may be identical to
44.Kan, K.S. and J.P. Thornber. 1976. The light-harvest- Bnd22, a drought-induced, 22-kilodalton protein in
ing chlorophyll a/b-protein of Chlamydomonas rein- rapeseed. Plant. Physiol. 115:841-846.
hardtii. Plant Physiol. 57:47-53. 61.Nußberger, S., K. Dörr, D.N. Wang, and W.
45.Knötzel, J., I. Svendsen, and D.J. Simpson. 1992. Kühlbrandt. 1993. Lipid-protein interactions in crys-
Identification of the photosystem-I antenna polypep- tals of plant light-harvesting complex. J. Mol. Biol.
tides in barley — isolation of 3 pigment-binding 234:347-356.
antenna complexes. Eur. J. Biochem. 206:209-215. 62.Pagano, A., G. Cinque, and R. Bassi. 1998. In vitro
46.Krupa, Z., N.P.A. Huner, J.P. Williams, E. Maissan, reconstitution of the recombinant photosystem II
and D.R. James. 1987. Development at cold-harden- light-harvesting complex CP24 and its spectroscopic
ing temperatures. The structure and composition of characterization. J. Biol. Chem. 273:17154-17165.
purified rye light harvesting complex II. Plant Physiol. 63.Palsson, L.O., S.E. Tjus, B. Andersson, and T. Gill-
84:19-24. bro. 1995. Ultrafast energy transfer dynamics resolved
47.Kügler, M., L. Jansch, V. Kruft, U.K. Schmitz, and in isolated spinach light-harvesting complex I and the
H.P. Braun. 1997. Analysis of the chloroplast protein LHC I-730 subpopulation. Biochim. Biophys. Acta
complexes by blue-native polyacrylamide gel elec- 1230:1-9.
trophoresis (BN-PAGE). Photosynth. Res. 53:35-44. 64.Parkes-Loach, P.S., J.R. Sprinkle, and P.A. Loach.
48.Kühlbrandt, W. 1984. Three-dimensional structure of 1988. Reconstitution of the B873 light-harvesting
the light-harvesting chlorophyll a/b-protein complex. complex of Rhodospirillum rubrum from the separately
Nature 307:478-480. isolated alpha and beta-polypeptides and bacteri-
49.Kühlbrandt, W., D.N. Wang, and Y. Fujiyoshi. 1994. ochlorophyll a. Biochemistry 27:2718-2727.
Atomic model of plant light-harvesting complex by 65.Patzlaff, J.S. and B.A. Barry. 1996. Pigment quantita-
electron crystallography. Nature 367:614-621. tion and analysis by HPLC reverse phase chromatogra-
50.Laemmli, U.K. 1970. Cleavage of structural proteins phy: a characterization of antenna size in oxygen-evolv-
during the assembly of the head of bacteriophage T4. ing photosystem II preparations from cyanobacteria
Nature 227:680-685. and plants. Biochemistry 35:7802-7811.
51.Lam, E., W. Ortiz, and R. Malkin. 1984. Chlorophyll 66.Paulsen, H., B. Finkenzeller, and N. Kühlein. 1993.
a/b proteins of photosystem I. FEBS Lett. 168:10-14. Pigments induce folding of light-harvesting chlorophyll
52.Martinson, T.A. and F.G. Plumley. 1995. One-step a/b-binding protein. Eur. J. Biochem. 215:809-816.
extraction and concentration of pigments and acyl 67.Paulsen, H. and S. Hobe. 1992. Pigment-binding
lipids by sec-butanol from in vitro and in vivo samples. properties of mutant light-harvesting chlorophyll a/b-
Anal. Biochem. 228:123-130. binding protein. Eur. J. Biochem. 205:71-76.
53.McDermott, G., S.M. Prince, A.A. Freer, A.M. 68.Paulsen, H., U. Rümler, and W. Rüdiger. 1990.
Hawthornthwaite-Lawless, M.Z. Papiz, R.J. Cogdell, Reconstitution of pigment-containing complexes from
and N.W. Isaacs. 1995. Crystal structure of an integral light-harvesting chlorophyll a/b-binding protein over-
membrane light-harvesting complex from photosyn- expressed in E. coli. Planta 181:204-211.
thetic bacteria. Nature 374:517-521. 69.Peter, G.F. and J.P. Thornber. 1991. Biochemical
54.Meyer, M. and C. Wilhelm. 1993. Reconstitution of composition and organization of higher plant photo-
light-harvesting complexes from Chlorella fusca system II light-harvesting pigment–proteins. J. Biol.
(Chlorophyceae) and Mantoniella squamata (Prasino- Chem. 266:16745-16754.
phyceae). Z. Naturforsch. C 48:461-473. 70.Peter, G.F. and J.P. Thornber. 1991. Electrophoretic
55.Meyer, M., C. Wilhelm and G. Garab. 1996. Pig- procedures for fractionation of photosystems I and II
ment-pigment interactions and secondary structure of pigment proteins of higher plants and for determina-

252
Analysis and Reconstitution of Chlorophyll–Proteins

tion of their subunit composition, p. 195-210. In L.G. 84.Sarvari, E. and P. Nyitrai. 1994. Separation of chloro-
Rogers (Ed.), Methods in Plant Biochemistry, Vol. 5. phyll-protein complexes by Deriphat polyacrylamide
Academic Press, New York. gradient gel electrophoresis. Electrophoresis 15:1068-
71.Peterson, G.L. 1977. A simplification of the protein 1071.
assay method of Lowry et al. which is more generally 85.Schmid, V. and C. Schäfer. 1994. Alterations of the
applicable. Anal. Biochem. 83:346-356. chlorophyll-protein pattern in chronically photo-
72.Plumley, F.G. and G.W. Schmidt. 1987. Reconstitu- inhibited Chenopodium rubrum cells. Planta 192:473-
tion of chlorophyll a/b light-harvesting complexes: 479.
xanthophyll-dependent assembly and energy transfer. 86.Schmid, V.H.R., K.V. Cammarata, B.U. Bruns, and
Proc. Natl. Acad. Sci. USA 84:146-150. G.W. Schmidt. 1997. In vitro reconstitution of the
73.Porra, R.J., W.A. Thompson, and P.E. Kriedemann. photosystem I light-harvesting complex LHCI-730:
1989. Determination of accurate extinction coeffi- heterodimerization is required for antenna pigment
cients and simultaneous equations for assaying chloro- organization. Proc. Nat. Acad. Sci. USA 94:7667-
phylls a and b extracted with four different solvents: 7672.
verification of the concentration of chlorophyll stan- 87.Setlikova, E., S. Ritter, R. Hienerwadel, J. Kopecky, J.
dards by atomic absorption spectroscopy. Biochim. Komenda, W. Welte, and I. Setlik. 1995. Purification
Biophys. Acta 975:384-394. of a photosystem II reaction center from a thermophilic
74.Rhee, K.H., E.P. Morris, J. Barber, and W. cyanobacterium using immobilized metal affinity chro-
Kühlbrandt. 1998. Three-dimensional structure of the matography. Photosynth. Res. 43:201-211.
plant photosystem II reaction centre at 8 Å resolution. 88.Smith, P.K., R.I. Krohn, G.T. Hermanson, A.K.
Nature 396:283-286. Mallia, F.H. Gartner, M.D. Provenzano, E.K. Fuji-
75.Rhee, K.H., E.P. Morris, D. Zheleva, B. Hankamer, moto, N.M. Goeke, B.J. Olson, and D.C. Klenk.
W. Kühlbrandt (Reprint Author), and J. Barber. 1985. Measurement of protein using bicinchoninic
1997. Two dimensional structure of plant photosystem acid. Anal. Biochem. 79:76-85.
II at 8 Å resolution. Nature 389:522-526. 89.Tan, S., G.R. Wolfe, F.X. Cunningham, and E.
76.Rhiel, E., W. Lange, and E. Mörschel. 1993. The Gantt. 1995. Decrease of polypeptides in the PS I
unusual light-harvesting complex of Mantoniella squa- antenna complex with increasing growth irradiance in
mata — supramolecular composition and assembly. the red alga Porphyridium cruentum. Photosynth. Res.
Biochim. Biophys. Acta 1143:163-172. 45:1-10.
77.Ritter, S., J. Komenda, E. Setlikova, I. Setlik, and W. 90.Thornber, J.P. 1995. Thirty years of fun with antenna
Welte. 1992. Immobilized metal affinity chromatogra- pigment–proteins and photochemical reaction centers:
phy for the separation of photosystem I and photosys- a tribute to the people who have influenced my career.
tem II from the thermophilic cyanobacterium Syne- Photosynth. Res. 44:3-22.
chococcus elongatus. J. Chromatogr. 625:21-31. 91.Tjus, S.E., M. Roobol-Boza, L.O. Palsson, and B.
78.Robinson, N.C., D. Wiginton, and L. Talbert. 1984. Andersson. 1995. Rapid isolation of Photosystem I
Phenyl sepharose-mediated detergent-exchange chro- chlorophyll-binding proteins by anion exchange perfu-
matography: its application to exchange of detergents sion chromatography. Photosynth. Res. 45:41-49.
bound to membrane proteins. Biochemistry 23:6121- 92.Todd, J.B., P.S. Parkes-Loach, J.F. Leykam, and P.A.
6126. Loach. 1998. In vitro reconstitution of the core and
79.Rogl, H., K. Kosemund, W. Kühlbrandt, and I. peripheral light-harvesting complexes of Rhodospiril-
Collinson. 1998. Refolding of Escherichia coli pro- lum molischianum from separately isolated compo-
duced membrane protein inclusion bodies immo- nents. Biochemistry 37:17458-17468.
bilised by nickel chelating chromatography. FEBS Lett. 93.Val, J., E. Monge, and N.R. Baker. 1994. An
432:21-26. improved HPLC method for rapid analysis of the xan-
80.Rögner, M., U. Mühlenhoff, E.J. Boekema, and H.T. thophyll cycle pigments. J. Chromatogr. Sci. 32:286-
Witt. 1990. Mono-, di- and trimeric PS I reaction cen- 289.
ter complexes isolated from the thermophilic 94.van Amerongen, H. and W.S. Struve. 1995. Polarized
cyanobacterium Synechococcus sp.* — size, shape and optical spectroscopy of chromoproteins. Biochem.
activity. Biochim. Biophys. Acta 1015:415-424. Spectroscopy 246:259-283.
81.Roobol-Boza, M. and B. Andersson. 1996. Isolation 95.van der Staay, G.W.M., A. Brouwer, R.L. Baard, F.
of hydrophobic membrane proteins by perfusion chro- Vanmourik, and H.C.P. Matthijs. 1992. Separation of
matography — purification of photosystem II reaction photosystem I and photosystem II from the oxy-
centers from spinach chloroplasts. Anal. Biochem. chlorobacterium (prochlorophyte) Prochlorothrix hol-
235:127-133. landica and association of chlorophyll-b binding anten-
82.Ros, F., R. Bassi, and H. Paulsen. 1998. Pigment- nae with photosystem II. Biochim. Biophys. Acta
binding properties of the recombinant photosystem II 1102:220-228.
subunit CP26 reconstituted in vitro. Eur. J. Biochem. 96.van Holde, K.E., C.E. Johnson, and P.S. Ho. 1998.
253:653-658. Physical Biochemistry. Prentice Hall, London.
83.Sandoná, D., R. Croce, A. Pagano, M. Crimi, and R. 97.Welte, C., R. Nickel, and A. Wild. 1995. Three-
Bassi. 1998. Higher plants light harvesting proteins. dimensional crystallization of the light-harvesting
Structure and function as revealed by mutation analysis complex from Mantoniella squamata (Prasinophyceae)
of either protein or chromophore moieties. Biochim. requires an adequate purification procedure. Biochim.
Biophys. Acta 1365:207-214. Biophys. Acta 1231:265-274.

253
Two-Dimensional Crystallization of
11 Chlorophyll Proteins

Georgios Tsiotis
Department of Chemistry, University of Crete, Heraklion, Greece

1. INTRODUCTION types of RCs probably have a common


evolutionary origin, as they apparently use
1.1. Photosynthesis Begins with the a special pair of chlorophyll (Chl) or bacte-
Chlorophylls riochlorophyll (BChl) molecules as the pri-
mary electron donor and a Chl or pheo-
The conversion of light energy into the phytin as the primary electron acceptor (2).
free energy of organic compounds is the The photosystems are large multisubunit
quintessence of photosynthesis. The pri- protein complexes integral to the thylakoid
mary process is the absorption of light by membrane.
the antenna complexes and its transfer to The initial step in the energy conversion
the reaction center (RC) where photo- process of photosynthesis is absorption of
chemical energy storage take places. These light by Chls (or BChls). Most of the Chl
are the most important proteins that cat- (or BChl) molecules in the photosynthetic
alyze the conversion of light energy into organisms serve as an antenna for light har-
chemical energy and are known as type-I vesting. Only one Chl molecule out of
and type-II RCs. Type-I RCs use iron–sul- approximately 500, and one BChl mole-
fur clusters and are exemplified by photo- cule out of 50 to 100, is directly connected
system I (PSI), found in plants, eukaryotic to the electron transfer chain through the
algae, and cyanobacteria. This complex reaction centers that trap excitation energy
catalyses the light-driven electron transfer by rapid electron transfer to an acceptor.
from reduced plastocyanin to oxidized The pigment molecules involved in anten-
ferredoxin. Type-II RCs use quinones as na light gathering function are bound to
electron acceptors, the best well-known proteins of relatively small size (10–30
example being photosystem II (PSII), kDa), known as light-harvesting (LH) pro-
which is found in all oxygen-evolving pho- teins, which are again integral membrane
tosynthetic organisms, where it carries out proteins (21).
the photochemical oxidation of water and In higher plants, algae, and cyanobacte-
produces reduced plastoquinone. The two ria, the two photosystems are linked by the

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

255
G. Tsiotis

cytochrome b6f complex. This is a plasto- proteins could be handled analogously to


quinol:plastocyanin oxidoreductase, which soluble proteins with respect to crystalliza-
is an integral membrane protein complex tion have not been realized. For this reason,
that participates in electron transfer and different attempts were undertaken to
the generation of an electrochemical pro- resolve this problem, such as the successful
ton gradient in oxygenic photosynthesis. co-crystallization of the cytochrome c oxi-
In this chapter, various techniques for dase with antibody fragments (39) and the
obtaining two-dimensional (2D) crystals of use of the cubic lipid phase to obtain high
chlorophyll a-containing membrane pro- quality 3D crystals of bacteriorhodopsin
teins and a cytochrome b6f complex are (29).
discussed. Electron crystallography of 2D crystals
offers a viable alternative to X-ray crystal-
1.2. Structural Analysis of Membrane lography. The inherent adaptation of mem-
Proteins brane proteins for the 2D environment of
the lipid bilayer suggests that the most
Membrane proteins are amphiphilic appropriate geometry for a regularly
macromolecules incorporated vectorially in packed protein would be a 2D crystal.
the fluid lipid membrane. Upon disruption Suitable 2D crystals are more easily pro-
of the membrane and removal of the lipids duced than 3D crystals, because membrane
by detergents, such proteins tend to aggre- proteins have a tendency to pack within a
gate nonspecifically and to precipitate. This lipid bilayer. The ultimate goal of electron
can be avoided, and the proteins can be microscopy-based techniques applied to
solubilized in aqueous solutions, when proteins is the production of images from
detergents replace the membrane lipids. which a clear depiction of the internal
While structure determination of soluble atomic structure can be derived. Methods
proteins by X-ray crystallography and and instruments have been developed
nuclear magnetic resonance (NMR) has which potentially allow analysis at near
progressed at a remarkable rate, this has atomic resolution (16,17).
been far less successful for membrane pro- There are several inherent advantages of
teins, as manifested by the small number electron microscopy when compared to
solved to atomic resolution. Three-dimen- X-ray crystallography for the study of mem-
sional (3D) crystals that allowed atomic brane proteins: (i) 2D crystals of membrane
resolution by X-ray crystallography have proteins form in a continuous lipid bilayer.
been obtained for the bacterial porins (25, Thus, the protein environment in the crys-
44,50), the photosynthetic RCs (1,34), tal is thought to be similar to that experi-
two cytochrome c oxidases (20,48), the enced in vivo; (ii) during 2D crystallization,
purple bacterium light-harvesting complex the time taken to effect crystallization is
(LHC) II (32), and bacteriorhodopsin short, hence opening up the possibility of
(40). The requirement that membrane pro- structural analysis on membrane proteins
teins must be solubilized in detergents not sufficiently stable for 3-D crystallization
leads to large complexes not suitable for trials; (iii) generally, 2D crystallization is
study by NMR. more readily achievable than 3D crystalliza-
In most cases, solubilized membrane tion; and (iv) phase information can be
proteins have been crystallized by conven- measured directly, eliminating the need to
tional methods that foster interaction of produce “heavy atom derivatives”, and is
hydrophilic protein surfaces. The initial generally of higher accuracy than those
hopes that detergent-solubilized membrane obtained in 3D crystallography.
256
Two-Dimensional Crystallization of Chlorophyll Proteins

Successful imaging of biological mole- gent is critical. Ideally, detergent and isola-
cules requires the adoption of experimental tion protocols should be selected not only
protocols that take into account the fragili- to yield a homogeneous structural state but
ty of such molecules when exposed to the also to preserve the protein in a unique
hostile environment encountered in an functional state (11,22). The pure protein
electron microscope. Initially, due to the is obtained in a detergent solution, often
high vacuum, it is essential to provide pro- with residual lipids. In fact, the latter often
tection against dehydration and stabiliza- contribute to the stability of the membrane
tion against molecular collapse. Methods protein and may be essential for successful
such as freezing in vitreous ice (8), sugar 2D crystallization.
embedding (16), and negative staining
(14) have been developed for this purpose. 2.1.1. Detergent Choice and
Frozen-hydrated and sugar-embedded Concentration
techniques yield information on internal
protein density, whereas negative staining Many membrane proteins are destabi-
provides information only on the molecu- lized on extraction from their native mem-
lar environment which is stain accessible. branes, especially when short-chain [high
critical micellar concentration (CMC)]
detergents are used. Although the proteins
2. TWO-DIMENSIONAL can be effectively solubilized with deter-
CRYSTALLIZATION gents that replace the lipid and keep the
hydrophobic surfaces of the protein shield-
2D crystals of membrane proteins have ed from water, delipidation may destabilize
been obtained in three different ways: (i) the protein. The choice of detergent is crit-
reconstitution of the purified membrane ical; there is a fine balance between disrup-
protein into a lipid bilayer at high protein tion of the membrane to solubilize a mem-
density; (ii) improvement of the packing of brane protein and preserving its structural
a highly abundant protein into regular integrity. As discussed above, reconstitu-
arrays in its native membrane; and (iii) tion is closely linked to the properties of
addition of precipitants to promote pro- the detergents used both during purifica-
tein–protein interactions, analogous to 3D tion and the reconstitution itself. There-
crystallization, but yielding 2D crystals. fore, the detergents used for purification
can be exchanged for a different detergent
2.1. Crystallization of Purified used for reconstitution. This makes possi-
Membrane Protein by ble the use of mild detergents for isolation
Reconstitution into Lipid Bilayers (most with long alkyl chain and low
CMC) and then to change to detergents
The crystallization of membrane pro- with high CMC for the crystallization.
teins in 2D arrays requires membrane pro- Among the detergents with an alkyl chain,
teins which are solubilized and purified to those with a charged head group (e.g.,
homogeneity. A unique oligomeric species dodecyl sulfate and hexatrimethylammoni-
possessing a high intrinsic molecular sym- um) are generally not suited for isolation of
metry is likely to favor the growth of “native” membrane proteins. Zwitterionic
crystalline arrays. Indeed, heterogeneous alkyl detergents (e.g., sulfobetains) and
protein solutions and the presence of dena- those with an N-oxide as polar head group
tured proteins greatly hinder the formation can be used with many proteins, but they
of crystalline patches. The choice of deter- are still too harsh for most of the mem-
257
G. Tsiotis

brane proteins. Among the detergents with of Chl a and b at 664 and 647 nm,
an alkyl chain, those with polyoxyethylene respectively.
(e.g., C12E8, Lubrol-series, Triton® X- Chl a (µM) = 13.19 × A664 - 2.57 × A647
series, and Brij-series) or a disaccharide Chl b (µM) = 21.10 × A647 - 5.26 × A664
head group are the mildest (33). Different Total Chl (µM) = 7.93 × A664 + 19.53 × A647
protocols have been developed for the For cyanobacteria, which contain only
exchange of detergent using ultrafiltration Chl a, the concentration of an 80% ace-
(e.g., with Centricon-100 concentrator tone extract can be calculated from A663
devices [Amicon Division, W.R. Grace, using the formula:
MA, USA]), gel filtration, anion or cation
Chl a (µg/mL) = 12.2 × A663 (ε663 =
exchange chromatography, sucrose density
82 mg-1.Chl a.L-1)
gradient, isoelectric focusing, and selective
absorption onto hydrophobic resin beads.
❖ Procedure 2. Bio-Rad Protein Assay
2.1.2. Determination of Chlorophyll This assay is based on the observation
and Protein Content that the absorbance maximum for an acidic
solution of Coomassie® Brilliant Blue G-
To set up optimal 2D crystallization trials,
250 changes from 465 to 595 nm when
accurate knowledge of both lipid and protein
bound to protein (5).
is required, and the following procedures
have been used with success in my laboratory. 1. Prepare a set of protein standards by
An advantage of Chl proteins is that the chro- diluting a stock of 2 mg/mL bovine
mophores that are present (see Procedure 1 serum albumin (BSA) standard in the
and Chapter 10 in this book by Paulsen and buffer used for the experimental sam-
Schmid) correlate with the protein content ples. Include a blank with no BSA.
(see Procedures 2 or 3 in this chapter). 2. Place 0.1 mL of each standard concen-
tration of BSA and appropriately dilut-
❖ Procedure 1. Determination of ed experimental samples in test tubes.
Chlorophyll Concentration 3. Add 5.0 mL of the Bio-Rad protein assay
dye reagent concentrate (Bio-Rad Labo-
Absorption spectroscopy is a quick and ratories, Hercules, CA, USA) (diluted 1:4
easy method for the estimation of chro- with water) to each test tube and vortex mix.
mophore content of Chl binding proteins. 4. After a standard period from 5 minutes
It is potentially nondestructive, although to 1 hour, measure the absorbance at
Chls are usually extracted from the protein 595 nm.
using organic solvents. 5. Plot the absorbance at 595 nm versus
1. Dilute 0.1 mL of the sample to 20 mL the protein concentration of standards.
(200-fold) with 80% acetone. 6. Estimate the protein concentration of
2. Mix well and centrifuge at 3000× g for experimental samples from the stan-
5 minutes. dard curve.
3. Measure the absorbance (A) of the ace-
tone extract at 664 and 647 nm in a 1- ❖ Procedure 3. Bicinchoninic Acid Assay
cm path length cuvette. to Determine Protein Concentration
4. Use the following equation to calculate
the Chl a, Chl b, and total Chl derived The water-soluble salt of bicinchoninic
from the known extinction coefficients acid (BCA) is a specific reagent for Cu+.

258
Two-Dimensional Crystallization of Chlorophyll Proteins

Peptide bonds and 4 amino acids (cysteine, and transition temperatures, and they also
cystine, tryptophan, and tyrosine) reduce provide mixtures of head group charges and
cupric (Cu2+) ions in alkaline medium to molecular geometries similar to membranes
Cu+ (Biuret reaction). The reaction prod- from which the protein originated. Howev-
uct of 2 molecules BCA and 1 Cu+ exhibits er, the complexity of such preparations may
a strong absorbance at 562 nm. Using preclude crystal formation. Nevertheless,
BCA protein assay reagent available from since synthetic lipids, Escherichia coli lipids,
Pierce Chemical (Rockford, IL, USA), the soybean lecithin, and egg lecithin have all
method below should be followed. been successfully used for 2D crystalliza-
1. Prepare a set of protein standards of tion, no general recommendations can be
known concentration by diluting a made on which lipid or lipid mixture is
stock 2 mg/mL BSA standard in the most suitable for any one particular mem-
buffer used for the experimental sam- brane protein. Commercially available
ples. Include a blank with no BSA. lipids (from Sigma [St. Louis, MO, USA]
2. Prepare a working reagent by mixing or Lipid Products [South Nutheld Redhill,
50 parts Reagent A with 1 part Reagent B. Surrey RH1 5 PG, UK]) are often stored as
chloroform solutions or a lyophilized pow-
3. Dilute 0.1 mL of each standard con-
der at -20°C where they are stable for long
centration of BSA and each experimen-
periods. Prior to use for 2D crystallization
tal sample to 2 mL in a working re-
trials, lipids have to be transferred to deter-
agent and incubate for 30 minutes at
gent-containing buffer solutions. The lipid
37°C or room temperature for 2 hours.
content of the reconstitution mixture is, in
4. Allow the samples to cool and measure general, a well-controlled parameter.
the absorbance at 562 nm. Lyophilized lipids can easily be weighed
5. Prepare a standard curve by plotting and redissolved in buffer at 1 to 10 mg/mL
the absorbance at 562 nm versus the containing a high concentration of deter-
protein concentration. Using the stan- gent. Lipids from chloroform can be han-
dard curve, determine the protein con- dled in the same way after removal of the
centration for each unknown protein organic solvent. The amount of detergent
sample. to solubilize completely a lipid stock can be
Since color development will continue calculated by use the following equation:
slowly, it is necessary that all absorbance Concentration of detergent (mol/L) =
readings be done immediately. Care must CMC (mol/L) + 3 × concentration of the
be taken to avoid the presence of reducing lipid (mol/L).
agents such as thiols [dithiothreitol (DTT) All organic solvent should be removed
or mercaptoethanol] or large amounts of before solubilization in detergent buffer, as
sugars, which will interfere with the assay. it interferes with the crystallization.

2.1.3. Lipid Mixture for Reconstitution 2.1.4. Lipid to Protein Ratio


The lipid mixture used for reconstitu- The reconstitution of membrane pro-
tion has an influence on crystallization teins into bilayers is achieved by mixing
results. Crystallization is more likely to lipids and protein, both solubilized in deter-
occur when the lipid bilayer is in the fluid gents, and decreasing the detergent concen-
phase and thus allows some lateral mobility tration. Figure 1 shows an example where
of the inserted membrane proteins. Native the concentration of octyl-polyoxyethylene
lipids are often ideal in terms of stability (8-POE) was decreased by dilution, and the
259
G. Tsiotis

formation of structures of different sizes was region of detergent concentration where


monitored using dynamic light scattering lipid bilayer and mixed micellar structures
(7). The dilution experiment led to the for- coexist. The micelle-bilayer transition
mation of vesicles with egg phosphatidyl- region (Stage II) was found to be the key to
choline (EggPC) or vesicles and 2D crystals reconstitution and, by implication, to 2D
with EggPC and the porin OmpF. The lat- crystallization (7,11). Cryo-electron
ter assembled only if the dilution rate was microscopy (9) has shown for several
slow. The relationship between detergent lipid–detergent systems that this transition
concentration and structure sizes can be involves the formation of worm-like extend-
described as the “3-stage” model of Lichten- ed lipid micelles, probably capped by deter-
berg et al. (30). Stage I (crystals/vesicles gents that must convert to vesicles on deter-
area) is characterized by a detergent concen- gent removal. Such rod-like structures are
tration too low to disrupt the lipid bilayer. therefore thought to be important interme-
Stage II (intermediate structures area) is the diates in the formation of 2D crystals.

Figure 1. Hydrodynamic radii on dilution of a mixed micellar suspension containing a detergent (8-POE) and either lipid
(EggPC) or lipid and a membrane protein (porin OmpF). Above 15 mM 8-POE, uniform micelles are seen. Below 15 mM 8-
POE, each treatment produces a range of structure radii with large crystals seen in the presence of OmpF. The dilution is repre-
sented as a function of detergent concentration to illustrate the 3-stage model. Large bilayer structures (crystals/vesicles) at low
detergent concentration (Stage I). Small micelles at high detergent concentration (Stage III). Mixture of structures at intermedi-
ate detergent concentration (Stage II). Black arrows indicate the saturation points, and white arrows indicate the solubilization
points. Data from Reference 7. Figure was modified from Reference 15.

260
Two-Dimensional Crystallization of Chlorophyll Proteins

At the start of a typical reconstitution all components by equal factors, until the
experiment, an excess of detergent ensures a free detergent concentration drops below
homogenous distribution of protein and saturation. Crystallization by the dilution
lipid in micelles. As detergent concentration method requires a significant dilution of
is decreased, lipids and proteins interact due the protein, and therefore, rather high ini-
to the exposure of their hydrophobic sur- tial concentrations are required. On the
faces. With an excess of lipid over protein, other hand, the dilution method allows the
the protein is mainly incorporated into lipid process to be arrested when the saturation
bilayers, similar to its native state. In an point is reached, extending the time in
excess of protein over lipid, the protein which an ordered assembly of the compo-
mostly ends up in amorphous aggregates, nents can take place. As an illustration of
perhaps denatured. An important parameter this, the following 2 procedures describe
is, therefore, the lipid:protein ratio (LPR), the preparation of 2D crystals of PSI com-
which should be low enough to promote plexes from the thermophilic cyanobacteri-
crystal contacts between protein molecules, um Synechococcus sp. OD 24 (Procedure 4)
but not so low that the protein is lost to and tubular crystals of spinach PSII (Pro-
aggregation. When the membrane protein is cedure 5).
reconstituted from a mixture of solubilized
components, crystal ordering of proteins ❖ Procedure 4. 2D Crystals of the PSI
may occur during reconstitution. For crystal Complex from Synechococcus sp. OD
packing during reconstitution, the LPR of 24 (7)
the reconstitution experiment must be as
low as possible to ensure close packing with- PSI complexes are isolated from Syne-
out leading to excessive aggregation. While chococcus sp. OD 24 according to Refer-
the lipid content of the reconstitution mix- ence 24.
ture is, in general, a well-controlled parame-
1. Make 600 µL of a starting mixture
ter, the content of monodisperse protein is
containing 1 mg/mL PSI, 1 mg/mL
sometimes unknown, because protein assays
lecithin, and 7.5 mg/mL octyl-β-thio-
do not indicate the amount of aggregates.
glucoside (OTG).
2. Dilute this mixture by the addition of
2.2. Crystallization Methods sequential 25-µL aliquots of 10 mM 2-
[N-morpholino]ethanesulfonic acid
The manner in which the detergent (MES), pH 6.0, 100 mM NaCl, 10
concentration is decreased for reconstitu- mM MgCl2 to achieve a slow dilution
tion and subsequent 2D crystallization is of PSI.
an important consideration. The common- 3. After the addition of 4 aliquots (i.e.,
ly used techniques for detergent removal 100 µL, thus reducing the concentra-
are dilution (7,47), dialysis (11,22), and tion of OTG to about 6 mM), large
selective adsorption of the detergent on interconnected protein–lipid aggre-
solid supports such as the hydrophobic gates can be observed.
resin beads (43). 4. Upon further dilution of detergent (ad-
dition of 12 aliquots, i.e., 300 µL), dis-
2.2.1. Dilution Method tinct vesicles can be seen. Crystalline
packing of PSI complexes should be
Diluting a solution of protein, lipid, and obtained after the addition of 20
detergent decreases the concentrations of aliquots (i.e., 500 µL).
261
G. Tsiotis

❖ Procedure 5. Tubular Crystals of maintained across the dialysis membrane,


PSII Complex from Spinach (47) which improves reproducibility. A draw-
back of the method is the long dialysis
1. Isolate PSII complexes from spinach times needed to remove low CMC deter-
leaves as described in Reference 36. gents, making it only practical for medium
2. Resuspend the complex in 50 mM to high CMC detergents (typically CMC >
MES, pH 6.0, 0.4 M sucrose, 10 mM 1 mM) and for proteins with high struc-
NaCl, 0.4% OTG. tural stability. Before use, it is necessary to
3. Adjust the Chl concentration to 1 carry out pretreatment of the dialysis mem-
mg/mL. branes (Procedure 6).
4. Mix with dimyristoyl phosphatidyl
choline (DMPC), which has been solu- ❖ Procedure 6. Pretreatment of the
bilized in 1.3% OTG to obtain a ratio Dialysis Membranes
of 1:1 with Chl.
5. Dilute with 50 mM N-[2-hydroxyethyl] 1. Heat the dialysis tubing (molecular
piperazine-N′-[2-ethanesulfonic acid] weight cutoff 6000–8000, Spectra/Por
(HEPES) over a 4× range. 1) in a 2-L beaker of boiling 50%
6. Incubate at 22°C for 1 day in the dark. ethanol for 1 hour. Use a 1-L beaker
A green pellet indicates the formation containing water to weigh down the
of aggregates. dialysis tubing.
7. Centrifuge for 5 minutes (4000× g) 2. Rinse the dialysis tubing well with sev-
and wash the pellet with the same eral changes of distilled water.
buffer to separate the crystals from the 3. Heat the dialysis tubing in boiling 10 mM
remaining material. Na2CO3, 1 mM EDTA for 1 hour.
8. Repeat the wash step twice more. 4. Rinse with distilled water as before.
The tubular crystals obtained have a 5. Heat the dialysis tubing in boiling dis-
length of 1 to 2 µm and a diameter of 72.9 tilled water for 1 hour.
nm. The rhombic unit cell (a = 16.2 nm, b 6. Store in distilled water containing
= 13.7 nm, γ = 142.4°) contains one PSII 0.05% NaN3 at 4°C.
complex.

2.2.2. Dialysis Method ❖ Procedure 7. 2D Crystals of PSI


Synechococcus sp. OD 24 (24)
Dialysis is the most widely used tech-
nique in 2D crystallization trials, usually in Trimer PSI complexes were isolated
the form of small sample compartments from the thermophilic cyanobacterium
dialyzed against large buffer volumes. To Synechococcus sp. OD 24 as described in
improve the reproducibility of crystalliza- Reference 12.
tion conditions, a temperature-controlled 1. Change the detergent Triton X-100 by
continuous flow dialysis apparatus was polyethyleneglycol (PEG) 6000/MgCl2
developed (22) (see Figure 2). The advan- precipitation.
tage of this system is a precise control of 2. Repeat this step 3 times.
the temperature profile that was found to 3. Resuspend the precipitated PSI in 10
be quite critical in the 2D crystallization of mM HEPES, pH 7.0, containing
membrane proteins. Additionally, a maxi- 0.5% OTG to a protein concentration
mal gradient of detergent concentration is of 2 mg/mL.
262
Two-Dimensional Crystallization of Chlorophyll Proteins

4. Add DMPC solution to achieve a LPR β-glucoside (OG), pH 7.8. After dialysis
of 1. against buffer containing 0.8% OG, 5 mM
5. Dialyze the mixture against a deter- MgCl2, and 50 mM NaCl in the dark at
gent-free buffer containing 25 mM 4°C for 5 days, 2D crystals were obtained,
ammonium ferric citrate. which had a hexagonal pattern with a lat-
Dialysis cell temperature: 26°C for tice constant of 12.3 nm. Modification of
24 hours; increase to 37°C over 12 hours; this procedure, by the use of sonicated vesi-
37°C for 24 hours; and decrease to 26°C cles of dioleoyl-9-10 phosphatidylcholine
over 10 hours. Total dialysis time is 70 lipid solubilized in OG in a ratio 1:1 to
hours. proteins, allowed the formation of crystals
Digital image processing of negatively which diffracted at 8.5 Å (23). Digital
stained and frozen-hydrated specimens pre- image processing of frozen-hydrated speci-
pared using Procedure 7 revealed ortho- mens revealed crystals with a p22121 sym-
rhombic crystals with unit cell dimensions a metry and unit cell with dimensions of a =
= 13.8 nm, b = 14.5 nm, and p121 symme- 12.8 nm, b = 19.4 nm.
try. The same procedure has also been used As an alternative to the dialysis appara-
with trimeric PSI isolated from mesophilic tus shown in Figure 2, an inexpensive mi-
cyanobacterium Synechococcus PCC 7002 by crodialysis arrangement with Eppendorf®
isoelectric focusing (46) to provide 2D crys- tubes or dialysis buttons may be used (Fig-
tals (Tsiotis, unpublished results). ure 3). This method enables the dialysis of
Purple sulfur and nonsulfur bacteria small volumes (<50 µL) of protein–lipid–
possess membrane-bound LH complexes, detergent. Another interesting microdialy-
which serve to transfer energy to the RC, sis device using a bent glass capillary tube,
where charge separation occurs. LH com- shown in Figure 4 has been described by
plex (170 µg), isolated from a carotenoid- Kuhlbrandt (26). A glass tube of 2.5 mm
less mutant of the purple nonsulfur bac- inner diameter and about 6.5 mm outer
terium Rhodospirillum rubrum G9 as diameter is bent by 90° near to one end.
described previously (13), was dissolved in The end is melted into a smooth surface,
100 µL of 50 mM NH4HCO3, 1% octyl- which forms a tight seal with the dialysis

Figure 2. Continuous dialysis system for the growing of 2D crystals.

263
G. Tsiotis

membrane. The dialysis membrane is fixed and the device is placed in a glass beaker
with a ring of silicon tubing, and 20 to 50 with dialysis buffer. One interesting advan-
mL dialysate is fed into the capillary from tage of this device is that a sample for elec-
the open end using a syringe (Figure 4). tron microscopy can be taken at anytime
The dialysate is shaken down against the with a finer glass capillary without disturb-
dialysis membrane to remove air bubbles, ing the progress of the experiment.

Figure 3. An Eppendorf cap


for the growing of 2D crys-
tals by microdialysis.

Figure 4. Glass capillary tube for


the growing of 2D crystals by
microdialysis.

264
Two-Dimensional Crystallization of Chlorophyll Proteins

❖ Procedure 8. 2D Crystals of a tories) is a simple alternative to conven-


Spinach PSII Subcomplex (38) tional dialysis for obtaining 2D crystals of
PSII membranes were isolated according integral membrane proteins (43). Hydro-
the method of Reference 10. phobic adsorption of detergents onto poly-
1. Concentrate the PSII complex to 0.5 styrene beads can be used for detergent
mg Chl/mL by ultrafiltration (Centri- removal independent of the respective
prep-10,™ MW cut off 10 kDa [Ami- CMC and has been used to generate high-
con Division, W.R. Grace, MA, USA]). ly ordered 2D crystals of membrane pro-
teins (43). Bio-Beads can be added to very
2. Microdialyze (12–14 kDa MW cut small sample volumes with almost no dilu-
off ) 30 µL against a buffer containing: tion of the protein or lipids. This property
40 mM MES, 20 mM NaCl, 1 mM CaCl2, is of great interest for membrane proteins
1 mM zinc acetate, 1 mM NaN3, for which purifying large quantities is par-
1 mM sodium ascorbate, 1 mg/mL ticularly difficult. However, it should be
butylated hydroxytoluene, pH 6.0, for noted that handling of small quantities of
4 days at 20°C.
Bio-Beads is difficult, because the beads
3. After dialysis, layer the crystalline sus- must not dry in the procedure. According
pension onto 7 mL 0.1 M sucrose, to Rigaud’s studies (43), the specific
40 mM MES, 20 mM NaCl, 1 mM adsorption of lipids is about 100 to 200
NaN3, pH 6.5. times lower than the specific adsorption of
4. Centrifuge at 2400× g for 5 minutes detergents (2–4 mg phospholipids/g Bio-
and collect the supernatant. Beads SM2). Since crystallization reactions
5. Centrifuge for 20 minutes at 35000× g are generally performed at low LPRs (0.2–
at 2°C to collect the 2D crystals from 0.5), weak lipid adsorption may have some
supernatant. effects. However, lipid adsorption can be
6. Resuspend the pellet in a buffer con- reduced by preincubation of the beads with
taining 40 mM MES, 20 mM NaCl, an excess of sonicated liposomes prior to
1 mM NaN3, pH 6.5. their use for detergent removal. 2D crystal-
The 2D crystals contain CP47, D1, D2, lization trials can be performed using either
cytochrome b-559, and the psbI subunit. a 1-step addition of Bio-Beads, resulting in
The unit cell is rectangular with dimen- quick and straightforward removal of the
sions of 16.7 and 15.3 nm containing 4 detergent, or by addition of the same mass
monomers and p22121 symmetry. of Bio-Beads in several steps, which will
Procedure 8 and its modification, using slow down the process. The rate of deter-
higher Chl concentration (1 mg/mL) and gent removal is directly linked to the
including of 30% glycerol in the dialysis weight of Bio-Beads used and also to the
buffer and 1 to 3 weeks dialysis time, working temperature. The rate of adsorp-
allowed the formation of well-ordered large tion of detergents doubles every 15°C.
tubular crystals of PSII complexes (42). Quantitation of the Bio-Beads can present
The unit cell is rectangular with dimen- some difficulties, because the beads should
sions of 16.6 and 16.3 nm containing 4 stay wet to keep a reproducible adsorption
monomers and belong to the same symme- property, and precise weighting has to be
try as the above mentioned crystals. performed on freshly blotted beads. Prior
to first use, wash the Bio-Beads with
2.2.3. Bio-Beads Method methanol and then with buffer (either in a
Adsorption of detergents to polystyrene batch procedure or packed on a column).
beads (Bio-Beads® SM2; Bio-Rad Labora- Washed beads can be stored at 4°C as a
265
G. Tsiotis

50% slurry in water supplemented with vantage is that these spontaneously formed
0.2% NaN3. Procedure 9 describes the use 2D crystals are rarely highly ordered
of this technique to crystallize the Chlamy- because other components can be trapped
domonas reinhardii cytochrome b6f com- and hinder the crystal growth. Another dis-
plex, purified as in Reference 41. advantage is that these methods are limited
to cases where the membrane protein
❖ Procedure 9. 2D Crystals of occurs at high density. However, the quali-
Cytochrome b6f Complex from ty of naturally occurring 2D crystals can be
C. reinhardii (37) improved by either detergent extraction,
fusion of crystalline patches, or incubation
1. Resuspend the purified cytochrome b6f with additives. Examples of naturally
complex in buffer containing: 6.8 mM occurring crystalline membranes are the
Tricine, 245 mM ammonium phos- photosynthetic membranes from purple
phate, 0.3 mM NaN3, 2 mM CaCl2, bacteria (35). PSII from higher plants is
1.1 mM benzamidine, 5.6 mM Â-ami- found in the thylakoid membrane at high
nocaproic acid, 0.2 mM phenylmethyl- density and sometimes in a regular lattice
sulfonyl fluoride (PMSF), 0.3% glyc- form (45). Detergent extraction of the thy-
erol, 20 mM Hecameg, pH 8.0. lakoid membrane under nonsolubilizing
2. Add a mixture of EggPC and di-C18:1- conditions promotes or improves the crys-
phosphatidylglycerol (1:1 ratio). The tallinity (3,19,31).
final protein concentration in reconsti- For example, crystals of PSII, which
tution mixture should be 0.5 mg/mL provided evidence for the presence of a
and the LPR 0.2. PSII dimer (3) were obtained as follows.
3. Preincubate the sample overnight in PSII membranes, isolated as in Reference
the cold room under gentle stirring. 4, were resuspended in 150 mM NaCl, 5
4. Treat with 200 mg/mL SM2 Bio-Beads. mM MgCl2, and 20 mM Tricine, pH 7.5,
at a final Chl concentration of 2 mg/mL.
5. Incubate for 12 hours. Triton X-100 was added to a concentration
6. Pipet off the reconstituted material and of 4% (wt/vol), and the mixture was incu-
keep for 24 hours at 4°C. bated at 20°C for 20 minutes in the dark.
7. Freeze in liquid nitrogen, then thaw It was then centrifuged for 30 minutes
the sample at 37°C 3 times. (45 000× g), and the pellet was washed
The crystals have a unit cell (a = 17.5 once with buffer containing 10 mM NaCl,
nm, b = 6.8 nm, and γ= 90°) and a p22121 5 mM MgCl2, and 10 mM HEPES, pH
symmetry. 7.6. The crystals obtained had a rectangu-
lar unit cell (a = 17.8 nm and b = 26.7
2.3. 2D Crystallization in Native nm). In contrast, similar crystals obtained
Membranes using dodecyl maltoside (0.06%) instead
of Triton X-100, and a subsequent sucrose
Some membrane proteins have a natural gradient (0–2 M) (18) had unit cell dimen-
propensity to form regular arrays within sions of a = 16.8 nm, b = 18.9 nm, with γ =
the native membrane. Since the membrane 91°, and a p1 symmetry group, and led to
protein does not dissociate from the lipid the proposal of the presence of a PSII
bilayer, its native orientation is maintained. monomer (19). Lastly, using a similar
Additionally, the proteins are not exposed approach but with altered conditions,
to harsh detergents and, therefore, are kept tubular PSII crystals have been obtained
under stabilizing conditions. One disad- from spinach, which indicate the presence
266
Two-Dimensional Crystallization of Chlorophyll Proteins

of 2 monomeric PSII complexes (31) (see the chloroplast, was resolved to atomic reso-
Procedure 10). lution after formation of 2D crystals in a
“batch method” (49) (see Procedure 11).
❖ Procedure 10. Tubular Crystals of The temperature profile proved to be critical
PSII from Spinach (31) for the crystallization of LHC-II. In addi-
tion, 2D crystals of some proteins with large
Thylakoid membranes were isolated as extramembrane domains can be obtained at
in Reference 35. a pH close to the isoelectric point (35) or by
1. Resuspend thylakoid membranes in 15 increasing the ionic strength (6,12). These
mM NaCl, 50 mM sucrose, 5 mM methods are best interpreted as variations of
MgCl2, and 20 mM HEPES, pH 7.5. 3D crystallization and not as proper recon-
2. Adjust Chl concentration to 0.8 stitution of membrane proteins into a
mg/mL. native-like environment.
3. Add Triton X-100 to give a detergent
to Chl ratio of 4.5:1. ❖ Procedure 11. 2D Crystals of LHC-II
4. Incubate for 20 minutes in ice with Complex from Pea Chloroplasts
stirring.
LHC-II complexes from pea chloroplas-
5. Centrifuge for 20 minutes (20 000× g) ts are isolated as described in Reference 27.
and resuspend the pellet in 15 mM
NaCl, 5 mM MgCl2, 20 mM HEPES, 1. An aliquot of LHC-II complex (with a
pH 7.5 Chl concentration of 4 mg/mL in
6. Determine the Chl concentration. 0.4% Triton X-100) is diluted 50 times
7. Add Triton X-100 to give a ratio of 3:1 with distilled water and KCl added to a
of detergent to Chl. final concentration of 300 mM.
8. Incubate for 10 minutes on ice. 2. Centrifuge and collect the pellet. Pro-
ceed with either Method A or B.
9. Centrifuge for 15 minutes (15 000× g)
and wash the pellet with the same Method A
buffer.
(i) Dissolve the pellet to a Chl concen-
Additional washing steps can be includ- tration of 0.7 mg/mL in Triton X-100
ed to separate the crystals from the remain- and glycerol at final concentrations of
ing material. 0.23% and 40%, respectively.
The tubular crystals have a length of 1
to 2 µm and a diameter of 0.2 µm, and the (ii) Incubate at 35° to 40°C for 2 hours.
unit cell has dimensions of a = 11.2 nm Method B
and b = 16.1 nm.
(i) Dissolve the pellet with 0.11% Triton
X-100 and 0.24% n-nonyl-β-gluco-
2.4. Crystallization of Purified Membrane
pyranoside.
Protein by Precipitation
(ii) Add glycine buffer (100 mM, pH
In a few cases, a solubilized membrane 7.0) and glycerol to a final concentra-
protein has been crystallized into 2D sheets tion of 10 mM and 40%, respective-
without detergent removal under condi- ly. The Chl concentration should be
tions similar to those used in 3D crystalliza- 0.75 mg/mL.
tion experiments. Pea thylakoid LHC-II, (iii) Incubate for 2 days at 25°C and then
the most abundant membrane protein in for 2 hours at 40°C.

267
G. Tsiotis

The crystals have a unit cell of a = 12.7 than biological atoms (C, H, O, N, P, and
nm, b = 12.7 nm, with γ = 60°. The plane S), the contrast is drastically increased but
group has a p321 symmetry (28). also inverted (hence the term “negative”
staining). In addition, the heavy atom salts
partially substitute the water in the native
3. ANALYSIS OF 2D CRYSTALLIZA- environment of the molecules, thus em-
TION REACTIONS bedding the specimen and protecting it
from collapse upon drying in air. The most
Even if 2D crystals of large membrane commonly used heavy atom salts are uranyl
proteins can sometimes be observed by acetate or formate and sodium or potassi-
light microscopy, the shape of the crystals um phosphotungstate. Uranyl salts are
and their degree of order can best be more suitable for more protein samples,
assessed by electron microscopy. Thus, whereas tungstate is useful for lipid struc-
screening of 2D crystallization experiments tures. The pH of tungstate solutions can be
requires access to an electron microscope. modified, whereas uranyl salts precipitate
The quickest specimen preparation meth- at pH greater than 4.5.
od for screening for 2D crystals is negative After negative staining, examination of
staining. This method is rapid, needs a the samples is carried out by transmission
small amount of sample (less than 5 µL), is electron microscopy. The presence of large
remarkably simple, and can provide struc- vesicles or sheets can be checked at low
tural information to a resolution of about 2 magnification (2000×–5000×) because of
nm. A negatively stained 2D crystal is the high contrast obtained when operating
embedded in a dry microcrystalline heavy at large underfocus even for very thin
atom replica. As heavy atoms used for neg- objects (Figure 5A). Further observations
ative staining scatter electrons much more are done at higher magnification (Figure

Figure 5. Negatively stained tubu-


lar crystals of PSII complex ob-
tained as described in Reference
47. (A) Low magnification (5000×).
Scale bar represents 500 nm. (B)
High magnification (50 000×).
Scale bar represents 100 nm.

268
Two-Dimensional Crystallization of Chlorophyll Proteins

5B). If no crystals are found, the presence ic acid]; LHC, light-harvesting complex;
of remaining detergent, single protein par- LPR, lipid:protein ratio; MES, 2-[N-mor-
ticles incorporated in lipid vesicles, solubi- pholino]ethanesulfonic acid; OG, octyl-β-
lized proteins, or protein aggregates can all glucoside; OTG, octyl-β-thioglucoside; 8-
be identified. This information may be POE, octyl-polyoxyethylene; PSI and PSII,
valuable for designing further crystalliza- photosystem I and II; RC, reaction center.
tion trials.
REFERENCES

4. CONCLUSIONS 1.Allen, J.P., G. Feher, T.O. Yeates, H. Komiya, and


D.C. Rees. 1987. Structure of the reaction center from
Rhodobacter sphaeroides R-26: the protein subunits.
Progress with the structural analysis of Proc. Natl. Acad. Sci. USA 84:6162.
membrane proteins is limited by difficulties 2.Barber, J. and B. Andersson. 1994. Revealing the blue-
print of photosynthesis. Nature 370:31-34.
in handling protein–detergent and pro- 3.Bassi, R., A.G. Magaldi, G. Tognon, G.M. Giacomet-
tein–lipid complexes. An alternative ap- ti, and K.R. Miller. 1989. Two-dimensional crystals of
proach to X-ray crystallography of 3D crys- the photosystem II reaction center complex from high-
er plants. Eur. J. Cell Biol. 50:84-93.
tals is the analysis of 2D crystals. 2D crystals 4.Berthold, D.A., G.T. Babcock, and C.F. Yocum.
have been obtained for many membrane 1981. A highly resolved, oxygen-evolving photosystem
proteins, indicating the natural tendency of II preparation from spinach thylakoid membranes:
EPR and electron transport properties. FEBS Lett.
these proteins to integrate into the 2D envi- 134:231-234.
ronment of the lipid bilayer. While tech- 5.Bradford, M. 1976. A rapid and sensitive method for
nologies for data acquisition and data analy- the quantitation of microgram quantities of protein
utilizing the principle of protein-dye binding. Anal.
sis are rapidly improving and are able to Biochem. 72:248-254.
provide atomic resolution, the bottle-neck 6.Dekker, J.P., S.D. Betts, C.F. Yocum, and E.J. Boeke-
still remains the crystallization process. ma. 1990. Characterization by electron microscopy of
isolated particles and two-dimensional crystals of the
CP47-D1-D2-cytochrome b-559 complex of photo-
system II. Biochemistry 29:3220-3225.
ACKNOWLEDGMENTS 7.Dolder, M., A. Engel, and M. Zulauf. 1996. The
micelle to vesicle transition of lipids and detergents
in the presence of a membrane protein: towards a
The author is indebted to A. Engel for rationale for 2D crystallization. FEBS Lett. 382:
203-208.
helpful discussions. Financial support from 8.Doubochet, J., J. Lepault, R. Freeman, J.A. Berri-
Maurice E. Müller Foundation of Switzer- man, and J.C. Homo. 1982. Electron microscopy of
land and the Swiss National Foundation frozen water and aqueous solutions. J. Microsc.
128:219-237.
for Scientific Research (Grant No. 31- 9.Egelhaaf, S.U. and P. Schurtenberger. 1994. Shape
46972.96) are gratefully acknowledged. transformations in the lecithin-bile salt system: from
cylinders to vesicles. J. Phys. Chem. 98:8560-8573.
10.Enami, I., K. Kamino, J.-R. Shen, K. Satoh, and S.
Katoh. 1989. Isolation and characterisation of photo-
ABBREVIATIONS system II complex which lack light-harvesting chloro-
phyll a/b proteins but retain three extrinsic proteins
related to oxygen evolution from spinach. Biochim.
2D and 3D, two- and three-dimension- Biophys. Acta 977:33-39.
al; BChl, bacteriochlorophyll; BCA, bicin- 11.Engel, A., A. Hoenger, A. Hefti, C. Henn, R.C. Ford,
choninic acid; BSA, bovine serum albu- J. Kistler, and M. Zulauf. 1992. Assembly of 2-D
membrane protein crystals: dynamics, crystal order,
min; Chl, chlorophyll; CMC, critical and fidelity of structure analysis by electron micro-
micellar concentration; DMPC, dimyris- scopy. J. Struct. Biol. 109:219-234.
toyl phosphatidyl choline; EggPC, egg 12.Ford, R.C., A. Hefti, and A. Engel. 1990. Ordered
arrays of the photosystem I reaction centre after recon-
phosphatidylcholine; HEPES, N-[2-hy- stitution: projections and surface reliefs of the complex
droxyethyl]piperazine-N′-[2-ethanesulfon- at 2 nm resolution. EMBO J. 9:3067-3075.

269
G. Tsiotis

13.Ghosh, R., A. Hoenger, A. Hardmeyer, D. Mihailes- 30.Lichtenberg, D., R.J. Robson, and E.A. Dennis.
cu, R. Bachofen, A. Engel, and J.P. Rosenbusch. 1993. 1983. Solubilization of phospholipids by detergents.
Two-dimensional crystallization of the light-harvesting Structural and kinetic aspects. Biochim. Biophys. Acta
complex from Rhodospirillum rubrum. J. Mol. Biol. 737:285-304.
231:501-504. 31.Lyon, M.K., K.M. Marr, and P.S. Furcinitti. 1993.
14.Haschenmeyer, R.H. and R.J. Meyers. 1972. Nega- Formation and characterization of 2-dimensional
tive staining, p. 101-107. In M.A. Hayat (Ed.), Princi- crystals of photosystem-II. J. Struct. Biol. 110:133-
ples and Techniques of Electron Microscopy. Vol. 2, 140.
Van Nostrand, New York. 32.McDermott, G., S.M. Prince, A.A. Freer, A.M. Haw-
15.Hasler, L., J.B. Heymann, T. Walz, J. Kistler, and A. thornthwaite-Lawless, M.Z. Papiz, R.J. Cogdell, and
Engel. 1998. 2D crystallization of membrane proteins: N.W. Isaacs. 1995. Crystal structure of an integral
rationales and examples. J. Struct. Biol. 121:162-172. membrane light-harvesting complex from photosyn-
16.Henderson, R. and P.N.T. Unwin. 1975. Three- thetic bacteria. Nature 374:517-521.
dimensional model of purple membrane obtained by 33.Michel, H. 1991. General and practical aspects of
electron microscopy. Nature 257:28-32. membrane protein crystallization, pp. 74-88. In
17.Henderson, R., J.M. Baldwin, K.H. Downing, J. Lep- H. Michel (Ed.), Crystallization of Membrane Pro-
ault, and F. Zemlin. 1986. Structure of purple mem- teins, CRC Press, Boca Raton.
brane from Halobacterium halobium: recording, mea- 34.Michel, H., K.A. Weyer, H. Gruenberg, and F.
surement and evaluation of electron micrographs at 3.5 Lottspeich. 1985. The ‘heavy’ subunit of the photo-
Å resolution. Ultramicroscopy 19:147-178. synthetic reaction centre from Rhodopseudomonas
18.Holzenburg, A., F.H. Wilson, M.E. Finbow, and R.C. vidris: isolation of the gene, nucleotide and amino acid
Ford. 1992. Structural investigations of membrane sequence. The EMBO Journal 4:1667-1672.
proteins: the versality of electron microscopy. Bio- 35.Miller, K.R. and J.S. Jacob. 1983. Two-dimensional
chem. Soc. Trans. 20:591-597. crystals formed from the photosynthetic reaction cen-
19.Holzenburg, A., M.C. Bewley, F.H. Wilson, W.V. ters. J. Cell Biol. 97:1266-1270.
Nicholson, and R.C. Ford. 1993. 3-dimensional struc- 36.Mishra, R.K. and D.F. Ghanotakis. 1994. Selective
ture of photosystem-II. Nature 363:470-472. extraction of CP 26 and CP 29 proteins without affect-
20.Iwata, S., C. Ostermeier, B. Ludwig, and H. Michel. ing the binding of the extrinsic proteins (33, 23 and 17
1995. Structure at 2.8 Å resolution of cytochrome c kDa) and the DCMU sensitivity of a photosystem II
oxidase from Paracoccus denitrificans. Nature 376:660- core complex. Photosynth. Res. 42:37-42.
669. 37.Mosser, G., C. Breyton, A. Olofsson, J.L. Popot,
21.Jansson, S. 1994. The light-harvesting chlorophyll a/b- and J.L. Rigaud. 1997. Projection map of cyto-
binding proteins. Biochim. Biophys. Acta 1184:1-19. chrome b6f complex at 8 Å resolution. J. Biol. Chem.
22.Jap, B.K., M. Zulauf, T. Scheybani, A. Hefti, W. 272:20263-8.
Baumeister, U. Aebi, and A. Engel. 1992. 2D crystal- 38.Nakazato, K., C. Toyoshima, I. Enami, and Y. Inoue.
lization: from art to science. Ultramicroscopy 46:45-84. 1996. Two-dimensional crystallization and cryo-elec-
23.Karrasch, S., P.A. Bullough, and R. Ghosh. 1995. The tron microscopy of photosystem II. J. Mol. Biol. 257:
8.5 Å projection map of the light-harvesting complex I 225-232.
from Rhodospirillum rubrum reveals a ring composed 39.Ostermeier, C., S. Iwata, B. Ludwig, and H. Michel.
of 16 subunits. EMBO J. 14:631-638. 1995. Fv fragment-mediated crystallization of the
24.Karrasch, S., D. Typke, T. Walz, M. Miller, G. Tsio- membrane protein bacterial cytochrome c oxidase.
tis, and A. Engel. 1996. Highly ordered two-dimen- Nat. Struct. Biol. 2:842-6.
sional crystals of photosystem I reaction center from 40.Pebay-Peyroula, E., G. Rummel, J.P. Rosenbusch, and
Synechococcus sp.: functional and structural analyses. E.M. Landau. 1997. X-ray structure of bacteri-
J. Mol. Biol. 262:336-348. orhodopsin at 2.5 angstroms from microcrystals grown
25.Kreusch, A., A. Neubuser, E. Schiltz, J. Weckesser, in lipidic cubic phases [see comments]. Science
and G. Schulz. 1994. Structure of the membrane 277:1676-81.
channel porin from Rhodopseudomonas blastica at 2.0 Å 41.Pierre, Y., C. Breyton, D. Kramer, and J.-L. Popot.
resolution. Prot. Sci. 3:58-63. 1995. Purification and characterization of the
26.Kühlbrandt, W. 1992. Two-dimensional crystalliza- cytochrome b6f complex from Chlamydomonas rein-
tion of membrane proteins. Q. Rev. Biophys. 25:1-49. hardtii. J. Biol. Chem. 270:29342-29349.
27.Kühlbrandt, W., T. Thaler, and E. Wehrli. 1983. The 42.Rhee, K.H., E.P. Morris, D. Zheleva, B. Hankamer,
structure of membrane crystals of the light-harvesting W. Kuhlbrandt, and J. Barber. 1997. Two-dimension-
chlorophyll a/b protein complex. J. Cell Biol. 96:1414- al structure of plant photosystem II at 8-angstrom res-
1424. olution. Nature 389:522-526.
28.Kühlbrandt, W. and D.N. Wang. 1991. Three- 43.Rigaud, J.L., G. Mosser, J.J. Lacapere, A. Olofsson,
dimensional structure of plant light-harvesting com- D. Levy, and J.L. Ranck. 1997. Bio-Beads: an effi-
plex determined by electron crystallography. Nature cient strategy for two-dimensional crystallization of
350:130-134. membrane proteins. J. Struct. Biol. 118:226-235.
29.Landau, E.M. and J.P. Rosenbusch. 1996. Lipidic 44.Schirmer, T., T. Keller, Y. Wang, and J. Rosenbusch.
cubic phases: a novel concept for the crystallization of 1995. Structural basis for sugar translocation through
membrane proteins. Proc. Natl. Acad. Sci. USA 93: maltoporin channels at 3.1 Å resolution [see com-
14532-14535. ments]. Science 267:512-514.

270
Two-Dimensional Crystallization of Chlorophyll Proteins

45.Staehelin, L.A. 1975. Chloroplast membrane struc- 48.Tsukihara, T., H. Aoyama, E. Yamashita, T. Tomiza-
ture. Intramembrenous particles of different sizes make ki, H. Yamaguchi, K. Shinzawa-Itoh, R. Nakashima,
contact in stacked membrane regions. Biochim. Bio- R. Yaono, and S. Yoshikawa. 1995. Structure of metal
phys. Acta 408:1-11. sites of oxidized bovine heart cytochrome c oxidase at
46.Tsiotis, G., W. Nitschke, W. Haase, and H. Michel. 2.8 Å. Science 269:1069-1074.
1993. Purification and crystallization of photosystem I 49.Wang, D.N. and W. Kühlbrandt. 1991. High-resolu-
complex from a phycobilisome-less mutant of the tion electron crystallography of light-harvesting
cyanobacterium Synechococcus PCC 7002. Photosynth. chlorophyll a/b-protein complex in three different
Res. 35:285-297. media. J. Mol. Biol. 217:691-699.
47.Tsiotis, G., T. Walz, A. Spyridaki, A. Lustig, A. 50.Weiss, M.S., A. Kreusch, E. Schiltz, U. Nestel, W.
Engel, and D. Ghanotakis. 1996. Tubular crystals of Welte, J. Weckesser, and G.E. Schulz. 1991. The
a photosystem II core complex. J. Mol. Biol. 259: structure of porin from Rhodobacter capsulatus at 1.8 Å
241-248. resolution. FEBS Lett. 280:379-382.

271
Biosynthesis and Analysis of Bilins
12
Matthew J. Terry
University of Southampton, Southampton, England, UK

1. THE DIVERSITY AND isms (25,26). The structures of two of the


BIOSYNTHESIS OF NATURALLY most common of these bilins, phyco-
OCCURRING BILINS cyanobilin (PCB) and phycoerythrobilin
(PEB), are also shown in Figure 1. Other
The term bilin is a collective one to light-harvesting pigments include phycovi-
describe a broad group of open chain olobilin, phycourobilin, 15,16-dihydro-
tetrapyrroles and derives from the name biliverdin (DHBV), and mesobiliverdin
“bile pigments” as the first of these com- (MBV) (25,26). In higher plants, the relat-
pounds to be characterized were isolated ed bilin, phytochromobilin (PΦB) (Figure
from animal bile. These bilins, biliverdin 1), serves as the chromophore of the phy-
(BV) and bilirubin (BR), are the sequential tochromes (17,58). This family of photore-
products of heme degradation (their green ceptors is important throughout plant
and yellow pigmentation can be detected development and regulates such diverse
during the discoloration of a bruise), with processes as germination, growth, flower-
BR being conjugated to glucuronic acid to ing, and the synthesis of the photosynthet-
expedite excretion. The structures of BV ic apparatus. Also, BV and BR may not
and BR are shown in Figure 1, and their just be waste products, since both have
biochemistry is still the best understood of been demonstrated to have potent antioxi-
the bilins today. However, we now know dant activities (51), and BR has even been
that there is a great diversity of naturally implicated in having a role in circadian reg-
occurring bilins that have a wide range of ulation in humans (41). BV is used for pig-
different functions. In cyanobacteria and mentation in reptiles (4), fishes (23), and
two groups of algae, the rhodophytes (red insects, where the insecticyanin protein is
algae) and the cryptomonads, a tremen- bound to BV IX rather than the IXα iso-
dous variety of bilins are utilized for light mer more commonly found in nature (27).
harvesting through covalent attachment to BV is also found in the eggshells of a wide
the phycobiliproteins, which comprise the variety of birds (31). More remarkably still,
photosynthetic apparatus of these organ- the marine snail Aplysia californica uses

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

273
M.J. Terry

PEB, which it obtains from a diet of red bilin 2,3-reductase, catalyses the formation
seaweed, as a defensive ink pigment (43). of the A-ring ethylidene group required for
The biosynthetic relationship between covalent assembly to apophytochrome (see
the bilins discussed in this chapter is shown Reference 55 and Chapter 13 in this text
in Figure 2. The first committed step in the by McDowell and Lagarias). The product
pathway for the synthesis of all bilins is the of this reaction, 3(Z)-PΦB (57), is subse-
oxidation of protoheme to BV IXα by the quently converted to 3(E)-PΦB, the imme-
enzyme heme oxygenase (5,34,42,53). Fol- diate precursor of the bound phytochrome
lowing this universal reaction, BV IXα can chromophore, by an, as yet unidentified,
be reduced at one of three different posi- bilin 31,32 cis-trans isomerase. In red algae,
tions in a somewhat phylogenetic-depen- the pathways for the synthesis of bilins are
dent manner. In animals, BV IXα is quite complex, but have been recently been
reduced at the C-10 position (see Figure 1) reviewed in some depth (5). Concentrating
by the enzyme BV reductase to give BR just on the central pathway, BV IXα is first
IXα (32,50). In higher plants, the plastid- reduced by a bilin 15,16-reductase to give
localized enzyme PΦB synthase, which is a 15,16-DHBV IXα (10) and subsequently

Figure 1. Chemical structures of the major bilins.

274
Biosynthesis and Analysis of Bilins

by a bilin 2,3-reductase to 3(Z)-PEB (9). ther increased by recent evidence that the
This later enzyme is presumably related to reduction of BV IXα is not simply related
PΦB synthase, as both enzymes accom- to broad phylogenetic group. The green
plish the same reaction. 3(Z)-PEB then alga Mesotaenium caldariorum is able to
undergoes a series of isomerizations to pro- synthesize 3(Z)-PCB directly from 3(Z)-
duce the 3(E)-isomers of both PEB and PΦB (67). Such a pathway suggests a rea-
PCB (5,9). sonable biosynthetic alternative to the red
The complexity of bilin synthesis is fur- algal pathway for PCB synthesis, and

Figure 2. The biosynthetic relationship of the bilins discussed in the text. The full pathway shown has been constructed from
biosynthetic pathways identified in a number of different organisms with no single organism containing all pathways. Heme oxy-
genase is present in animals, plants, algae, and cyanobacteria. Of the four reductases, BV reductase has been detected in animals
and cyanobacteria, PΦB synthase (bilin 2,3-reductase) in plants and green algae, bilin 15,16-reductase in red algae and possibly
cyanobacteria, and bilin 181,182-reductase in green algae alone. There are two isomerase activities. 3(Z),3(E) cis-trans-isomerase
(31,32-isomerase) appears to be present in plants and algae, while (15,16),(181,182)-isomerase activity has only been detected in
red algae. Broken arrows indicate possible biosynthetic routes that have yet to be shown experimentally. Abbreviations: BR, biliru-
bin; BV, biliverdin; DHBV, dihydrobiliverdin; PCB, phycocyanobilin; PEB, phycoerythrobilin; PΦB, phytochromobilin.

275
M.J. Terry

indeed the authors report the unpublished more cost-effective method of obtaining
observation that Cyanidium caldarium has BV is to purify it from a mixture of bile
PΦB synthase activity. Since the yeast acids, which can be purchased quite cheap-
Pichia pastoris also has this activity (66), ly from both Sigma and ICN Pharmaceuti-
bilin 2,3-reductases may be considerably cals. An alternative method of obtaining
more widespread than first thought. To BV IXα is by oxidation of the BR IXα
complicate matters further, it now appears with 2,3-dichloro-5,6-dicyano-1,4-benzo-
that cyanobacteria also possess a BV (bilin quinone (22,36). This method is also cost-
10,11-)reductase (49), while two more effective if large quantities of BV IXα are
novel enzymes (a bilin 4,5-reductase and a required and is the most suitable for
bilin 121,122-dehydrogenase) have been obtaining (14C)-labeled BV IXα. In addi-
proposed to account for the synthesis of tion, it can be used for the synthesis of the
the five additional phycobilins identified to BV IIIα and XIIIα isomers from the corre-
date (5). sponding BR isomers or for MBV IXα
While the diversity of bilins and their from mesobiliverdin (MBR) IXα. Exten-
biosynthesis is a fascinating topic, this sive methods for the synthesis of a wide
chapter is primarily concerned with the range of BV dimethyl esters, including
methods required to study bilin biosynthe- DHBV dimethyl ester, have also been pub-
sis in general. In particular, I will focus on lished (52).
a few specific steps in bilin synthesis in
photosynthetic organisms, only addressing 2.2. Phycobilins
the biochemistry of these pathways in ani-
mals for comparative purposes. It is intend- The phycobilins can most easily be
ed that the methods described here could obtained by cleavage from the appropriate
be applied equally well to the biosynthetic phycobiliprotein using a HgCl2-assisted
steps that have yet to be characterized. methanolysis protocol. A method for the
preparation of PCB from lyophilized,
mixed Spirulina species is shown in Proce-
2. ISOLATION AND PREPARATION dure 1. This method has been described in
OF BILINS Terry et al. (56) and is based on the earlier
methods of Beale and Cornejo (9) for the
2.1. Biliverdins isolation of phycobilins from Porphyridium
cruentum and Arciero et al. (1) who used
BV can be purchased as a dihydrochlo- Synechococcus.
ride of about 80% purity from ICN Phar-
maceuticals (Costa Mesa, CA, USA) and, ❖ Procedure 1. Isolation and
until very recently, from Sigma (St. Louis, Purification of PCB from
MO, USA). One problem with these sam- Cyanobacteria
ples is that they contain a mixture of iso-
mers making them unsuitable for many Reagents
purposes. An alternative is to buy a more
pure sample of the IXα isomer (the isomer • Lyophilized mixed Spirulina species
that predominates in biological systems) (Sigma)
from Porphyrin Products (Logan, UT, • C18 cartridge e.g., Sep-Pak (Waters-
USA). The IXα isomer can then be further Millipore, Milford, MA, USA) or
purified by high-performance liquid chro- Bond Elut (Varian Instruments, Santa
matography (HPLC) (see section 3.1). A Clarita, CA, USA).
276
Biosynthesis and Analysis of Bilins

• Acetonitrile, 2-mercaptoethanol, HgCl2, HgCl2 and centrifuge at 30 000× g for


methanol, and trichloroacetic acid. 10 minutes. The PCB, which remains
• 0.1% (vol/vol) trifluoroacetic acid in the supernatant, can be further
(TFA). purified and concentrated using a C18
• Acetonitrile:0.1% TFA (20:80, vol/ cartridge.
vol). Note: Alternatively, PCB can be
• Acetonitrile:0.1% TFA (60:40, vol/ extracted into methylene chloride:1-
vol). butanol (2:1 vol/vol) and purified by
diethylaminoethyl (DEAE)-Sepha-
rose chromatography (Amersham
Method Pharmacia Biotech, Piscataway, NJ,
USA) (8).
1. Rehydrate the Spirulina powder in 30
mL/g dry weight of deionized water 9. Precondition the C18 cartridge with
and incubate for 10 minutes at room the sequential addition of acetonitrile
temperature. (to aid hydration and remove any con-
taminating compounds that might
2. Centrifuge at 30 000× g for 20 minutes
and collect the deep-blue supernatant otherwise be present in the final elu-
containing the phycocyanin protein. tion step), water, and 0.1% (vol/vol)
TFA (to equilibrate the column to the
3. Add trichloroacetic acid to give a final sample pH).
concentration of 1% (wt/vol) and
incubate for 1 hour at 4°C in the dark 10. Dilute the crude PCB solution 10-fold
to precipitate the protein. in 0.1% TFA and apply to the precon-
ditioned cartridge.
4. Centrifuge at 30 000× g for 20 minutes
and discard the supernatant. 11. Wash cartridge twice with 0.1% TFA
followed by acetonitrile:0.1% TFA
5. Wash the pellet repeatedly in methanol (20:80 vol/vol).
(20 mL/g Spirulina powder), discard-
ing the supernatant each time, until all 12. Elute PCB with a minimum, but
of the chlorophyll has been removed. known, volume of acetonitrile:0.1%
TFA (60:40 vol/vol).
6. Resuspend the pellet in methanol (2
mL/g Spirulina powder) containing 1 13. Aliquot and remove solvent under vac-
mg/mL HgCl2, and incubate the sam- uum (without heat).
ple for 20 hours at 42°C in darkness to 14. Store sample at -20°C, or below, in the
cleave the covalently-bound PCB dark until required.
chromophore from the phycocyanin HPLC analysis of the PCB sample iso-
protein. lated from Spirulina using Procedure 1 (see
Note: Samples containing free bilins section 3.1.) indicates that it is predomi-
should be kept as far as possible in low nantly 3(E)-PCB (>90%) with the 3(Z)-
light (or even safe light) conditions to isomer comprising the major additional
prevent photodegradation. This is a product. These isomers can be collected
general rule that is applicable to all and reconcentrated by diluting 10-fold in
work with pigments. 0.1% (vol/vol) TFA and applying to a C18
7. Remove the protein by centrifugation cartridge exactly as described above (Proce-
at 10 000× g for 10 minutes. dure 1, steps 9–14).
8 Add 1 µL/mL 2-mercaptoethanol to PEB has been isolated both by whole
the supernatant to precipitate the cell methanolysis (17) and by methanolysis
277
M.J. Terry

of phycobiliprotein fractions (3,40). For steps 9–14). An alternative procedure for


whole cell methanolysis, either the red alga, isolating PEB has recently been reported,
P. cruentum, or other phycoerythrin-con- in which the freeze-dried seaweed nori
taining organisms, such as the cyanobac- (Porphyra yezoensis ueda) was used as the
terium Calothrix sp. PCC 7601 can been starting material (40). The powdered mate-
used (17). P. cruentum is grown at 27°C in rial was extracted twice with deionized
a liquid suspension culture continuously water, and the suspension was filtered
provided with 1% (vol/vol) CO2. It is gen- through cheesecloth before the phyco-
erally slow growing, but we have found biliproteins were precipitated with 65%
that the ASP2 medium described in Prova- ammonium sulfate. Standard methanolysis
soli et al. (44) greatly improves the growth and purification protocols were then used
rate compared with other reported media (40).
(17). Calothrix sp. PCC 7601 can be Although methanolysis has proved to be
grown in BG-11 medium (47) containing the most widely adopted method of bilin
15 mM N-tris(hydroxymethyl)methyl-2- isolation, other procedures have been
aminoethanesulfonic acid (TES) (pH 8.2) reported. One interesting method that has
and 50 mM dextrose at 23°C (17). In both been described in the literature comes from
cases, an equal mixture of cool white and the observation that C. caldarium cells
red fluorescent lamps was used (17). For excrete large quantities of PCB when fed
Porphyridium, cells were extracted with the tetrapyrrole precursor 5-aminolevulinic
dimethyl sulfoxide (DMSO), and the solu- acid (ALA) (59). Another alternative is
tion was diluted 8-fold with acetone. Fol- chemical synthesis, which has now been
lowing centrifugation, the cells were accomplished for the free acid (30). How-
extracted 5 times with DMSO:acetone ever, this is not a trivial procedure, and a
(1:8 vol/vol) and then a further 3 times better approach for most laboratories will
with methanol, until the supernatant was be to let the enzymes themselves accom-
colorless (17). However, extracting with plish the required chemistry. Many of these
acetone alone followed by methanol was enzymes have now been partially purified
sufficient. Calothrix cells can be extracted (5) (described later in text) and could
directly with methanol (17). The HgCl2- already be used to synthesize phycobilins
assisted methanolysis procedure is essen- from BV IXα or heme. However, perhaps
tially identical to that described for PCB in the real breakthrough will come from the
Procedure 1, but using a ratio of 8 mL cloning and expression of these enzymes,
methanol/g cells. The method outlined in which will eventually permit the produc-
Procedure 1 (steps 9–14) is also suitable for tion of specific phycobilins to order. A sim-
the purification of PEB, but requires a fur- ilar strategy for the intermediates of heme
ther HPLC step to separate PEB from and corrin synthesis is described in Chap-
other bilins in the sample. Details of ter 4 in this text by Warren and Shoolin-
HPLC purification methods for bilins are gin-Jordan.
given below in section 3.1. As with the iso-
lation of PCB, the 3(E)-isomer of PEB is 2.3. Phytochromobilins
the principal product of this procedure,
but the 3(Z)-isomer can also be obtained. As it is difficult to obtain enough phy-
HPLC-purified samples can be concentrat- tochrome protein for methanolytic cleav-
ed by diluting 10-fold in 0.1% (vol/vol) age of the bound chromophore, there are
TFA and applying to a C18 cartridge really only two practical approaches for
exactly as described for PCB (Procedure 1, purifying PΦB. The first takes advantage
278
Biosynthesis and Analysis of Bilins

of the observation that the acetone treat- fonic acid (HEPES)-NaOH, pH 7.7,
ment required for extraction of P. cruentum 1% (wt/vol) polyvinylpyrrolidone (solu-
cells prior to phycoerythrin methanolysis ble PVP), 2 mM MgCl2, 2 mM
leads to oxidation of the bound PEB to EDTA (free acid), 2 mM EDTA (di-
give PΦB, which is then cleaved by the Na salt)].
normal methanolysis procedure (17). Phy- • Bovine serum albumin (BSA) and
coerythrin-containing cells extracted with cysteine.
methanol do not produce PΦB. The • Assay buffer [500 mM sorbitol, 20
methanolysis and purification procedures mM TES, 10 mM HEPES-NaOH,
are exactly as described above, and the yield pH 7.7, with 1 mM phenylmethyl-
of, predominantly, 3(E)-PΦB was estimat- sulphonyl fluoride (PMSF), 2 M leu-
ed to be approximately one third the yield peptin, 0.5 mM dithiothreitol (DTT)
of PEB (17). added fresh] 300 000 U/mL catalase in
A reasonable alternative is to synthesize 5 mM citrate buffer, pH 7.5.
PΦB enzymatically by incubating the BV
• NADPH regenerating system (12 mM
IXa precursor with isolated plastids (57,
NADP+, 100 mM glucose-6-phos-
62). In principle, plastid preparations from
phate, and 15 U/mL glucose-6-phos-
a wide variety of tissues or species could be
phate dehydrogenase).
used, but in my experience, there is consid-
erable variation in the yields of PΦB that • 1 mM BV IXα stock solution in
can be obtained. Of the plants that have DMSO.
been investigated, the best starting materi-
al appears to be pea and oat seedlings, Method
while tomato, cucumber, and Arabidopsis
are much less suitable. It is also generally 1. Grow pea seedlings in moist vermi-
better to use etioplasts than chloroplasts, as culite in the dark for 8 to 10 days at
bilin recovery is greater from these prepa- 25°C.
rations. The procedure is relatively straight- 2. Prepare homogenization buffer from
forward and only requires crude etioplast stock solution by diluting 2-fold with
preparations. Indeed, attempts to purify water. Add BSA and cysteine to final
plastids further with Percoll gradients led concentrations of 0.2% (wt/vol) and 5
to decreased bilin yields, possibly because mM, respectively, and adjust back to
of interactions between the bilins and the pH 7.7.
Percoll itself. Procedure 2 shows a simple 3. Under green safelight, harvest approxi-
procedure for obtaining PΦB from BV mately 30 g apical tissue (top 3–4 cm
using isolated pea (Pisum sativum L.) etio- of about 200 seedlings) and homoge-
plasts (55,62). nize in a prechilled mortar and pestle
in 2 mL/g fresh weight ice-cold
homogenization buffer. The sample
❖ Procedure 2. Synthesis of PΦB from
should be kept chilled (4°C) through-
BV Using Isolated Pea Etioplasts
out the isolation procedure.
4. Filter homogenate through 4 layers of
Reagents
muslin.
• Homogenization stock solution [1 M 5. Centrifuge for 1 minute at 8000× g.
sorbitol, 40 mM TES, 20 mM 4-(2- 6. Wipe away any starch from the side of
hydroxyethyl)-1-piperazineethanesul- the tube with a tissue and gently sus-

279
M.J. Terry

pend pellet in homogenization medi- prior to the application of the sample.


um (approximately 1 mL/g fresh 16. Purify 3(Z)- and 3(E)-PΦB by HPLC
weight) using a paintbrush to tease the as described in section 3.1.
pellet away from the centrifuge tube. The method outlined in Procedure 2
7. Centrifuge for 1 minute at 100× g to also produces a mixture of isomers,
remove unbroken cells and other although in this case, the yield of 3(Z)-
debris. PΦB greatly exceeds that of 3(E)-PΦB.
8. Centrifuge supernatant for 2 minutes Again, purification of both isomers can be
at 1500× g. readily accomplished by HPLC.
9. Wash the plastids by resuspending the With all bilin samples, it will generally
pellet in a small volume of the assay be necessary to prepare stock solutions
buffer stock solution and centrifuge which can then be used for various bio-
again for 2 minutes at 1500× g. chemical experiments such as assembly of
10. Resuspend the crude etioplast pellet in phytochrome (see Chapter 13 in this text
1 mL assay buffer. by McDowell and Lagarias) or phyco-
biliproteins (see Chapter 14 in this text by
11. Prepare a 2 mL reaction mixture con-
Bryant and Schluchter) or as standards for
taining 1 mL etioplasts (the excess
biosynthesis studies. These solutions are
should be saved for protein determina-
routinely prepared by dissolving the dried
tion; a final plastid protein concentra-
bilin samples in DMSO. The concentra-
tion of about 1 mg/mL is recommend-
tion should be adjusted to 1 mM following
ed), 20 µL catalase, 200 µL NADPH
spectrophotometric determination of the
regenerating system, and 560 µL assay
sample concentration (see section 3.2. and
buffer in a 25-mL Erlenmeyer flask.
Table 1), and samples should then be
12. Start the reaction by adding 20 µL of 1 stored at -80°C in the dark.
mM BV IXα (final concentration 10
µM), deplete oxygen from the flask by
replacing air with argon, and seal the 3. ANALYSIS OF BILINS
flask.
13. Incubate in the dark for 3 hours at
A wide range of techniques has been
28°C with gentle shaking.
employed for the analysis of bilins, and the
Note: In order to maximize the yield methods employed will depend on the
of PΦB, it is important to drive the information required. Bilins prepared by
reaction to completion. However, the large-scale isolation procedures
excessively long incubation periods described above will need to be analyzed
can result in nonspecific bilin degrada- for purity. HPLC can be used to determine
tion and care needs to be taken when the purity of samples and also to confirm
choosing incubation times. the identity of the sample by co-injection
14. Remove the etioplasts by centrifuga- of known standards. The absorption spec-
tion at top speed in a benchtop trum of the sample should also be taken, as
microfuge for 10 minutes. the absorption properties of bilins are char-
15. Partially purify bilins on a C18 car- acteristically different and can also be used
tridge as described in Procedure 1 to confirm the identity of samples. In addi-
(steps 9–14) except that the cartridge is tion, absorption spectroscopy can be used
conditioned with 50 mM N-methyl- to quantify purified bilin samples. If the
morpholine–acetate buffer, pH 7.7, sample to be analyzed contains an

280
Biosynthesis and Analysis of Bilins

unknown bilin(s), then a wider range of with this system BV IXα and 3(Z)-PΦB
techniques may be appropriate. Although still elute close to each other, and further
HPLC and absorption spectroscopy will improvements in separation have been
again prove useful, it may be necessary to achieved using a mobile phase of ace-
determine the chemical structure in order tone:20 mM formic acid (50:50 vol/vol).
to confirm or elucidate the identity of the Interestingly, this fairly small change in sol-
unknown bilin. In this case, 1H nuclear vent system leads to a complete reversal in
magnetic resonance (NMR) spectroscopy the order that the bilins elute (67).
is the most appropriate technique. Another problem associated with most
of these solvent systems is that retention
3.1. HPLC Analysis and Purification times vary considerably from day to day
and even during a series of runs. This prob-
Bilins are readily separated by C18 ably reflects a pronounced temperature
reverse phase HPLC using isocratic solvent sensitivity of these systems, but may also be
systems, and their distinctive absorption related in part to the condition of the col-
spectra make them easy to detect in the vis- umn itself. This variation highlights the
ible wavelength range. The most common- need to identify peaks by co-injection of
ly used solvent system for analyzing most known standards rather than by retention
bilins is that of Beale and Cornejo (8), who time. Perhaps the single biggest difficulty
separated phycobilins on an Altex Ultras- of using HPLC for bilin analysis is that
phere octadecylsilane (ODS) column (250 there is tremendous variation between
mm long × 46 mm diameter, 5 µm particle columns, with new columns, apparently
size). Separation was achieved using a identical to columns already in use, being
mobile phase of acetone:ethanol:water: completely unsuitable for bilin separation.
acetic acid (38:50:11:1 vol/vol/vol/vol) This normally manifests itself as an inabil-
with a flow rate of 4 mL/minute at 30°C ity to retain the bilins on the column, and
(8,17). An almost identical system (ace- this difference in performance is even
tone:ethanol:water:acetic acid, 34:48:17:1 apparent between columns from the same
vol/vol/vol/vol) has also proved suitable for company. However, ODS columns from a
separating phytochromobilins, in this case number of companies (some of which have
using an Ultrasphere ODS column (250 × been named above) have been used success-
10 mm, 5 µm particle size; Beckman Coul- fully, and researchers new to the field
ter, Fullerton, CA, USA) at room tempera- should persevere in finding a suitable one.
ture (57). However, one potentially serious The solvent systems described above
problem with this solvent system is that have not only been used for separating
there is almost no resolution of BV IXα phycobilins, but are also suitable for ana-
and 3(Z)-PΦB. To solve this problem, lyzing both BV IXα and MBV IXα syn-
some improvements on this system have thesized from heme (54,62) and mesoheme
been developed in the Lagarias laboratory. (unpublished results), respectively. Howev-
The first of these, changing the mobile er, a simpler system comprising 95%
phase to acetone:ethanol:100 mM formic (vol/vol) aqueous ethanol:acetic acid:water
acid (25:65:10 vol/vol/vol), has been used (92:1:7 vol/vol/vol) has also commonly
successfully for the analysis of PΦB synthe- been used (16,45). Both systems are also
sis using a Supercosil LC-18 ODS column capable of separating the 4 BV IX isomers.
(250 × 4.6 mm, 5 µm particle size; Supel- For the analysis of BR IXα formation from
co, Bellefonte, PA, USA) with a flow rate BV IXα by cyanobacterial BV reductase,
of 1.5 mL/minute (54,62). However, even Schluchter and Glazer (49) used a linear
281
M.J. Terry

Table 1. Spectroscopic Properties of Bilins Discussed in the Text


1H-NMR
Bilina λmax (nm)b ε (M-1cm-1)c spectrad

biliverdin IXα 377, 696 (37) ε377 = 66, 200 (37) 9, 57


15,16-dihydrobiliverdin IXα 335, 560 (10) ND 10
mesobiliverdin IXα 359, 685 (7) ε359 = 78, 600 (7) 2, 13e

3(Z)-phycocyanobilin 369, 686 (9) ε368 = 46, 774 (63)e 63e,f, 9e,f
3(E)-phycocyanobilin 375, 692 (9) ε374 = 47, 900 (13)e 19, 2, 30, 28e

3(Z)-phycoerythrobilin 327, 591 (9) ND 9


3(E)-phycoerythrobilin 329, 592 (9) ε594 = 25, 200 (12)e 20, 9

3(Z)-phytochromobilin 381, 698 (67) ε382 = 38, 019 (63)e 57


3(E)-phytochromobilin 386, 700 (17) ε386 = 64, 565 (63)e 17
aValues are given for the free acids unless otherwise stated.
bAbsorption maxima are in HCl/methanol unless otherwise stated, with references in
parentheses.
cMolar absorption (extinction) coefficients are in HCl/methanol unless otherwise stated, with
references in parentheses.
dReferences are for 1H-NMR spectra recorded in pyridine-D unless otherwise stated.
5
eDimethyl ester.
fSpectrum recorded in CDCl .
3
ND, not determined.

gradient from 50% solvent A (water), 50% 3.2. Absorption Spectroscopy


solvent B (acetone:ethanol:water:acetic
acid, 50:38:11:1 vol/vol/vol/vol) to 100% The standard solvent for recording visi-
solvent B over 30 minutes. ble absorption spectra is methanol:36%
One further consideration for the (wt/vol) aqueous HCl (49:1 vol/vol),
HPLC analysis of bilins is the most suit- although other solvents such as acetoni-
trile:0.1% TFA (60:40 vol/vol) can and
able wavelength for detection. As men-
have been used. Absorption maxima for a
tioned above, all these compounds absorb
range of common bilins in methanol/HCl
strongly in visible regions of the radiation are shown in Table 1. BV, PΦB, and PCB
spectrum making detection relatively all have a Soret peak between about 370 to
straightforward. The wavelength of choice 390 nm and a broader peak at approxi-
will depend on the range of bilins to be mately 700 nm. The shorter wavelength
detected. Information about the absorp- peak is more reliable for quantitation, as its
tion spectra of the common bilins is given position and height are less dependent on
in Table 1, but in general, detection at 370 factors such as solvent composition and
or 380 nm is suitable for most bilins. The temperature. Table 1 also gives molar
exception is PEB, which does not absorb absorption (extinction) coefficients for this
strongly in this region. For detection of a peak, and these can be used for quantita-
mixture of phycobilins that includes PEB, tion of bilin samples using Beer’s law:
600 nm is more suitable (8,17). A = εcl

282
Biosynthesis and Analysis of Bilins

where A is absorbance or optical density at known should be analyzed at the same


a specific wavelength, ε is the molar time, and these can be used, together with
absorption coefficient at the same wave- published spectra, for the determination of
length, c is the concentration of the sample the unknown structure. Table 1 provides a
in M, and l is the light path in centimeters. list of references containing information on
It should be noted that for quantitation, 1H NMR spectra for a range of bilins.
absorption spectra should be recorded in
the same solvent used to calculate the 3.4. Other Spectroscopic Techniques
molar absorption coefficient, as absorption
properties are highly solvent dependent. A number of other spectroscopic tech-
PEB has a different spectrum with a niques have also proved useful. Structural
major peak at about 590 nm and a smaller information can also be obtained by mass
one near 330 nm. In this case, the longer spectrometry, and this technique has been
wavelength peak is more suitable for quan- used in the analysis of BV IXα (37), PCB
titation as absorbance is much greater at (24), and the dimethyl esters of PCB (14),
this wavelength. BR is not soluble in PEB (12), and various BVs (14,52). Circu-
methanol, but has a single major peak of lar dichroism spectroscopy can provide
454 nm in chloroform (ε454 = 62 600 M-1 information on the configuration of chiral
cm-1) (36). carbons. Circular dichroism spectra are
also generally taken in HCl/methanol, as
3.3. 1H NMR Spectroscopy fully protonated bilins are free from helical
conformations that can hinder the inter-
1H NMR spectroscopy is the most pretation of the data. 3(E)-PΦB has a sin-
appropriate technique for determining the gle chiral carbon at position 2 (see Figure
chemical structure of bilins. Obtaining and 1), and this was shown to be in the R con-
interpreting 1H NMR spectra are special- figuration (17). 3(E)-PCB (11) and 3(E)-
ized procedures that require considerable PEB (29) isolated from phycobiliproteins
experience and are outside the scope of this also have a 2(R) configuration.
review. However, help with these aspects of Fluorescence spectroscopy has not been
the technique can generally be provided by widely used, as free bilins fluoresce very
an NMR facility. For the researcher, the poorly. However, chelation with zinc
most important factor in obtaining good results in a highly fluorescent compound,
spectra is the quality of the sample, and and this property has been used to study
great care should be taken during sample holophytochrome assembly (see Chapter
preparation. Small amounts of contamina- 13 in this text by McDowell and Lagarias).
tion by other compounds, or even dirt or More imaginative still has been the
grease, can severely affect the quality of the exploitation of the fluorescent properties of
spectra. If glassware is involved, this should PEB bound to phytochrome apoprotein to
be washed in nitric acid and thoroughly generate a new series of fluorescent protein
rinsed in deionized water before use. Puri- probes, the phytofluors (see Reference 40
fied samples should be dissolved in pyri- and Chapter 13 in this text by McDowell
dine-D5 (100.0 atom % D; Sigma), and Lagarias).
although other solvents such as CDCl3
have also been used. The interpretation of
spectra is aided by comparison with known 4. BILIN BIOSYNTHESIS
spectra. For this reason, spectra of related
compounds for which the structure is Much of the original work that estab-
283
M.J. Terry

lished the pathway for bilin biosynthesis in partially purified using differential ammo-
photosynthetic organisms has utilized the nium sulfate precipitation and DEAE-cel-
classical biochemical approach of feeding lulose and blue-Sepharose affinity chro-
putative bilin precursors. Using these tech- matography steps (16). This procedure was
niques, and in particular taking advantage subsequently extended to include a ferre-
of radiolabeled precursors, it was estab- doxin-Sepharose affinity chromatography
lished that the phycobilins were derived step to give a 200-fold purification of heme
from heme synthesized from ALA by the oxygenase activity (46).
main tetrapyrrole pathway. These experi- The employment of a ferredoxin affinity
ments are discussed by Beale (5). Similarly, purification step highlights one of the dis-
ALA and BV IXα were also shown to be tinguishing features of heme oxygenases
precursors of phytochromobilin (reviewed from photosynthetic organisms: the
in Reference 58), though only recently was requirement for reduced ferredoxin, pro-
the intermediacy of heme confirmed (62). duced by ferredoxin-NADP+ oxidoreduc-
While this type of approach will continue tase and NADPH, for enzyme activity. In
to be important in bilin research, this sec- contrast, mammalian heme oxygenase (first
tion will concentrate on assaying the described in Reference 53), found associat-
enzymes committed to bilin synthesis. ed with microsomal membranes, utilizes an
NADPH-cytochrome P450 reductase (35,
4.1. Heme Oxygenase 69). A second distinguishing feature is that
the algal enzyme requires a second reduc-
Heme oxygenase catalyses the synthesis tant in addition to ferredoxin (16): ascor-
of BV IXα from protoheme in a reaction bate appears to be the most effective with
that requires O2 and reducing power, and Trolox (6-hydroxy-2,5,7,8-tetramethyl-
liberates CO and Fe2+ (Figure 3) (34,42). chroman-2-carboxylic acid) also proving
In photosynthetic organisms, the most suitable (46). The reason that the reaction
extensively characterized heme oxygenase requires a second reductant is currently
system is that of C. caldarium (5). Heme unknown.
oxygenase activity was first measured in The heme oxygenase assay in extracts or
Cyanidium extracts by Beale and Cornejo partially purified fractions of Cyanidium is
(7). The enzyme is soluble and has been an HPLC-based assay that takes advantage

Figure 3. The reaction catalyzed by the enzyme heme oxygenase. Substrates and products are not shown stoichiometrically.

284
Biosynthesis and Analysis of Bilins

of the observation that MBV IXα, in con- (18). However, in most other respects, the
trast to BV IXα, is not further metabolized cyanobacterial heme oxygenase appears to
in these preparations. A typical reaction be similar to its algal counterpart (15,18).
buffer contains 10 µM mesohemin IX in One important consideration when
20 mM Tris-HCl, pH 7.6, with 5 mM L- assaying heme oxygenase is product identi-
ascorbate, 0.1 mg/mL catalase, 0.5 mg/mL fication. Heme readily undergoes coupled
BSA, 10% (wt/vol) glycerol, and reducing oxidation to a mixture of BV IXα, β, γ,
power provided by an NADPH-regenerat- and δ (particularly in the presence of ascor-
ing system (10 mM glucose 6-phosphate, bate), and it is necessary to establish that
0.5 mM NADP+, and 0.3 U/mL glucose- the reaction product is exclusively the IXα
6-phosphate dehydrogenase), together with isomer in order to demonstrate that BV
5 µg/mL Porphyra umbilicalis ferredoxin, synthesis is enzymatic. This can be done by
and 0.01 U/mL spinach ferredoxin- HPLC as described in section 3.1. To pre-
NADP+ reductase (46). O2 is also required vent the formation of additional BV IX
for the reaction to proceed. Mesohemin isomers from H2O2 generated by the reac-
can be obtained from Porphyrin Products, tion of ascorbate and O2, catalase is rou-
while all the other components are readily tinely included in the reaction mixture (7).
available from Sigma. In the Beale labora- Heme oxygenase can be stimulated by the
tory, assays are performed at 42°C for 1 addition of strong iron chelators such as
hour in the dark, and the bilin products are desferrioxamine or Tiron (4,5-dihydroxy-
extracted into dichloromethane:1-butanol 1,3-benzene disulphonic acid). These are
(2:1 vol/vol) and concentrated on DEAE- thought to aid the dissociation of an
Sepharose prior to HPLC analysis (46). Fe(III)-BV IXα complex that may be the
The electrons required for the reaction primary product in vitro (46). A number
are transferred directly from the ferredox- of inhibitors are also available. Sn-proto-
in, as evidenced by the observation that porphyrin IX is a potent inhibitor of the
light-driven ferredoxin reduction via a algal enzyme (16), while the animal
spinach photosystem I fraction can support enzyme is inhibited by a wide range of
heme oxygenase activity (45). As there is metal porphyrins (60) that have yet to be
likely to be a direct interaction between tested on heme oxygenases from photosyn-
heme oxygenase and the ferredoxin, it is thetic organisms. Heme oxygenase is also
unsurprising that the source of ferredoxin inhibited by diethylpyrocarbonate, proba-
is important for maximum activity. Por- bly through binding to active-site his-
phyra ferredoxin is almost as good as a tidines, but is insensitive to the sulfhydryl
ferredoxin-containing fraction from Cyani- reagent p-hydroxymercuribenzoate (16).
dium (86%), but spinach ferredoxin only In preparations from higher plants,
resulted in 20% of activity (16). There is heme oxygenase has only been assayed in
also considerable variation in the effective- isolated plastids (62). The method is essen-
ness of different second reductants. L- and tially the same as that described in Proce-
D-ascorbate were equally effective, while dure 2, except that 10 µM heme replaces
dehydroascorbate could not support any the BV, and the O2 depletion step is not
activity. Trolox (29% of the activity with required. Under these assay conditions, BV
ascorbate), dihydroquinone (16%), and IXα is completely metabolized to 3(Z)-
DTT (15%) were all partially active (46). PΦB. However, as for the algal system,
In the cyanobacterium Synechocystis sp. mesoheme is also a substrate for the plant
PCC 6701, Trolox was actually more effec- heme oxygenase, while the product MBV
tive as a second reductant than ascorbate IXα is not further metabolized (unpub-
285
M.J. Terry

lished results). In tomato etioplasts, BV thereby allowing the determination of ini-


IXα is the main product after incubation tial reaction rates and is commonly per-
with heme (54). In this case, activities are formed as a coupled assay with excess BV
quite low, and the reaction can only pro- reductase allowing BR synthesis to be mea-
ceed if 5 mM sodium ascorbate is included sured. However, it is also possible to detect
in the assay. This leads to problems from BV IXα formation directly by using the
coupled oxidation, and care needs to be longer wavelength peak.
taken to establish that BV IXα is the pri- Using a coupled-assay approach, recom-
mary isomer produced. binant plant heme oxygenase was assayed
Substantial progress has recently been under the following conditions: 100 mM
made in our understanding of heme oxyge- Tris-HCl, pH 7.8, containing 15 µM
nases by the cloning and expression of hemin, 0.15 mg/mL BSA, recombinant
recombinant heme oxygenases from both cyanobacterial BV reductase (with an activ-
Synechocystis sp. PCC 6803 (18) and Ara- ity of 45 nmol BR/hour), 250 µM
bidopsis (39). Two genes were identified in NADPH, 50 µg/mL spinach ferredoxin,
Synechocystis, only one of which appeared 0.025 U/mL spinach ferredoxin-NADP+
to be expressed under the conditions tested reductase, and 2 mM Tiron (39). The assay
(18). This gene encoded a protein of 27 was performed at 25°C with reaction rates
kDa that was designated HO1 (18). Two determined by following the formation of
genes have also been identified in higher BR at 450 nm. It can be seen from the
plants (21,39). One gene, designated HY1 composition of the assay buffer that the
after the mutant that led to its identifica- plastidic plant enzyme has similar proper-
tion, but also known as AtHO1, encodes a ties to the enzymes from algae and
protein of 32.6 kDa (including a chloro- cyanobacteria. This similarity extends to
plast target sequence of 6 kDa) and is plas- the requirement for a second reductant
tid localized (39). This protein shows sub- such as ascorbate for maximal activity (T.
stantial similarity to specific conserved Muramoto, M.J. Terry, A. Yokota, and T.
regions of mammalian heme oxygenases Kohchi, unpublished results). One inter-
and has been shown to have heme oxyge- esting difference is that there is a fairly
nase activity (39). Heme oxygenase activity strict requirement for an iron chelator for
of AtHO2, encoded by the second gene the reaction to proceed (39). Recombinant
with sequence similarity to known heme heme oxygenase has also been studied from
oxygenases, could not be established (21). the pathogenic bacterium Corynebacterium
Genes encoding heme oxygenases have diptheriae (65). This heme oxygenase
now been expressed in Escherichia coli appears to utilize a third type of redox part-
resulting in large quantities of enzyme that ner, as activity was most efficient with an
can be readily purified by conventional NADH-dependent putidaredoxin–puti-
methods (64,65) or by attaching a purifi- daredoxin reductase system (65).
cation tag such as polyhistidine (18) or glu- There are also a number of additional
tathione S-transferase (39). One benefit assays for heme oxygenase activity. An
from the overexpression of plant heme oxy- alternative HPLC-based assay was recently
genase is that it has permitted the use of a described in some detail (48). This assay is
spectrophotometric assay for heme oxyge- suitable for measuring heme oxygenase in
nase which had not been possible previous- crude samples from mammalian tissues
ly because of interference by cellular pig- and therefore provides a suitable alternative
ments. This assay has the advantage that to the standard spectrophotometric assay
the reaction can be followed in real time, (see above) as it overcomes problems such
286
Biosynthesis and Analysis of Bilins

as spectral interference and the effect of glucose-6-phosphate, and 0.1 U/mL


protein composition on the molar absorp- glucose-6-phosphate dehydrogenase,
tion coefficient of BR. In addition, it is also type XII from Sigma)].
possible to measure heme oxygenase activi- • Assay buffer B [0.1 M potassium
ty by the release of CO instead of BV IXα phosphate buffer, pH 7.0, containing
synthesis. The production of CO in crude 1 mg/mL BSA, 10 µM BV IXα, and
samples has been measured by gas chro- an NADH-regenerating system (0.2
matography (61), although this method mM NAD+, 1 mM glucose-6-phos-
has obvious problems of specificity. Alter- phate, and 0.2 U/mL glucose-6-phos-
natively, the release of 14CO can be mea- phate dehydrogenase, type XXIV from
sured following the incubation of purified Sigma)].
heme oxygenase with 14C-heme (70). • 20 to 80 µg/mL BV reductase.
However, a simpler method of CO detec-
tion in purified samples is to follow the
change in the absorbance maximum of Method
myoglobin following CO binding (65).
1. Place 495 µL of assay buffer A or B
into a cuvette and zero the baseline
4.2. Bilin Reductases absorbance.
Bilin reductases catalyzing four different 2. Start the reaction by adding 5 µL BV
reactions have been reported to date (see reductase. Mix thoroughly and quickly
Figure 2). By far the most extensively char- using a pipet.
acterized of these enzymes is mammalian 3. Follow the appearance of BR by
BV reductase, which reduces BV IXα to recording the absorbance changes at
BR IXα. This enzyme was first assayed 466 nm (pH 8.7) or 458 nm (pH 7.0).
directly by Singleton and Laster (50) who 4. Determine the rate of BV reductase
followed the disappearance of BV IXα activity using the derived absorption
spectrophotometrically. However, it is now coefficients for BR in these buffers of
standard practice to assay BV reductase by ε466 = 64 100 M-1 cm-1 at pH 8.7 and
following the appearance of BR IXα. The ε458 = 55 800 M-1 cm-1 at pH 7.0 (see
enzyme is unusual in that it has two differ- Reference 56 and section 3.2.).
ent pH optima for NADPH and NADH, Interestingly, in addition to BV IXα,
both of which it uses directly (32). A stan- PΦB, PCB, and PEB are all substrates for
dard assay for mammalian BV reductase, BV reductase (56), and this property has
adapted by Terry et al. (56) from Kutty and been used creatively to produce phy-
Maines (32), is shown in Procedure 3. tochrome chromophore-deficient plants
through overexpression of the mammalian
❖ Procedure 3. Spectrophotometric BV reductase gene (33). The enzyme from
Assay for Mammalian BV Reductase human liver is not, however, capable of
reducing BV IXβ, and it now appears that
Reagents a second enzyme is present which has sub-
stantial activity with BV IXβ, γ, and δ, but
• Assay buffer A [0.1 M Tris-HCl, pH cannot reduce the IXα isomer (68). Sur-
8.7, containing 1 mg/mL BSA, 10 µM prisingly, BV IXα reductase is also present
BV IXα, and an NADPH-regenerat- in the cyanobacteria, Synechocystis sp. PCC
ing system (0.1 mM NADP+, 1 mM 6803 (49), although the significance of this

287
M.J. Terry

is not entirely clear, and it is unknown activity has been measured in isolated plas-
whether algae or higher plants also have tids from oat (57), pea (62), and tomato
this enzyme. (54). The conditions for measuring this
Algae appear to have at least four differ- enzyme in isolated plastids have been
ent bilin-reducing enzymes for which the described in Procedure 2. Recently,
substrate specificity has yet to be clearly progress has been made in purifying PΦB
defined (Figure 2). The best characterized synthase (38). This work has revealed that
of these are the enzymes from the red alga the enzyme is soluble, has an apparent
C. caldarium. Beale and Cornejo (6) molecular mass of 29 kDa, and functions
showed that cell-free extracts of Cyanidium in the same ferredoxin-dependent manner
reduced BV IXα to give the final products as the algal (45) and cyanobacterial (15)
of 3(Z)- and 3(E)-PCB. Subsequently, reductases.
these authors identified two soluble activi-
ties, one of which reduced BV IXα to 4.3. Bilin Isomerases
15,16 DHBV, while a second reduced
15,16 DHBV to 3(Z)-PEB (9,10). 3(Z)- Little is known about the bilin isomeras-
PEB is then further isomerized to PCB (see es. Two types of isomerase activity have
section 4.3.). In contrast to BV reductase, been reported. A glutathione-dependent
the two bilin reductases required reduced 3(Z) to 3(E) cis-trans isomerase was identi-
ferredoxin for activity (8,45). The assay fied in Cyanidium extracts (9), and a simi-
conditions used were as follows: 25 mM lar activity has been proposed to exist in
HEPES buffer, pH 7.3, 10 µM BV IXα, plants as incubation of BV IXα with plas-
10% (vol/vol) glycerol, 1 mM MgCl2, tids results in the synthesis, first of 3(Z)-
approximately 1 mg/mL BSA, 3000 U/mL PΦB and then of 3(E)-PΦB (57). It is still
catalase, and reducing power in the form of not entirely clear whether this is an
an NADPH-regenerating system together enzyme-catalyzed reaction, as it may sim-
with ferredoxin and ferredoxin NADP+ ply reflect the fact that the (Z)-isomer is
reductase (see conditions for the heme oxy- chemically less stable than the (E)-isomer.
genase assay in section 4.1). Following However, cyanobacterial extracts synthe-
incubation at 40°C for 30 minutes, the sized 3(Z)-PCB, but not 3(E)-PCB, from
bilin products were extracted and analyzed BV IXα supporting a role for a specific
by HPLC as described in section 3.1. 3(Ζ) to 3(E) cis-trans isomerase in other
Incubation of BV IXα with soluble pro- organisms (15). The second isomerase
tein extracts from plastids of the green alga activity converts PEB to PCB with both
M. caldariorum under similar assay condi- the (Z)- and (E)-isomers serving as sub-
tions resulted in completely different prod- strates (9). In this case, the reaction was
ucts. In this case, 3(Z)-PΦB and 3(Z)-PCB catalyzed by a high molecular weight
were synthesized sequentially (67). This (>60 kDa) protein fraction, but did not
result suggests the presence of a homolog require NADPH or reduced ferredoxin for
of the plant enzyme, PΦB synthase, and a activity (9).
previously undescribed 181,182-reductase
(see Figures 1 and 2). PΦB synthase activi-
ty was first detected in cucumber using a ABBREVIATIONS
coupled assay to measure PΦB synthesis by
assembly with apophytochrome (55). This ALA, 5-aminolevulinic acid; BV,
assay has now been superceded by an biliverdin; BR, bilirubin; BSA, bovine
HPLC-based assay and PΦB synthase serum albumin; DHBV, dihydrobiliverdin;
288
Biosynthesis and Analysis of Bilins

DMSO, dimethyl sulfoxide; MBV, meso- 10.Beale, S.I. and J. Cornejo. 1991. Biosynthesis of phy-
biliverdin; PCB, phycocyanobilin; PEB, cobilins. 15,16-dihydrobiliverdin IXα is a partially
reduced intermediate in the formation of phycobilins
phycoerythrobilin; PΦB, phytochromo- from biliverdin IXα. J. Biol. Chem. 266:22341-
bilin; TFA, trifluoroacetic acid. 22345.
11.Brockmann, H., Jr. and G. Knobloch. 1973. Die
absolute Konfiguration des 2E-Äthyliden-3-methyl-
succinimids. Ein Beitrag zur Bestimmung der
ACKNOWLEDGMENTS absoluten Konfiguration von Phycobilinen und Phy-
tochrom. Chem. Ber. 106:803-811.
12.Chapman, D.J., W.J. Cole, and H.W. Siegelman.
I would like to thank the Royal Society 1967. The structure of phycoerythrobilin. J. Am.
for their support through a Royal Society Chem. Soc. 89:5976-5977.
13.Cole, W.J., D.J. Chapman, and H.W. Siegelman.
University Research Fellowship, Professors 1967. The structure of phycocyanobilin. J. Am. Chem.
Sam Beale and Peter Shoolingin-Jordan for Soc. 89:3643-3645.
reading this manuscript prior to publica- 14.Cole, W.J., D.J. Chapman, and H.W. Siegelman.
1968. The structure and properties of phyco-
tion, Mark Milford for checking the Proce- cyanobilin and related bilatrienes. Biochem. 7:2929-
dures, and Professor Clark Lagarias for the 2935.
opportunity to learn about the wonderful 15.Cornejo, J. and S.I. Beale. 1997. Phycobilin biosyn-
thetic reactions in extracts of cyanobacteria. Photosyn.
world of bilins. Res. 51:223-230.
16.Cornejo, J. and S.I. Beale. 1988. Algal heme oxyge-
nase from Cyanidium caldarium. Partial purification
REFERENCES and fractionation into three required protein compo-
nents. J. Biol. Chem. 263:11915-11921.
1.Arciero, D.M., D.A. Bryant, and A.N. Glazer. 1988. 17.Cornejo, J., S.I. Beale, M.J. Terry, and J.C. Lagarias.
In vitro attachment of bilins to apophycocyanin. I. 1992. Phytochrome assembly. The structure and bio-
Specific covalent adduct formation at cysteinyl residues logical activity of 2(R),3(E)-phytochromobilin derived
involved in phycocyanobilin binding in C-phyco- from phycobiliproteins. J. Biol. Chem. 267:14790-
cyanin. J. Biol. Chem. 263:18343-18349. 14798.
2.Arciero, D.M., J.L. Dallas, and A.N. Glazer. 1988. 18.Cornejo, J., R.D. Willows, and S.I. Beale. 1998.
In vitro attachment of bilins to apophycocyanin. II. Phytobilin biosynthesis: cloning and expression of a
Determination of the structures of tryptic bilin pep- gene encoding soluble ferredoxin-dependent heme
tides derived from the phycocyanobilin adduct. J. Biol. oxygenase from Synechocystis sp. PCC 6803. Plant J.
Chem. 263:18350-18357. 15:99-107.
3.Arciero, D.M., J.L. Dallas, and A.N. Glazer. 1988. 19.Crespi, H.L., L.J. Boucher, G.D. Norman, J.J. Katz,
In vitro attachment of bilins to apophycocyanin. III. and R.C. Dougherty. 1967. Structure of phyco-
Properties of the phycoerythrobilin adduct. J. Biol. cyanobilin. J. Am. Chem. Soc. 89:3642-3643.
Chem. 263:18358-18363. 20.Crespi, H.L. and J.J. Katz. 1969. Exchangeable hydro-
4.Austin, C.C. and K.W. Jessing. 1994. Green-blood gen in phycoerythrobilin. Phytochem. 8:759-761.
pigmentation in lizards. Comp. Biochem. Physiol. 21.Davis, S.J., J. Kurepa, and R.D. Vierstra. 1999. The
109A:619-626. Arabidopsis thaliana HY1 locus, required for phy-
5.Beale, S.I. 1993. Biosynthesis of phycobilins. Chem. tochrome-chromophore biosynthesis, encodes a pro-
Rev. 93:785-802. tein related to heme oxygenases. Proc. Natl. Acad. Sci.
6.Beale, S.I. and J. Cornejo. 1984. Enzymic Transfor- USA 96:6541-6546.
mation of biliverdin to phycocyanobilin by extracts of 22.Elich, T.D., A.F. McDonagh, L.A. Palma, and J.C.
the unicellular red alga Cyanidium caldarium. Plant Lagarias. 1989. Phytochrome chromophore biosynthe-
Physiol. 76:7-15. sis. Treatment of tetrapyrrole-deficient Avena explants
7.Beale, S.I. and J. Cornejo. 1984. Enzymatic heme with natural and non-natural bilatrienes leads to for-
oxygenase activity in soluble extracts of the unicellular mation of spectrally active holoproteins. J. Biol. Chem.
red alga, Cyanidium caldarium. Arch. Biochem. Bio- 264:183-189.
phys. 235:371-384. 23.Fang, L.-S. and J.L. Bada. 1990. The blue-green
8.Beale, S.I. and J. Cornejo. 1991. Biosynthesis of phy- blood plasma of marine fish. Comp. Biochem. Physiol.
cobilins. Ferredoxin-mediated reduction of biliverdin 97B:37-45.
catalyzed by extracts of Cyanidium caldarium. J. Biol. 24.Fu, E., L. Friedman, and H.W. Siegelman. 1979.
Chem. 266:22328-22332. Mass-spectral identification and purification of phy-
9.Beale, S.I. and J. Cornejo. 1991. Biosynthesis of phy- coerythrobilin and phycocyanobilin. Biochem. J. 179:
cobilins. 3(Z)-phycoerythrobilin and 3(Z)-phyco- 1-6.
cyanobilin are intermediates in the formation of 3(E)- 25.Glazer, A.N. 1989. Light guides. Directional energy
phycocyanobilin from biliverdin IXα. J. Biol. Chem. transfer in a photosynthetic antenna. J. Biol. Chem.
266:22333-22340. 264:1-4.

289
M.J. Terry

26.Glazer, A.N. and G.J. Wedemayer. 1995. Cryptomon- californica: concentration and storage of phycoerythro-
ad biliproteins—an evolutionary perspective. Photosyn. bilin in the ink gland. J. Exp. Biol. 201:1595-1613.
Res. 46:93-105. 44.Provasoli, L., J.J.A. McLaughlin, and M.R. Droop.
27.Goodman, W.G., B. Adams, and J.T. Trost. 1985. 1957. The development of artificial media for marine
Purification and characterization of a biliverdin-associ- algae. Archiv. Mikrobiol. 25:392-428.
ated protein from the hemolymph of Manduca Sexta. 45.Rhie, G. and S.I. Beale. 1992. Biosynthesis of Phyco-
Biochem. 24:1168-1175. bilins. Ferredoxin-supported NADPH-independent
28.Gossauer, A. and W. Hirsch. 1974. Totalsynthese des heme oxygenase and phycobilin-forming activities
racemischen Phycocyanobilins (Phycobiliverdins) from Cyanidium caldarium. J. Biol. Chem. 267:16088-
sowie eines “Homophycobiliverdins”. Liebigs Ann. 16093.
Chem. 1974:1496-1513. 46.Rhie, G. and S.I. Beale. 1995. Phycobilin biosynthesis:
29.Gossauer, A. and J.-P. Weller. 1978. Chemical total reductant requirements and product identification for
synthesis of (+)-(2R,16R)- and (+)-(2S, 16R)-phycoery- heme oxygenase from Cyanidium caldarium. Arch.
throbilin dimethyl ester. J. Am. Chem. Soc. 100:5928- Biochem. Biophys. 320:182-194.
5933. 47.Rippka, R., J. Deruelles, J.B. Waterbury, M. Herd-
30.Kakiuchi, T., H. Kato, K.P. Jayasundera, T. Higashi, man, and R.Y. Stanier. 1979. Generic assignments,
K. Watabe, D. Sawamoto, H. Kinoshita, and K. Ino- strain histories and properties of pure cultures of
mata. 1998. Total syntheses of (±)-phycocyanobilin cyanobacteria. J. Gen. Microbiol. 111:1-61.
and its derivatives bearing a photoreactive group at D- 48.Ryter, S., E. Kvam, and R.M. Tyrell. 1999. Heme
ring. Chem. Lett. 1998:1001-1002. oxygenase activity determination by high-performance
31.Kennedy, G.Y. and H.G. Vevers. 1976. A survey of liquid chromatography. Methods Enzymol. 300:322-
avian eggshell pigments. Comp. Biochem. Physiol. 336.
55B:117-123. 49.Schluchter, W.M. and A.N. Glazer. 1997. Character-
32.Kutty, R.K. and M.D. Maines. 1981. Purification and ization of cyanobacterial biliverdin reductase. J. Biol.
characterization of biliverdin reductase from rat liver. Chem. 272:13562-13569.
J. Biol. Chem. 256:3956-3962. 50.Singleton, J.W. and L. Laster. 1965. Biliverdin reduc-
33.Lagarias, D.M., M.W. Crepeau, M.D. Maines, and tase of guinea pig liver. J. Biol. Chem. 240:4780-4789.
J.C. Lagarias. 1997. Regulation of photomorphogen- 51.Stocker, R., Y. Yamamoto, A.F. McDonagh, A.N.
esis by expression of mammalian biliverdin reductase in Glazer, and B.N. Ames. 1987. Science 235:1043-
transgenic Arabidopsis plants. Plant Cell 9:675-688. 1046.
34.Maines, M.D. 1988. Heme oxygenase: function, mul- 52.Stoll, M.S. and C.H. Gray. 1977. The preparation
tiplicity, regulatory mechanisms, and clinical applica- and characterization of bile pigments. Biochem. J.
tions. FASEB J. 2:2557-2568. 163:59-101.
35.Maines, M.D., N.G. Ibrahim, and A. Kappas. 1977. 53.Tenhunen, R., H.S. Marver, and R. Schmid. 1968.
Solubilization and partial purification of heme oxyge- The enzymatic conversion of heme to bilirubin by
nase from rat liver. J. Biol. Chem. 252:5900-5903. microsomal heme oxygenase. Proc. Natl. Acad. Sci.
36.McDonagh, A.F. and F. Assisi. 1971. Commercial USA 61:748-755.
bilirubin: a trinity of isomers. FEBS Lett. 18:315-317. 54.Terry M.J. and R.E. Kendrick. 1996. The aurea and
37.McDonagh, A.F. and L.A. Palma. 1980. Preparation yellow-green-2 mutants of tomato are deficient in phy-
and properties of crystalline biliverdin IXα. Simple tochrome chromophore synthesis. J. Biol. Chem.
methods for preparing isomerically homogenous 271:21681-21686.
biliverdin and (14C) biliverdin by using 2,3-dichloro- 55.Terry, M.J. and J.C. Lagarias. 1991. Holophy-
5,6-dicyanobenzoquinone. Biochem. J. 189:193-208. tochrome assembly. Coupled assay for phytochromo-
38.McDowell, M.D. and J.C. Lagarias. 1997. Partial bilin synthesis in organello. J. Biol. Chem. 266:22215-
purification, photoaffinity labeling, and characteriza- 22221.
tion of phytochromobilin synthase. Plant Physiol. 56.Terry, M.J., M.D. Maines, and J.C. Lagarias. 1993.
114:S739. Inactivation of phytochrome- and phycobiliprotein-
39.Muramoto, T., T. Kohchi, A. Yokota, I. Hwang, and chromophore precursors by rat liver biliverdin reduc-
H.M. Goodman. 1999. The Arabidopsis photomor- tase. J. Biol. Chem. 268:26099-26106.
phogenic mutant hy1 is deficient in phytochrome 57.Terry, M.J., M.D. McDowell, and J.C. Lagarias.
chromophore biosynthesis as a result of a mutation in a 1995. (3Z)- and (3E)-phytochromobilin are interme-
plastid heme oxygenase. Plant Cell 11:335-347. diates in the biosynthesis of the phytochrome chro-
40.Murphy, J.T. and J.C. Lagarias. 1997. The phyto- mophore. J. Biol. Chem. 270:11111-11119.
fluors: a new class of fluorescent protein probes. Curr. 58.Terry M.J., J.A. Wahleithner, and J.C. Lagarias.
Biol. 7:870-876. 1993. Biosynthesis of the plant photoreceptor phy-
41.Oren, D.A. 1997. Bilirubin, rem sleep, and photo- tochrome. Arch. Biochem. Biophys. 306:1-15.
transduction of environmental time cues. A Hypothe- 59.Turner, L., J.D. Houghton, and S.B. Brown. 1992.
sis. Chronobiol. Int. 14:319-329. Isolation and partial purification of phycocyanin
42.Ortiz de Montellano, P.R. 1998. Heme oxygenase apoprotein and its role in studies of bilin-apoprotein
mechanism: evidence for an electrophilic, ferric perox- attachment. Plant Physiol. Biochem. 30:309-314.
ide species. Acc. Chem. Res. 31:543-549. 60.Vreman, H.J., D.A. Cipkala, and D.K. Stevenson.
43.Prince, J., T.G. Nolen, and L. Coelho. 1998. Defen- 1996. Characterization of porphyrin heme oxygenase
sive ink pigment processing and secretion in Aplysia inhibitors. Can. J. Physiol. Pharmacol. 74:278-285.

290
Biosynthesis and Analysis of Bilins

61.Vreman, H.J. and D.K. Stevenson. 1988. Heme oxy- ly active chromophore precursor of the plant photore-
genase activity as measured by carbon monoxide pro- ceptor phytochrome. Proc. Natl. Acad. Sci. USA
duction. Anal. Biochem. 168:31-38. 93:8989-8994.
62.Weller, J.L., M.J. Terry, C. Rameau, J.B. Reid, and 67.Wu, S.-H., M.T. McDowell, and J.C. Lagarias.
R.E. Kendrick. 1996. The phytochrome-deficient pcd1 1997. Phycocyanobilin is the natural precursor of
mutant of pea is unable to convert heme to biliverdin the phytochrome chromophore in the green alga
IXa. Plant Cell 8:55-67. Mesotaenium caldariorum. J. Biol. Chem. 272:25700-
63.Weller, J.-P. and A. Gossauer. 1980. Synthese und 25705.
photoisomerisierung des racem. Phytochromobilin- 68.Yamaguchi, T., Y. Komoda, and H. Nakajima.
dimethylesters. Chem. Ber. 113:1603-1611. 1994. Biliverdin-IXα and biliverdin IXβ reductase
64.Wilks, A. and P.R. Ortiz de Montellano. 1993. Rat from human liver. J. Biol. Chem. 269:24343-
liver heme oxygenase. High level expression of a trun- 24348.
cated soluble form and nature of the meso-hydroxylat- 69.Yoshida, T. and G. Kikuchi. 1978. Features of the
ing species. J. Biol. Chem. 268:22357-22362. reaction of heme degradation catalyzed by the recon-
65.Wilks, A. and M.P. Schmitt. 1998. Expression and stituted microsomal heme oxygenase system. J. Biol.
characterization of a heme oxygenase (Hmu O) from Chem. 253:4230-4236.
Corynebacterium diphtheriae. Iron acquisition requires 70.Yoshida, T., M. Noguchi, and G. Kikuchi. 1982. The
oxidative cleavage of the heme macrocycle. J. Biol. step of carbon monoxide liberation in the sequence of
Chem. 273:837-841. heme degradation catalyzed by the reconstituted
66.Wu, S.-H. and J.C. Lagarias. 1996. The methy- microsomal heme oxygenase system. J. Biol. Chem.
lotrophic yeast Pichia pastoris synthesizes a functional- 257:9345-9348.

291
Analysis and Reconstitution of
13 Phytochromes

Michael T. McDowell and J. Clark Lagarias


University of California-Davis, Davis, CA, USA

1. THE PHYTOCHROME FAMILY via a thioether to an invariant cysteine resi-


due. Biochemical and molecular cloning
Photosynthetic organisms, from bacteria studies indicate that the basic architecture
to higher plants, possess light sensing mol- of eukaryotic phytochromes has been pre-
ecules that enable adaptation to fluctua- served, while a growing family of phyto-
tions in intensity, direction, duration, chrome-related genes in cyanobacteria
polarization and spectral quality of light encode polypeptides considerably more
from their environment (26). The most divergent in structure. Experimental meth-
well known of these photoreceptors are the ods outlined in this chapter discuss 2 major
phytochromes, which sense the ambient tools, difference spectroscopy and holo-
light conditions via their ability to pho- phytochrome assembly, necessary for estab-
tointerconvert between red (Pr) and far-red lishing whether candidate genes encode
(Pfr) light absorbing forms (17,47,51,57). bonafide phytochromes.
This unique property of phytochromes is With only 2 known exceptions, eukaryo-
conferred by a linear tetrapyrrole (bilin) tic phytochromes are soluble homodimeric
prosthetic group that is covalently linked proteins with a subunit roughly 1100 amino
to a large polypeptide. First discovered in acids in length (Figure 1). The bilin pros-
plants, phytochrome-like molecules also thetic group is associated with a highly con-
have been identified in lower eukaryotic served photosensory domain at the protein’s
plant species (i.e. green algae, mosses, and N terminus, which is readily cleaved from
ferns) (27,64) and, more recently, in cya- the C-terminal region by limited proteolysis
nobacteria (21,25,66,70). It is well estab- to yield a photochemically active 60 to 70
lished that higher plants possess multiple kDa monomer (24). The more diverged
phytochromes that are encoded by a small 500 amino acid C terminus of eukaryotic
nuclear gene family termed PHYA-F (5, phytochromes specifies the high affinity
45). All phytochrome proteins share a subunit–subunit interaction (23) and also
highly conserved photosensory domain, in possesses 2 regulatory subdomains that
which the bilin prosthetic group is linked genetic studies have established to be critical

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

293
M.T. McDowell and J.C. Lagarias

Figure 1. Domain structure of eukaryotic and cyanobacterial phytochromes and phytofluors. (A) Phytochromes exist in pho-
tointerconvertible red light absorbing (Pr) and far-red light absorbing (Pfr) forms. Phytochromes possess either phytochromobilin
or phycocyanobilin prosthetic groups bound to a conserved cysteine residue indicated with an asterisk. (B) Phytofluors are orange
fluorescent biliproteins consisting of phycoerythrobilin thioether-linked to the conserved cysteine residue on an apophytochrome.
Regulatory domains include the PAS-related domain (PRD), which contain 2 direct repeats shown as dark boxes, and the histi-
dine kinase domain (HKD) and the histidine kinase-related domain (HKRD), which are depicted with cross hatching.

294
Analysis and Reconstitution of Phytochromes

for transducing the light signal (reviewed in 2. DIFFERENCE SPECTROSCOPY


References 47 and 63). These include the FOR PHYTOCHROME
PRD, a domain related to the PAS domain QUANTITATION
found on eukaryotic regulatory proteins
(30,59), and the HKRD, a domain related 2.1. History and Development
to histidine kinase transmitter domains of 2
component sensor proteins from bacteria The application of difference spec-
(56). Like eukaryotic phytochromes, the troscopy dates back to the early stages of
cyanobacterial phytochrome Cph1 from phytochrome research with the discovery
Synechocystis sp PCC 6803 (21,70) possesses of the photoreversible nature of the biology
the conserved N-terminal photosensory of phytochrome. Action spectroscopy
domain and histidine kinase domain helped to define further the key wave-
(HKD), but lacks the PRD regulatory sub- lengths of light that phytochrome uses for
domain (Figure 1). Recent investigations its light switching mechanism (53). The
support the hypothesis that both Cph1 and key observation that led ultimately to the
eukaryotic phytochromes transduce the development of difference spectroscopy
light signal perceived by the photosensory was that the physiological effects of red
domain via changes in protein kinase activi- light irradiation on higher plants and algae
ty of the regulatory domains (69,70). could, in many cases, be reversed or nulli-
It was not until the purification of native fied by a subsequent irradiation with far-
phytochrome from plants that a chemical red light. The result of these observations
examination of the chromophore of phy- has become a key diagnostic test for phy-
tochrome took place. 1H-Nuclear magnetic tochrome-mediated responses: red, far-red
resonance (NMR) analysis of chro- light photoreversibility. For the purifica-
mopeptides from oat phytochrome A tion and characterization of phytochrome
revealed that its chromophore and linkage proteins, this led to the development of the
to the apoprotein were very similar to those difference spectrum. The two forms of
found in phycobiliproteins (34,52). These phytochrome, Pr and Pfr, are spectrally dif-
studies also revealed that oat phytochrome ferent (Figure 2A). When the Pfr absorp-
possessed a phytochromobilin (PΦB) chro- tion spectrum is subtracted from the Pr
mophore (Figure 1), confirming earlier absorption spectrum, the result is a differ-
investigations of bilins obtained from phy- ence spectrum with a very characteristic
tochromes by chemical cleavage (50). The wave appearance (Figure 2B). The differ-
precursors of the chromophores of the ence between the maximum and the mini-
other phytochrome genes within a given mum is then used to quantitate the
plant species (i.e., PHYB-E) have not been amount of spectrally active phytochrome
directly determined, but they are assumed present in the sample.
to be PΦB. One exception is phytochrome
from the green alga Mesotaenium caldario- 2.2. Difference Spectral Assay for
rum, which possesses the phycobiliprotein Holophytochrome
chromophore precursor phycocyanobilin
(PCB) (68). The natural chromophore pre- The following methodology refers only
cursor of Cph1 has not yet been deter- to the measurement of phytochrome in
mined. PCB has been proposed as a likely extracts and is based on the methods devel-
candidate owing to its intermediacy in the oped in this laboratory (31). For a discus-
phycobiliprotein chromophore biosynthetic sion of in vivo phytochrome photoassay,
pathway in Synechocystis sp. PCC 6803 (6). we refer the reader to the following refer-
295
M.T. McDowell and J.C. Lagarias

ences (19,46). Two major caveats about be performed in a laboratory and kept
phytochrome measurements need to be under dim green safelight (54). Second, the
made at this point. First, to avoid continu- validity of the phytochrome difference
ous photocycling, all measurements should assay depends upon the lack of other pig-

Figure 2. Absorption and difference spectra of purified recombinant oat phytochrome. (A) Absorption spectra of Pr and Pfr
forms of PΦB and PCB adducts of recombinant oat apophytochrome A (AsphyA). (B) Difference spectra of PΦB and PCB
adducts of recombinant AsphyA. Adapted from Murphy and Lagarias (44).

296
Analysis and Reconstitution of Phytochromes

ments that absorb in the 500 to 800 nm absorption maximum) no longer chan-
wavelength range. For this reason, chloro- ges with further illumination. For puri-
phyll must be removed from extracts, from fied phytochrome preparations, absor-
light-grown plant extracts which can be bances between 250 and 800 nm are
accomplished by precipitation with poly- recorded; for crude samples, a 500 to
ethyleneimine (41). The latter is not a con- 800 nm range is used. The wavelength
cern with recombinant phytochromes pre- of red light needed is dependent on the
pared from bacterial, yeast, or insect cell species of phytochrome under exami-
extracts. Our standard method for phy- nation. For phytochromes containing
tochrome photoassay is outlined in Proce- the PΦB chromophore, the red actinic
dure 1. Because actinic illumination and light should be 660 nm (± 5 nm), and
sample measurement can be performed for PCB-containing phytochrome, the
simultaneously, we recommend the use of actinic light should be less than 650
a diode array spectrophotometer, such as nm (e.g., we use 636 ± 5 nm). Actinic
the Model 8453 from Hewlett Packard light is typically provided by a 250 W
(Wilmington, DE, USA). The length of quartz halogen lamp filtered through
irradiation will also depend on the light an appropriate interference filter (Cori-
source. Typically, for a focused 250 W on, Franklin, MA, USA).
quartz halogen lamp, a 200-second irradia- 3. The sample is then irradiated with sat-
tion is sufficient. The limit of sensitivity for urating far-red light, and another ab-
this assay is approximately 10 nM phy- sorption spectrum is taken. Far-red
tochrome (i.e., 1 µg/mL protein). light is obtained using a 250 W quartz
halogen lamp filtered through an FRS
❖ Procedure 1. Phytochrome Photoassay 700 black plexiglas filter (Rohm and
Hass, Philadelphia, PA, USA).
1. The protein sample, either from plant 4. The difference spectrum and quantita-
tissue or reconstituted recombinant tion of photoactive phytochrome is
phytochrome (see later section), is obtained by mathematically subtract-
diluted or reconstituted in the desired ing the spectrum of the red light-irra-
amount of TEGE buffer [25 mM Tris- diated sample from the spectrum of the
HCl, pH 7.8, containing 25% (vol/ far-red light-irradiated sample (see Fig-
vol) ethylene glycol, 1 mM EDTA, 2 ure 2B). The quantitation is obtained
mM phenylmethylsulfonyl fluoride by measuring the ∆∆A from the Amax
(PMSF) and 1 mM dithiothreitol and the Amin of the difference spec-
(DTT)], typically 500 µL. During irra- trum. To correct for the incomplete
diation and the subsequent analysis, photoconversion of Pr to Pfr and back,
the sample is kept at 10° to 16°C. the ∆∆A is multiplied by 0.86. To cal-
2. The sample is transferred to a 1-cm culate the molar concentration of
pathlength spectrophotometric cu- phytochrome, the corrected ∆∆A value
vette, placed in the spectrophotometer, is entered into the Lambert-Beers Law
irradiated with saturating red light, and equation of Acorr = εcl, where ε665 =
an absorption spectrum is taken. Satu- 132 000 M-1cm-1 for purified oat phy-
rating illumination is defined as the tochrome A (31).
amount of illumination needed to pro- 5. To determine the relative purity for a
duce a photostationary state and is particular phytochrome preparation,
determined empirically when the ab- the specific absorbance ratio (SAR) is
sorbance at 650 to 665 nm (i.e., Pr’s determined. To do this, the full spec-
297
M.T. McDowell and J.C. Lagarias

trum of the Pr form of phytochrome higher plant apophytochrome (21,70).


(i.e., sample irradiated with saturating This discovery led us to develop the cou-
far-red light) is obtained, and the pled assay for the purification of PΦB syn-
absorption of the red light maximum thase which follows. This method is essen-
(i.e., 660 nm) is divided by the absorp- tially that described by Terry and Lagarias
tion of the protein maximum (i.e., 280 (60), with the exceptions that Cph1 apo-
nm). Purified full-length phytochrome phytochrome is substituted for oat apophy-
A preparations typically exhibit SARs tochrome A, and apophytochrome is
of 1.0 to 1.1 (31,36). added only after the assay is complete.

2.3. Coupled Difference Spectral Assay ❖ Procedure 2. PΦB Synthase Assay


for PΦB Synthase Activity Coupled to Phytochrome
Another use for the phytochrome differ- 1. Crude plastid preparations from dark
ence spectrum was first described by Elich grown oats were typically used as a
and Lagarias (13,15) and more recently source of phytochromobilin synthase
was exploited by Terry and Lagarias (60). activity (42,60). Dilute protein samples
This application was developed for the in a 1-mL final volume of 50 mM
study of phytochrome chromophore bio- TES/KOH (pH 7.3) containing an
synthesis. At the time of its development, NADPH regenerating system (6.5 mM
there was no reliable way to assay directly glucose 6-phosphate, 0.82 mM
the bilin intermediates in the biosynthesis NADP+, 1.1 U/mL Torula yeast Type
of the phytochrome chromophore. This XII glucose-6-phosphate dehydroge-
application suffers from two drawbacks, nase EC 1.1.1.49), a ferredoxin, ferre-
however. The first is that the reaction can- doxin-reducing system (4.6 µM puri-
not be used to study the kinetics of the fied spinach ferredoxin, 0.025 U/mL
biosynthetic reactions. The other is, until ferredoxin:NADP+ oxidoreductase EC
recently, the lack of an abundant source of 1.18.1.2), and 10 µM bovine serum
recombinant apophytochrome, as the sys- albumin (BSA) (fraction V, heat shock).
tems previously developed for expression of
recombinant higher plant apophytochrome 2. Assays are initiated by adding biliverdin
could only produce small amounts of pro- (BV) IXα in 10 µL of dimethyl sulfox-
tein. Subsequently, high-performance liq- ide (DMSO). Usually, the final concen-
uid chromatography (HPLC) methods tration of BV in an assay was 5 µM.
were developed to assay linear tetrapyrrole 3. Assay mixtures were incubated in a
metabolism more directly (see Chapter 12 28°C water bath under green safelight
in this text by Terry). While HPLC analy- or subdued light for the desired
ses have facilitated the purification and amount of time and stopped by placing
biochemical characterization of phytochro- them on ice. For assays of intact plastid
mobilin synthase (42), they are somewhat fractions or membrane fractions, the
tedious and time-consuming, making assays were clarified by centrifugation
analysis of column fractions a chore. The at 12 000× g for 15 minutes at 4°C
recent discovery of a Cph1 from Synecho- prior to analysis.
cystis sp. PCC 6803 had a significant side 4. PΦB synthase assays can be analyzed
benefit; recombinant Cph1 apoprotein either by HPLC (as described in Chap-
could be expressed in Escherichia coli and ter 12 in this text by Terry) or by differ-
obtained in much larger amounts than ence spectroscopy. To obtain a difference
298
Analysis and Reconstitution of Phytochromes

spectrum, a small aliquot of concen- examined to date can catalyze thioether for-
trated recombinant Cph1 is added to mation with PΦB, as well as the phyco-
the PΦB synthase assay mixture. bilins, PCB, and phycoerythrobilin (PEB),
5. The assay was incubated an additional both precursors of the phycobiliproteins
20 to 30 minutes at room temperature (37). This work is in striking contrast with
to facilitate assembly, and then a differ- phycobiliprotein assembly, whose bilin liga-
ence spectrum is obtained as outlined tion requires separate lyases for proper
above. assembly (see Chapter 14 in this text by
Schluchter and Bryant). Analyses of bilin
assembly to apophytochrome have relied
3. ASSAYS FOR HOLOPHYTO- on 3 major tools: (i) difference spectro-
CHROME ASSEMBLY scopy (described above); (ii) zinc blot ana-
lysis; and (iii) fluorescence spectroscopy. In
3.1. History and Development the sections that follow, the latter 2 tools
will be highlighted.
Based on the pioneering work of Gard-
ner and collaborators (22), it is well estab-
lished that the chromophore and apophy- 3.2. Zinc Blot Assay for
tochrome biosynthetic pathways are Bilin Attachment
essentially independent. Plants treated with
inhibitors of 4-aminolevulinic acid (ALA) Early examination of bile pigments re-
biosynthesis, such as gabaculine or amino- vealed that bilatrienes form intensely fluo-
5-hexynoic acid, possess reduced amount rescent complexes with zinc ions in the
of photoactive holophytochrome, but accu- presence of iodine (1). This and related
mulate near normal amounts of apophy- observations have led to the examination of
tochrome (12,14,22). Using these in- fluorescent zinc complexes of bilin-linked
hibitors, it was shown that co-incubating polypeptides. The initial work was per-
the plants with ALA and BV IXα, as well formed using standard sodium dodecyl sul-
as the unnatural isomer BV XIIIα, could fate polyacrylamide gel electrophoresis
restore the levels of spectrally active phy- (SDS-PAGE) gels with the fluorescent
tochrome (15). The phycobiliprotein chro- products being observed following UV
mophore precursor PCB was also found to illumination (2). The purpose of SDS-
restore the levels of spectrally active phy- PAGE is to remove the unbound bilin
tochrome, albeit with a blue-shifted differ- from the protein-bound bilin. This
ence spectrum (see Figure 2B). Follow-up methodology, which can be used for
investigations utilizing apophytochrome phycobiliproteins as well (48), has been
isolated from inhibitor-treated plants indi- further improved by the extension to
cated that either the lyase enzyme copuri- electroblotted protein samples or the
fies with apophytochrome, or that apophy- “zinc blot” (37). The zinc blot is used as a
tochrome is itself a bilin lyase (13). The general diagnostic technique to assess the
development of recombinant apophyto- ligation competency of a particular
chrome expression systems by many labo- apophytochrome. The limit of sensitivity of
ratories (7,10,13,18,20,21,29,32,33,35,55, this technique is approximately 50 ng/cm
62,65) have corroborated the latter hypo- of phytochrome per lane for gels and
thesis that apophytochrome is a bilin lyase 12 ng/cm of phytochrome per lane for
that catalyzes thioether linkage formation blots. Procedure 2, for the zinc blot assay,
with bilins. All bonafide apophytochromes is based on 2 references (2,37).
299
M.T. McDowell and J.C. Lagarias

❖ Procedure 3. The Zinc Blot Assay 3.3. Fluorescence Assay for


for Bilins Holophytochrome Assembly

1. Protein samples to be analyzed are elec- The natural biological function of phy-
trophoresed in a SDS-polyacrylamide tochrome is to act as a light switch due to
gel using any standard procedure. After its ability to photointerconvert between the
electrophoresis, the gel is transblotted Pr and Pfr light-absorbing forms. Based on
to a polyvinylidene difluoride (PVDF) NMR and resonance Raman spectroscopic
membrane using any standard proce- analysis, this photoconversion has been
dure for electroblotting. proposed to involve the Z to E isomeriza-
NOTE: Nylon and nitrocellulose are con- tion of the C15 methine bridge double
siderably less effective owing to their bond (16,52). While the 2 known phy-
greater autofluorescence background tochrome chromophore precursors, PΦB
and large UV absorption, respectively. and PCB, possess this double bond, PEB
does not. Indeed, PEB adducts of apophy-
2. After electroblotting, the membrane is tochromes are inactive photochemically, a
washed briefly with deionized water. result that led to the hypothesis that such
3. After the water wash, the blot is trans- adducts might be fluorescent. That PEB
ferred to 1.3 M zinc acetate in deion- incubation with apophytochromes produce
ized water and incubated at room tem- intensely fluorescent adducts was first docu-
perature for roughly 30 minutes under mented in 1995 (39). This finding led to
reduced light. the development of a real-time kinetic assay
4. Just prior to visualization, the blot is for the study of phytochrome assembly that
rinsed with deionized water to remove is described below. This assay has not only
excess Zn2+ ion. enabled the determination of both bilin
5. The blot is visualized by placing on a binding and catalytic rate constants for the
long wavelength UV transilluminator reconstitution of holophytochrome with its
and photographing with Technical Pan natural chromophore (i.e., Kd ∼ 1 µM, kcat
film (Type 4415; Eastman Kodak, ∼ 0.25–0.3 s-1), but it has also facilitated
Rochester, NY, USA) using an RG-630 the analysis of potential inhibitors of this
cutoff filter (Schott, Laurel, NJ, USA) process, such as the PΦB precursor BV (i.e.,
(2-min exposure). Alternatively, the Ki ∼ 1 mM). The procedures for these
blot can be imaged using a Storm assays, summarized below, are based on the
860 PhosphorImager® (Molecular work of Li, Murphy, and Lagarias (39). Two
Dynamics, Sunnyvale, CA, USA) with major fluorescent assay methodologies are
the red laser in fluorescence mode. The described below, the standard and the com-
image obtained using the Storm can be petitive assays. The only difference between
analyzed using the ImageQuant soft- the kinetic assays is the data analysis. The
ware or converted to tagged image for- data analysis for the standard assay is out-
mat files (TIFF) and analyzed using a lined in Scheme 1. The analyses of the 2
types of competitive fluorescence assays are
program such as National Institutes of
outlined in Schemes 2 and 3, respectively.
Health (NIH) Image. For quantitative
analyses, a dilution series of known ❖ Procedure 4. Fluorescence Assay for
quantities of holophytochrome (or Holophytochrome Assembly
phycobiliprotein) should be included
on each blot. 1. The formation of the fluorescent PEB-

300
Analysis and Reconstitution of Phytochromes

phytochrome adduct is initiated in a mM EDTA, 1 mM PMSF, and 1 mM


semimicrofluorescence cuvette by addi- DTT]. The PEB is dissolved in Me2SO,
tion of apophytochrome (10 nM final with the final Me2SO concentration in
concentration) from a concentrated the assay no greater than 2% (vol/vol).
stock solution to a greater-than or equal The assay mixture is rapidly mixed and
to 70-fold molar excess of PEB (typical- placed in a fluorescence spectropho-
ly 0.5 to 15 µM final concentration) in tometer. Typically, measurements are
TEGE buffer [10 mM Tris-HCl, pH performed with samples maintained at
8.0, 25% (vol/vol) ethylene glycol, 1 room temperature (22°–25°C).

Scheme 1. Kinetic analysis of holophytochrome assembly (adapted from Reference 39). The enzyme reaction of a bilin, typical-
ly PEB, with apoPC to produce a fluorescent product is shown in the diagram. Equation 1 is the integrated rate equation describ-
ing this reaction. Equation 2 defines the kinetics of bilin–apophytochrome adduct formation. If the bilin precursor concentration
is kept essentially constant, by providing a large excess, semilog plots of the fraction of apophytochrome remaining are expected to
be linear as described in Equation 3. Equation 4 and its reciprocal, Equation 5, provide a means for determining the affinity of
apophytochrome for a particular bilin, Kbilin, and the rate constant, or turnover, of apophytochrome for a particular bilin, k2.

301
M.T. McDowell and J.C. Lagarias

2. For time-based measurements, samples Experimentally, the amount of com-


were excited with 570 nm light with 2 petitor bilin is estimated from the
nm bandpass. Fluorescence emission degree of fluorescence inhibition rela-
data was collected at 586 nm with 16 tive to the control reaction with no
nm bandpass. Data was collected with inhibitor. Since both the concentration
1-second integration for 15 to 30 min- of PEB-phytochrome (PC) adduct and
utes. Saturated fluorescence intensity the kapp for PEB-PC formation are
(i.e., 100% assembly) is determined in known, the kIapp can be calculated
parallel by incubating a control sample using Equation 13 (Scheme 3). The
of phytochrome with a very large KIbilin is obtained following a double
excess of PEB (>500-fold molar excess) reciprocal plot of the kIapp versus the
for more than 1 hour. bilin concentration as shown in Equa-
3. The equations outlining the analysis of tion 15 (Scheme 3).
data for the standard kinetic assay with
PEB and apophytochrome are outlined
4. BILIN AND APOPHYTOCHROME
in Scheme 1, and example data is shown
SPECIFICITY FOR THIOETHER
in Figure 3. Raw fluorescence data is
LINKAGE FORMATION
transformed using Equation 3 in
Scheme 1. When data is replotted on a
semilog graph, kapp values for each 4.1. Bilin Specificity for Thioether
assembly reaction are determined from Linkage Formation
the slope of the line. According to Equa-
tion 5 (Scheme 1), 1/kapp values for the The question of bilin chromophore pre-
different assemblies are then plotted ver- cursor specificity for holophytochrome
sus 1/(PEB). The x- and y-intercepts for assembly has been addressed using zinc
this data provide the Kbilin and kcat, or blot analysis, difference spectroscopy, and
k2, respectively, for PEB. fluorescence spectroscopy (15,37,40). The
requirements for assembly are an A-ring
Variations:
ethylidene at the C3 position, as is present
4. The analysis of data for the competitive in PΦB, PCB, and PEB (37), and a C10
assay using a reversible inhibitor of methine bridge. The former conclusion is
PEB-phytochrome formation such as based on in vivo feeding of BVs IIIα, IXα,
BV (see Scheme 2), is carried out in and XIIIα; BV IXα and XIIIα feeding
much the same manner as outlined for restored levels of spectrally active phy-
the standard assay above. The raw fluo- tochrome, while BV IIIα had no apparent
rescence data is transformed using effect (15). Assembly of a bilin with an eth-
Equation 8 (Scheme 2). This data is ylidene at the C2 position cannot be ruled
graphed on a semilog plot to obtain the out, but this compound is not readily avail-
kapp as before. The KI for BV, or KBV, able, and based on BV IIIα feeding experi-
is estimated using the x-intercept of the ments, it is probably not biologically rele-
plot of the 1/kapp versus the BV con- vant. The requirement for a C10 methine
centration (Equation 10 in Scheme 2). bridge is based on the inability of rubins,
5. The analysis of the data when using an including those possessing an A-ring eth-
irreversible inhibitor of PEB-apophy- ylidene moiety, such as phycocyanorubin,
tochrome adduct formation, such as to assemble with apophytochrome (61).
PΦB or PCB, is much different from The observations that the D-ring can be
the previous examples (Scheme 3). modified including 18-vinyl reduction,
302
Analysis and Reconstitution of Phytochromes

Scheme 2. Reversible competitive inhibition of PEB phytochrome assembly. A kinetic model for PEB adduct formation in the
presence of a reversible competitive inhibitor such as BV. The kinetics of PEB adduct formation should be pseudo-first-order as
predicted by Equation 7. The raw fluorescence data is transformed and replotted as described by Equation 8. The slopes of these
semilog replots yield kapp values. These values are used to construct the plot described by Equation 10. The x-intercept of Equa-
tion 10 yields an estimate of the equilibrium dissociation constant for the reversible competitive inhibitor.

303
M.T. McDowell and J.C. Lagarias

Scheme 3. Irreversible inhibition of PEB phytochrome assembly. A kinetic model for PEB adduct formation in the presence of
an irreversible competitive inhibitor such as PΦB. The formation of both PEB-phytochrome and competitor bilin–phytochrome
are described by Equations 11 and 12. In the presence of large molar excesses of all bilins, these equations are first-order expres-
sions. The kappi values are calculated using Equation 13, then plotted as a function of the competitive inhibitor according to
Equations 14 and 15. A plot of Equation 15 yields the dissociation constant (Ki) and the catalytic rate constant (k4) for the com-
petitive inhibitor of fluorescent adduct formation.

304
Analysis and Reconstitution of Phytochromes

switching of the C17 and C18 methyl and required for assembly, as demonstrated by
vinyl moieties and elaboration of the C18 the binding of PEB to apophytochome.
side chain reveal that the bilin binding BVs and bilirubins, which lack the A-ring
pocket of apophytochrome is not very dis- ethylidene moiety, also do not form cova-
cerning with regard to the C18 substituent lent adducts with phytochrome, although
(15,37,40). The C15 methine bridge is not the former are capable of noncovalent

Figure 3. Fluorescence assay for holophytochrome assembly. Representative data for standard fluorescence analysis of PEB
attachment to recombinant oat phytochrome A, after Li et al. (39). The upper panel shows raw fluorescence kinetic data as a func-
tion of increasing PEB concentration. The middle panel is a replot of the same data according to Equation 3 (Scheme 1), from
which kapp values were estimated. The bottom panel depicts a replot of 1/kapp versus 1/(PEB) according to Equation 5 (Scheme
1). See text and Reference 39 for details.

305
M.T. McDowell and J.C. Lagarias

interaction with phytochrome and, there- presently unresolved. However, the observed
fore, can act as reversible competitive bilin lyase activity recombinant pea apophy-
inhibitors as discussed above. The require- tochrome mutants, in which this histidine
ment for both propionic acid moieties has residue was changed to a glutamine or argi-
been established by the inability of bilin– nine residue, suggest otherwise (3,58).
esters to bind to apophytochrome (3). Ongoing studies to identify catalytically
important residues for bilin lyase activity
4.2. Apophytochrome Specificity for will take advantage of the growing family of
Thioether Linkage Formation phytochrome-related proteins in cyanobac-
teria, in which deletions, insertions, and
While bilin specificity has been actively amino acid substitutions, which influence
addressed, less is known about the regions of bilin ligation, can be assessed.
the apophytochrome required for bilin
attachment. Thus far, the only unequivocal 4.3. Assembly of Recombinant
requirement is the conserved cysteine, Phytochromes In Vivo
through which the bilin forms its thioether
linkage (cys-321 in the case of oat PHYA3) Recently, recombinant expression of
(3,4). Much effort has been directed at try- phytochrome led to a novel application
ing to determine other amino acid residues (11,28,38,67). Phytochrome expressed in
or regions of the protein that are involved a heterologous system such as Saccharo-
either directly or indirectly with the lyase myces cerevisiae could be assembled in vivo
activity. Deletion analysis of phytochrome if the chromophore was supplied exoge-
and expression of the truncated proteins in nously. The key stumbling block was get-
either E. coli or yeast have established that ting the chromophore into living cells.
neither the first 68 amino acids nor the This could be accomplished by dissolving
entire C-terminal domain are required for the chromophore precursor in DMSO,
the autocatalytic assemble of recombinant which was added to a minimal buffer
phytochromes (10,20). With recombinant medium at a final concentration of 50 µM
rice apophytochrome however, the deletion (38). The dissolved chromophore was then
of the first 80 residues abolished bilin bind- diluted in the appropriate buffer to no
ing (62). Site-directed mutagenesis of the more than 10% (vol/vol). The cells were
region surrounding the conserved cysteine able to take up the bilin that assembled
attachment site has been undertaken by sev- with the recombinant phytochrome, while
eral groups (3,9,49,58). These experiments the cells remained viable. The ability to
have so far failed to identify other residues reconstitute holophytochromes in living
essential for bilin assembly, although an cells provides a powerful tool for struc-
important role for the histidine residue adja- ture–function analysis of this photorecep-
cent to the conserved cysteine (i.e., H324 in tor family in nonplant cell systems and has
pea PHYA) has been proposed based of the also led to the development of a new fami-
loss-of-function of site-directed mutants of ly of apophytochrome-based fluorescent
this histidine residue (3,9,49). Interestingly, probes called phytofluors (43).
for the Cph1-related bacteriophytochrome
BphP from Deinococcus radiodurans, which 4.4. Phytofluors: A New Class of
lacks the conserved cysteine residue, this Fluorescent Protein Probe
adjacent histidine appears to be the site of
bilin binding (8). Whether this histidine Phytofluors are intensely orange fluores-
represents a catalytically important residue is cent adducts that are formed spontaneous-
306
Analysis and Reconstitution of Phytochromes

ly upon co-incubation of apophytochrome ABBREVIATIONS


with PEB (see Figure 1B and Reference
43). The intense molar absorption coeffi- ALA, 5-aminolevulinic acid; BV, bili-
cient of PEB-apophytochrome adducts and verdin IXα; HKRD, histidine kinase-relat-
its spectrofluorometric properties (i.e., ed domain; Me2SO, dimethyl sulfoxide;
photostability, very sharp excitation, and PCB, phycocyanobilin; PEB, phycoery-
emission maxima at 576 and 586 nm, throbilin; PΦB, phytochromobilin; Pr, red
respectively) make phytofluors ideal candi- light absorbing form of phytochrome; Pfr,
dates as in vivo fluorescent protein tags. far-red light absorbing form of phyto-
PEB can be fed to organisms that are chrome; PRD, PAS-related domain.
expressing an apophytochrome gene. PEB
is taken up by plant cells and autocatalyti-
cally assembles with apophytochrome to REFERENCES
produce a fluorescent adduct that can be 1.Auche, A. 1908. Comptes Rendus De Societe Biologie
detected by techniques such as confocal 64:297-298.
microscopy (43). No central method has 2.Berkelman, T.R. and J.C. Lagarias. 1986. Visualiza-
tion of bilin-linked peptides and proteins in polyacry-
been developed for phytofluors that is lamide gels. Anal. Biochem. 156:194-201.
broadly applicable to all possible uses of 3.Bhoo, S.H., T. Hirano, H.Y. Jeong, J.G. Lee, M.
this novel fluorescent label. There are 2 Furuya, and P.S. Song. 1997. Phytochrome pho-
tochromism probed by site-directed mutations and
requirements for the use of phytofluors: (i) chromophore esterification. J. Am. Chem. Soc.
ligation-competent apophytochrome, and 119:11717-11718.
(ii) PEB. Transgenic expression of various 4.Boylan, M. and P. Quail. 1989. Oat Phytochrome is
biologically active in transgenic tomatoes. Plant Cell
phytochromes in a variety of bacteria, 1:765-773.
yeast, and mammalian cells has been 5.Clack, T., S. Mathews, and R.A. Sharrock. 1994. The
demonstrated. The key limitations for the phytochrome apoprotein family in Arabidopsis is
encoded by five genes—the sequences and expression
application of this technique at present are of PhyD and PhyE. Plant Mol. Biol. 25:413-427.
PEB uptake and catabolism by different 6.Cornejo, J. and S.I. Beale. 1997. Phycobilin biosyn-
types of cells and the commercial availabil- thetic reactions in extracts of cyanobacteria. Photo-
synth. Res. 51:223-230.
ity of free PEB. In all the examples of the 7.Cornejo, J., S.I. Beale, M.J. Terry, and J.C. Lagarias.
phytofluor technology, PEB has been sup- 1992. Phytochrome assembly—the structure and bio-
logical activity of 2(R),3(E)-phytochromobilin derived
plied exogenously in a buffered Me2SO from phycobiliproteins. J. Biol. Chem. 267:14790-
solution. One goal for further development 14798.
of this technology is the coexpression of 8.Davis, S.J., A.V. Vener, and R.D. Vierstra. 1999. Bac-
teriophytochromes: phytochrome-like photoreceptors
bilin biosynthetic enzymes with apophy- from nonphotosynthetic eubacteria. Science 286:
tochrome. Research toward this end is 2517-2520.
ongoing in a number of laboratories. 9.Deforce, L., M. Furuya, and P.S. Song. 1993. Muta-
tional analysis of the pea phytochrome a chromophore
pocket—chromophore assembly with apophytochrome
A and photoreversibility. Biochemistry 32:14165-
ACKNOWLEDGMENTS 14172.
10.Deforce, L., K.I. Tomizawa, N. Ito, D. Farrens, P.S.
Song, and M. Furuya. 1991. In vitro assembly of
We thank Beronda Montgomery, Nicole apophytochrome and apophytochrome deletion
Frankenberg, and Jihong Wang for helpful mutants expressed in yeast with phycocyanobilin. Proc.
Natl. Acad. Sci. USA 88:10392-10396.
comments regarding this manuscript. We 11.Eichenberg, K., T. Kunkel, T. Kretsch, V. Speth, and
also gratefully acknowledge the support E. Schafer. 1999. In vivo characterization of chimeric
from the United States Department of phytochromes in yeast. J. Biol. Chem. 274:354-359.
12.Elich, T.D. and J.C. Lagarias. 1988. 4-Amino-5-
Agriculture Competitive Research Grant hexynoic acid—a potent inhibitor of tetrapyrrole
No. AMD-9801768 to J.C.L. biosynthesis in plants. Plant Physiol. 88:747-751.

307
M.T. McDowell and J.C. Lagarias

13.Elich, T.D. and J.C. Lagarias. 1989. Formation of a 29.Kunkel, T., K. Tomizawa, R. Kern, M. Furuya, N.H.
photoreversible phycocyanobilin-apophytochrome Chua, and E. Schafer. 1993. In vitro formation of a
adduct in vitro . J. Biol. Chem. 264:12902-12908. photoreversible adduct of phycocyanobilin and tobac-
14.Elich, T.D. and J.C. Lagarias. 1987. Phytochrome co apophytochrome B. Eur. J. Biochem. 215:587-594.
chromophore biosynthesis. Both 5-aminolevulinic acid 30.Lagarias, D.M., S.H. Wu, and J.C. Lagarias. 1995.
and biliverdin overcome inhibition by gabaculine in Atypical phytochrome gene structure in the green alga
etiolated Avena sativa L. seedlings. Plant Physiol. mesotaenium caldariorum. Plant Mol. Biol. 29:1127-
84:304-310. 1142.
15.Elich, T.D., A.F. McDonagh, L.A. Palma, and J.C. 31.Lagarias, J.C., J.M. Kelly, K.L. Cyr, and W.O. Smith,
Lagarias. 1989. Phytochrome chromophore biosyn- Jr. 1987. Comparative photochemical analysis of high-
thesis. Treatment of tetrapyrrole-deficient Avena explants ly purified 124 kilodalton oat and rye phytochromes in
with natural and non-natural bilatrienes leads to for- vitro. Photochem. Photobiol. 46:5-13.
mation of spectrally active holoproteins. J. Biol. Chem. 32.Lagarias, J.C. and D.M. Lagarias. 1989. Self assembly
264:183-189. of synthetic phytochrome holoprotein in vitro. Proc.
16.Fodor, S.P.A., J.C. Lagarias, and R.A. Mathies. 1990. Natl. Acad. Sci. USA 86:5778-5780.
Resonance Raman analysis of the Pr and Pfr forms of 33.Lagarias, J.C. and F.M. Mercurio. 1985. Structure
phytochrome. Biochemistry 29:11141-11146. function studies on phytochrome. Identification of
17.Furuya, M. 1993. Phytochromes—their molecular light-induced conformational changes in 124-kDa
species, gene families, and functions. Ann. Rev. Plant Avena phytochrome in vitro. J. Biol. Chem. 260:2415-
Physiol. Plant Mol. Biol. 44:617-645. 2423.
18.Grimm, R., G.K. Donaldson, SM Vandervies, E. 34.Lagarias, J.C. and H. Rapoport. 1980. Chromopep-
Schafer, and A.A. Gatenby. 1993. Chaperonin-medi- tides from phytochrome. The structure and linkage of
ated reconstitution of the phytochrome photoreceptor. the Pr form of the phytochrome chromophore. J. Am.
J. Biol. Chem. 268:5220-5226. Chem. Soc. 102:4821-4828.
19.Gross, J., M. Seyfried, L. Fukshansky, and E. Schaefer. 35.Lamparter, T., F Mittmann, W. Gartner, T. Borner,
1984. In vivo spectrophotometry, p. 131-158. In H. E. Hartmann, and J. Hughes. 1997. Characterization
Smith and M.G. Holmes (Eds.), Techniques in Photo- of recombinant phytochrome from the cyanobacteri-
morphogenesis. Academic Press, New York. um Synechocystis. Proc. Natl. Acad. Sci. USA
20.Hill, C., W. Gartner, P. Towner, S.E. Braslavsky, and 94:11792-11797.
K. Schaffner. 1994. Expression of phytochrome 36.Lapko, V.N. and P.S. Song. 1995. A simple and
apoprotein from Avena sativa in Escherichia coli and improved method of isolation and purification for
formation of photoactive chromoproteins by assembly native oat phytochrome. Photochem. Photobiol.
with phycocyanobilin. Eur. J. Biochem. 223:69-77. 62:194-198.
21.Hughes, J., T. Lamparter, F. Mittmann, E. Hart- 37.Li, L. and JC Lagarias. 1992. Phytochrome assem-
mann, W. Gartner, A. Wilde, and T. Borner. 1997. A bly—defining chromophore structural requirements
prokaryotic phytochrome. Nature 386:663-663. for covalent attachment and photoreversibility. J. Biol.
22.Jones, A.M., C.D. Allen, G. Gardner, and P.H. Quail. Chem. 267:19204-19210.
1986. Synthesis of phytochrome apoprotein and chro- 38.Li, L. and J.C. Lagarias. 1994. Phytochrome assembly
mophore are not coupled obligatorily. Plant Physiol. in living cells of the yeast Saccharomyces cerevisiae. Proc.
81:1014-1016. Natl. Acad. Sci. USA 91:12535-12539.
23.Jones, A.M., and M.D. Edgerton. 1994. The anatomy 39.Li, L., J.T. Murphy, and J.C. Lagarias. 1995. Contin-
of phytochrome, a unique photoreceptor in plants. uous fluorescence assay of phytochrome assembly in
Sem. Cell Biol. 5:295-302. vitro. Biochem. 34:7923-7930.
24.Jones, A.M., R.D. Vierstra, S.M. Daniels, and P.H. 40.Lindner, I., B. Knipp, S.E. Braslavsky, W. Gartner,
Quail. 1985. The role of separate domains in the struc- and K. Schaffner. 1998. A novel chromophore selec-
ture of phytochrome from etiolated Avena sativa L. tively modifies the spectral properties of one of the two
Planta. 164:505-506. stable states of the plant photoreceptor phytochrome.
25.Kehoe, D.M. and A.R. Grossman. 1996. Similarity of Angew. Chem., Int. Ed. 37:1843-1846.
a chromatic adaptation sensor to phytochrome and 41.Litts, J.C., J.M. Kelly, and J.C. Lagarias. 1983. Struc-
ethylene receptors. Science 273:1409-1412. ture-function studies on phytochrome. Preliminary
26.Kendrick, R.E. and G.H.M. Kronenberg (Eds.). characterization of highly purified phytochrome from
1994. Photomorphogenesis in Plants, 2nd ed. Marti- Avena sativa enriched in the 124-kilodalton species. J.
nus Nijhoff Publishers, Dordrecht, The Netherlands. Biol. Chem. 258:11025-11031.
27.Kolukisaoglu, H.U., S. Marx, C. Wiegmann, S. 42.McDowell, M.T. and J.C. Lagarias. Purification and
Hanelt, and H.A.W. Schneider-Poetsch. 1995. Diver- properties of phytochromobilin synthase from etiolat-
gence of the phytochrome gene family predates ed oat seedlings. Plant Physiol. (In press).
angiosperm evolution and suggests that Selaginella and 43.Murphy, J.T. and J.C. Lagarias. 1997. The phyto-
Equisetum arose prior to Psilotum. J. Mol. Evol. fluors: a new class of fluorescent protein probes. Curr.
41:329-337. Biol. 7:870-876.
28.Kunkel, T., V. Speth, C. Buche, and E. Schafer. 44.Murphy, J.T. and J.C. Lagarias. 1997. Purification and
1995. In vivo characterization of phytochrome-phyco- characterization of recombinant affinity peptide-tagged
cyanobilin adducts in yeast. J. Biol. Chem. 270:20193- oat phytochrome A. Photochem. Photobiol. 65:750-
20200. 758.

308
Analysis and Reconstitution of Phytochromes

45.Pratt, L.H., M.M. Cordonnier-Pratt, P.M. Kelmen- 58.Song, P.S., M.H. Park, and M. Furuya. 1997. Chro-
son, G.I. Lazarova, T. Kubota, and R.M. Alba. 1997. mophore: apoprotein interactions in phytochrome A.
The phytochrome gene family in tomato (Solanum Plant Cell Environ. 20:707-712.
lycopersicum L). Plant Cell Environ. 20:672-677. 59.Taylor, B.L. and I.B. Zhulin. 1999. PAS domains:
46.Pratt, L.H., J.E. Wampler, and E.S. Rich, Jr. 1984. internal sensors of oxygen, redox potential, and light.
An automated dual-wavelength spectrophotometer Microbiol. Mol. Biol. Rev. 63:479-506.
optimized for phytochrome assay. Anal. Instrum. 60.Terry, M.J. and J.C. Lagarias. 1991. Holophyto-
13:269-287. chrome assembly—coupled assay for phytochromobi-
47.Quail, P.H., M.T. Boylan, B.M. Parks, T.W. Short, Y. lin synthase in organello. J. Biol. Chem. 266:22215-
Xu, and D. Wagner. 1995. Phytochromes: photosen- 22221.
sory perception and signal transduction. Science 61.Terry, M.J., M.D. Maines, and J.C. Lagarias.
268:675-680. 1993. Inactivation of phytochrome-chromophore
48.Raps, S. 1990. Differentiation between phycobilipro- and phycobiliprotein-chromophore precursors by rat
tein and colorless linker polypeptides by fluorescence liver biliverdin reductase. J. Biol. Chem. 268:26099-
in the presence of ZnSO4. Plant Physiol. 92:358-362. 26106.
49.Remberg, A., P. Schmidt, S.E. Braslavsky, W. Gartner, 62.Tomizawa, K., J. Stockhaus, N.H. Chua, and M.
and K. Schaffner. 1999. Differential effects of muta- Furuya. 1995. Spectrophotometric and molecular
tions in the chromophore pocket of recombinant phy- properties of mutated rice phytochrome A. Plant Cell
tochrome on chromoprotein assembly and Pr-to-Pfr Physiol. 36:511-516.
photoconversion. Eur. J. Biochem. 266:201-208. 63.Vierstra, R.D. 1993. Illuminating phytochrome func-
50.Rudiger, W., T. Brandlmeier, I. Blos, A. Gossauer, and tions. Plant Physiol. 103:679-684.
J.P. Weller. 1980. Isolation of the phytochrome chro- 64.Wada, M., T. Kanegae, K. Nozue, and S. Fukuda.
mophore. The cleavage reaction with hydrogen bro- 1997. Cryptogam phytochromes. Plant Cell Environ.
mide. Z. Naturforsch. 35:763-769. 20:685-690.
51.Rudiger, W. and F. Lopez-Figueroa. 1992. Photore- 65.Wahleithner, J.A., L. Li, and J.C. Lagarias. 1991.
ceptors in algae. Photochem. Photobiol. 55:949-954. Expression and assembly of spectrally active recombi-
52.Rudiger, W., F. Thummler, E. Cmiel, and S. Schnei- nant holophytochrome. Proc. Natl. Acad. Sci. USA
der. 1983. Chromophore structure of the physiologi- 88:10387-10391.
cally active form (Pfr) of phytochrome. Proc. Natl. 66.Wilde, A., Y. Churin, H. Schubert, and T. Borner.
Acad. Sci. USA 80:6244-6248. 1997. Disruption of a Synechocystis sp. PCC 6803 gene
53.Sage, L.C. 1992. Pigment of the Imagination: A History with partial similarity to phytochrome genes alters
of Phytochrome Research. Academic Press, San Diego. growth under changing light qualities. FEBS Lett.
54.Schiff, J.A. 1972. A green safelight for the study of 406:89-92.
chloroplast development and other photomorpho- 67.Wu, S.H. and J.C. Lagarias. 1996. The methy-
genetic phenomena. Methods Enzymol. 24B:321-322. lotrophic yeast synthesizes a functionally active
55.Schmidt, P., U.H. Westphal, K. Worm, S. Braslavsky, chromophore precursor of the plant photoreceptor
W. Gartner, and K. Schaffner. 1996. Chromophore- phytochrome. Proc. Natl. Acad. Sci. USA 93:8989-
protein interaction controls the complexity of the phy- 8994.
tochrome photocycle. J. Photochem. Photobiol. B. 68.Wu, S.H., M.T. McDowell, and J.C. Lagarias. 1997.
Biol. 34:73-77. Phycocyanobilin is the natural chromophore precursor
56.Schneider-Poetsch, H.A.W., B. Braun, S. Marx, and of phytochrome from the green alga Mesotaenium cal-
A. Schaumburg. 1991. Phytochromes and bacterial dariorum. J. Biol. Chem. 272:25700-25705.
sensor proteins are related by structural and functional 69.Yeh, K.C. and J.C. Lagarias. 1998. Eukaryotic phy-
homologies—hypothesis on phytochrome-mediated tochromes: light-regulated serine/threonine protein
signal-transduction. FEBS Lett. 281:245-249. kinases with histidine kinase ancestry. Proc. Natl. Acad.
57.Schneider-Poetsch, H.A.W., S. Marx, H.U. Koluk- Sci. USA 95:13976-13981.
isaoglu, S. Hanelt, and B. Braun. 1994. Phytochrome 70.Yeh, K.C., S.H. Wu, J.T. Murphy, and J.C. Lagarias.
evolution—phytochrome genes in ferns and mosses. 1997. A cyanobacterial phytochrome two-component
Physiol. Plant. 91:241-250. light sensory system. Science 277:1505-1508.

309
Analysis and Reconstitution of
14 Phycobiliproteins: Methods for
the Characterization of Bilin
Attachment Reactions

Wendy M. Schluchter1 and Donald A. Bryant2


1Department of Biological Sciences, University of New Orleans,

New Orleans, LA, and 2Department of Biochemistry and Molecular


Biology, The Pennsylvania State University, University Park, PA, USA

1. INTRODUCTION phycobiliproteins have a different composi-


tion and structural organization and will
Phycobiliproteins are a homologous not be discussed further in this chapter (for
family of light-harvesting accessory pro- reviews on cryptomonad phycobilipro-
teins present in cyanobacteria (25,51), red teins, see References 36, 52, 53, and 73).
algae (25), cryptomonads (36,52), and There are three major types of phyco-
some species of prochlorophytes (41,48). biliproteins, each having unique spectro-
The blue, violet, red, or yellow colors of scopic properties: (i) phycoerythrins (PEs;
the phycobiliproteins are due to linear λmax approximately 565 nm); (ii) phyco-
tetrapyrrole chromophores called bilins cyanins (PCs; λmax approximately 620
that are covalently attached at cysteine nm); and (iii) allophycocyanins (APs; λmax
residues (25). These water-soluble proteins approximately 650 nm) (27). These three
are composed of α and β subunits. The αβ proteins differ in both the numbers and
monomers form (αβ)3 trimers which fur- the types of bilins that are associated with
ther stack into (αβ)6 hexamers. These disc- each αβ monomer. Cyanobacterial phyco-
shaped trimers and hexamers can be stabi- biliproteins are formed by the interaction
lized or organized into larger structures by of the apoprotein subunits with one or
linker proteins. Through the association of more of four different types of isomeric
several types of phycobiliproteins with bilins: phycourobilin (PUB), phycoery-
these linker proteins [69), the large light- throbilin (PEB), phycobiliviolin (PXB),
harvesting complex called the phycobili- and phycocyanobilin (PCB) (see Figure 1
some is formed (51,63). Cryptomonad and Reference 27). Although cyanobacteria

Heme, Chlorophyll, and Bilins: Methods and Protocols


Edited by A.G. Smith and M. Witty
©2002 Humana Press, Totowa, NJ

311
W.M. Schluchter and D.A. Bryant

have been shown to contain proteins simi- biliproteins (26,35,63,65). Recently, the X-
lar to eukaryotic phytochrome (44,47, ray structure of the AP trimers carrying the
64,77,78), definitive evidence for the core linker polypeptide was solved (65).
occurrence of phytochromobilin (PφB) in This structure shows that this AP linker
cyanobacteria has not yet been obtained. (and probably the related rod linker that
The phycobilisome is composed of an interacts with phycocyanin) modifies the
AP core that is surrounded by rods con- spectroscopic properties of the phyco-
taining PC that radiate outwards from this biliprotein with which it is associated by
core. In some organisms, PE is also a com- causing slight shifts in bilin conformation
ponent of these peripheral rods and is as well as by bringing two bilins closer
found distal to the PC (28). In other together within the trimer. The linker pro-
cyanobacteria, phycoerythrocyanin (PEC) tein is located between two of the three
is present in small to moderate amounts β-AP subunits in the trimer and directly
under low light-intensity conditions and is interacts with the PCBs of these two sub-
likewise found distal to phycocyanin in the units (65). Approximately half of the sur-
peripheral rods (9,12). face of the linker protein is located within
The α and β subunits that compose the cavity of the trimer (65).
each phycobiliprotein share amino acid In some strains of marine cyanobacteria,
sequence similarity to each other and to three different bilins may occur on their
the subunits of other phycobiliproteins, phycobiliproteins (55), whereas in other
and this observation supports the hypothe- strains, such as Synechococcus sp. PCC
sis that this family of proteins evolved 7002 and Synechocystis sp. PCC 6803, only
through gene duplication (63). Indeed, it is PCB is present. Even in those strains which
the β subunit of phycoerythrin that is only contain PCB, two different stereoiso-
thought to be the ancestral phycobilipro- mers occur on the C-phycocyanin β sub-
tein from which all others evolved (36, 71). unit at the C3′ of the bilin: the R configu-
The three-dimensional structures of at least ration is found for the PCB attached at
one member of each of the major spectro- cysteine β-82 and the S configuration is
scopic classes of phycobiliproteins have found for the PCB attached at cysteine
been determined (8,13,18,19,23,24, β-153 (62). The biochemical basis for how
59–62,65), and these structures show that the biosynthesis of the phycobiliproteins is
the amino acid similarity translates into controlled, such that the correct bilin is
remarkable structural conservation (5). attached to the proper cysteine residue
The subunit structure for this family of with the appropriate stereochemistry, is a
proteins resembles that of members of the fascinating but incompletely understood
globin family with a predominance of α process. Since autocatalytic reactions with
helices and the complete absence of apophycobiliproteins and free bilins have
β-pleated sheets (61). The unique spectro- yielded nonnatural products (2,4,20), all
scopic properties of each phycobiliprotein evidence currently indicates that bilins are
are believed to be due to the type(s) of enzymatically attached to the appropriate
bilin(s) attached, to the immediate electri- apophycobiliprotein.
cal charge and polarity of the environment Phycobiliproteins have been studied for
of the chromophore, and to the way by more than a century and a half now, and
which the phycobiliprotein subunits hold they have captured the imaginations of
the bilins in a stretched conformation many scientists because of their brilliant
(26,35,63). Linker proteins also affect the colors. These proteins are relatively easy to
spectroscopic properties of the phyco- isolate and purify because they comprise
312
Analysis and Reconstitution of Phycobiliproteins

such a large proportion of the total protein tion, and quantitation of phycobiliproteins
in many cells. Much is known about their utilizing reverse-phase high-performance
structure and function, but much less is liquid chromatography (HPLC) was devel-
known about the biosynthesis of the indi- oped (66). This method has been exten-
vidual proteins and the assembly of the sively used in the characterization of phy-
macromolecular phycobilisome. Most cobiliproteins from newly discovered
approaches toward understanding how organisms (29) and from mutants defective
these proteins are synthesized have been in phycobiliprotein biosynthesis (42,68).
made in the attempts to reconstitute them. The methods necessary for the reconstitu-
Most of these reconstitution studies have tion of phycobiliproteins are summarized
taken place in the last 10 years when below.
recombinant DNA technology has allowed
one to overproduce the apoproteins for Nomenclature
such studies and to generate mutants. The
majority of work done on phycobiliprotein The nomenclature for phycobiliproteins
reconstitution has been performed using can be somewhat confusing and reflects, in
cyanobacterial proteins. Therefore, this part, historical developments in the study
chapter will summarize some of the many of phycobiliproteins. PCs and PEs were all
methods that have been developed for ana- originally given the prefix C- or R- to des-
lyzing and reconstituting phycobiliproteins ignate whether they were purified from
from these organisms. However, it is hoped cyanophytes (cyanobacteria) or rho-
that this information will serve as a good dophytes (red algae). The designation B-
starting point for researchers who are inter- was later introduced for a distinct type of
ested in studying the reconstitution and PE from the red alga Smithora naiadum,
biosynthesis of phycobiliproteins from red which is a member of the order Bangiales
algae, cyanobacteria, or cryptomonads. (1,37). The three major types of Class I PE
Also, since the last review on phycobilipro- (those that contain five bilins per αβ
tein purification was written (27), a new monomer; see below) differ in their
method for the separation, characteriza- absorbance properties due to the types of

Figure 1. Structures of the four singly-linked peptide-linked bilins present in the phycobiliproteins of cyanobacteria. The num-
bering scheme for the carbon atoms is indicated.

313
W.M. Schluchter and D.A. Bryant

bilins present on their αβ subunits. These entum, contains PEB at cysteine β-155 and
proteins exhibit one, two, or three distinct PCB at the other two positions (33). R-PC
peaks in the visible region of the spectrum II, isolated from several unicellular marine
and are called C-phycoerythrin (C-PE; cyanobacterial strains, contains PEB at cys-
containing only PEB), B-phycoerythrin teines α-84 and β-155 and PCB at cysteine
(B-PE), and R-phycoerythrin (R-PE), β-82 (55,56). R-PC-III was isolated from
respectively, regardless of the group from Synechococcus sp. WH7805 and has a
which they have been isolated (References PCB:PEB ratio of 2:1, but differences in
33, 37, and references therein). These des- the absorption properties of this PC sug-
ignations seemed sufficient until marine gest that the chromophores are distributed
unicellular cyanobacteria were shown to differently than in R-PC-I (57). The fourth
contain two forms of PE, PE I, and PE II, form of PC, R-PC-IV, was isolated from
in the rods of their phycobilisomes (55, Synechococcus sp. WH8501 and was found
67). PE I was less abundant than PE II. PE to contain PUB attached at α-84 and PCB
II has an extra bilin (PUB) on the α sub- at the other two positions on the β subunit
unit (α-75) and contains both PEB and (67). Finally, PEC is structurally more sim-
PUB. Thus far, only two Synechococcus ilar to PCs than to PEs, but is found distal
strains (WH8020 and WH8103) have to PC in the phycobilisome rods of some
been shown to contain a PE with six chro- cyanobacterial strains. PEC carries PXB at
mophores per αβ monomer (55). There- α-84 and PCB at both positions on the β
fore, these two PEs are members of a new subunit (9,12). Spectroscopic variants of
class of PE, dubbed Class II PE. However, AP (which contains one PCB on each sub-
PE I is more like other PEs that have been unit) have not yet been identified. Thus,
characterized, in that it contains only five the nomenclature for biliproteins devised
bilins per αβ monomer and contains either previously has been rather haphazard and
only PEB or both PEB and PUB (55). confusing. A new form of nomenclature
Thus far, no red algal PE has been shown has been suggested (51), but has not been
to be a member of Class II PE. B-PE, R- widely used thus far.
PE, and PE II complexes carry bilins on
their associated linker protein, called γ (34,
45). A recently discovered red algal PE 2. HOLO-PHYCOBILIPROTEINS
(from Audouinella macrospora) contains a
PE with PCB, PEB, and PUB chro- 2.1. Phycobiliprotein Purification
mophores and is more like B- and R-PEs in
that it contains 5 bilins per αβ monomer Phycobiliproteins may exist in different
and has bilins present on its γ subunit (29). aggregation states depending upon the
This PE is a Class I member, but is not by individual type of biliprotein, the organism
definition an R-PE, which have been from which it was isolated, the composi-
shown to contain PUB and PEB that con- tion of the solution containing it (pH,
tribute to the three absorption peaks in the ionic strength), and such factors as temper-
visible region. ature and protein concentration. The
Four major types of PC have been char- purification of individual phycobiliproteins
acterized (15,63), and all PC types contain has been summarized previously (27). A
PCB as the terminal acceptor bilin at cys- few minor improvements have been intro-
teine β-82. C-PC contains PCB at all three duced using fast protein, peptide, and
cysteines (25,46,60). R-PC-I, present in polynucleotide liquid chromatography
some red algae including Porphyridium cru- (FPLC) (Mono Q; Amersham Pharmacia
314
Analysis and Reconstitution of Phycobiliproteins

Biotech, Piscataway, NJ, USA) (66), but tributions of each bilin type at various
for the most part, the conventional chro- wavelengths must be considered. The con-
matographic methods are still widely used tributions of the various chromophores at
today. Therefore, this chapter will primari- different wavelengths are listed in Table 1.
ly summarize the methods for separation In some cases, the concentration of the
and purification of individual α and β sub- phycobiliprotein sample may be limiting;
units. for example, this is often the case when
isolating phycoerythrins from field sam-
2.2. Storage and Recovery of Purified ples of red algae. The spectra for PEs in
Phycobiliproteins 20% acetic acid (vol/vol) have been deter-
mined to be identical to those for the
Phycobiliproteins are very stable when same protein dissolved in 8 M urea, pH
stored in phosphate buffer at pH 7.0 in the 3.0 (37). This was also found to be true
presence of a reducing agent [1–5 mM for AP and PC (A.N. Glazer, personal
β-mercaptoethanol or dithiothreitol communication).
(DTT)] and sodium azide (1 mM) in the
dark at 4°C. For long-term storage, ammo- 2.4. Purification of Individual Subunits
nium sulfate may be added to 65% satura- by Conventional Chromatographic
tion at 4°C. When sealed at 4°C in the Methods
dark, such slurries–precipitates can be
stored indefinitely. The phycobiliproteins The first method for the separation and
can be recovered by centrifugation at purification of phycobiliprotein subunits
27 000× g for 15 minutes or by centrifuga- was developed by Glazer and Fang and was
tion in a microcentrifuge at 13 000× g for based upon methods used for the separa-
15 minutes. The phycobiliprotein pellet tion of the subunits of hemoglobin
should be resuspended in 5 mM phosphate (30,31). All methods described thus far are
buffer, pH 7.0, 1 mM β-mercaptoethanol, performed under denaturing and acidic
and dialyzed against the same buffer at 4°C conditions which limit oxidation and other
prior to use. side reactions that can modify the bilins.
Each procedure described will include the
2.3. Concentration Determination cyanobacterial source for the phycobilipro-
tein. Most of these conditions have been
Because the absorption properties of the shown to work successfully for the separa-
phycobiliproteins are highly dependent on tion of phycobiliproteins from a wide vari-
the aggregation state, pH, ionic strength, ety of sources, but some optimization of
and protein concentration (26), the most the method may be required if the user is
reliable method to determine the concen- attempting to adapt the method to the
tration of phycobiliprotein solutions is to purification of subunits from a different
measure the absorption spectrum of the phycobiliprotein (see Procedure 1). Each
peptide-bound bilins by dissolving an subunit, when renatured without its part-
aliquot of the protein in 8 M urea, pH 1.9, ner, is much less stable and tends to aggre-
or in 10 mM TFA (trifluoroacetic acid); its gate over time when in solution. Proce-
concentration can then be determined by dures 2 and 3 describe renaturation of
using the extinction coefficients given in phycocyanin subunits. Both renaturation
Table 1 (Reference 27 and references there- procedures, followed by the last diethyl-
in). For phycobiliproteins that contain 2 or aminoethyl (DEAE) chromatography step,
more different bilins per subunit, the con- have a recovery rate for renatured protein
315
W.M. Schluchter and D.A. Bryant

of approximately 25% (32). In contrast, The PC subunits should adsorb to the


when the α and β subunits are renatured top of the column.
together in a 1:1 molar ratio, the yield of 4. Wash the column extensively with 2 M
reconstituted phycocyanin is between urea, 10 mM β-mercaptoethanol, pH
40%–60% (31). The absorption spectra of 3.0.
the renatured α and β subunits purified 5. Elute PC subunit by a stepwise increase
from the PC of Synechococcus sp. PCC in urea concentration (approximately
6301 are shown in Figure 2. one column volume each of 4.0 M, 6.0
M, 8.0 M, and 9.0 M urea, pH 3.0,
❖ Procedure 1. Separation of PC and 10 mM β-mercaptoethanol). The
Subunits α subunit typically elutes with 8.0 M
urea, while elution of the β subunit
1. Prepare 25 to 50 mg of Anabaena sp. requires 9.0 M urea.
PCC 6411 PC in 100 mM Na-phos-
6. For long-term storage, subunits can be
phate buffer, pH 7.0 2. See section 2.1.
dialyzed extensively against water,
2. Adjust pH to 3.0 by addition of glacial lyophilized, and subsequently rena-
acetic acid with reductant present (10 tured using either Procedure 2 or 3
mM β-mercaptoethanol). below.
3. Apply this mixture to a column of Bio-
Rex70 resin (weak cation exchange, ❖ Procedure 2. Renaturing Fresh
minus 400 mesh, 2.2 × 13 cm; Bio- Phycocyanin Subunits (31)
Rad Laboratories, Hercules, CA, USA)
that has previously been equilibrated 1. Dilute PC subunits from procedure 1
with 0.4% acetic acid, pH 3.0 (31). to 0.1 to 0.4 mg/mL protein with 8 M

Figure 2 Absorption spectra of the


individual renatured α and β sub-
units of phycocyanin purified
from Synechococcus sp. PCC 6301.
Spectra were determined in 50 mM
Na phosphate, 1 mM β-mercap-
toethanol, pH 7.0, and at α protein
concentration of 1.05 × 10-5 M for
the α-subunit and 1.11 × 10-5 M
for the β subunit. The λmax was
620 nm for the α subunit and 608
nm for the β subunit. This figure
was modified with permission from
Reference 32.

316
Analysis and Reconstitution of Phycobiliproteins

Table 1. Millimolar Extinction Coefficients of β-mercaptoethanol, pH 7.0. A large pro-


Peptide-Linked Bilinsa portion of blue nonfluorescent material
is typically retained at the top of this
Bilin ε495 ε495 ε495 ε495 column. This extra step ensures that
properly folded subunits are recovered.
PUB 94.0 0 0 0 Sedimentation analyses of α and β sub-
PEB 18.3 53.7 8.5 0 units at neutral pH indicate that each puri-
fied subunit has a tendency to dimerize at
PXB 6.8 28.4 38.6 0 higher protein concentrations (greater than
PCB 1.45 6.0 16.2 35.4 0.2 mg/mL). Protein concentrations can be
calculated from the absorption at 662.5
nm in 8 M urea, pH 1.9, using the molar
aExtinction coefficients are mM-1cm-1 at the extinction coefficient values of 33.2
wavelength indicated. The absorption mM-1cm-1 for the α subunit and 69.5
spectra of peptide-linked bilins were mM-1cm-1 for the β subunit (32).
measured in 10 mM TFA or 8 M urea, pH The first separation method for AP sub-
1.9. These values are taken from units was developed by Gysi and Zuber for
References 7, 33, and 44. the protein from the thermophilic
cyanobacterium Mastigocladus laminosus
(40) and is described in Procedure 4.
urea, 5 mM β-mercaptoethanol, pH
8.0. ❖ Procedure 4. Separation of
2. Dialyzed against 3 M urea, 5 mM Allophycocyanin from Mastigocladus
β-mercaptoethanol, 6 mM Na-phos- laminosus
phate, pH 6.7, at 4°C.
3. Dialyze against two changes of 10 mM 1. Prepare 10 mg of allophycocyanin in
Na-phosphate, 5 mM β-mercapto- 20 mM phosphate buffer, 8.0 M urea,
ethanol, pH 6.5, at 4°C. 10 mM β-mercaptoethanol, pH 8.0,
4. Dialyze against 5 mM Na-phosphate, and allowed to incubate for 2.5 hours
pH 7.0, at 4°C. at 37°C. See section 2.1.
2. Apply this mixture to a DEAE
❖ Procedure 3. Renaturing Lyophilized Sephadex A-50 column (2.5 × 45 cm;
Phycocyanin Subunits (32) Amersham Pharmacia Biotech) at
room temperature, equilibrated in the
1. Dissolve lyophilized PC in 5 mM Na-
same buffer.
phosphate, 1 mM β-mercaptoethanol,
3. Elute the AP subunits with a linear gra-
pH 7.0, and allow to stand overnight at
dient (400 mL) of KCl (50 to 300
4°C.
mM) in 20 mM phosphate, 8.0 M
2. Remove insoluble material by centrifu- urea, 10 mM β-mercaptoethanol, pH
gation. 8.0. The β subunit of AP elutes first
3. Loaded the protein solution onto a followed by the α subunit.
DEAE cellulose DE-52 column (0.5 × 4. Fractions containing these purified
5 cm; Whatman, Clifton, NJ, USA) subunits should be pooled and dialyzed
equilibrated in 5 mM Na-phosphate, 1 exhaustively against 20 mM Na-phos-
mM β-mercaptoethanol, pH 7.0. phate buffer, pH 8.0.
4. Subunits can be eluted immediately This procedure was slightly modified for
with 200 mM Na-phosphate, 1 mM the purification of AP subunits from Syne-
317
W.M. Schluchter and D.A. Bryant

chococcus sp. PCC 6301 and Synechocystis The Bio-Rex 70 column (1.5 × 15 cm)
sp.; (22663; ATCC, Manassas, VA, USA) with the PE subunits adsorbed was washed
also called Microcystis aeruginosa (14) (Pro- with 15 mL of 2.0 M urea, 30 mL of 4.0
cedure 5). M urea, and 50 mL of 6.0 M urea before
development with a linear gradient of 6.0
❖ Procedure 5. Separation of to 10.0 M urea, pH 3.0 (20 mL total vol-
Allophycocyanin Subunits from ume). The α subunit eluted first followed
Synechococcus sp. PCC 6301 and by the β subunit. Subunits were renatured
Synechocystis sp. by exhaustive dialysis against 50 mM K-
phosphate buffer at pH 7.0 at room tem-
1. Dissolve 325 mg purified and lyo- perature.
philized AP in 50 mL of 10 mM K- Separation of the subunits of Anabaena
phosphate, 8 M urea, 10 mM β-mer- variabilis PEC was first demonstrated by
captoethanol, pH 8.0, and equilibrate Bryant et al. using a modification of the
for 1 hour at room temperature. See method developed for the separation of PC
section 2.1. subunits described above (12). The Bio-
2. Load the material onto a DEAE Rex 70 column (3.9 × 51 cm) was subject-
Sephadex A-50 column (3.5 × 12 cm) ed to incremental step gradients of acidic
and wash with equilibration buffer. urea as described previously, followed by
3. Use 200 mL of equilibration buffer elution of the α subunit by addition of
plus 50 mM KCl to elute the elute the 7.4 M urea, pH 3.0. Once the elution of the
β subunit of AP. α subunit was complete, elution of the
β subunit was accomplished by addition
4. Residual β subunit is eluted by repeat-
of 9.0 M urea, pH 3.0. Subunits were dia-
ed washes with 150 mL equilibration
lyzed against 50 mM ammonium acetate,
buffer plus 80 mM KCl.
pH 6.8.
5. Use equilibration buffer plus 180 mM
KCl to elute the α subunit of AP.
2.5. Purification of Phycobiliproteins by
6. Pool fractions of each subunit from HPLC
steps 3 and 5 for dialysis against 25
mM ammonium acetate, pH 6.8, and In 1987, HPLC was used to verify the
concentration by ultrafiltration using an purity of PC and AP preparations from M.
Amicon cell with a 10 000 MWCO aeruginosa (58); however, the method also
membrane (Millipore, Bedford, MA, showed that the AP and PC subunits could
USA). be separated on a C18 reverse-phase col-
A method similar to the one developed umn. In 1990, Swanson and Glazer intro-
for the separation of the PC subunits was duced a method for separation of phyco-
successfully used in the separation of the α, biliprotein subunits using C4 reverse-phase
β, and γ subunits of phycoerythrin from P. HPLC (66). These HPLC methods have
cruentum (34). The only significant differ- several advantages over the conventional
ence was in the development of the col- chromatographic methods. They are more
umn. The γ subunit was eluted with 7.4 M rapid and require much less starting mater-
urea, the α subunit with 8.0 M urea, and ial. When used in conjunction with a pho-
the β subunit with 9.0 M urea. Similar todiode array detector, these methods also
conditions were used to separate the sub- give immediate spectroscopic information
units of phycoerythrin II (PE II) from the about bilin content and subunit stoichiom-
cyanobacterium Gloeobacter violaceus (10). etry. The method of Swanson and Glazer
318
Analysis and Reconstitution of Phycobiliproteins

has also successfully been used to separate The method of Swanson and Glazer
phycobiliproteins obtained directly from uses a C4 reverse-phase analytical column
purified phycobilisomes, giving quantita- (250 × 10 mm) and a solvent system con-
tive information regarding phycobilipro- sisting of 0.1% TFA in water (Buffer A)
tein stoichiometry and content in these and a 2:1 acetonitrile: isopropanol mixture
mixtures (39). When both HPLC methods (Buffer B). This purification procedure has
were compared, the method of Swanson been very successful in the separation and
and Glazer gave better resolution of phyco- resolution of diverse types and mixtures of
biliproteins isolated from Arthrospira max- phycobiliproteins. The purified phyco-
ima (38,39). The use of reverse-phase biliprotein or phycobiliprotein mixture,
HPLC is clearly a better choice than con- typically 100 to 1500 µg in 200 to 500 µL
ventional chromatographic procedures for in 5 mM Na-phosphate, pH 7.0, 1 mM β-
determining stoichiometric information mercaptoethanol is combined with an
when the amount of starting material and equal volume of 9.0 M urea, pH 2.0 (fresh-
speed are primary concerns. ly prepared), and subjected to centrifuga-

Figure 3. HPLC separation of cyanobacterial C-PC subunits. Purified PC from Synechococcus sp. PCC 6301 (top panel) or
Anabaena sp. PCC 7120 (bottom panel) was separated on a C4 reverse-phase HPLC column as described in the text. Elution of
subunits was monitored at 660 nm in order to follow the absorbance of peptide-linked PCB. In each case, the α subunit elutes
prior to the β subunit. This figure was modified with permission from Reference 66.

319
W.M. Schluchter and D.A. Bryant

tion in a microcentrifuge for 5 minutes very important to wash the column exten-
prior to injection on the column. A Hi- sively between injections using a linear gra-
Pore RP304 column (Bio-Rad Laborato- dient to 100% Buffer B over 5 minutes fol-
ries) equilibrated in 65% Buffer A and lowed by at least 5 to 10 minutes of
35% Buffer B (1.5 mL/min) has typically washing the column with 100% Buffer B.
been used. After injection of the sample, The β subunits of phycobiliproteins are
proteins are eluted with a linear gradient to sometimes retained on the column, and
30% Buffer A and 70% Buffer B over 35 these will usually be eluted by this treat-
to 40 minutes depending upon the source ment. If careful quantitation of a sample is
of the phycobiliprotein (see Figure 3). required, it is wise to perform a blank
With a few alterations of the elution gradi- injection between each run with samples in
ent profile, this method has been success- order to insure that the column is entirely
fully employed in the separation of a wide free of residual phycobiliprotein subunits.
variety of phycobiliproteins from cyano- Preparative separation of phycobilipro-
bacteria, red algae, and cryptomonads tein subunits can be accomplished using
(17,29,39,66,74). In fact, researchers have this method in conjunction with a semi-
had success in the separation of phyco- preparative C4 reverse-phase column (or by
biliproteins from phycobilisome samples employing multiple runs on an analytical
taken directly from sucrose gradients (after column). Subunits can be collected as they
dialysis against 5 mM Na-phosphate, pH elute from the column, and the solvents
7.0 followed by combination with an equal can be removed by rotary evaporation. The
volume of 9.0 M urea, pH 1.9, prior to aqueous subunits can then be diluted 2:9
injection) (see Figure 4). Toole et al. com- with 9.0 M urea, pH 2.0, 10 mM β-mer-
bined the phycobilisomes taken directly captoethanol, followed by dialysis against
from sucrose gradients (in sucrose–phos- 50 mM Na-phosphate, pH 7.0 (22).
phate) with an equal volume of 8.4 M
guanidine hydrochloride, pH 6.4 (followed 2.6. Methods for Analyzing the Quality
by centrifugation), prior to loading on the of the Renatured Subunits
C4 column (Vydac/The Separations
Group, Hesperia, CA, USA) using the gra- The best method to analyze the quality
dient conditions described above (72). This of renatured subunits is to compare the
method has also been successfully used to absorption spectrum of a dilute solution
characterize the linker polypeptide and containing the subunit with the fluores-
phycobiliprotein stoichiometry in phyco- cence excitation spectrum of the same solu-
bilisomes from A. maxima (38,39). tion. In order to obtain an accurate excita-
Some technical considerations to keep tion spectrum, the absorbance at the
in mind for each separation include the long-wavelength maximum should be less
need to use higher concentrations of urea than 0.05 OD so that reabsorption of
to solubilize phycobiliprotein mixtures that emitted light will be minimized. If the two
may contain any given apophycobilipro- spectra differ significantly, then it is likely
tein. It has been observed that apophyco- that the renatured subunit is not folded
biliprotein subunits often do not bind as properly or that the chromophore(s) may
well as holo-subunits under these condi- have been chemically modified during
tions, but that addition of urea to at least 6 purification and renaturation. If the major-
M final concentration in the solution to be ity of the protein has been oxidized, it is
injected greatly increases the yield of unlikely that the sample will be a good
apophycobiliprotein material (22). It is also source of bilin in bilin transfer assays.
320
Analysis and Reconstitution of Phycobiliproteins

3. APOPHYCOBILIPROTEINS When the α and β subunits of PC are


produced together, a high yield of αβ
In order to understand how phyco- monomer is produced (2,11). After
biliproteins are biosynthesized, one must removal of unbroken cells and large cell
have an effective assay system. One such membrane fragments by centrifugation
system has successfully been developed and (31 000× g for 30 min), apo-αβ-PC can be
shown to be effective for the reconstitution precipitated by addition of ammonium sul-
of the α subunit of phycocyanin as fate to 38% saturation. Following centrifu-
described below. However, the overproduc- gation at 18 000 × g, the pellet should be
tion of various apophycobiliproteins has resuspended in a large volume of 50 mM
been successfully accomplished, and this Na-phosphate buffer, pH 7.0 (approxi-
information is also described below. mately three times the initial volume of the
cell-free supernatant). This mixture should
be immediately loaded on a DEAE cellu-
3.1. Overproduction of lose DE-52 column, and the flow-through
Apophycobiliprotein Subunits should be collected and pooled after rins-
3.1.1. Apophycocyanin ing with two column volumes of the phos-
phate buffer (2). The apo-αβ-PC subunits
The first successful overproduction of can be precipitated with ammonium sul-
apophycobiliproteins was accomplished fate added to 50% saturation. The pellet
with the α and β subunits of phycocyanin from this precipitation should be resus-
(11). The cpcBcpcA genes encoding the β pended in a small volume of phosphate
and α subunits, respectively, from the buffer with 2 mM β-mercaptoethanol.
cyanobacterium Synechococcus sp. PCC This mixture can be desalted and further
7002 were cloned into a vector and purified by loading onto a gel filtration col-
expressed in Escherichia coli using their umn (Sephadex G-100) run at room tem-
native promoter (2,11). Both subunits perature. Fractions should be collected and
were produced at a low level throughout monitored for purity by sodium dodecyl
growth of the E. coli culture. A lower level sulfate polyacrylamide gel electrophoresis
of expression of phycocyanin and allophy- (SDS-PAGE). These subunits should be
cocyanin subunits throughout the growth stored under N2 or degassed in Na-phos-
of the culture typically seems to yield pro- phate buffer at 4°C containing a reducing
teins that are properly folded. When the agent (10 mM β-mercaptoethanol or
T7/pET vector system was used for the DTT) to prevent oxidation of cysteine
expression of the cpcA gene in BL21 DE3 residues.
pLysS cells, a significant proportion of apo-
α-PE was present in inclusion bodies 3.1.2. Apophycoerythrin
(W.M. Schluchter and A.N. Glazer,
unpublished observations). Although the Expression of Calothrix sp. PCC 7601
apo-α-PC subunit could be renatured from cpeAcpeB genes in E. coli resulted in the
these inclusion bodies, the soluble subunit production of large amounts of insoluble
produced in E. coli expression cultures was apo-αβ-PE subunits, which were found in
always a better substrate for in vitro addi- inclusion bodies (20). These proteins could
tion reactions than the product of these be successfully solubilized in acid urea (9
renaturation experiments (W.M. Sch- M urea-HCl, 10 mM DTT, pH 2.5). After
luchter and A.N. Glazer, unpublished dialysis against 3 M urea-HCl, 10 mM β-
results). mercaptoethanol, pH 2.5 at 4°C, insoluble
321
W.M. Schluchter and D.A. Bryant

material was removed by ultracentrifuga- it is much easier to store frozen E. coli cells
tion at 100 000× g for 30 minutes. The containing the overproduced apophyco-
supernatant containing both subunits was biliprotein fusion and purify small batches
applied to a Bio-Gel P100 gel filtration of protein when one needs it. This insures
column (5 × 75 cm; Bio-Rad Laboratories) that the substrate for in vitro addition reac-
with 3 M urea-HCl, 10 mM β-mercap- tions is properly folded and contains fully
toethanol, pH 2.5, as the buffer at room reduced cysteines.
temperature. The β subunit eluted first,
followed by fractions containing both α 3.1.4. Attaching Apophycobiliproteins to
and β subunits, and finally followed by Agarose Beads
fractions containing only the α subunit.
Attempts to renature the β subunit were The covalent attachment of apo-α-PC
unsuccessful. However, the α subunit to agarose beads greatly facilitated reconsti-
could be renatured as long as the protein tution studies because it was possible to
concentration remained below 0.1 mg/mL. perform addition reactions in a small
Dialysis against 50 mM Na-phosphate, 1.0 microcentrifuge tube, to wash away excess
mM DTT, pH 7.0, and 0.1 mM NaN3 bilin after the reaction was terminated if
resulted in renaturation of some apo-α-PE. necessary, and then to measure the fluores-
cence of the sample after this process
3.1.3. Producing Apophycobiliproteins as (21,22). The apoprotein in 50 mM Na-
Fusions phosphate, pH 7.0, 5 mM EDTA was
mixed with Affi-Gel 15 (Bio-Rad Labo-
Several different phycobiliprotein struc- ratories) at 1 mg of protein per mL of
tural genes have been successfully fused beads (22). The covalent attachment of the
with the genes encoding other proteins, protein to the beads continued for 30 min-
and this has allowed the purification pro- utes at 4°C until the reaction was stopped
cedure to be simplified to a single affinity by the addition of 0.05 volumes of 1 M
chromatography step (Y.A. Cai, W.M. glycylglycine, pH 7.0 (incubated for 1
Schluchter, and A.N. Glazer, unpublished hour at 4°C). To remove excess unbound
results). The maltose binding protein has protein, the beads were washed with 50
been employed in such fusions, as well as a mM Na-phosphate, pH 7.0, 5 mM
domain of 24 amino acids containing 6 EDTA, 0.5 M NaCl, followed by 50 mM
contiguous histidine residues that has usu- Na-phosphate, pH 7.0, 5 mM EDTA. The
ally been fused to the N termini of several air was evacuated out of the flask contain-
phycobiliprotein subunits (including α- ing the beads, and the beads were stored at
PC, β-PC, α-AP, and β-AP) from several 4°C in the same buffer with the addition
different cyanobacteria. Following the of 5 mM DTT.
manufacturer’s procedures for purification
of the fusion proteins, high yields of prod- 4. RECONSTITUTION OF
ucts were generally obtained. An important HOLOPHYCOBILIPROTEINS
factor to remember is to add reductant
throughout the purification procedure in 4.1. Nonenzymatic Assays
order to keep the cysteines reduced. It is
also best to purify only as much protein as The first evidence that enzymes might
is needed in the next week. Within 2 weeks be required for bilin addition to phyco-
at 4°C, these proteins tend to oxidize and biliproteins was revealed through the
begin denaturing. As a matter of practice, experiments of Arciero et al. with apo-PC
322
Analysis and Reconstitution of Phycobiliproteins

(2–4). When either PCB or PEB was must take place in vivo in organisms which
added to apo-αβ-PC, covalent addition contain more than one bilin attached to
took place at the α-84 and β-82 sites, but phycobiliproteins.
not at the β-153 site. The primary prod-
ucts of those nonenzymatic additions were 4.1.1. Assay Conditions
bilins at a higher oxidation state, with an
extra double bond between C2-C3 of ring The single most important factor in
A (see Figure 1 for numbering scheme). these assays is that the apoprotein be fully
Mesobiliverdin (MBV) was the product reduced prior to addition of the bilin sub-
when PCB was added, and 15,16 dihydro- strate. This is accomplished by using fresh-
biliverdin was the product when PEB was ly purified apoprotein, adding DTT to 10
added. mM, and incubating this mixture for 30
Nonenzymatic addition reactions have minutes at room temperature (22). The
also been performed with apo-α-PC (20) DTT should be removed by gel filtration
and with apoallophycocyanin subunits prior to addition of bilin. It has been
(W.M. Schluchter and A.N. Glazer, observed that bilins will react with DTT
unpublished results). In all cases, a phyco- when this compound is present at high
biliprotein adduct is formed, and there is concentrations (W.M. Schluchter and
no discrimination between the bilin iso- A.N. Glazer, unpublished results) (20).
mers observed in such in vitro addition Generally, apophycobiliproteins are very
experiments. Such discrimination clearly stable when present in 5 to 50 mM Na-

Figure 4. HPLC separation of


phycobiliproteins present in the
phycobilisomes purified from
Synechocystis sp. PCC 6803. Phy-
cobilisomes from sucrose gradients
were dialyzed extensively against 5
mM Na-phosphate, pH 7.0, prior
to injection on a C4 reverse-phase
column (see text for details). The
elution of polypeptides was moni-
tored at 280 nm (upper panel) and
680 nm (lower panel). The α-AP
subunit is poorly resolved from the
β-PC subunit.
323
W.M. Schluchter and D.A. Bryant

phosphate, pH 7.0, or 10 to 50 mM Tris- 4.2. Enzymatic Assays


HCl, pH 8.0, 75 mM NaCl, and therefore
these conditions have been used in non- The evidence that enzymes were
enzymatic assays. involved in bilin attachment to phyco-
biliproteins came from the characterization
of the products of two genes, cpcE and
❖ Procedure 6. Nonenzymatic Assay of cpcF, that occur downstream of cpcBA, the
Adduct Formation structural genes for the β and α subunits of
phycocyanin, in Synechococcus sp. PCC
1. The bilin, after dissolution in dimethyl
7002 (68,80). Insertional inactivation of
sulfoxide (DMSO), is added to a final
either gene affected only PCB addition to
concentration of 10 to 50 µM to the
the α subunit of PC. Fairchild et al. later
reduced apoprotein that is present at a
showed that these two proteins acted
similar concentration (10–50 µM).
together as a heterodimeric PC α subunit
Generally, the majority of the bilin
PCB lyase (22). Other cpcE and/or cpcF
combines with the apoprotein within 1
mutants have been characterized in Syne-
hour (2).
chococcus sp. PCC 7942 (6), Anabaena sp.
2. The reaction should be protected from PCC 7120 (W.M. Schluchter and A.N.
the light at room temperature, but Glazer, unpublished results), and in
reaction mixtures can be purged with Calothrix sp. PCC 7601 (70). In all of
N2 and left overnight at room temper- these cases, the mutants produce signifi-
ature in the dark. cantly reduced amounts of PC. Jung et al.
3. At the end of the incubation period, (42) showed that a mutation in one or
the phycobiliprotein should be separat- both of the pecE and pecF genes of Anabae-
ed from unreacted bilin, and this can na sp. PCC 7120, whose products show a
be accomplished by one of several high degree of sequence similarity with
methods. CpcE and CpcF, affected the level of the
4. In instances in which nonaffinity PEC holo-α-subunit. The PEC α subunit
tagged apoprotein is used, the reac- that could be purified from a pecEF mutant
tion mixture should be loaded onto a was found to contain a PCB adduct instead
small Sephadex G-25 column equili- of the PXB (see Figure 1) chromophore
brated with the same buffer used in that is normally present in wild-type cells.
the reaction. Bilins will bind to the These results suggest that PecE and PecF
surface of the resin [and require 10% form a heterodimeric PEC α subunit PXB
(vol/vol) acetic acid to be released; lyase, and that in the absence of PecE and
the resin can usually be regenerated PecF, another lyase, possibly CpcE and
by standard procedures for reuse], CpcF, recognizes this site and adds PCB to
while the phycobiliprotein will elute the α-PEC subunit (42). Very recently,
immediately (2). Zhao et al. have shown that PecE and PecF
5. If the phycobiliprotein has been affini- from M. laminosus act together to attach
ty-tagged, one can proceed directly and isomerize PCB to PXB to the α sub-
with the procedure for purification rec- unit of PEC (79). This reaction required
ommended by the manufacturer. the presense of both subunits, because
6. If the phycobiliprotein is covalently when one or both PecE and PecF were
attached to agarose beads, the beads absent, the only product was MBV-α-
can be washed exhaustively to remove PEC. In the cyanobacterium Fremyella
any trace bilins from the protein. diplosiphon, a mutation in cpeY, the prod-
324
Analysis and Reconstitution of Phycobiliproteins

uct of which shows limited sequence simi- 8.0. Washing entails full resuspension,
larity to the family of putative lyases preferably using a tissue homogenizer,
including CpcE and which is located followed by centrifugation at 8000× g;
downstream of the structural genes encod- the inclusion bodies containing
ing PE, produced markedly lower levels of CpcE/CpcF will be in the pellet frac-
PE. These observations suggest that CpeY tion.
is a lyase subunit as well (43). 4. The inclusion body proteins are solubi-
The activities of only a few lyases have lized with 9.0 M urea-HCl, pH 1.9, 1
been tested in vitro, and to date, the most mM DTT. The concentrations of each
extensive analyses have been performed protein should be determined spec-
using Synechococcus sp. PCC 7002 CpcE trophotometrically using the ε280 nm
and CpcF. So far, no lyase that can specifi- for each protein (calculated from the
cally attach bilins to the β subunit of any Trp [ε = 5540 M-1cm-1] and Tyr [ε =
phycobiliprotein has been identified. 1480 M-1cm-1] content of the pro-
Methods for assaying these enzymes will be teins) (54).
summarized here in the hopes that this will 5. This estimate should be compared with
encourage additional research in this area. the staining intensities of diluted
aliquots of each urea-solubilized pro-
4.2.1. CpcE CpcF Expression tein on SDS-PAGE. The ε280 nm for
Synechococcus sp. PCC 7002 CpcE and
Recombinant CpcE and CpcF were pro-
CpcF under denaturing conditions are
duced in both soluble form and in the
35 640 M-1cm-1 and 20 220 M-1cm-1,
form of inclusion bodies in E. coli. Howev-
respectively (22).
er, Fairchild et al. showed that corenatura-
tion of these two proteins in a 1:1 ratio led 6. These proteins should be mixed in a
to the most activity (22). 1:1 molar ratio at a concentration of
0.15 to 0.3 mg/mL prior to renatura-
❖ Procedure 7. Purification of tion.
Recombinant CpcE and CpcF Several methods have been used success-
fully to renature these proteins. A concen-
1. The cpcE and cpcF genes overexpressed trated mixture can be diluted approximate-
using a T7/pET vector system and the ly 1:10 with 50 mM Tris-HCl, 75 mM
majority of the recombinant proteins NaCl, pH 8.0; the dilution is followed by
are found in inclusion bodies. extensive dialysis against the same buffer at
2. The inclusion bodies are collected by 4°C. This procedure yielded renatured het-
low-speed centrifugation after the cells erodimeric CpcECpcF, but direct dialysis
have been lysed by passage through a of the diluted proteins in 9.0 M urea
French pressure cell. The inclusion against the same Tris-NaCl buffer pro-
bodies appear as a chalky white pellet duced similar results. In both cases, the
and are easily differentiated from yield of renatured CpcECpcF was approxi-
unbroken cells which usually appear mately 50%. The extinction coefficients
more tan or brownish in color. for native CpcE and CpcF were calculated
3. The inclusion bodies should be washed (from the Trp and Tyr content of each pro-
extensively using the following solu- tein) to be 38 440 M-1cm-1 and 21 060
tions: 50 mM Tris-HCl, 5 mM EDTA, M-1cm-1, respectively. After filter steriliza-
pH 8.0; 50 mM Tris-HCl, pH 8.0, 1% tion through a 0.2 µm membrane to pre-
Triton X-100; 50 mM Tris-HCl, pH vent microbial growth, these proteins were

325
W.M. Schluchter and D.A. Bryant

stable for weeks at 4°C. Although other elsewhere in this volume (see Chapter 8)
purification procedures have been utilized and in References 2 and 22. There is
for preparations of proteins for more rigor- presently no reported method for the
ous kinetic analyses (21), the procedure purification of the precursor of peptide-
described above yielded a preparation of linked PUB or of the doubly-linked forms
enzyme with high activity. of PEB and PUB. However, it has been
shown that CpcECpcF from Synechococcus
4.2.2. Bilin Donors sp. PCC 7002 (13) and Anabaena sp. PCC
7120 (C.F. Chan, W.M. Schluchter, and
PEB and PCB can be cleaved from holo- A.N. Glazer, unpublished results) will
phycobiliproteins and purified as described transfer the bilin from a holo-α-PC sub-

Figure 5. Bilin addition assays with Anabaena sp. PCC 7120 apo-α-PC resin. Assay conditions were as follows. Approximately
300 µL of settled resin (containing Anabaena sp. PCC 7120 apo-α-PC subunit covalently attached as described in Reference
22 was in a 1.5-mL microcentrifuge tube containing 0.8 mL of reaction assay buffer (50 mM Tris-HCl, pH 8.0, 75 mM NaCl,
1 mM MgCl2, 1 mM Na pyrophosphate, 1 mM thioglycollate). The enzyme to be tested was Anabaena sp. PCC 7120
CpcECpcF (overproduced and purified as described in this chapter; W.M. Schluchter, C. Chan, and A.N. Glazer, manuscript in
preparation). In assays where the enzyme was added (+CpcEF), Anabaena sp. PCC 7120 CpcECpcF were present at 0.25 µM. In
control assays, the same volume of reaction assay buffer was added in place of CpcECpcF (-CpcEF). The reaction was initiated by
the addition of the bilin donor. After incubation at 37°C in the dark for 1 hour, the resin was washed extensively as described in
the text to remove any remaining donor bilin. The fluorescence emission of the resin present in each assay was measured at 640
nm because this is the peak of fluorescence emission for the native holo-α-PC. The donor bilin was purified PCB (11.6 µM;
labeled as PCB), purified holophycocyanin from Anabaena sp. PCC 7120 (0.92 µM; labeled as 7120 PC), or purified holophy-
cocyanin from Synechococcus sp. PCC 7002 (1.0 µM; labeled as 7002 PC). The Anabaena sp. PCC 7120 CpcECpcF lyase cat-
alyzed the addition of free PCB to Anabaena sp. PCC 7120 apo-α-PC. However, this enzyme also catalyzed the reverse reaction
by transferring bilin from the α-PC subunit (purified either from Anabaena sp. PCC 7120 or from Synechococcus sp. PCC 7002;
W.M. Schluchter, C. Chan, and A.N. Glazer, unpublished results).

326
Analysis and Reconstitution of Phycobiliproteins

unit to an apo-α-PC subunit. It is concentrations between 0.8 to 2 mM


unknown whether all lyases have this trans- and added to the reaction to a final
fer activity. However, it is possible that concentration of 10 to 20 µM. If the
many of these enzymes also serve as repair source of the bilin is to be a holophy-
enzymes or as part of the phycobiliprotein cobiliprotein, then this protein should
degradation pathway under nutrient star- be added to a final concentration of 1
vation conditions (16). to 10 µM.
5. The reaction should be incubated in
4.2.3. Enzyme Assay Conditions the dark at 37°C for 1 hour.

The first assays performed to test the 4.3. Methods for Analysis: Detection of
activity of Synechococcus sp. PCC 7002 Covalent Products
CpcECpcF used apo-α-PC bound to resin
as the substrate (22). This greatly facilitated Enzymatic bilin addition reactions
the removal of unreacted bilins or the holo- should always be compared with control
α-PC substrate and the enzyme prior to nonenzymatic reactions using one or more
the measurement of the incorporation of of the following methods. If holophyco-
PCB onto the α-PC resin. However, affin- biliproteins were the source of bilin for the
ity-tagged apophycobiliproteins have been addition reaction, care must be taken to
successfully used as substrates in these same insure that all residual holophycobilipro-
reactions (W.M. Schluchter and A.N. tein has been removed. This is most easily
Glazer, unpublished results). accomplished when the apophycobilipro-
tein is attached to a solid support. The
❖ Procedure 8. Enzymatic Assay of beads are washed extensively with 9.0 M
Adduct Formation urea-HCl, pH 2.5, followed by 50 mM
Na-phosphate, pH 7.0. When the apo-sub-
1. The fully prereduced apophycobilipro- unit has been affinity tagged, it is very like-
tein is added to a microcentrifuge tube. ly that any holo-phycobiliprotein added as
If the subunit is affinity tagged, a source of bilin can dimerize with either
approximately 0.3 to 0.6 mg is used. affinity-tagged apo-subunits or affinity-
However, if the subunit is attached to a tagged enzyme-mediated bilin adducts and
solid support, an aliquot corresponding will copurify with the affinity-tagged sub-
to approximately 300 to 500 µL of set- unit. Therefore, purification of the affini-
tled beads is added. ty-tagged protein must be performed
2. The buffer conditions (as determined according the manufacturer’s procedure
for optimal activity of the Synechococ- under denaturing conditions whenever
cus sp. PCC 7002 CpcECpcF) are 50 possible. If this is not possible, then anoth-
mM Tris-HCl, pH 8.0, 75 mM NaCl, er method for the detection and separation
1 mM MgCl2, 1 mM disodium pyro- of these two subunits should be used (see
phosphate, and 1 mM thioglycolate. HPLC separation below).
3. The enzyme to be tested should be
added to a final concentration of 0.1 to 4.3.1. Absorbance
0.4 µM. Absorbance is the easiest and most
4. The reaction is usually initiated by the straightforward method to detect an addi-
addition of the bilin substrate. Free tion product. Unfortunately, this is the
bilin should be dissolved in DMSO at method that gives one the least amount of
327
W.M. Schluchter and D.A. Bryant

information about the product. Although absorbance and fluorescence are red-shift-
it is a good starting point, this method ed relative to the PCB product. The MBV
should never be used as the only indicator adduct, with a fluorescence emission maxi-
of which product(s) is present. When PCB mum at 668 nm, is much less fluorescent
is added to apo-α-PC in the absence of than the PCB adduct, which has a fluores-
CpcECpcF, the unnatural MBV adduct cence emission maximum at 643 nm
predominates and can be easily distin- (3,22). Additionally, the extinction coeffi-
guished from the PCB product. The cients for the long wavelength absorbing
absorbance maximum of MBV attached to species of MBV peptides in 10 mM TFA
the native PC subunit occurs at 647 nm, were determined to be 40% lower than
whereas the absorbance maximum for the those of the naturally occurring PCB-bear-
proper PCB adduct occurs at 622 nm (2). ing peptides (2).
However, in cases in which multiple prod- Much less is known about the fluores-
ucts may be attached at the same site, the cence properties of the unnatural DBV
absorbance spectrum of the addition prod- adduct formed when PEB is added to apo-
uct will usually be difficult to interpret PC or apo-α-PE (4,20). The use of fluores-
(20). The absorbance of the peptide-bound cence to monitor product accumulation
bilins present can be determined by denat- with putative lyases that attach PEB may
uration of the addition product using one be complicated by the fact that multiple
of the methods described above. For PEB products accumulate in nonenzymatic
addition experiments, the nonenzymatical- reactions. Therefore, absorbance and fluo-
ly favored product, DBV, was found to rescence spectroscopy may not work as well
accumulate (4,20). DBV exhibits charac- as one of the following methods for the
teristic absorbance maxima at 606 and 330 characterization of enzymatic bilin addi-
nm in native proteins (74); for denatured tion to apo-PE.
subunits in acidic urea solutions, a 330 nm
absorbance peak is diagnostic of peptide- 4.3.3. HPLC Separation and Detection
linked DBV, whereas a 308 nm peak is
characteristic of peptide-linked PEB If the holo- and apo-subunits, which
(33,74). might be produced or used as substrates in
an enzymatic reaction, can be separated by
4.3.2. Fluorescence C4 reverse-phase chromatography as
described above, then this method provides
Free bilins exhibit little fluorescence in an excellent way to detect the transfer of
solution but become highly fluorescent bilin from a holophycobiliprotein to an
once they have been covalently attached to apo-subunit. Such separations are usually
phycobiliproteins, because they are rigidly best achieved if the source of the holo-sub-
held in a stretched conformation that does unit is from another organism. The trans-
not facilitate nonradiative decay of the fer reaction of PCB from Anabaena sp.
excited state (22). Therefore, the fluores- PCC 7120 holo-α-PC to Synechococcus sp.
cence emission spectrum of both control PCC 7002 apo-α-PC mediated by Syne-
and enzymatic reactions can be measured chococcus sp. PCC 7002 CpcECpcF was
as a way of monitoring the products of the detected using this method (22). Syne-
reaction (see Figure 5). The MBV product chococcus sp. PCC 7002 CpcECpcF pro-
of nonenzymatic PCB addition to apophy- teins can also transfer a bilin from Syne-
cocyanin is easily distinguished from the chococcus sp. PCC 7002 holo-PC to
natural PCB product, because both the Anabaena sp. PCC 7120 apo-α-PC sub-
328
Analysis and Reconstitution of Phycobiliproteins

unit (see Figure 6). aliquot of trypsin is added, and the incuba-
tion is continued for another 2 hours
4.3.4. Characterization of the Product by under the same conditions. The reaction is
Tryptic Digestion stopped by the addition of glacial acetic
acid to 30% (vol/vol). If a large amount of
This is the most quantitative method of protein is being digested, then fractiona-
characterization of the bilin product tion on Sephadex G-50 in 30% acetic acid
(2,3,20). The addition product is cleaved (vol/vol) is a good method to separate
using trypsin, and tryptic peptides are sep- undigested material from tryptic peptides.
arated on a C18 reverse-phase column (45). If the amount of material is scaled for ana-
Tryptic peptides can be collected, their lytical purposes, then the colored material
absorption spectra in 10 mM TFA deter- can be collected and loaded directly onto a
mined, and their composition evaluated by SepPak C18 cartridge. The cartridge can be
amino acid analysis or sequencing to show washed with 0.1% TFA followed by elu-
rigorously which bilin was added to a par- tion by 60% acetonitrile, 40% 0.1% TFA.
ticular site(s) on the apophycobiliprotein The eluate should be collected, dried under
subunit. If multiple products are present, N2, and redissolved in 10 mM TFA prior
this is the best method to determine how to HPLC separation. However, if the
many products have been formed and to amount of material is scaled for preparative
quantitate their relative amounts. Keep in purposes, the colored material in the eluate
mind that for each phycobiliprotein, diges- from the gel exclusion chromatography in
tion by more than one protease may be 30% acetic acid should then be concentrat-
required to obtain a fragment sufficiently ed under N2 before dilution with 50 mM
small to allow its isolation and characteri- Na-phosphate, pH 2.5. The mixture
zation. Digestion procedures for each type should then be fractionated on an ion-
of phycobiliprotein have been published exchange column (SP-Sephadex G-25, 2 ×
(7,49,50,55,67,74–76), and it is recom- 6.5 cm) and eluted with a linear gradient
mended that the user refer to the opti- of 0 to 0.6 M NaCl in 50 mM Na-phos-
mized procedure for the particular phyco- phate, pH 2.5. Fractions containing col-
biliprotein with which he/she is working. ored material should be collected, desalted
The procedure described below was used on the SepPak C18 cartridge as described
successfully on C-PC and R-PE (2,45). above, before separation by HPLC.
The addition product should be separat- The conditions used for separating the
ed from unreacted bilin by chromatogra- tryptic peptides of phycocyanin follow.
phy on Sephadex G-25. The phycobilipro- However, for each phycobiliprotein, differ-
tein should then be fully denatured by ent gradient conditions may be required,
acidification with 1 N HCl to pH 2.0 and and optimization of these conditions
stored under N2 for 45 minutes. Trypsin should be pursued prior to preparative-
(TCPK-treated; Worthington Biochemical, scale analyses. For the phycocyanin of
Lakewood, NJ, USA), dissolved in 1 mM Synechococcus sp. PCC 7002, a C18 reverse-
HCl at 5 mg/mL concentration, is added phase analytical column (5 µm, 4.6 × 250
to 2% (wt/wt) to the denatured phyco- mm) should be used for separation of tryp-
biliprotein in HCl. This mixture is titrated tic peptides (see Figure 7). The solvent sys-
to pH 7.5 with 1 N NaOH after the addi- tem is 0.1 M Na-phosphate, pH 2.1
tion of ammonium bicarbonate to 100 (Buffer A) and acetonitrile (Buffer B) with
mM. After incubation of this mixture for 2 flow rates of 1.5/mL min. Peptides are
hours at 30°C in the dark, an additional loaded at 20% Buffer B (80% Buffer A)
329
W.M. Schluchter and D.A. Bryant

Figure 6. Monitoring the transfer of bilin from Synechococcus sp. PCC 7002 PC to Anabaena sp. PCC 7120 apo-α-PC by
C4 reverse-phase HPLC. Each assay contained 100 µg of Anabaena sp. PCC 7120 apo-α-PC, 75 µg of Synechococcus sp. PCC 7002
PC, 0.2 µM Synechococcus sp. PCC 7002 CpcECpcF (if present) in a volume of 400 µL (reaction assay buffer conditions are as
described in Figure 5). Reactions were allowed to proceed for 16 hours at room temperature in the dark. Each reaction was com-
bined with 800 µL of 9 M urea, pH 1.9, mixed, and centrifuged prior to injection on the C4 column (as described in this chap-
ter). After injection, buffer conditions (buffers are those from Swanson and Glazer; Reference 66) are as follows: 2 minutes at 35%
Buffer B (65% Buffer A), a 1-minute linear gradient to 53% Buffer B (47% Buffer A), followed by a linear gradient to 63% Buffer
B over 20 minutes (22). Each assay was monitored at 280 nm (reflecting protein content) and 680 nm (reflecting bilin content).
Retention times for various components are as follows: Anabaena sp. PCC 7120 apo-α-PC, 9.5 minutes; Anabaena sp. PCC
7120 holo-α-PC, 10 minutes; Synechococcus sp. PCC 7002 apo-α-PC, 11.7 minutes; Synechococcus sp. PCC 7002 holo-α-PC,
12.2 minutes; Synechococcus sp. PCC 7002 holoβ-PC, 15.8 minutes. Synechococcus sp. PCC 7002 CpcECpcF is capable of trans-
ferring bilin from 7002 holo-α-PC to Anabaena sp. PCC 7120 apo-α-PC (W.M. Schluchter and A.N. Glazer, unpublished
results).
330
Analysis and Reconstitution of Phycobiliproteins

and eluted with a linear gradient to 40% ACKNOWLEDGMENTS


Buffer B (60% Buffer A) over 20 minutes
(2). We thank Dr. Alexander N. Glazer for
helpful comments. This research was sup-
ported in part by United States Public
5. CONCLUDING REMARKS Health Service (USPHS) Grant No.
GM-31625 (to D.A.B.), a National
Although phycobiliproteins were among Research Service Award Grant No. GM-
the first proteins to be characterized and 16935 (to W.M.S.), and the LA Board of
much is known about their structures, rela- Regents Grant No. LEQSF(1999-2002)-
tively little is still known concerning the RD-A-45 (to W.M.S.).
details of chromophore attachment to this
large and highly diverse protein family. ABBREVIATIONS
This situation has not improved dramati-
cally in spite of the availability of the com- AP, allophycocyanin; DBV, 15,16 dihy-
plete genomic sequence of the cyanobac- drobiliverdin; DTT, dithiothreitol; EDTA,
terium Synechocystis sp. PCC 6803. It is ethylenediamine tetraacetate; HPLC,
hoped that the procedures described above high-performance liquid chromatography;
for the production of substrate proteins MBV, mesobiliverdin; PC, phycocyanin;
and for the characterization of bilin attach- PCB, phycocyanobilin; PE, phycoerythrin;
ment reactions will aid other researchers PEB, phycoerythrobilin; PEC, phycoery-
interested in the characterization of new throcyanin; PUB, phycourobilin; PXB,
phycobiliproteins or in the characterization phycobiliviolin; PφB, phytochromobilin;
of the biosynthesis of phycobiliproteins. TFA, trifluoroacetic acid.

Figure 7. HPLC elution profile


from a C18-reverse phase column
of a tryptic digest of a preparation
of Synechococcus sp. PCC 7002
apophycocyanin after reaction
with free PCB, in the absence of
enzymes. The major products are
MBV at the α-84 (α-1MBV) and
β-82 (β-1MBV) sites with some
PCB forming at the β-82 site (β-
1PCB; the amount of this product
is variable). The elution of bilin-
linked peptides was monitored at
660 nm. This figure was modified
with permission from Reference 2.
331
W.M. Schluchter and D.A. Bryant

REFERENCES 16.Dolganov, N. and A.R. Grossman. 1999. A polypep-


tide with similarity to phycocyanin α-subunit phyco-
1.Airth, R.L. and L.R. Blinks. 1956. A new phycoery- cyanobilin lyase involved in degradation of phycobili-
thrin from Porphyra naiadum. Biol. Bull. 111:321-327. somes. J. Bacteriol. 181:610-617.
2.Arciero, D.M., D.A. Bryant, and A.N. Glazer. 1988. 17.Ducret, A., W. Sidler, G. Frank, and H. Zuber. 1994.
In vitro attachment of bilins to apophycocyanin I: spe- The complete amino acid sequence of R-Phycocyanin-
cific covalent adduct formation at cysteinyl residues I α and β subunits from the red alga Porphyridium cru-
involved in phycocyanobilin binding in C-phyco- entum: structural and phylogenetic relationships of the
cyanin. J. Biol. Chem. 263:18343-18349. phycocyanins within the phycobiliproteins families.
3.Arciero, D.M., J.L. Dallas, and A.N. Glazer. 1988. In Eur. J. Biochem. 221:563-580.
vitro attachment of bilins to apophycocyanin: II: deter- 18.Duerring, M., R. Huber, W. Bode, R. Ruembeli, and
mination of the structures of tryptic bilin peptides H. Zuber. 1990. Refined three-dimensional structure
derived from the phycocyanobilin adduct. J. Biol. of phycoerythrocyanin from the cyanobacterium
Chem. 263:18350-18357. Mastigocladus laminosus at 2.7 Å. J. Mol. Biol.
4.Arciero, D.M., J.L. Dallas, and A.N. Glazer. 1988. In 211:633-644.
vitro attachment of bilins to apophycocyanin: III: 19.Duerring, M., G.B. Schmidt, and R. Huber. 1991.
properties of the phycoerythrobilin adduct. J. Biol. Isolation, crystallization, crystal structure analysis and
Chem. 263:18358-18363. refinement of constitutive C-phycocyanin from the
5.Betz, M. 1997. One century of protein crystallogra- chromatically adapting cyanobacterium Fremyella
phy: the phycobiliproteins. Biol. Chem. 378:167-176. diplosiphon at 1.66 Å resolution. J. Mol. Biol. 217:577-
6.Bhalerao, R.P., L.K. Kind, and P. Gustafsson. 1994. 592.
Cloning of the cpcE and cpcF genes from Synechococcus 20.Fairchild, C.D., and A.N. Glazer. 1994. Nonenzymat-
sp. PCC 6301 and their inactivation in Synechococcus ic bilin addition to the a-subunit of an apophycoery-
sp. PCC 7942. Plant Mol. Biol. 26:313-326. thrin. J. Biol. Chem. 269:28988-28996.
7.Bishop, J.E., H. Rapoport, A.V. Klotz, C.F. Chan, 21.Fairchild, C.D., and A.N. Glazer. 1994. Oligomeric
A.N. Glazer, P. Guglistaller, and H. Zuber. 1987. structure, enzyme kinetics, and substrate specificity of
Chromopeptides from phycoerythrocyanin: structure the phycocyanin α subunit phycocyanobilin lyase. J.
and linkage of the three bilin groups. J. Am. Chem. Biol. Chem. 269:8686-8694.
Soc. 109:875-881. 22.Fairchild, C.D., J. Zhao, J. Zhou, S.E. Colson, D.A.
8.Brejc, K., R. Ficner, R. Huber, and S. Steinbacher. Bryant, and A.N. Glazer. 1992. Phycocyanin a sub-
1995. Isolation, crystallization, crystal structure analy- unit phycocyanobilin lyase. Proc. Natl. Acad. Sci. USA
sis and refinement of allophycocyanin form the 89:7017-7021.
cyanobacterium Spirulina platensis at 2.3 Å resolution. 23.Ficner, R. and R. Huber. 1993. Refined crystal struc-
J. Mol. Biol. 249:424-440. ture of phycoerythrin from Porphyridium cruentum at
9.Bryant, D.A. 1982. Phycoerythrocyanin and phyco- 0.23 nm resolution and localization of the g subunit.
erythrin: properties and occurrence in cyanobacteria. J. Eur. J. Biochem. 218:103-106.
Gen. Microbiol. 128:835-844. 24.Ficner, R., K. Lobeck, G. Schmidt, and R. Huber.
10.Bryant, D.A., G. Cohen-Bazire, and A.N. Glazer. 1992. Isolation, crystallization, crystal structure analy-
1981. Characterization of the biliproteins of Gloeobac- sis and refinement of B-phycoerythrin from the red
ter violaceus: chromophore content of a cyanobacterial alga Porphyridium sordidum at 2.2 Å resolution. J. Mol.
phycoerythrin carrying phycourobilin chromophore. Biol. 228:935-950.
Arch. Microbiol. 129:190-198. 25.Glazer, A.N. 1989. Light guides: directional energy
11.Bryant, D.A., J.M. Dubbs, P.I. Fields, R.D. Porter, transfer in a photosynthetic antenna. J. Biol. Chem.
and R. de Lorimier. 1985. Expression of phyoco- 264:1-4.
biliprotein genes in Escherichia coli. FEMS Microbiol. 26.Glazer, A.N. 1981. Photosynthetic accessory proteins
Lett. 29:343-349. with bilin prosthetic groups, p. 51-96. In M.D. Hatch
12.Bryant, D.A., A.N. Glazer, and F.A. Eiserling. 1976. and N.K. Boardman (Eds.), The Biochemistry of
Characterization and structural properties of the major Plants, vol. 8. Academic Press, New York.
phycobiliproteins of Anabaena sp. Arch. Microbiol. 27.Glazer, A.N. 1988. Phycobiliproteins. Methods Enzy-
110:61-75. mol. 167:291-303.
13.Chang, W., T. Jiang, Z. Wan, J. Zhang, Z. Yang, and 28.Glazer, A.N. 1988. Phycobilisomes. Methods Enzy-
D. Liang. 1996. Crystal structure of R-phycoerythrin mol. 167:304-312.
from Polysiphonia urceolata at 2.8 Å resolution. J. Mol. 29.Glazer, A.N., C.F. Chan, and J.A. West. 1997. An
Biol. 262:721-731. unusual phycocyanobilin-containing phycoerythrin of
14.Cohen-Bazire, G., S. Béguin, S. Rimon, A.N. Glazer, several bluish-colored, arcochaetioid, freshwater red
and D. M. Brown. 1977. Physico-chemical and algal species. J. Phycol. 33:617-624.
immunological properties of allophycocyanins. Arch. 30.Glazer, A.N. and S. Fang. 1973. Chromophore con-
Microbiol. 111:225-238. tent of blue-green algal phycobiliproteins. J. Biol.
15.de Lorimier, R., S.M. Wilbanks, and A.N. Glazer. Chem. 248:659-662.
1993. Genes of the R-phycocyanin II locus of marine 31.Glazer, A.N. and S. Fang. 1973. Formation of hybrid
Synechococcus spp. and comparison of protein-chro- proteins from the α and β subunits of phycocyanins of
mophore interactions in phycocyanins differing in unicellular and filamentous blue-green algae. J. Biol.
bilin composition. Plant Mol. Biol. 21:225-237. Chem. 248:663-671.

332
Analysis and Reconstitution of Phycobiliproteins

32.Glazer, A.N., S. Fang, and D.M. Brown. 1973. Spec- 48.Lokstein, H., C. Steglich, and W.R. Hess. 1999.
troscopic properties of C-phycocyanin and of its α and Light-harvesting antenna function of phycoerythrin in
β subunits. J. Biol. Chem. 248:5679-5685. Prochlorococcus marinus. Biochim. Biophys. Acta.
33.Glazer, A.N. and C.S. Hixson. 1975. Characterization 1410:97-98.
of R-phycocyanin: chromophore content of R-phyco- 49.Lundell, D.J. and A.N. Glazer. 1981. Allophyco-
cyanin and C-phycoerythrin. J. Biol. Chem. 250: cyanin B: α common β subunit in Synechococcus allophy-
5487-5495. cocyanin B (λmax 670 nm) and allophycocyanin (λmax
34.Glazer, A.N. and C.S. Hixson. 1977. Subunit struc- 650 nm). J. Biol. Chem. 246:12600-12606.
ture and chromophore composition of rhodophytan 50.Lundell, D.J., A.N. Glazer, R.J. DeLange, and D.M.
phycoerythrins: Porphyridium cruentum B-phyco- Brown. 1984. Bilin attachment sites in the α and β
erythrin and b-phycoerythrin. J. Biol. Chem. 252: subunits of B-phycoerythrin: amino acid sequence
32-42. studies. J. Biol. Chem. 259:5472-5480.
35.Glazer, A.N., D.J. Lundell, G. Yamanaka, and R.C. 51.MacColl, R. 1998. Cyanobacterial phycobilisomes. J.
Williams. 1983. The structure of a “simple” phycobil- Struct. Biol. 124:311-334.
isome. Ann. Microbiol. (Inst. Pasteur). 134B:159-180. 52.MacColl, R., L.E. Eisele, M. Dhar, J.-P. Ecuyer, S.
36.Glazer, A.N. and G.J. Wedemayer. 1995. Cryptomon- Hopkins, J. Marrone, R. Barnard, H. Malak, and A.J.
ad biliproteins—an evolutionary perspective. Photo- Lewitus. 1999. Bilin organization in cryptomonad
syn. Res. 46:93-105. biliproteins. Biochemistry 38:4097-4105.
37.Glazer, A.N., J.A. West, and C. Chan. 1982. Phyco- 53.MacColl, R. and D. Guard-Friar. 1987. Phycobilipro-
erythrins as chemotaxonomic markers in red algae: a teins. CRC Press, Boca Raton.
survey. Biochem. Sys. Ecol. 10:203-215. 54.Mach, H., C.R. Middaugh, and R.V. Lewis. 1992.
38.Gómez-Lojero, C., B. Pérez-Gómez, D.W. Krog- Statistical determination of the average values of the
mann, and A. Peña-Diaz. 1997. The tricylindrical core extinction coefficients of tryptophan and tyrosine in
of the phycobilisome of the cyanobacterium Arthrospi- native proteins. Anal. Biochem. 200:74-80.
ra (Spirulina) maxima. Int. J. Biochem. Cell Biol. 55.Ong, L.J. and A.N. Glazer. 1991. Phycoerythrins of
29:959-970. marine unicellular cyanobacteria I: bilin types and
39.Gómez-Lojero, C., B. Pérez-Gómez, G. Prado-Flores, locations and energy transfer pathways in Synechococ-
D.W. Krogmann, A. Cárabez-Rejo, and A. Peña- cus sp. phycoerythrins. J. Biol. Chem. 266:9515-9527.
Diaz. 1997. The phycobilisomes of the cyanobacteri- 56.Ong, L. J. and A.N. Glazer. 1987. R-phycocyanin II, a
um Arthrospira (Spirulina) maxima. Int. J. Biochem. new phycocyanin occurring in marine Synechococcus
Cell Biol. 29:1191-1205. species. Identification of the terminal energy acceptor
40.Gysi, J. and H. Zuber. 1974. Isolation and character- bilin in phycocyanins. J. Biol. Chem. 262:6323-6327.
ization of allophycocyanin from the thermophilic blue- 57.Ong, L.J. and A.N. Glazer. 1988. Structural studies of
green alga Mastigocladus laminosus cohn. FEBS Lett. phycobiliproteins in unicellular marine cyanobacteria,
48:209-213. p. 102-121. In S.E. Stevens, Jr. and D.A. Bryant
41.Hess, W.R., F. Partensky, G.W.M. van der Staay, (Eds.), Light-Energy Transduction in Photosynthesis:
J.M. Garcia-Fernandez, T. Börner, and D. Vaulot. Higher Plant and Bacterial Models. American Society
1996. Coexistence of phycoerythrin and a chlorophyll of Plant Physiologists, Rockville, MD.
α/β antenna in a marine prokaryote. Proc. Natl. Acad. 58.Padgett, M.P. and D.W. Krogmann. 1987. Large scale
Sci. USA 93:11126-11130. preparation of pure phycobiliproteins. Photosyn. Res.
42.Jung, L.J., C.F. Chan, and A.N. Glazer. 1995. Candi- 11:225-235.
date genes for the phycoerythrocyanin α subunit lyase: 59.Reuter, W., G. Wiegand, R. Huber, and M.E. Than.
biochemical analysis of pecE and pecF interposon 1999. Structural analysis at 2.2 Å of orthorhombic
mutants. J. Biol. Chem. 270:12877-12884. crystals presents the asymmetry of the allophyco-
43.Kahn, K., D. Mazel, J. Houmard, N. Tandeau de cyanin-linker complex, AP-LC7.8, from phycobilisomes
Marsac, and M.R. Schaefer. 1997. A role for cpeYZ in of Mastigocladus laminosus. Proc. Natl. Acad. Sci. USA
cyanobacterial phycoerythrin biosynthesis. J. Bacteriol. 96:1363-1368.
179: 998-1006. 60.Schirmer, T.,W. Bode, and R. Huber. 1987. Refined
44.Kehoe, D. and A. Grossman. 1996. Similarity of a three-dimensional structures of two cyanobacterial C-
chromatic adaptation sensor to phytochrome and eth- phycocyanins at 2.1 and 2.5 Å resolution. J. Mol. Biol.
ylene receptors. Science 273:1409-1412. 196:677-695.
45.Klotz, A.V. and A.N. Glazer. 1985. Characterization 61.Schirmer, T., W. Bode, R. Huber, W. Sidler, and H.
of the bilin attachment sites in R-phycoerythrin. J. Zuber. 1985. X-ray crystallographyic structure of the
Biol. Chem. 260:4856-4863. light-harvesting biliprotein C-phycocyanin from the
46.Lagarias, J.C., A.V. Klotz, J.L. Dallas, A.N. Glazer, thermophilic cyanobacterium Mastigocladus laminosus
J.E. Bishop, J.F. O’Connell, and H. Rapoport. 1988. and its resemblance to globin structures. J. Mol. Biol.
Exclusive A-ring linkage for singly attached phyco- 184:257-277.
cyanobilins and phycoerythrobilins in phycobilipro- 62.Schirmer, T., R. Huber, M. Schneider, W. Bode, M.
teins. J. Biol. Chem. 263:12977-12985. Miller, and M.L. Hackert. 1986. Crystal structure
47.Lamparter, T., F. Mittmann, W. Gartner, T. Börner, E. analysis and refinement at 2.5 Å of hexameric C-phy-
Hartmann, and J. Hughes. 1997. Characterization of cocyanin from the cyanobacterium Agmenellum qua-
recombinant phytochrome from the cyanobacterium druplicatum: the molecular model and its implications
Synechocystis. Proc. Natl. Acad. Sci. USA 94:11792-7. from light-harvesting. J. Mol. Biol. 188:651-676.

333
W.M. Schluchter and D.A. Bryant

63.Sidler, W.A. 1994. Phycobilisome and phycobilipro- in cyanobacterial light-harvesting proteins. Mol.
tein structure, p. 139-216. In D.A. Bryant (Ed.), The Microbiol. 30:475-486.
Molecular Biology of Cyanobacteria, Vol. 1. Kluwer 73.Wedemayer, G.J., D.G. Kidd, and A.N. Glazer. 1996.
Academic, Dordrecht, The Netherlands. Cryptomonad biliproteins: bilin types and locations.
64.Sineshchekov, V., J. Hughes, E. Hartmann, and T. Photosyn. Res. 48:163-170.
Lamparter. 1998. Fluorescence and photochemistry of 74.Wedemayer, G.J., D.G. Kidd, D.E. Wemmer, and
recombinant phytochrome from the cyanobacterium A.N. Glazer. 1992. Phycobilins of cryptophycean
Synechocystis. Photochem. Photobiol. 67:263-267. algae: occurrence of dihydrobiliverdin and meso-
65.Stec, B., R.F. Troxler, and M.M. Teeter. 1999. Crystal biliverdin in cryptomonad biliproteins. J. Biol. Chem.
structure of C-phyocyanin from Cyanidium caldarium 267:7315-7331.
provides a new perspective on phycobilisome assembly. 75.Wemmer, D.E., G.J. Wedemayer, and A.N. Glazer.
Biophys. J. 76:2912-2921. 1993. Phycobilins of cryptophycean algae: novel link-
66.Swanson, R.V. and A.N. Glazer. 1990. Separation of age of dihydrobiliverdin in a phycoerythrin 555 and a
phycobiliprotein subunits by reverse-phase high pressure phycocyanin 645. J. Biol. Chem. 268:1658-1669.
liquid chromatography. Anal. Biochem. 188:295-299. 76.Wilbanks, S.M. and A.N. Glazer. 1993. Rod struc-
67.Swanson, R.V., L.J. Ong, S.M. Wilbanks, and A.N. ture of a phycoerythrin II-containing phycobilisome
Glazer. 1991. Phycoerythrins of marine unicellular II: complete sequence and bilin attachment site of a
cyanobacteria II: characterization of phycobiliproteins phycoerythrin g subunit. J. Biol. Chem. 268:1236-
with unusually high phycourobilin content. J. Biol. 1241.
Chem. 266:9528-9534. 77.Wilde, A., Y. Churin, H. Schubert, and T. Börner.
68.Swanson, R.V., J. Zhou, J.A. Leary, T. Williams, R. de 1997. Disruption of a Synechocystis sp. PCC 6803 gene
Lorimier, D.A. Bryant, and A.N. Glazer. 1992. Char- with partial similarity to phytochrome genes alters
acterization of phycocyanin produced by cpcE and cpcF growth under changing light qualities. FEBS Lett. 406:
mutants and identification of an intergenic suppressor 89-92.
of the defect in bilin attachment. J. Biol. Chem. 78.Yeh, K. C., S.-H. Wu, J.T. Murphy, and J.C.
267:16146-16154. Lagarias. 1997. A cyanobacterial phytochrome two-
69.Tandeau de Marsac, N. and G. Cohen-Bazire. 1977. component light sensory system. Science 277:1505-
Molecular composition of cyanobacterial phycobili- 1508.
somes. Proc. Natl. Acad. Sci. USA 74:1635-1639. 79.Zhao, K.H., M.G. Deng, M. Zheng, M. Zhou, A.
70.Tandeau de Marsac, N., D. Mazel, V. Capuano, T. Parbel, M. Storf, M. Meyer, B. Strohmann, and H.
Damerval, and J. Houmard. 1990. Genetic analysis Scheer. 2000. Novel activity of a phycobiliprotein
of the cyanobacterial light-harvesting antenna complex, lyase: both the attachment of phycocyanobilin and the
p. 143-153. In D. Drews and E.A. Dawes (Eds.), Mol- isomerization to phycobiliviolin are catalyzed by the
ecular Biology of Membrane-Bound Complexes in proteins PecE and PecF encoded by the phycoerythro-
Phototropic Bacteria. Plenum, New York. cyanin operon. FEBS Lett. 469:9-13.
71.Thomas, J.-C. and C. Passquet. 1999. Characteriza- 80.Zhou, J., G.E. Gasparich, V.L. Stirewalt, R. de Lorim-
tion of a phycoerythrin without α-subunits from a uni- ier, and D.A. Bryant. 1992. The cpcE and cpcF genes
cellular red alga. J. Biol. Chem. 274:2472-2482. of Synechococcus sp. PCC 7002: construction and phe-
72.Toole, C.M., T.L. Plank, A.R. Grossman, and L.K. notypic characterization of interposon mutants. J. Biol.
Anderson. 1998. Bilin deletions and subunit stability Chem. 267:16138-16145.

334
Index 335

Index

A
ALA (see Aminolevulinic acid)
ALA synthase, 9, 70, 72, 73–76, 79
ALAD preparation (procedure), 77
ALAS (see ALA synthase)
ALAS preparation (procedure), 73
Allophycocyanin, 311, 317–318, 321, 323
Aminolevulinic acid, 4, 71, 72–76, 95–96, 278, 299
Ammoniacal extraction (procedure), 112
AP (see Allophycocyanin)
Aqueous two phase partitioning procedure, 198–200
B
Bacteriochlorophyll, 6, 237, 255
Bacteriocide, 195, 196
Bacteriophytochrome, 5, 306
Bacteriorhodopsin, 210, 218, 256
BChl (see Bacteriochlorophyll)
Bilatrienes, 299
Bile, 273, 276, 299
Bilin adduct assay (procedure), 324, 327
Bilirubin, 273–275
Biliverdin reductase assay (procedure), 287
Biliverdin, 10, 161, 173, 175, 177, 273-276, 282, 298, 323
Binodal curve, 188–189, 190, 199
Blood, 8, 10, 15, 17, 171, 172, 193
BR (see Bilirubin)
BV (see Biliverdin)
C
Capillary electrophoresis, 95, 108
Carotenoid, 97, 122, 237, 241–245, 263
CD (see Circular dichroism)
CE (see Capillary electrophoresis)
Chl (see Chlorophyll)
Chlide (see Chlorophyllide)
Chlorophyll determination (procedure), 258

335
336 Index

Chlorophyll extraction (procedure), 243–245


Chlorophyll, 2, 15, 20–24, 113, 121, 146, 235–237
Chlorophyllide, 6, 23, 112, 114, 120-121, 132, 135, 141
Chloroplast, 10, 89, 224
Circular dichroism, 241
Cobalamin, 71, 82
Collidine, 23, 24
Copro (see Coproporphyrin)
Coproporphyrin 20, 55, 87, 100, 104, 115
Coproporphyrinogen oxidase, 70, 88–89, 90
CPO (see Coproporphyrinogen oxidase)
Cross point procedure, 203
Cytochrome preparation (procedure), 166
D
DBV (see Dihydrobiliverdin)
DDQ (see Dichloro-dicyanobenzoquinone)
Detergent exchange (procedure), 266–247
Detergent removal (procedure), 265
DHBV (see Dihydrobiliverdin)
DHGG (see Dihydrogeranylgeraniol)
Dialysis membrane pretreatment (procedure), 262
Dichloro-dicyanobenzoquinone, 26, 33, 51, 52, 63
Dihydrobiliverdin, 274, 275, 282, 288, 328
Dihydrogeranylgeraniol, 139, 140, 147, 150
Dissolving porphyrins 59, 92
E
Electrospray ionization mass spectrometry, 96, 107
ESIMS (see Electrospray ionization mass spectrometry)
Ether extraction (procedure), 118
Ethyl diethylprrole carboxylate synthesis (procedure), 27
ETIO-I (see Etioporphyrin-I)
Etioporphyrin I synthesis (procedure), 34
Etioporphyrin-I, 15, 34, 36, 41
F
Feces (porphyrin extraction procedure), 97
Ferrochelatase preparation (procedure), 91
Freezing specimens (procedure), 218
G
Geranylgeraniol, 139, 140, 147, 150
GG (see Geranylgeraniol)
H
HEAR (procedure), 114
HEAR (see Hexane extracted acetone residue)
Heavy metal shadowing procedure, 216
Index 337

Hematuria, 172
Heme chemiluminescence procedure, 171
Heme detection (procedure), 168
Heme, 8, 9, 17, 100, 102, 158, 175, 179, 209, 273, 284
Hemin, 47, 55, 165, 166–167, 285, 286
Hemoglobin preparation (procedure), 163, 164
Hemoglobin, 3, 9, 10, 15, 172, 193, 200, 204, 315
Hemoprotein spectral analysis, 169, 170, 178
Hexane extracted acetone residue, 114, 116–119, 122, 123,
126–129, 131, 133, 134, 136-138, 141-143, 145, 151, 152
High performance liquid chromatography, 17, 45–46, 57,
58, 87, 89, 95, 96, 100, 102, 103, 105–108, 116, 121–123,
132, 134, 138, 140, 149, 152, 164, 237, 243, 245, 277, 278,
280–282, 284–286, 288, 298, 318-319, 323, 327–330
HO-1 (see human heme oxygenase isozyme 1)
Horse radish peroxidase, 161, 171
H-PHEN+, 63
HPLC (see High pressure/performance liquid chromatography)
HRP (see Horse radish peroxidase)
HSAP (see Hemoprotein spectral analysis)
Human heme oxygenase isozyme 1, 173–178
Hydroxymethylbilane (also called Preuroporphyrinogen),
4, 71, 80-82
I
Insecticyanin, 273
Iron octaethylporphyrin chromatography (procedure), 43
I
LCFA (see Long chain fatty alcohol)
Leghemoglobin, 8
LHC (see Light harvesting complex)
LHC preparation (procedure), 245–246
Light harvesting complex, 111, 235, 236, 238–243, 245–249,
256, 267
Long chain fatty alcohol, 119-121, 149, 150
Lutein, 242, 245
M
Magnetic circular dichroism, 173–174
MBV (see Mesobiliverdin)
MCD (see Magnetic circular dichroism)
Mesobiliverdin, 273, 276, 281, 285, 323, 328
Methine group, 1, 34, 106, 300, 302, 305
Methyl para-toluenesulfonate, 52, 61, 63
Methyl pheophorbide isolation (procedure), 26
Mg-protoporphyrin IX monomethyl ester, 114, 116, 117,
123, 126, 127, 130-132, 134, 139, 142, 143, 145
338 Index

Micrograph resolution (procedure), 226


Mitochondrion, 9, 88, 90
Mpe (see Mg-protoporphyrin IX monomethyl ester)
MTS (see Methyl para-toluenesulfonate)
Myoglobin preparation (procedure), 163
N
Negative staining procedure, 215–216
Neoxanthin, 245
NMR (see Nuclear magnetic resonance)
Nuclear magnetic resonance, 57, 177, 209, 256, 282, 283, 300
O
Octaethylporphyrin iron incorporation (procedure), 63
Octaethylporphyrin, 26, 33, 34, 63
Octylglucoside, 238, 240, 244, 247, 263
OEP (see Octaethylporphyrin)
OEP synthesis (procedure), 34
OG (see Octylglucoside)
P
PAGE (see Polyacrylamide gel electrophoresis)
Partition coefficient procedure, 201–202
PBG (see Porphobilinogen)
PBGD (see Porphobilinogen deaminase)
PBGD preparation (procedure), 80
PC (see Phycocyanin)
PCA (see Principal component analysis)
PCB (see Phycocyanobilin)
PCB (see Phycocyanobilin)
PCB preparation (procedure), 276–278
Pchlide (see Protochlorophyllide)
Pchlide E (see Protochlorophyllide ester)
PDT (see Photodynamic therapy)
PE (see Phycoerythrin)
PEB (see Phycoerythrobilin)
PEC (see Phycoerythrocyanin)
PEG derivatization, 196
Pheophorbide, 25, 55, 116, 151, 152
Pheophytin, 20, 21, 23, 26, 55, 116, 125, 147, 149, 150, 151–152,
240, 243, 244
Pheophytin, 20, 23, 26, 55, 116, 149, 151–152, 240, 243, 244
Photodynamic therapy, 10, 50, 54
Phycobiliviolin, 311
Phycocyanin preparation (procedure), 317–318, 325
Phycocyanin subunit renaturation (procedure), 316–317
Phycocyanin subunit separation (procedure), 316
Index 339

Phycocyanin, 277, 312, 314, 316, 321


Phycocyanobilin, 273–276, 282, 311
Phycoerythrin, 278, 279, 312, 314
Phycoerythrobilin, 273–275, 311
Phycoerythrocyanin, 312, 314, 324
Phycourobilin, 273, 311
Phytochrome assay (procedure), 297
Phytochrome assembly assay (procedure), 300
Phytochrome, 274, 293
Phytochromobilin preparation (procedure), 279–280
Phytochromobilin synthase assay (procedure),298–299
Phytochromobilin, 4, 273–274, 278, 281, 282, 294
Phytol, 121, 139, 140, 147, 149, 150
Polyacrylamide gel electrophoresis, 75, 81, 83, 90, 161,
170–172, 299, 321, 325, 247–248
POR (see Protochlorophyllide oxidoreductase)
Porphobilinogen deaminase, 70, 80, 84
Porphobilinogen, 4, 71, 76, 95
Porphyria, 55, 87, 89, 90, 98, 102
Porphyrinogen preparation (procedure), 106
PPO (see Protoporphyrinogen oxidase)
PPO preparation (procedure), 89
Preuroporphyrinogen (see Hydroxymethylbilane)
Principal component analysis, 169
Protein determination (procedure), 258–259
Proto (see protoporphyrin )
Protochlorophyllide ester, 139
Protochlorophyllide oxidoreductase, 136
Protochlorophyllide, 6, 120, 132
Protoheme IX 3, 8, 71, 90-92, 167, 168
Protoporphyrin IX dimethyl ester recrystallization (procedure),
47
Protoporphyrin, 4, 14, 17, 20, 27, 43, 49, 55, 57, 59, 88–90,
92, 100–101, 106, 118, 123, 178
Protoporphyrinogen oxidase, 70, 89–90
PUB (see Phycourobilin)
Purpurin, 23, 27
PXB (see Phycobiliviolin)
Pyridine hemochrome procedure, 167
Pyrrole, 1, 6, 27, 29, 30, 33–36, 51
PfB (see Phytochromobilin)
R
Reactive oxygen species, 4, 8–9
Rhodoporphyrin, 23
ROS (see Reactive oxygen species)
340 Index

S
Shemin pathway, 4, 72
Siroheme, 4, 87,
Sucrose density gradient, 248–249
T
TAPP, 49, 50, 52, 63
Tetrahydrogeranylgeraniol, 139, 140, 147, 150
Tetrakis(2-amino-phenyl)porphyrin TLC (procedure), 45
Tetramethylbenzidine, 170
Tetraphenylporphyrin, 30, 31, 33, 57, 63
Thaumatin, 186
THGG (see Tetrahydrogeranylgeraniol)
Thylakoid, 111, 224, 238, 242, 266, 267
TMBZ (see Tetramethylbenzidine)
TMBZ PAGE staining procedure, 170
TMPyP(X), 48, 63
TPP (see Tetraphenylporphyrin)
TPP synthesis (procedure), 33
TPP, 26, 30, 31, 33, 42, 51, 54, 57
TPPC4, 49, 54, 63
TPPS1, 54, 63
TPPS2, 54, 63
TPPS3, 54, 56, 57, 63
TPPS4, 48, 49, 50, 54, 57, 63
TPyP(X), 49, 50, 63
Turacin, 10
Turacoverdin, 10
Two-dimensional crystal growth procedure, 212, 261–263,
265, 266–268
U
Urine, 97
Uro (see Uroporphyrin)
Uroporphyrin, 10, 98
Uroporphyrinogen III, 4, 20, 80, 81–85
UROS preparation (procedure), 83
V
Violaxanthin, 245
X
Xanthophyll, 148, 243, 244–45, 246–247
Z
Zeaxanthin, 242
Zinc blot assay (procedure), 300
Heme, Chlorophyll, and Bilins
Methods and Protocols
Edited by
Alison G. Smith
Department of Plant Sciences, University of Cambridge, UK

Michael Witty
Department of Biochemistry, University of Cambridge, UK

Although researchers can profitably investigate heme, chlorophyll, and related tetrapyrroles in a wide
range of academic and medical research programs, the handling and manipulation of these delicate com-
pounds requires considerable skill and cross-boundary knowledge. In Heme, Chlorophyll, and Bilins: Methods
and Protocols, an interdisciplinary panel of hands-on investigators overcomes these limitations by describing in
detail how to work successfully with chlorophyll, heme, and bilins in biological, medical, chemical, and bio-
chemical research. Each method is presented by a researcher who actually uses it on a daily basis and includes
step-by-step instructions and pertinent tricks-of-the-trade that often make the difference between laboratory
success and failure. Topics range from methods for the analysis of tetrapyrroles, heme, and hemoproteins, to
the biosynthesis and the analysis of chlorophyll and bilins.
Timely and highly practical, Heme, Chlorophyll, and Bilins: Methods and Protocols is a gold-standard
collection of readily reproducible techniques suitable for a wide range of researchers, whether it be a clinician
studying photodynamic therapy, an ecologist studying the chlorophyll composition of leaves in a tropical
forest, or a cell biologist investigating the function of specific hemoproteins.

Features

• Detailed step-by-step protocols that have been • Time-saving techniques that even a highly
optimized for robust results skilled researcher will find helpful
• Numerous tricks-of-the-trade that often make • Troubleshooting tips, alternative ways of doing
the difference between success and failure things, and informative explanations

Contents
Laboratory Methods for the Study of Tetrapyrroles. Syn- Phase Systems. Structural Study of Heme Proteins by Elec-
theses of Tetrapyrroles. General Laboratory Methods for tron Microscopy of 2-Dimensional Crystals. Analysis and
Tetrapyrroles. Enzymatic Preparation of Tetrapyrrole In- Reconstitution of Chlorophyll–Proteins. Two-Dimensional
termediates. Analysis of Biosynthetic Intermediates, 5- Crystallization of Chlorophyll Proteins. Biosynthesis and
Aminolevulinic Acid to Heme. Analysis of Intermediates Analysis of Bilins. Analysis and Reconstitution of Phyto-
and End Products of the Chlorophyll Biosynthetic Path- chromes. Analysis and Reconstitution of Phycobiliproteins:
way. Analysis of Heme and Hemoproteins. Hemoproteins Methods for the Characterization of Bilin Attachment Reac-
Purification and Characterization by Using Aqueous Two- tions. Index.

90000

Heme, Chlorophyll, and Bilins: Methods and Protocols


ISBN: 1-58829-111-1 9 781588 291110

Вам также может понравиться