Вы находитесь на странице: 1из 52

CHAPTER

Post-transcriptional
Gene Control
Portion of a "lampbrush chromosome" from an oocyte of the newt
Nophtha/mus viridescens; hnRNP protein associated with nascent RNA
transcripts fluoresces red after staining with a monoclonal antibody.
[Courtesy of M. Roth and J. Gall.]

n the previous chapter, we saw that most genes are regulated that -95 percent of human genes give rise to alternatively

I at the first step in gene expression, transcription, by regu-


lating the assembly of the transcription pre-initiation com-
plex on a promoter DNA sequence and regulating transcription
spliced mRNAs. These alternatively spliced mRNAs encode
related proteins with differences in sequences limited to spe-
cific functional domains. In many cases, alternative RNA
elongation in the promoter proximal region . Once transcrip- splicing is regulated to meet the need for a specific protein
tion has been initiated, synthesis of the encoded RNA re- isoform in a specific cell type. Given the complexity of pre-
quires that RNA polymerase transcribe the entire gene and mRNA splicing, it is not surprising that mistakes are occasion-
not terminate prematurely. Moreover, the initial primary ally made, giving rise to mRNA precursors with improperly
transcripts produced from eukaryotic genes must undergo spliced exons. However, eukaryotic cells have evolved RNA
various processing reactions to yield the corresponding func- surveillance mechanisms that prevent the transport of mcor-
tional RNAs. For mRNAs, the 5' cap structure necessary for rectly processed RNAs to the cytoplasm or lead to their deg-
translation must be added (see Figure 4-14), introns must be radation if they are transported.
spliced out of pre-mRNAs (Table 8-1), and the 3' end must Additional control of gene expression can occur in the
be polyadenylated (see Figure 4-1 5 ). Once formed in the nu- cytoplasm. In the case of protein-coding genes, for instance,
cleus, mature, functional RNAs are exported to the cyto- the amount of protein produced depends on the stability of
plasm as components of ribonucleoproteins. Both processing the corresponding mRNAs in the cytoplasm and the rate of
of RNAs and their export from the nucleus offer opportuni- their translation. For example, during an immune response,
ties for further regulating gene expression after the initiation lymphocytes communicate by secreting polypeptide hor-
of transcription. mones called cytokines that signal neighboring lymphocytes
Recently, the vast ampunt of sequence data on human through cytokine receptors that span their plasma mem-
mRNAs expressed in different tissues and at various times branes (Chapter 23). lt is important for lymphocytes to syn-
during embryogenesis and cellular differentiation has revealed thesize and secrete cytokines in short bursts. This is possible

OUTLINE

8.1 Processing of Eukaryotic Pre-mRNA 348 8.4 Cytoplasmic Mechanisms of Post -transcriptional
Control 370
8.2 Regulation of Pre-mRNA Processing 360
8.5 Processing of rRNA and tRNA 384
8.3 Transport of mRNA Across the Nuclear Envelope 365
FiJ:i!J:ii RNAs Discussed in Chapter 8

mRNA Fully processed messenger RNA with 5' cap, introns removed b} RNA splicing, and a poly(A) tail

pre-mRNA An mRNA precursor conraming introns and not cleaved at the poly(A) site

hnRNA Heterogeneous nuclear RNAs. These include pre-mRNAs and RNA processing intermediates containing one or
more inrrons.

snR~A Five small nuclear RNAs that function in the removal of introns from pre-mRNAs by RNA splicing, plus rwo
small nuclear RNAs that substitute for the first two at rare inrrons

pre-tRNA A tRNA precursor containing additional transcribed bases at the 5' and 3' ends compared to the mature tRNA.
Some pre-tRNAs also contain an intron in the anti-codon loop.

pre rR~A The precursor to mature J8S, 5.8S, and 28S ribosomal RNAs. The mature rRNAs are processed from this long
precursor RNA molecule by cleavage, removal of bases from the ends of the cleaved products, and modification
of specific bases. '

snoRNA Small nucleolar RNAs. These base-pair with complementary regions of the pre-RNA molecule, directing
cleavage of the RNA chain and modification of bases during maturation of the rRNAs.

siRNA Short interfering RNAs, -22 bases long, that arc each perfectly complementary to a sequence in an mRNA.
Together with associated proteins, siRNAs cause cleavage of the "target" RNA, leading to its ra~id degradation.

m1RNA M1cro RNAs, -22 bases long, that base-pa1r extensively, but not completely, with mRNAs, especially over the
SIX base pairs at the 5' end of the miRNA. Th1s inhibits translation of the "target" mRNA.

because cytokine mRNAs are extremely unstable. Conse- expressed in the multiple types of human cells. Although some
quently, the concentration of the mRNA in the cytoplasm have recently been discovered to function through inhibition
falls rapidly once its synthesis is stopped. In contrast, mRNAs of target gene expression in the appropriate tissue and at the
encoding proteins required in large amounts that function appropriate time in development, the functions of the vast
over long period.s, such as ribosomal proteins, are extremely majority of human miRNAs are unknown and are the subject
stable so that multiple polypeptides are transcribed from each of a growing new area of research. If most miRNAs do indeed
mRNA. have significant functions, miRNA genes constitute an impor-
In addition to regulation of pre-mRNA processing, nu - tant subset of the -25,000 human gene~- A closely related
clear export, and translation, the cellular locations of many, process called RNA interference (RNAi) leads to the degrada-
if not most, mRNAs are regulated so that newly synthesized tion of viral RNAs in infected cells and the degradation of
protein is concentrated where it is needed. Particularly strik- transposon-encoded RNAs in many eukaryotes. This is of tre-
ing examples of this occur in the nervous systems of multi- mendous significance to biological researchers because it is
cellular animals. Some neurons in the human brain generate possible to design short interfering RNAs (siRNA) to inhibit
more than 1000 separate synapses with other neurons. Dur- the translation of specific mRNAs experimentally by a process
ing the process of learning, synapses that fire more frequently called RNA knockdown. This makes it possible to inhibit the
than others increase in size many times, while other synapses function of any desired gene, even in organisms that arc not
made by the same neuron do not. This can occur because amenable to classic genetic methods for isolating mutants.
mRNAs encoding proteins crittcal for synapse enlargement We refer to all the mechanisms that regulate gene expres-
are stored at all synapses, but translation of these localized, sion following transcription as post-transcriptional gene con-
stored mRNAs is regulated at each synapse independently by trol (Figure 8-1 ). Since the stability and translation rate of an
the frequency at which it initiates firing. In this way, synthe- mRNA contribute to the amount of protein expressed from a
sis of synapse-associated protein<, c;1n hP regulated independ- gene, these post-transcriptional processes are important com-
ently at each of the many synapses made by the same neuron. ponents of gene control. Indeed, the protein output of a gene
Another type of gene regulation that has recently come to is regulated at every step in the life of an m RNA from the ini-
light involves micro RNAs (miRNAs), which regulate the sta- tiation of its synthesis to its degradation. Thus genetic regula -
bility and translation of specific target mRNAs in multicellu- tory processes act on RNA as well as DNA. In this chapter,
lar animals and plants. Analyses of these short miRNAs in we consider the events that occur in the processing of mRNA
vanous human tissues indicate that there are -500 miRNAs following transcription initiation and promoter proximal

346 CHAPTER 8 Post-transcriptional Gene Co ntrol


Nucleolus

Pre-rR NA
Pre-mRNA transcription
transcription 5S rRNA

~
\
~~p..
Pre-tRNA
transcription
Ribosomal
subunit ,Sf)
Exc1sed
synthesis in
nucleolus pre-rRNA
a q ~ =~~i~Tn~NA mD
.......-----------~-
1'U:111 polyadenylation
Cleavage'
~

.-------------~-
...-
Improperly
processed Exosome
mRNA ~

~0 m ~d
pre-tRNA
mANA
export Nucleus

~AAAAA

liJ iI
Cytoplasmic
Poly(A) pol
Cytoplasmic
exosome

~
Cytoplasmic
De~~j CJ
enzyme Deadenylase

~ liJ
polyadenylation

~AAAAA
miRNA \ IJ /
i
~~AAAAA
miRNA
t ranslation inhibition Translation Cytoplasmic
initiation deadenylation

FIGURE 8-1 Overview of RNA processing and post- by degradation by cytoplasmic exosomes. The degradation rate of each
transcriptional gene control. Nearly all cytoplasmic RNAs are mRNA is controlled, thereby regu lating the mRNA concentration and,
processed from primary transcripts in the nucleus before t hey are consequently, the amount of protein translated. Some mRNAs are
exported to the cytoplasm. For protein-coding genes transcribed by synthesized without long poly(A) tails. Their translation is regulated by
RNA polymerase II, gene cont rol can be exerted through 0 the choice [iJ controlling the synthesis of a long poly(A) tail by a cytoplasmic
of alternative exons during pre-mRNA splicing and f) the choice of poly(A) polymerase. fJ Translation is also regulated by other mecha-
alternative poly(A) sites. Improperly processed mRNAs are blocked nisms including miRNAs. When expressed, t hese - 22-nucleotide RNAs
from export to the cytoplasm and deqraded 11 by a large complex inhibit translation of mRNAs to whic.h they hybridize, usually in the
called the exosome that contains multiple ribonucleases. Once 3'-untranslated region. tRNAs and rRNAs are also synthesized as
exported to the cytop l asm, ~ translation initiation factors bind to t he precursor RNAs that must be (;) processed before they are functional.
mRNA 5' -cap cooperatively with poly(A)-binding protein I bound to Regions of precursors cleaved from the mature RNAs are degraded by
m
the poly(A) tail and initiate translation (see Figure 4-28). mRNA is nuclear exosomes ~ . [Adapted from Houseley, et. al., 2006, Nat. Rev. Mol. Cell
degraded in the cytoplasm by de-adenylation and decapping followed Bioi. 7:529.)

CH A PTER 8 Post-transcriptional Gene Control 347


elongation and the various mechanisms t hat are known to precursor is being transcribed. Thus pre-mRNA processing is
regulate these events. In the last section, we briefly discuss the co-transcriptional. As the RNA emerges from the surface of
processing of primary transcripts produced from genes encod- RNA polymerase II, its 5' end is immediately modified by the
mg rRNAs and tRNAs. addition of the 5' cap structure found on all mRNAs (see Fig-
ure 4-14). As the nascent pre-mRNA continues to emerge from
the surface of the polymerase, it is immediately bound by
members of a complex group of RNA-binding proteins that
8.1 Processing of Eukaryotic Pre-mRNA
assist in RNA splicing and export of the fully processed mRNA
In this section, we take a closer luuk at huw t:ukaryoric cells through nuclear pore complexes into the cytoplasm. ~orne of
convert the initial primary transcript synthesized by RNA these proteins remain associated with the mRNA in the cyto-
polymerase II into a functional mRNA. T h ree major events plasm, but most either remain in the nucleus or shuttle back
occur during the process: 5' capping, 3' cleavagelpolyadenyla- into the nucleus shortly after the mRNA is exported to the
tion, and RNA splicing (Figure 8-2). Adding these specific cytoplasm. Cytoplasmic RNA-bindi ng proteins are exchanged
modifications to the 5 ' and 3' ends of the pre-mRNA is impor- for the nuclear ones. Consequently, mRNAs never occur as
tant to protect it from enzymes that quickly digest uncapped free RNA molecules in the cell but are always associated with
RNAs generated by RNA processing, such as spliced-out in- protein as 1ibonucleoprotein (RNP) complexes, first as nascent
trons and RNA transcribed downstream from a polyadenyla- pre-mRNPs that are capped and- spliced as they are tran-
tion site. The 5' cap and 3' poly(A) tail distinguish pre-mRNA scribed. Then, following cleavage and polyadenylation, they
molecules from the many other kinds of RNAs in the nucleus. are referred to as nuclear mRNPs. Following the exchange of
Pre-mRNA molecules (see Table 8-1 ) are bound by nuclear proteins that accompany export to the cytoplasm, they are
proteins that function in mRNA export to the cytoplasm. After called cytofJ!asmic mRNPs. Although we frequently refer to
mRNAs are exported to the cytoplasm, they are bound by a set pre-mRNAs and mRNAs, it is important to remember that
of cytoplasmic proteins that stimulate translation and are criti- they are always associated with proteins as RNP complexes.
cal for mRNA stabi lity in the cytoplasm. Prior to nuclear ex-
port, introns must be removed to generate the correct coding
The 5 ' Cap Is Added to Nascent RNAs
region of the mRNA. In higher eukaryotes, including humans,
alternative splicing is intricately regulated in order to substitute Shortly After Transcription Initiation
different functional domains into proteins, producing a consid- As the nascent RNA transcript emerges from the RNA chan -
erable expansion of the proteome of these organisms. nel of RNA polymerase II and reaches a length of - 25 nu-
The pre-mRNA processing events of capping, splicing, and cleotides, a protective cap composed of 7 -methylguanosine
polyadenylation occur in the nucleus as the nascent mRNA and methylated riboses is added to the 5' end of eukaryotic

0 ANIMATION: Life Cycle of an mRNA

Poly(A) Termination
site sites

DNA

Primary RNA
Cap
tII Transcription, 5' capping
3'
transcript
5'
Endo~ucle
tfJ Cleavage at poly(A) site
"'J
ro
3:xJ
5'
A polymetase
tIJ Polyadenylation
3'
z
l>
'0
0
5'
tIIJ RNA splicing
A1oo-2so3' C)
(!)
(/)

!:'!.
::J
co
mRNA 5' A100-2so 3 ' J
FIGURE 8 - 2 Overview of mRNA processing in eukaryotes. Shortly adenosine (A) residues is added (step iJ). The poly(A) tail contains - 250 A
after RNA polymerase II initiate~ trdmc.ription at the first nucleotide of the residues in mammals, - 1SO in insects, and - 100 in yeasts. For short
first exon of a gene, the 5' end of the nascent RNA is capped with primary transcripts with few introns, splicing (step 9 ) usually follows cleav-
7-methylguanylate (step 0 ). Transcription by RNA polymerase II age and polyadenylation, as shown. For large genes with multiple introns,
terminates at any one of multiple termination sites downstream from the introns often are spliced out of the nascent RNA during its transcription,
poly(A) site, which is located at the 3' end of the final exon. After the i.e., before transcription of the gene is complete. Note that the 5 ' cap and
primary transcript is cleaved at the poly(A) site (step f)), a string of sequence adjacent to the poly(A) tail are retained in mature mRNAs.

348 CHAPTER B Post -transcriptional Gene Control


5' end of RNA activation of the capping enzyme by the phosphorylated
CTD result in specific capping of R0!As transcribed by RNA
N -J@ijj,J;J~t;.i polymerase II.
One subunit of the capping enzyme removes the-y-
phosphate from the 5' end of the nascent RNA (Figure 8-3).
Another domain of this subunit transfers the GMP moiety
from GTP to the 5 '-diphosphate of the nascent transcript,
a ~ y ~ a
creating the unusual guanosine 5'-5' -triphosphate structure.
G 0
P P + ~ fl N-i@ij .. @r!J
'----v----' In the final steps, separate eu.qmes transfer methyl groups
GTP from S-adenosylmethionine to the N7 position of the guanine
and one or two 2' oxygens of riboses at the 5' end of the
nascent RNA.
Considerable evidence indicates that capping of the nas-
N -i@ij,J;J~f!l
cent transcript is coupled to elongation by RNA polymerase

l
Gua~ine 7-mot~ +CH 3 from
II so that all of its transcripts are capped during the earliest
t ansfe S-Ado-Met phase of elongation. As discussed in Chapter 7, in metazoans,
during the initial phase of transcription the polymerase elon-
gates the nascent transcript very slowly due to association of
N-i@ij,I;J@f!l
NELF (negative elongation factor) with RNA polymerase II

l+CH 3 from in the promoter proximal region (see Figure 7-20). Once the
S-Ado-Met 5' end of the nascent RNA is capped, phosphorylation of the
RNA polymerase CTD at position 2 in the heptapeptide re-
peat and of NELF and DSIF by the CDK9-cyclin T protein
kinase causes the release of NELF. This allows RNA polymer-
FIGURE 8-3 Synthesis of 5' -cap on eukaryotic mRNAs. The 5' end ase Il to enter into a faster mode of elongat_ion that rapidly
of a nascent RNA contains a 5 '-triphosphate from the initiating NTP. transcribes away from the promoter. The net effect of this
The-y-phosphate is removed in the first step of capping, while the mechanism is that the polymerase waits for the nascent RNA
remaining a- and (3-phosphates (orange) remain associated with the to be capped before elongating at a rapid rate.
cap. The third phosphate of the 5',5'-triphosphate bond is derived
from the ex-phosphate ofthe GTP that donates the guanine. The
methyl donor for methylation of the cap guanine and the first one
or two riboses of the mRNA is 5-adenosylmethionine (5-Ado-Met). A Diverse Set of Proteins with Conserved
[From S. Venkatesan and B. Moss, 1982, Proc. Nor/. Acad. Sci. USA 79:304.] RNA-Binding Domains Associate
with Pre-mRNAs
As noted earlier, neither nascent RNA transcripts from protein-
mRNAs (see Figure 4-14 ). The 5' cap marks RNA molecules coding genes nor the intermediates of mRJ:\A processing, col-
as mRNA precursors and protects them from RNA-digesting lectively referred to as pre-mRNA, exist as free RNA molecules
enzymes (5' -exoribonucleascs) in the nucleus and cytoplasm. in the nuclei of eukaryotic cells. From the time nascent tran-
This initial step in RNA processing is catalyzed by a dimeric scripts first emerge from RNA polymerase II until mature
capping enzyme, which associates with the phosphorylated mRNAs are transported into the cytoplasm, the RNA mole-
carboxyl-terminal domain (CTD) of RNA polymerase II. Re- cules are associated with an abundant set of nuclear proteins.
call that the CTD becomes phosphorylated by the TFIIH These are the major protein components of heterogeneous ribo-
general transcription facto.r at multiple serines at the 5 posi- nucleoprotein particles (lmRNPs), which contain heterogene-
tion in the CTD heptapeptide repeat during transcription ous nuclear RNA (hnRNA), a collective term referring to
initiation (see Figure 7-17). Binding to the phosphorylated pre-mRNA and other nuclear RNAs of various sizes. These
CTD stimulates the activity of the capping enzymes so that hnRNP proteins contribute to further steps in RNA processing,
they are focused on R NAs containing a 5'-triphosphate that including splicing, polyadenylation, and export through nu-
emerge from RNA polymerase II and nor on RNAs tran- clear pore complexes to the cytoplasm.
scribed by RNA polymerases I or III, which do not have a Researchers identified hnRNP proteins by first exposing
CTD. This is important because pre-mRNA synthesis ac- cultured cells to high-dose UV irradiation, which causes cova-
counts for only -80 percent of the total RNA synthesized in lent cross-links to form between RNA bases and closely associ-
replicating cells. About 20 percent is preribosomal RNA, ated proteins. Chromatography of nuclear extracts from treated
which is transcribed by RNA polymerase I, and 55 rRNA, cells on an oligo-dT cellulose column, which binds RNAs with
tRNAs, and other stable small RNAs, which are transcribed a poly( A) tail, was used to recover the proteins that had become
by RNA polymerase IlL The two mechanisms of (1) binding cross-linked to nuclear polyadenylared RNA. Subsequent treat-
of the capping enzyme to initiated RNA polymerase II spe- ment of cell extracts from un-irradiated cells with monoclonal
cifically through its unique, phosphorylated CTD and (2) antibodies specific for the major proteins identified by this

8.1 Processing of Eukaryotic Pre-mRNA 349


~ VIDEO: hnRNP A 1 Nucleocytoplasmic Shuttling

(a) (b) (c)

.
......
.~!

FIGURE 8-4 Human hnRNP A1 protein can cycle in and out of the right of the oval-shaped Xenopus nucleus. (b, c) When the same
cytoplasm, but human hnRNP C protein cannot. Cultured Hela cells preparation was viewed by fluorescence microscopy, the stained
and Xenopus cells were fused by treatment with polyethylene glycol, hnRNP C protein appeared green and thi stained hnRNP A 1 protein
producing heterokaryons containing nuclei from each cell type. The appeared red. Note that the unfused Xenopus cell on the left is
hybrid cells were treated with cycloheximide immediately after fusion unstained, confirming that the antibodies are specific for the human
to prevent protein synthesis. After 2 hours, the cells were fixed and proteins. In the heterokaryon, hnRNP C protein appears only in the
stained with fluorescent-labeled antibodies specific for human hnRNP Hela-cell nucleus (b), whereas the A 1 protein appears in both the
C and A 1 proteins. These antibodies do not bind to the homologous Hela-cell nucleus and the Xenopus nucleus (c). Since protein synthesis
Xenopus proteins. (a) A fixed preparation viewed by phase-contrast was blocked after cell fusion, some of the human hnRNP A 1 protein
microscopy includes unfused Hela cells (a rrowhead) and Xenopus cells must have left the Hela-cell nucleus, moved through the cytoplasm,
(dotted arrow), as well as fused heterokaryons (solid arrow). ln the and entered the Xenopus nucleus in the heterokaryon. [SeeS. Pinoi-Roma
heterokaryon in this micrograph, the round Hela-cell nucleus is to the and G. Dreyfuss, 1992, Nature 355:730; courtesy of G. Dreyfuss.]

cross-linking technique revealed a complex set of abundant out of the cytoplasm, suggesting that they function in the
hnRJ\.rp proteins ranging in size from -30 to - 120 kDa. transport of mRNA (Figure 8-4).
Like transcription factors, most hnR, P proteins have a
modular structure. They contain one or more RNA-binding Conserved RNA-Binding Motifs The RNA recognztion motif
domains and at least one other domain that interacts with (RRM), also called the RNP motif and the RNA-binding do-
other proteins. Several different RNA-binding motifs have main (RBD), is the most common RNA-binding domain in
been identified by creating hnRNP proteins with missing hnRNP proteins. This -SO-residue domain, which occurs in
amino acid seq uences and testing their ability to bind RNA. many other RNA-binding proteins, contains two highly con-
served sequences (RNP1 and RNP2) that are found across
Functions of hnRNP Proteins The association of pre-mRNAs organisms ranging from yeast to human-indicating that
with hnRNP proteins prevents the pre-mRNAs from forming like many D A-binding domains, they evolved early in
short secondary structures dependent on base pairing of com- eukaryotic evolution.
plementary regions, thereby making the pre-mRNAs accessi- Structural analyses have shown that the RRM domain con-
ble for interaction with other RNA molecules or proteins. sists of a four-stranded ~sheet flanked on one side by two a
Pre-mRNAs associated with hnRNP proteins present a more helices. To interact with the negatively charged RNA phos-
uniform substrate for subsequent processing steps than would phates, the~ sheet forms a positively charged surface. The con-
free, unbound pre-mRNAs, in which each mRNA forms a served RNPl and RNP2 sequences lie side by side on the two
unique secondary structure due to its specific sequence. central ~ strands, and their side chains make multiple contacts
Binding studies with purified hnRNP proteins indicate that with a single-stranded region of RNA that lies across the sur-
different hnRNP proteins associate with different regions of a face of the 13 sheet (Figure 8-5).
newly made pre-mRNA molecule. For example, the hnRNP The 45-residue KH motif is found in the hnRNP K pro-
proteins A1, C, and D bind preferentially to the pyrimidine- tein and several other RNA-binding proteins. The three-
rich sequences at the 3' ends of introns (see Figure 8-7). Some dimensional structure of representative KH domains is similar
hnRNP proteins interact with the RNA sequences that specify to that of the RRM domain but smaller, consisting of a
RNA splicing or cleavage/polyadenylation and contribute to three-stranded 13 sheet supported from one side by two a
the structure recognized by RNA-processing factors. Finally, helices. Nonetheless, the KH domain interacts with RNA
cell-fusion experiments have shown that some hnRNP proteins much differently than does the RRM domain. RNA binds to
remain localized in the nucleus, whereas others cycle in and the KH domain by interacting with a hydrophobic surface

350 CHAPTER 8 Post-transcriptional Gene Control


(a) RNA recognition motif (RRM) (b) Sex-lethal (Sxl) RRM domains (c) Polypyrimidine tract binding protein {PTB)

FIGURE 8-5 Structure of the RRM domain and its interaction with regions, in shades of red. The pre-mRNA is bound to the surfaces of the
RNA. {a) Diagram of the RRM domain showin g the two n helices positively charged [3 sheets, making most of its contacts with the RNP 1
{green) and four [3 strands {red) that characterize this motif. The and RNP2 regions of each RRM. {c) Strikingly different orientation of
conserved RNPl and RNP2 regions are located in the two central RRM domains in a different hnRNP, the polypyrimidine tract binding
[3 stra nds. (b) Surface representation of the two RRM domains in (PTB) protein, illustrating that RRMs are oriented in different relative
Drosophila Sex-lethal (Sxl) protein, which bind a nine-base sequence in positions in different hnRNPs; colors are as in (b). Polypyrimidine (p{Y))
transformer pre-mRNA (yellow). The two RRMs are oriented like the two single-stranded RNA is bound to the upward (RRM3) and downward
parts of an open pair of castanets, with the [3 sheet of RRMl facing (RRM4) facing [3-sheets. RNA is shown in yellow. [Part (a) adapted from
upward and the [3 sheet of RRM2 facing downward. Positively charged K. Nagai et al., 1995, Trends Biochem. Sci. 20:235. Part (b) after N. Harada et al.,
regions in Sxl protein are shown in shad es of blue; negatively charged 1999, Nature 398:579. Part (c) after F. C. Oberstrass et al., 2006, Science 309:2054.)

formed by the o: helices and one 13 strand. The RGG box, the polyadenylated end of mRNA processing intermediates
another RNA-binding motif found in hnRNP proteins, con- in hnRNA are retained in the mature mRNA in the cyto-
tains five Arg-Gly-Gly (RGG} repeats with several interspersed plasm. The solution to this apparent conundrum came from
aromatic amino acids. Although the structure of this motif the discovery of introns by electron microscopy of RNA-
has not yet been determined, its arginine-rich nature is simi- DNA hybrids of adenovirus DNA and the mRNA encoding
lar to the RNA-oinding domains of the HIV Tat protein. KH hexon, a major virion capsid protein (Figure 8-6). Other
domains and RGG repeats are often interspersed in two or studies revealed nuclear viral RNAs that were colinear with
more sets in a single RNA-binding protein. the viral DNA (primary transcripts), and RNAs with one or
two of the introns removed (processing intermediates). These
Splicing Occurs at Short, Conserved results, together with the earlier findings that the 5' cap and
3' poly(A) tai l at each end of long mRNA precursors are re-
Sequences in Pre-mRNAs via Two tained in shorter mature cytoplasmic mRNAs, led to the re-
Transesterification Reactions alization that introns are removed from primary transcripts
During formation of a mature, functional mRNA, the introns as exons are spliced together.
are removed and exons are spliced together. For short tran- The location of splzce sites-that is, exon-inrron junc-
scription units, RNA splicing usua lly follows cleavage and tions-in a pre-mRNA can be determined by comparing the
polyadenylation of the 3' end of the primary transcript, as sequence of genomic DNA with that of the eDNA prepared
depicted in Figure 8-2. However, for lo ng transcription units from the corresponding mRNA (see Figure 5-15). Sequences
containing multiple exons, splicing of exons in the nascent that are present in the genomic DNA but absent from the
RNA begins before transcription of the gene is complete. eDNA represent introns ;:~nd indicate the positions of splice
Early pioneenng research on the nuclear processing of sites. Such analysis of a large number of different mRNAs
mRNAs revealed that mRNAs are initially transcribed as revealed moderately conserved, short consensus sequences at
much longer RNA molecules than the mature mRNAs in the the splice sites flanking inrrons in eukaryotic pre-mRNAs; a
cytoplasm. It was also shown that RNA sequences near the pyrimidine-rich region just upstream of the 3' splice site also
5' cap added shortly after transcription initiation are re- is common (Figure 8-7). Studies of mutant genes with dele-
tained in the mature mRNA, and that RNA sequences near tions introduced into introns have shown that much of the

8.1 Processing of Eukaryotic Pre-mRNA 351


G) PODCAST: Discovery of lntrons

EXPERIMENTAL F GURE 8-6 Electron (a) Adenovirus hexon gene


microscopy of mRNA-template DNA hybrids shows
that introns are spliced out during pre-mRNA
processing. (a) Diagram of the EcoRI A fragment of 5' ====1
1~A IIT
s JI
E=~c~==:J-=---#3'
adenovirus DNA, which extends from the left end of ~------------------EcoRIA------------------~
f--i
the genome to just before the end of the final exon
Exons 0 lntrons lkb
of the hexon gene. The gene consists of three short
(b)
exons and one long (- 3.5 kb) exon separated by
three introns of 1, 2.5, and 9 kb. (b) Electron
micrograph (left) and schematic drawing (right) of a
hybrid between an EcoRI A DNA fragment and a
hexon mRNA. The loops marked A, B, and C corre-
spond to the introns indicated in (a). Since these
intron sequences in the viral genomic DNA are not
present in the mature hexon mRNA, they loop out
between the exon sequences that hybridize to their
complementary sequences in the mRNA. [Micrograph
from S.M. Berget et al., 1977, Proc. Not'/. Acad. Sci. USA
74:3171; courtesy of P. A. Sharp.]

\
mRNA

center portion of introns can be removed without affecting net result of these two reactions is that two exons are ligated
splicing; generally only 30-40 nucleotides at each end of an and the intervening intron is released as a branched lariat
1ntron are necessary for splicing to occur at normal rates. structure.
Analysis of the intermediates formed during splicing of
pre-mRNAs in vitro led to the discovery that splicing of exons
proceeds via two sequential transesterification reactions (Fig-
During Splicing, snRNAs Base-Pair
ure 8-8). lntrons are removed as a lariat structure in which the
5' G of the intron is joined in an unusual2' ,5 '-phosphodiester with Pre-mRNA
bond to an adenosine ncar the 3' end of the intron. This A Splicing requires the presence of small n uclear RNAs
residue is called the branch point A because it forms an RNA (snRNAs), important for base pairing with the pre-mRNA,
branch in the lariat structure. In each transestcrification re- and - 170 associated proteins. Five U-rich snRNAs, desig-
action, one phosphoester bond is exchanged for another. nated U1, U2, U4, US, and U6, participate in pre-mRNA
Since the number of phosphoester bonds in the molecule is splicing. Ranging in length from 107-210 nucleotides, these
not changed in either reaction, no energy is consumed. The snRNAs are associated with 6- 10 proteins each in the many

5' splice site Branch point 3' splice site

Pre-mRNA

Frequency of 70 60 80 100 100 95 70 80 45 80 90 80 100 80 80 100 100 60


occurrence (%) 1+---------- 20- 50 b ----------~

FIGURE 8 -7 Consensus se quences around splice sites in adenosine, also invariant, usually is 20-50 bases from the 3' splice site.
vertebrate pre-mRNAs. The only nearly invariant bases are the 5' GU The central region of the intron, which may range from 40 bases to
and the 3 ' AG of the intron (blue), although the flanking bases 50 kilo bases in length, generally is unnecessary for splicing to occur.
indicated are found at frequencies higher than expected based on a [SeeR. A. Padgett et al., 1986, Ann. Rev. Biochem. 55:1119, and E. B. Keller and
random distribution. A pyrimidine-rich region (hatch marked) near the W. A. Noon, 1984, Proc. Not'/. Acad. Sci. USA 81 :7417.]
3' end of the intron is found in most cases. The branch-point

352 CHAPTER 8 Post-transcriptional Gene Control


lntron (Figure 8-9b). Involvement of U2 snRNA in splicing initially
was suspected when it was found to have an internal se-
( 2' A
quence that is largely complementary to the consensus se-
I quence flanking the branch point in pre-mRNAs (see Figure
5'0 H- 3' 8-7). Compensating mutation experiments, similar to those
1/
O=P-o- -o-P=O
I conducted with Ul snRNA and 5' splice sites, demonstrated
1 I that base pairing between U2 snRNA and the branch-point
5'- - - -0 0 - Exon 2 - 3 ' sequence in pre-mRNA also is critical to splicing.
- - - - 3' 5'
Figure 8-9a illu~trates the general structures of the U1 and
0 = 3' oxygen of
exon 1
l First transesterification
U2 snRNAs and how they base-pair with pre-mRNA during
splicing. Significantly, the branch-point A itself, which is not
0 = 2' oxygen of base-paired to U2 snRNA, "bulges out" (Figure 8-lOa), al-
branch point A lowing its 2' hydroxyl to participate in the first transesterifi-
I. =3' oxygen of cation reaction of RNA splicing (see Figure 8-8).
intron Similar studies with other snRNAs demonstrated that
5'0 ba se pairing between them also occurs during splicing.
I 2> A Moreover, rearrangements in these RNA-RNA interactions
O=P-0 are critical in the splicing pathway, as we describe next.
I
0 3'
f
-o- P=O Spliceosomes, Assembled from snRNPs
. ~I
5'- - - -0 -H 0- - 3' and a Pre-mRNA, Carry Out Splicing
3' 5'
The five splicing snRNPs and other proteins involved in splicing
l Second transesterification
assemble on a pre-mRNA, forming a large ribonucleoprotein
complex called a spliceosome (Figure 8-11 ). '.fhe spliceosome
has a mass similar to that of a ribosome. Assembly of a spliceo-
some begins with the base pairing of the Ul snRNA to the
0 5' splice site and the cooperative binding of protein SFl (splic-
I
+ 5'- - - -0 - P - 0 - io--3' ing factor l ) to the branch point A and the heterodimeric pro-
5'0 11
tein U2AF (U2 associated factor) to the pyrimidine tract and
I 2> A 0
O=P- 0 Spliced exons the 3' AG of the intron via its large and small subunits, respec-
I 3' tively. The U2 snRNP then base-pairs with the branch point
o- OH
region (Figure 8-9a) as Sfl is released. Extensive base pairing
Excised lariat intron
between the snRNAs in the U4 and U6 snRNPs forms a
FIGURE 8-8 Two transesterification reactions that result in complex that associates with U5 snRNP. The U4/U6/U5
splicing of exons in pre-mRNA. In the first reaction, the ester bond "tri-snRNP" then associates with the previously formed U1/
between the 5' pho51Jhorus of the intron and the 3' oxygen (dark red)
U2/pre-mRNA complex to generate a spliceosome.
of exon 1 is exchanged for an ester bond with the 2' oxygen (blue) of the
After formation of the spliceosome, extensive rearrange-
branch-site A residue. In the second reaction, the ester bond between
ments in the pairing of snRNAs and the pre-mRNA lead to
the 5' phosphorus of exon 2 and the 3' oxygen (orange) of the intron is
exchanged for an ester bond with the 3' oxygen of exon 1, releasing the
release of the U1 snRNP. Figure 8-10b shows a cryoelectron-
intron as a lariat structure and joining the two exons. Arrows show where
microscopy structure of this intermediate in the splicing pro-
activated hydroxyl oxygens react with phosphorus atoms. cess. A further rearrangement of spliceosomal components
occurs with the loss of U4 snRNP. This generates a complex
that catalyzes the first transesterification reaction that forms
small nuclear ribonucleoprotein particles (s nRNPs) in the the 2' ,5 '-phosphodiester bond between the 2' hydroxyl on the
nuclei of eukaryotic cells. branch point A and the phosphate at the 5' end of the intron
Definitive evidence for the role of Ul snRNA in splicing (Figure 8-9). Following another rearrangement of the snRNPs,
came from experiments indicating that base pairing between the second transesterification reaction ligates the two exons in
the 5' splice site of a pre-mRNA and the 5' region ofUl snRNA a standard 3' ,5' -phosphodiester bond, releasing the intron as
is required for RNA splicing (Figure 8-9a). In vitro experiments a lariat structure associated with the snRNPs. This final intron-
showed that a synthetic oligonucleotide that hybridizes with snRNP complex rapidly dissociates, and the individual snRNPs
the 5' -end region of U 1 snRNA blocks RNA splicing. In vivo released can participate in a new cycle of splicing. The excised
experiments showed that base-pairing-disrupting mutations intron is then rapidly degraded by a debranching enzyme and
in the pre-mRNA 5' splice site also block RNA splicing; in other nuclear RNases discussed later.
this case, however, splicing can be restored by expression of As mentioned above, a spliceosome is roughly the sile of a
a mutant Ul snRNA with a compensating mutation that re- ribosome and is composed of - 170 proteins, including -100
stores base pairing to the mutant pre-mRNA 5' splice site "splicing factors" in addition ro the proteins associated with the

8.1 Processing of Eukaryotic Pre-mRNA 353


~ FOCUS ANIMATION: mRNA Splicing

(a)

~Ji~1snRNA
l
l
U2 snRNA
Sm
3' --

cap 5 '
Sm
3' - Gl - _ ca 5'
-----l.JJ. IIIIII P I I II II Py
5 ' ~UAAGU-- ----------UACUAC CAG G Exon 2 <-- 3'
PremRNA A
'-...._B ranch point

(b)
W.t. U1 snRNA 3 ' 1 - . cap 5' Mutant U1 snRNA 3 ' ( _ _ . . . U." cap 5'

Mutant premRNA 5'~~liA~--- - 3' Mutant premRNA


-----l.JJ.ll I I I I
5' ~ U AAAU - 3'

Mutation in pre-m RNA 5' splice site Compensatory mutat ion in U 1


blocks splicing restores splicing
FIGURE 8 -9 Base pairing between pre-mRNA, U1 snRNA, and U2 characterizing components of the splicing reaction."(b) Only t he 5' ends
snRNA early in the splicing process. (a) In this diagram, secondary of U 1 snRNAs and 5' splice sites in pre-mRNAs are shown. (Left) A
structures in the snRNAs that are not altered during splicing are mutation (A) in a pre-mRNA splice site that interferes with base pairing
depicted schematically. The yeast branch-point sequence is shown to the 5' end of Ul snRNA blocks splicing. (Right) Expression of a Ul
here. Note that U2 snRNA base-pa irs w ith a sequence that includes the snRNA w ith a compensating mutation (U) that restores base pairing
branch-point A, although this residue is not base-paired. The purple also restores splicing of the mutant pre-mRNA. [Part (a) adapted from
rectangles represent sequences that bind snRNP proteins recognized M. J. Moore et al., 1993, in R. Gesteland and J. Atkins, eds., The RNA World, Cold
by anti-Sm antibodies. For unknown reasons, antisera from patients Spring Harbor Press, pp. 303-357. Part (b) see Y. Zhuang and A.M. Weiner, 1986,
with the autoimmune disease systemic lup us eryt hematosus (SLE) Ce// 46:827.]
contain antibodies to snRNP p roteins, w h ich have been useful in

t,;) VIDEO: Dynamic Nature of Pre-mRNA Splicing Factor Movement in Living Cells

FIGURE 8-10 Structures of a bulged A in an RNA RNA helix and (a) Self-complementary (c) Spliceosome structure
an intermediate in the splicing process. (a) The structure of an RNA sequence w ith bulging A
duplex w ith the sequence shown, cont aining bulged A residues (red) at
5'UACUA ACGU AGUA
position 5 in the RNA helix, was determined by x-ray crystallography.
AUGA UGCA UCAU 5'
(b) The bulged A residues extend from the side of an A-form RNA-RNA A
helix. The phosphate backbone of one strand is shown in green; the
other strand in blue. The structure on th e right is turned 90 degrees for (b) X-ray crystallog ra phy
a view down the axis o f the helix. (c) 40 A reso lution structure of a structure
spliceosomal splicing intermediate containing U2, U4, US, and U6 37oA
snRNPs, determined by cryoelectron microscopy and image recon-
struction. The U4/U6/US tri-snRNP complex has a similar structure to
th~ triangu lar body of this complex, suggesting that t hese snRNPs are
at the bottom o f the structure shown here and t hat the head is
composed largely of U2 snRNP. [Parts (a) and (b) from J. A. Berglund et al., As As
2001, RNA 7:682. Part (c) from D. Boehringer et al., 2004, Nor. Srruct. Mol. Bioi. (top) (bottom)
11 :463. See also H. Stark and R. Luhrmann, 2006, Annu. Rev. Biophys. Biomol.
Srruct. 35:435.]

354 CHAPTER 8 Post-transcriptional Gene Control


0 FOCUS ANIMATION: mRNA Splicing
FIGURE 8-11 Model of spliceosome-mediated splicing of SF1 U2AF
pre-mRNA. Step 0 : After U1 base pairs with the consensus S'splice U1
site, SF1 (splicing factor 1) binds the branch point A; U2AF (U2 snRNP 5- pG - "A Yn.rJ.G 3

associated factor) associates with the polypyrimidine tract and 3'splice


site; and the U2 snRNP associates with the branch point A via base-
pairing interactions shown in Figure 8-9, displacing SF1 . Step IFJ: A D~ SFl
trim eric snRNP complex of U4, US, and U6 joins the initial complex
to form the spliccosome. Step ~ : Rearrang~ments of base-pa1rmg U1
interactions between snRNAs converts the spliceosome into a 5- pG ___. - A p 3
catalytically active conformation and destabilizes the U1 and U4 U2
snRNPs, which are released. Step~: The catalytic core, thought to
be formed by U6 and U2, then catalyzes the first transesterification fJ ~ U4/ U6/ U5
reaction, forming the intermediate containing a 2'.5'-phosphodiester
bond as shown in Figure 8-8. Step I,'J: Following further rearrangements
between the snRNPs, the second transesterification reaction joins the
two exons by a standard 3 ',S'-phosphodiester bond and releases the U1 \ A
I U2
intron as a lariat structure and the remaining snRNPs. Step r;J: The U6
"-..:.:p 3'
excised lariat intron is converted into a linear RNA by a debranching
enzyme. [Adapted from T. Villa et al .. 2002, Ce// 109:149.] U5 Spliceosome

five snRNPs. This makes RNA splicing comparable in complex-


~~~ Ul, U4
ity to initiation of transcription and protein synthesis. Some of
the splicing factors are associated with snRNPs, but others are ~
not. For instance, the 65-kD subunit of the U2-associated factor U6 A./
OH
(U2AF) binds to the pyrimidine-rich region near the 3' end of G/ ) .' U2
s P;.__/
introns and to the U2 snRNP. The 35-kD subunit of U2AF U5 P 3'
binds to the AG dinucleotide at the 3' end of the intron and also -
interacts with the larger U2AF subunit bound nearby. These two
U2AF subunits act together with SF1 to help specify the 3' splice
site by promoting interaction of U2 snRNP with the branch
IJ 1 First transesterification

G
point (see Figure 8-11, step 0 ). Some splicing factors also ex-
hibit sequence homologies to known RNA helicases; these are
probably necessary for the base-pairing rearrangements that
U6 (
occur in snRNAs during the spliceosomal splicing cycle.
. Following RNA splicing, a specific set of hnRNP proteins --o~P 3
remain bound to the spliced RNA approximately 20 nucle- s U5
otides 5' to each exon-exon junction, thus forming an exon-
junction complex. One of the hnRNP proteins associated with
the exon-junction complex is the RNA export factor (REF),
II 1 Second transesterification

which functions in the export of fully processed mRNPs from


the nucleus to the cytoplaslll, as discussed in Section 8.3. Other + U2, U5, U6
proteins associated with the exon-junction complex function Spliced exons
in a quality-control mechanism that leads to the degradation of
improperly spliced mRNAs, known as nonsense-mediated
decay (Section 8.4 ).
A small fraction of pre-mRNAs (< 1% in humans) con-
tain introns whose splice sites do not conform to the standard 5 pG- - - -A - OH3
consensus sequence. This class of introns begins with AU and Linear intron RNA
ends with AC rather than following the usual "GU-AG rule"
(see Figure 8-7). Splicmg ot this special class of introns occurs
via a splicing cycle analogous to that shown in Figure 8-11, sponding pre-mRNA by removal of internal intron s and
except that four novel, low-abundance snRNPs, together spl icing of exons. However, in two types of protozoans-
with the standard US snRNP, are involved. trypanosomes and euglenoids-mRNAs are constructed by
Nea rly all functional mRNAs in vertebrate, insect, and splicing together separate RNA molecules. This process, re-
plant cells are derived from a single molecule of the corre- ferred to as trans-splicing, is also used in the synthesis of

8.1 Processing of Eukaryotic Pre -mRNA 355


RNA polymerase II

~
~. Linker CTD

I< 28 nm 65 nm --------------+1
FIGURE 812 Schematic diagram of RNA polymerase II with the polymerase. The CTD of mammalian RNA polymerase II is twice as long.
CTD ext ended. The length of the fully extended yeast RNA polymerase In its extended form, the CTD can associate with mu ltiple RNA-processing
II carboxyl-terminal domain (CTD) and the linker region that connects it factors simultaneously. [From P. Cramer, D. A. Bushnell, and R. D. Kornberg,
to the polymerase is shown relative to the globular domain of the 2001, Science 292:1863.)

10-15 percent of the mRNAs in the nematode (roundworm) (-3500 bases). The longest introns contain upward of 500 kb!
Caenorhabditis elegans, an important model organism for Because the sequences of 5' and 3' splice sites and branch
studying embryonic development. Trans-splicing is carried po.ints are so degenerate, multiple copies are likely ro occur
out by snRNPs by a process similar to the splicing of exons random ly in long introns. Consequently, additional sequence
m a single pre-mRNA. information is required to define the exons that should be
spliced together in higher organisms with long introns.
Chain Elongation by RNA Polymerase II The information for defining the splice sites that demar-
cate exons is encoded within the sequences of the exons. A
Is Coupled to the Presence of RNA- family of RNA-binding proteins, the SR proteins, interact
Processing Factors with sequences within exons called exonic spJicing enhancers.
I fow is RNA processing efficiently coupled with the transcrip- SR proteins are a subset of the hnRNP proteins discussed
tion of a pre-mRNA? The key lies in the long carboxyl-terminal earlier and so contain one or more RRM RNA-binding do-
domain (CTD) of RNA polymerase II, which, as discussed in mains. They also contain several protein-protein interaction
Chapter 7, is composed of multiple repeats of a seven-residue domains rich in arginine (R ) and serine (S) residues called RS
(heptapeptide) sequence. When fully extended, the CTD do- domains. When bound to exonic splicing enhancers, SR pro-
main in the yeast enzyme is about 65 nm long (Figure 8-12); teins mediate the cooperative binding of Ul snRNP to a true
the CTD in human RNA polymerase II is about twice as long. 5' splice site and U2 snRNP to a branch point through a net-
The remarkable length of the CTD apparently allows multiple work of protein-protein interactions that span an exon (Fig- _I
proteins to associate simultaneously with a single RNA ure 8-13). The complex of SR proteins, snRNPs, and other
polymerase II molecule. For instance, as mentioned earlier, the splicing factors (e.g., U2AF) that assemble across an exon,
enzymes that add the 5' cap to nascent transcripts associate which has been called a cross-exon recognition complex, per-
with the CTD phosphorylated on multiple serines at the fifth mits precise specification of exons in long pre-mRNAs.
position in the heptapeptide repeat (Ser-5) during or shortly
after transcription initiation by a subunit of TFIIH. In addi- In the transcription units of higher organisms with
tion, RNA splicing and polyadenylation factors have been long introns, exons not only encode the amino acid
found to associate with the phosphorylated CTD. As a conse- sequences of different portions of a protein but also contain
quence, these processing factors are present at high local con- binding sites for SR proteins. Mutations that interfere with
centrations when splice sites and poly( A) signals are transcribed the binding of an SR protein to an exonic splicing enhancer,
hy the polymerase, enhancing the rate and specificity of RNA even if they do not change the encoded amino acid sequence,
processing. In a reciprocal fashion, the association of hnRNP prevent formation of the cross-exon recognition complex. As
proteins with the nascent RNA enhances the interaction of a result, the affected exon is "skipped" during splicing and is
RNA polymerase II with elongation factors such as DSIF and not included in the final processed mRNA. The truncated
CDK9-cyclin T (P-TEFb) (Figure 7-20), increasing the rate of mRNA produced in this case is either degraded or translated
transcription. As a consequence, the rate of transcription is into a mutant, abnormally functioning protein. This type of
coordinated with the rate of nascent RNA association with mutation occurs in some human genetic diseases. For exam-
hnRNPs and RNA-processing factors. This mechanism may ple, spinal muscular atrophy is one of the most common ge-
ensure that a pre-mRNA is not synthesized unless the machin- netic causes of childhood mortality. This disease results from
ery for processing it is properly positioned. mutations in a region of [he genumt: cumaining two closely
related genes, SMNJ and SMN2, that arose by gene duplica-
tion. SMN2 encodes a protein identical with SMNJ. SMN2
SR Proteins Contribute to Exon Definition
is expressed at a much lower level because a silent mutation
in Long Pre-mRNAs in one exon interferes with the binding of an SR protein.
The average length of an exon in the human genome is - 150 This leads to exon skipping in most of the SMN2 mRNAs.
bases, whereas the average length of an intron is much longer The homologous SMN gene in the mouse, where there is

356 CHAPTER 8 Post-transcriptional Gene Control


Spliceosome

U2 U1

~---11"-
-
1 \
- -.l...
Jr
U
2
U2AF65
A ..-yyyy. AG
<.....:f
3~ Iu GU
1

__;L.._
7r-
3'

sranch point 3' splice Site ESE s splice site

Cross-exon recognition complex Cross-exon recognition complex


FIGURE 8-13 Exon recognition through cooperative binding of spans an exon and activates the correct splice sites for RNA splicing.
SR proteins and splicing factors to pre-mRNA. The correct 5' GU and Note that the Ul and U2 snRNPs in this unit do not become part of the
3' AG splice sites are recognized by splicing factors on the basis oftheir same spliceosome. The U2 snRNP on the right forms a spliceosome
proximity to exons. The exons contain exonic splicing enhancers (ESEs) with the Ul snRNP bound to the 5' end of the same intron. The Ul
that are binding sites for SR proteins. When bound to ESEs, the SR snRNP shown on the right forms a spliceosome with the U2 snRNP
proteins interact with one another and promote the cooperative bound to the branch point of the downstream intron (not shown), and
binding of the Ul snRNP to the 5' splice site of the downstream intron, the U2 snRNP on the left forms a spliceosome with a Ul snRNP bound
SFl and then the U2 snRNP to the branch point of the upstream intron, to the 5' splice site of the upstream intron (not shown). Double-headed
the 65- and 35-kD subunits of U2AF to the pyrimidine-rich region and arrows indicate protein-protein interactions. [Adapted from T. Maniatis,
AG 3' splice site ofthe upstream intron, and other splicing factors (not 2002, Nature 41 8:236; see also S.M. Berget, 1995, J. Bioi. Chern. 270:2411.)
shown). The resulting RNA-protein cross-exon recognition complex

only a single copy, is essential for cell viability. Spinal mus- fungi. Discovery of the catalytic activity of self-splicing in-
cular atrophy in huma ns resu lts from homozygous muta- trans revolutionized concepts about the functions of RNA.
tions that inactivate SMNl. The low level of protein translated As discussed in Chapter 4, RNA is now kn.own to catalyze
from the small fraction of SMN2 mRNAs that are correctly peptide-bond formation during protein synthesis in ribo-
spliced is sufficient to maintain cell viability during embryo- somes. Here we discuss the probable role of group II introns,
genesis and fetal development, but it is not sufficient to now found only in mitochondrial and chloroplast DNA, in
maintain viability of spi nal cord motor neurons in childhood, the evolution of snRNAs; the functioning of group I introns
resulting in their death and the associated disease. is considered in the later section on rRNA processing.
Approximately 15 percent of the single-base-pair muta- Even though their precise sequences are not highly con-
tions that cause human genetic diseases interfere with proper served, all group II introns fo ld into a conserved, complex
exon definition. Some of these mutations occur in 5' or 3' secondary structure containing numerous stem loops (Fig-
splice sites, often resu lting in the use of nearby alternative ure 8-14a). Self-splicing by a group II intron occurs via two
"cryptic" splice sites present in the normal gene sequence. ln transesterification reactions, involving intermediates and
the absence of the normal splice site, the cross-exon recogni- products analogous to those found in nuclear pre-mRNA
tion complex recognizes these alternative sites . Other muta- splicing. The mechanistic similarities between group II intron
tions that cause abnormal splicing result in a new consensus self-splicing and spliceosomal splicing led to the hypothesis
splice-site sequence that becomes recognized in place of the that snRNAs function analogously to the stem loops in the
normal splice site. Finally, some mutations can interfere with secondary structure of group II imrons. According to this hy-
the binding of specific SR proteins to pre-mRNAs. These pothesis, snRNAs interact with 5' and 3' splice sites of pre-
mutations inhibit splicing at normal splice sites, as in the case mRNAs and with each other to produce a three-dimensional
of the SMN2 gene, and thus lead to exon skipping. RNA structure functionally analogous to that of group II self-
splicing introns (Figure 8-14b).
An extension of this hypothesis is that introns in ancient
pre-mRNAs evolved from group II self-splicing introns
Self-Splicing Group II lntrons Provide
through the progressive loss of internal RNA structures,
Clues to the Evolution of snRNAs which concurrently evolved into trans-acting snRNAs that
Under certain nonphysio logical in vitro conditions, pure perform the same functions. Support for this type of evolu-
preparations of some RNA transcripts slowly splice out in- tionary model comes from experiments with group I! intron
trans in the absence of any protein. This observation led to mutants in which domain V and part of domain I are deleted.
the recognition that some introns are self-splicing. Two types Rl'\A transcripts containing such mutant introns are defective
of self-splicing introns have been discovered: group I introns, in self-splicing, but when RNA molecules equivalent to the
present in nuclear rRNA genes of protozoans, and group IT deleted regions are added to the in vitro reaction, self-splicing
introns, present in protein-coding genes and some rRNA and occurs. This finding demonstrates that these domains in group
tRNA genes in mitochondria and chloroplasts of plants and II introns can be trans-acting, like snRNAs.

8.1 Processing of Eukaryotic Pre-mRNA 357


(a) Group II intron (b) U sn RNAs in spliceosome processing that involves cleavage but not polyadenylation.
Specialized RNA-binding proteins that help to regulate his-
tone mRNA translation bind to the 3' end generated by this
specialized system.) Early studies of pulse-labeled adenovirus
and SV40 RNA demonstrated that the viral primary tran-
scripts extend beyond the site from which the poly(A) tail
extends. These results suggested that A residues are added to
a 3' hydroxyl generated by endonucleolytic cleavage of a
longer transcript, but the predicted downstream RNA frag-
5' 3'
- Pre-mRNA
ments never were detected in vivo, presumably because of
intron their rapid degradation. However, detection of both pre-
FIGURE 8-14 Comparison of group II self-splicing introns and
dicted cleavage products was observed in in vitro processing
the spliceosome. The schematic diagrams compare the secondary reactions performed with nuclear extracts of cultured human
structures of (a) group II self-splicing introns and (b) U snRNAs present cells. The cleavage/polyadenylation process and degradation
in the spliceosome. The first transesterification reaction is indicated by of the RNA downstream of the cleavage site occurs much
light green arrows; the second reaction, by blue arrows. The branch- more slowly in these in vitro reactions, simplifying detection
point A is boldfaced. The similarity in these structures suggests that the of the downstream cleavage prodl1ct.
spliceosomal snRNAs evolved from group II introns, with the trans-acting Early sequencing of eDNA clones from animal cells showed
snRNAs being functionally analogous to the corresponding domains in that nearly all mRNAs contain the sequence AAUAAA 10-35
group II introns. The colored bars flanking the introns in (a) and nucleotides upstream from the poly(A) tail (Figure 8-15). Poly-
(b) represent exons. [Adapted from P. A. Sharp, 1991, Science 254:663.] adenylation of RNA transcripts is virtually eliminated when
the corresponding sequence in the template DNA is mutated to
any other sequence except one encoding a closely related se-
The similarity in the mechanisms of group II intron self- quence (AUUAAA). The unprocessed RNA transcripts pro-
splteing and spliceosomal splicing of pre-mRNAs also sug- duced from such mutant templates do not accumulate in nuclei
gests that the splicing reaction is catalyzed by the snRNA, not but are rapidly degraded. Further mutagenesis studies revealed
the protein, components of spliceosomes. Although group II that a second signal downstream from the cleavage site is re-
introns can self-splice in vitro at elevated temperatures and quired for efficient cleavage and polyadenylation of most pre-
Mg2 concentrations, under in vivo conditions proteins called mRNAs in animal cells. This downstream signal is not a
maturases, which bind to group II intron RNA, are required specific sequence but rather a GU-rich or simply a U-rich re-
for rapid splicing. Maturases are thought to stabilize the pre- gion within - 50 nucleotides of the cleavage site.
cise three-dimensional interactions of the intron RNA re- Identification and purification of the proteins required for
quired to catalyze the two splicing transesterification reactions. cleavage and polyadenylation of pre-mRNA have led to the
By analogy, snRNP proteins in spliceosomes are thought to model shown in Figure 8-15. A 360-kDa cleavage and poly-
stabilize the precise geometry of snRNAs and intron nucle- adenylation specificity factor (CPSF), composed of five differ-
otides required to catalyze pre-mRNA splicing. ent polypeptides, first forms an unstable complex with the
The evolution of snRNAs may have been an important upstream AAUAAA poly(A) signal. Then at least three addi-
step in the rapid evolution of higher eukaryotes. As internal tional proteins bind to the CPSF-RNA complex: a 200-kDa
intron sequences were lost and their functions in RNA splicing heterotrimer called cleavage stimulatory factor (CStF), which
supplanted by trans-acting snRNAs, the remaining intron se- interacts with the G/U-rich sequence; a 150-kDa hetero-
quences would be free to diverge. This in turn likely facilitated tetramer called cleavage factor I (CFI); and a second het-
the evolution of new genes through exon shuffling since there erodimeric cleavage factor (CFII). An -150-kDa protein
are few constraints on the sequence of new introns generated called symplekin is thought to form a scaffold on which these
in the process (see Figures 6-18 and 6-19). It also permitted the cleavage/polyadenylation factors assemble. Finally, poly(A)
increase in protein diversity that results from alternative RNA polymerase (PAP) binds to the complex before cleavage can
splicing and an additional level of gene control resulting from occur. This requirement for PAP binding links cleavage and
regulated RNA splicing. polyadenylation, so that the free 3' end generated is rapidly
polyadenylated and no essential information is lost to exonu-
clease degradation of an unprotected 3' end.
Assembly of the large, multiprotein cleavage/polyadenyla-
3' Cleavage and Polyadenylation
tion complex around the AU-rid1 poly(A) signal in a pre-mRNA
of Pre-mRNAs Are Tightly Coupled IS analogous in many ways to formation of the transcription-
In eukaryotic cells, all mRNAs, except histone mRNAs, have preinitiation complex at the AT-rich TATA box of a template
a 3' poly(A) tail. (The major histone mRNAs are transcribed DNA molecule (see Figure 7-16). In both cases, multiprotein
from repeated genes at prodigious levels in replicating cells complexes assemble cooperatively through a network of specific
during the S phase. They undergo a special form of 3'-end protein-nucleic acid and protein-protein interactions.

358 CHAPTER 8 Post-transcriptional Gene Control


Poly(A) Poly(A) FIGURE 8-15 Model for cleavage and polyadenylat ion o f
signal Poly(A) site signal pre-mRNAs in mammalian cells. Cleavage and polyadenylation

5' e>o-----!I~~~U~AAA~I--*L--~ 3' specificity factor (CPSF) binds to the upstream AAUAAA poly(A) signal.
CStF interacts with a downstream GU- or U-rich sequence and with
CPSF, CStF, CFI, CFII i p,~mRNA bound CPSF, forming a loop in the RNA; binding of CFI and CFII helps
stabilize the complex. Binding of poly(A) polymerase (PAP) then
stimulates cleavage at a poly(A) site, which usually is 10-35 nucleotides
3' of the upstream poly(A) signal. The cleavage factors are released, as
CPSF is the downstream RNA cleavage product, which is rapidly degraded.
Bound PAP then adds - 12 A residues at a slow rate to the 3'-hydroxyl
group generated b y the cleavage reaction. Binding of poly(A)-binding
protein II (PABPII) to the initial short poly(A) tail accelerates the rate of
addition by PAP. After 200-250 A residues have been added, PABPII
signals PAP to stop polym erization.

Following cleavage at the poly(A) site, polyadenylation pro-


ceeds in two phases. Add ition of the first 12 or so A residues
5'
occurs slowly, followed by rapid addition of up to 200-250
more A residues. The rapid phase requires the binding of mul-
tiple copies of a poly(A)-binding protein containing the RRM
motif. This protein is designated PABPII to distinguish it from

1
3'
the poly(A)-bind ing protein present in the cytoplasm. PABPII
c....... binds to the short A tail initially added by PAP, stimulating the
rate of polymerization of additional A residues by PAP, result-
ing in the fast phase of polyadenylation (Figur.e 8-15). PABPII is
also responsible for signaling poly(A) polymerase to terminate
polymerization when the poly(A) tail reaches a length of 200-
250 residues, although the mechanism for controlling the length
of the ta il is not yet understood. Bi nding of PABPII to the
poly(A) tail is essential for mRNA export into the cytoplasm.

p~
J' + ATP pp.
Slow
polyadenylation Nuclear Exonucleases Degrade RNA
+ CStF, CFI, CFII I That Is Processed Out of Pre-mRNAs
Because the h uman genome contains long introns, only - 5
percent of the nucleotides that are polymerized by RNA
polymerase II during transcription are retained in mature,
5' A
processed mRNAs. Although this process appears ineffi-
cient, it probably evolved in mu lticellular organisms because
PABPIIi the process of exon shuffling facilitated the evolution of new
genes in organisms with long introns (Chapter 6). The in-
trons that are spliced out and the region downstream from
the cleavage and polyadenylation site are degraded by nu-
5' clear exoribonucleases that hydrolyze one base at a time
from either the 5 ' or 3' end of an RNA strand .
As mentioned earlier, the 2',5'-phosphodiester bond in
PABPIIF ATP Rapid excised introns is hydrolyzed by a debranching enzyme,
polyadenylation
PP; yielding a linear molecule with unprotected ends that can be
attacked by exonucleases (see Figure 8-11 ). The predomi-
nant nuclear decay pathway is 3'-4S' hydrolysis by 11 exo-
nucleases that associate with one another in a large protein
5' H 3' complex called the exosome. O ther proteins in the complex
include RNA helicases that disr upt base pairing and RNA-
protein interactions that wou ld o therwise impede the exonu-
cleases. Exosomes also function in the cytoplasm, as discussed

8.1 Processi ng of Eukaryotic Pre-mRNA 359


later. In addition to introns, the exosome appears to degrade
pre-mRNAs that have not been properly spliced or poly- come associated with the CTD, where they are poised to inter-
adenylated. It is not yet clear how the exosome recognizes act with the nascent prc-mRNA as it emerges from the surface
improperly processed pre-mRNAs. But in yeast cells with of the polymerase.
temperature-sensitive mutant poly(A) polymerase (Figure Five different snRNPs interact via base pairing with one
8-15), pre-mRNAs are retained at their sites of transcription another and with pre-mRNA to form the spliceosome (see
in the nucleus at the nonpermissive temperature. These ab- Figure 8-11 ). This very large ribonucleoprotein complex
normally processed pre-mRNAs are released in cells with a catalyzes two transesterification reactions that join two ex-
second mutation in a subunit uf the exosome found only in ons and remove the intron as a lariat structure, wh ich is sub-
nuclear and not in cytoplasmic exosomes (PM-Scl; 100 kD sequently degraded (see Figure 8-8).
in humans). Also, exosomes are found concentrated at sites
SR proteins that bind to exonic splicing enhancer sequences in
of transcription in Drosophila polytene chromosomes,
exons are critical in defining exons in the large pre-mRNAs of
where they are associated with RNA polymerase II elonga-
higher organisms. A network of interactions between SR pro- , '
tion factors. These results suggest that the exosome partici-
teins, snRNPs, and splicing factors forms a cross-exon recogni-
pates in an as yet poorly understood quality-control mechanism
tion complex that specifies correct splice sites (see Figure 8- 13 ).
that recognizes aberrantly processed pre-mRNAs, prevent-
ing their export to the cytoplasm and ultimately leading to The snRNAs in the spliccosorn.e are thought to have an
their degradation. overall tertiary structure similar to that of group II self-splicing
To avoid being degraded by nuclear exonucleases, nascent introns.
transcripts, pre-mRNA processing intermediates, and mature For long transcription units in higher organisms, splicing of
mRNAs in the nucleus must have their ends protected. As dis- exons usually begins as the pre-mRNA is still being formed.
cussed above, the 5' end of a nascent transcript is protected by Cleavage and polyadenylation to form the 3' end of the mRNA
addition of the 5' cap structure as soon as the 5' end emerges occur after the poly(A) site is transcribed.
from the polymerase. The 5' cap is protected because it is
In most protein-coding genes, a conserved AAUAAA poly(A)
bound by a nuclear cap-binding complex, which protects it
signal lies slightly upstream from a poly(A) site where cleavage
from 5' exonucleases and also functions in export of mRNA
and polyadenylation occur. A GU- or U-rich sequence down-
to the cytoplasm. The 3' end of a nascent transcript lies within
stream from the poly(A) site contributes to the efficiency of
the RNA polymerase and thus is inaccessible to exonucleases
cleavage and polyadenylation.
(see figure 4-12). As discussed previously, the free 3' end gen-
erated by cleavage of a pre-mRNA downstream from the A multiprotein complex that includes poly(A) polymerase
poly(A) signal is rapidly polyadenylated by the poly(A) (PAP) carries out the cleavage and polyadenylation of a pre-
polymerase associated with the other 3' processing factors, mRNA. A nuclear poly(A)-binding protein, PABPII, stimu-
and the resulting poly(A) rail is bound by PABPII (Figure lates addition of A residues by PAP and stops addition once
8-15). This tigh~ coupling of cleavage and polyadenylation the poly(A) tail reaches 200-250 residues (see Figure 8-15).
protects the 3' end from exonuclease attack. Excised introns and RNA downstream from the cleavage/
poly(A) site arc degraded primarily by exosomes, multipro-
tcin complexes that contain eleven 3 '-75' exonucleases as
well as RNA helicases. Exosomes also degrade improperly
KEY CONCEPTS of Section 8.1 processed pre-mRNAs.
Processing of Eukaryotic Pre-mRNA
In the nucleus of eukaryotic cells, pre-mRNAs are associated
with hnRNP proteins and processed by 5' capping, 3' cleavage
and polyadcnylation, and splicing before being transported to 8.2 Regulation of Pre-mRNA Processing
the cytoplasm (see Figure 8-2). Now that we've seen how pre-mRNAs are processed into ma-
Shortly after transcription initiation, capping enzymes asso- ture, functional mRNAs, we consider how regulation of this
ciated with the carboxyl-terminal domain (CTD) of RNA process can contribute to gene control. Recall from Chapter 6
polymerase II, which is phosphorylated multiple rimes at ser- that higher eukaryotes contain both simple and complex tran-
ine 5 of the heptapeptide repeat by TFIIH during transcription scription units encoded in their DNA. The primary transcripts
initiation, add the 5' cap to the nascent transcript. Other RNA- produced from the former contain one poly(A) site and exhibit
processing factors involved in RNA splicing, 3' cleavage, ami only one pattern of RNA splicing, even if multiple introm are
polyadenylation associate with the CTD when it is phosphory- present; thus simple transcription units encode a single mRNA.
lated at serine 2 of the hcptapeptidc repeat, increasing the rate In contrast, the primary transcripts produced from complex
of transcription elongation. Consequently, transcription does transcription units (-92-94 % of all human transcription
not proceed at a high rate until RNA-processing factors be- units) can be processed in alternative ways to yield different
mRNAs that encode distinct proteins (see Figure 6-3).

360 CHAPTER 8 Post-transcriptional Gene Control


Alternative Splicing Generates Transcripts A Cascade of Regulated RNA Splicing Controls
with Different Combinations of Exons Drosophila Sexual Differentiation
The discovery that a large fraction of transcription units in One of the earliest examples of regulated alternative splicing
higher organisms encode alternatively spliced mRNAs and of pre-mRNA came from studies of sexual differentiation in
that differently spliced mRNAs are expressed in different cell Drosophila. Genes required for normal Drosophila sexual dif-
types revealed that regulation of RNA splicing is an impor- ferentiation were first characterized by isolating Drosophila
tant gene-control mechanism in higher eukaryotes. Although mutants defective in the process. When the proteins encoded
many examples of cleavage at alternative poly(A) sites in pre- by the wild-type genes were characterized biochemically, two
mRNA!. an: known, alternative splicing of different exons is of them were found to regulate a cascade of alternative RNA
the more common mechanism for expressing different pro- splicing in Drosophila embryos. More recent research has
teins from one complex transcnption unit. In Chapter 4, for provided insight into how these proteins regulate RNA
example, we mentioned that fibroblasts produce one type of processing and ultimately lead to the creation of two different
the extracellular protein fibronectin, whereas hepatocytes sex-specific transcriptional repressors that suppress the devel-
produce another type. Both fibronectin isoforms are encoded opment of characteristics of the opposite sex.
by the same transcription unit, which is spliced differently in The Sxl protein, encoded by the sex-lethal gene, is the
... the two cell types to yield two different mRNAs (see Figure first protein to act in the cascade (Figure 8-16). The Sxl pro-
4-16). In other cases, alternative processing may occur simul- tein is present only in female embryos. Early in development,
taneously in the same cell type in response to different devel- the gene is transcribed from a promoter that functions only in
opmental or environmental signals. First we discuss one of female embryos. Later in development, this female-specific
the best-understood examples of regulated RNA processing promoter is shut off and another promoter for sex-lethal be-
and then we briefly consider the consequences of RNA splic- comes active in both male and female embryos. However, in
ing in the development of the nervous system. the absence of early Sxl protein, the sex-lethal pre-mRNA in

Pre-mRNAs mRNAs

<j> / -i 2 I 4 ~ -+ Sxl protein

'I!/
(a) sxl

' v / 'v /

------------------------
(b) tra 5~ ~
' v /

Rbp1 + Tca2'~' ---


Dsx protein
(c) dsx 4
Dsx protein

FIGURE 8 - 16 Cascade of regulated splicing that controls sex Rbpl and Tra2, activates splicing between exons 3 and 4 and cleavage/
determination in Drosophila embryos. For clarity, only the exons polyadenylation(An)at the 3' end of exon 4 in dsx pre-mRNA in female
(boxes) and introns (black lines) where regulated splicing occurs are embryos. In male embryos, which lack functional Tra, the SR proteins
shown. Splicing is indicated by red dashed lines above (female) and do not bind to exon 4, and consequently exon 3 is spliced to exon S.
blue dashed lines below (male) the pre-mRNAs. Vertical red lines in The distinct Dsx proteins produced in female and male embryos as the
exons indicate in-frame stop codons, which prevent synthesis of result of this cascade of regulated splicing repress transcription of
functional protein. Only female embryos produce functional Sxl genes required for sexual differentiation of the opposite sex. [Adapted
protein, which represses splicing between exons 2 and 3 in sxl pre- from M. J. Moore et al., 1993, in R. Gesteland and J. Atkins, eds., The RNA World,
mRNA (a) and between exons 1 and 2 in tra pre-mRNA (b). (c) In Cold Spring Harbor Press, pp. 303- 357.]
contrast, the cooperative binding ofTra protein and two SR proteins,

8.2 Regulation of Pre -mRNA Processing 361


male embryos is spliced to produce an mRNA that contains
a stop codon early in the sequence. The net result is that
male embryos produce no functional Sxl protein either earl}
or later in development.
ln contrast, the Sxl protein expressed in early female em-
bryos directs splicing of the sex-lethal pre-mRNA so that a
functional sex-lethal mRNA is produced (Figure 8-16a). Sxl
accomplishes this by binding to a sequence in the pre-mRNA
near the 3' end of the intron between exon 2 and exon 3,
thereby blocking the proper association of U2AF and U2 5' 3
snRNP (Figure 8-1 I). As a consequence, the Ul snRNP bound
to the 3' end of exon 2 assembles into a spliceosome with U2
snRNP bound to the branch point at the 3' end of the intron FIGURE 8-17 Model of splicing activation by Tra protein and the
between exons 3 and 4, leading to splicing of exon 2 to exon SR proteins Rbpl and Tra2. In female Drosophila embryos, splicing of
4 and skipping of exon 3. The binding site for Sxl in the Sxl exons 3 and 4 in dsx pre-mRNA is activated by binding ofTrafTra2/
pre-mRNA is called an intronic SfJlicing silencer because of its Rbpl complexes to six sites in exon 4. Because Rbpl and Tra2 cannot
location in an intron and its function in blocking, or "silenc- bind to the pre-mRNA in the absence onra, exon 4 is skip ped in male
ing," use of a splice site. The resulting female-specific sex-lethal embryos. See the text for discussion. An= polyadenylation. [Adapted
mRNA is translated into functional Sxl protein, which rein- from T. Maniatis and B. Tasic, 2002. Nature 418:236.]
forces its own expression in female embryos by continuing to
cause skipping of exon 3. The absence of Sxl protein in male for female development. Conversely, the female Dsx protein
embryos allows the inclusion of exon 3 and, consequently, of represses transcription of genes required for male development.
the stop codon that prevents translation of functional Sxl Figure 8- I 7 illustrates how the Tra/Tra2/Rbpl complex is
protein. thought to interact with double-sex (dsx) pre-mRNA. Rbpl
Sxl protein also regulates alternative RNA splicing of the and T ra2 are SR proteins, but they do not interact with exon 4
transformer gene pre-mRNA (Figure 8-16b). In male embryos, in the absence of the Tra protein. Tra protein interacts with
where no Sxl is expressed, exon l is spliced to exon 2, which Rbpl and Tra2, resulting in the cooperative binding of all
contains a stop codon that prevents synthesis of a functional three proteins to six exonic splicing enhancers in exon 4. The
transformer protein. ln female embryos, however, binding of bound Tra2 and Rbpl proteins then promote the binding of
Sxl protein to an intronic splicing silencer at the 3' end of the U2AF and U2 snRNP to the 3' end of the intron between
inrron between exons 1 and 2 blocks binding of U2AF ar this exons 3 and 4, just as other SR proteins do for constitutively
site. The interaction of Sxl with transformer pre-mRNA is me- spliced exons (see Figure 8-13). The Tra/Tra2/Rbp1 complexes
diated by two RRM domains in the protein (see Figure 8-5). may also enhance binding of the cleavage/polyadenylation
When Sxl is bound, U2AF binds to a lower-affinity site farther complex to the 3' end of cxon 4.
3' in the pre-mRNA; as a result exon 1 is spliced to this alter-
native 3' splice c;ite, eliminating exon 2 with irs stop codon.
Splicing Repressors and Activators Control
The resulting female-specific transformer mRNA, which con-
tains additional constitutively spliced exons, is translated into Splicing at Alternative Sites
functional Transformer (Tra) protein. As is evident from Figure 8-16, the Drosophila Sxl protein
Finally, Tra protein regulates the alternative processing and Tra protein have opposite effects: Sxl prevents splicing,
of pre-mRNA transcribed from the double-sex gene (Figure causing exons to be skipped, whereas Tra promotes splicing.
8 16c). In female embryos, a complex ofT ra and two consti- The action of similar proteins may explain the cell-type-specific
tutively expressed proteins, Rbpl and Tra2, directs spl icing expression of fibronectin isoforms in humans. For instance,
of exon 3 to exon 4 and also promotes cleavage/polyaden- an Sxl-like splicing repressor expressed in hepatocytes might
ylation at the alternative poly(A) site at the 3' end of exon bind to splice sites for the EIIIA and EIIIB exons in the fi -
4-leading to a short, female-specific version of the Dsx pro- bronectin pre-mRNA, causing them to be skipped during
tein. In male embryos, which produce no Tra protein, exon RNA splicing (see Figure 4-16). Alternatively, a Tra-like
4 is skipped, so that exon 3 is spliced to exon 5. Exon 5 is splicing activator expressed in fibroblasts might activate the
constitutively spliced to exon 6, which is polyadenylated at splice sites associated with the fibronectin EIIIA and ElliB
its 3' end-leading to a longer, male-specific version of the Dsx exons, leading to inclusion of these exons in the mature
protein. The RNA sequence ro which Tra binds in exon 4 is mRNA. Experimental examination in some systems has re-
called an exomc splicing enhancer since it enhances splicing at vealed that inclusion of an exon in some cell types versus
a nearby splice site. skipping of the same exon in other cell types results from the
As a result of the cascade of regulated RNA processing de- combined influence of several splicing repressors and en-
picted in Figure 8-16, different Dsx proteins are expressed in hancers. RNA binding sites for repressors, usually hnRNP
male and female embryos. The male Dsx protein is a transcrip- proteins, can also occur in exons, where they are called exonic
tional repressor that inhibits the expression of genes required splicing silencers. And binding sites for splicing activators,

362 CHAPTERs Post-transcriptional Gene Control


usually SR proteins, can also occur in introns, where they arc (a)
called intronic splicing enhancers.
Alternative splicing of cxons is especially common in the
nervous system, generating multiple isoforms of many proteins
required for neuronal development and function in both verte-
brates and invertebrates. The primary transcripts from these ./"' ~~'7'1Wl':mY
genes often show complex splicing patterns that can generate Apical Auditory
hair cell nerve Basal
several different mRNAs, with different spliced forms expressed (50 Hz) cell body Auditory hair cell
. ' in different anatomical locations within the cenrr::~l nervous sys- nP.rve (5000 Hz)

tem. We consider two remarkable examples that illustrate the


(b)
critical role of this process in neural function. N
Exterior
Expression of K+ -Channel Proteins in Vertebrate Hair Cells In
the inner car of verteb rates, individual "hair cells," which
are ciliated neurons, respond most strongly to a specific fre-
quency of sound. Cells tuned to low frequency (-50 Hz) arc
Cytosol
found at one end of the tubular cochlea that makes up the inner
car; cells responding to high frequency (-5000Hz) are found
at the other end (Figure 8-18a). Cells in between the ends
respond to a gradient of frequencies between these extremes.
One component in the tuning of hair cells in reptiles and
birds is the opening of K + ion channels in response to in-
creased intracellular Ca 2 concentrations. The Cal+ concen-
tration at which the channel opens determines the frequency
with which the membrane potential oscillates and hence the
frequency to which the cell is tuned. FIGURE 8-18 Role of alternative splicing in the perception of
The gene encoding this Ca2 + -activated K channel is ex- sounds of different frequency. (a) The chicken cochlea, a 5-mm-long
pressed as multiple, alternatively spliced mRNAs. The vari- tube, contains an epithelium of auditory hair cells that are tuned to a
ous proteins encoded by these alternative mRNAs open at gradient of vibrational frequencies from SO Hz at the apical end (left)
different Ca 2 + concentrations. Hair cells with differcnr re- 2
to 5000 Hz at the basal end (right). (b) The Ca -activated K channel
sponse frequencies express different isoforms of the channel contains seven transmembrane a helices (S0-56), which associate to
protein depending on their position along the length of the form the channel. The cytosolic domain, which includes four hydropho-
cochlea. The sequence variation in the protein is very com- bic regions (57-510), regulates opening of the channel in response
plex: there are at least eight regions in the mRNA where alter- to Ca 2 +. lsoforms of the channel, encoded by alternatively spliced
native exons are utilized, permitting the expression of 576 mRNAs produced from the same primary transcript, open at different
possible isoforms (Figure 8-18b). PCR analysis of mRNAs Ca 2 concentrations and thus respond to different frequencies.
from individ ual "hair cells has shown that each hair cell ex- Red numbers refer to regions where alternative splicing produces
different amino acid sequences in the various isoforms. [Adapted from
presses a mixture of different alternative Ca 2 +-activated K+-
K. P. Rosenblatt et al., 1997, Neuron 19:1061.]
channel mRNAs, with different forms predominating in
different cells according to their position along the cochlea.
This remarkable arrangement suggests that splicing of the
Ca 2 -activated K+-channel pre-mRNA is regulated in re- Many examples of genes similar to those that encode
sponse to extracellular signals that inform the cell of its posi- the cochlear K+-channel have been observed in verte-
tion along the cochlea. brate neurons; alternatively spliced mRNAs co-expressed
Other studies demonstrated that splicing at one of the al- from a specific gene in one type of neuron are expressed at
ternative splice sites in the Ca 2 -activated K+ -channel pre- different relative concentrations in different regions of the
mRNA in the rat is suppressed when a specific protein kinase central nervous system. Expansions in the number of micro-
is activated by neuron depolarization in response to synaptic satellite repeats within the transcribed regions of genes ex-
activity from interacting neurons. This observation raises the pressed in neurons can cause an alteration in the relative
possibility that a splicing repressor specific for this site may be concentrations of alternatively spliced mRNAs transcribed
activated when it is phosphorylated by this protein kinase, from multiple genes. In Chapter l'l, we discussed how back-
whose activity in turn is regulated by synaptic activity. Since ward slippage during DNA replication can lead to expansion
hnRNP and SR proteins are extensively modified by phos- of a microsatellite repeat (see Figure 6-5). At least 14 different
phorylation and other post-translational modifications, it types of neurological disease result from expansion of micro-
seems likely that complex regulation of alternative RNA splic- satellite regions within transcription units expressed in neu-
ing through post-translational modifications of splicing fac- rons. The resulting long regions of repeated simple sequences
tors plays a significant role in modulating neuron function. in nuclear RNAs of these neurons result in abnormalities in

8.2 Regulation of Pre-mRNA Processing 363


CAA TAA
~
~:::::t:t:
. ~ F:'f=
l ::::;:
1 ::t-:!~~
.UJ~---+~-~
apoB
3'
gene !
i =

apoB
mRNA 5'
...
CAA
...
UAA
An 5'
CAA-+UAA UAA

~ ~
1 4536 1 2152

Proteins NH21 d cooH NH 2 ll--~-~....j~ COOH


ApoB-100 ApoB-48
FIGURE 819 RNA editing of apo-8 pre-mRNA. The apoB mRNA receptors on cell membranes. In the apo-8 mRNA produced in the
produced in the liver has the same sequence as the exons in the intestine, the CAA codon in exon 26 is edited to a UAA stop codon.
primary transcript. Th is mRNA is translated into apoB-1 00, which has As a result, intestinal cells produce apoB-48, which corresponds to the
two functional domains: anN-terminal domain (green) that associates N-terminal domain of apoB-100. [Adapted from P. Hodges and J. Scott,
,
with lipids and a (-terminal domain (orange) that binds to LDL 1992, Trends Biochem. Sci. 17:77.]

the relative concentrations of alternatively spliced mRNAs. RNA Editing Alters the Sequences
For example, the most common of these types of diseases, of Some Pre-mRNAs
myotonic dystrophy, is characterized by paralysis, cognitive
impairment, and personality and behavior disorders. Myo- In the mid-1980s, sequencing of numerous eDNA clones and
tonic dystrophy results from increased copies of either CUG corresponding genomic DNAs from multiple organisms led
repeats in one transcript, in some patients, or CCUG repeats to the unexpected discovery of another type of pre-mRNA
in another transcript, in other patients. When the number of processing. In this type of processing, called RNA editing, the
these repeats increases to 10 or more times the normal num- sequence of a pre-mRNA is altered; as a result, the sequence
ber of repeats in these genes, abnormalities are observed in of the corresponding mature mRNA differs from the exons
the level of two hnRNP proteins that hind to these repeated encoding it in genomic DNA.
sequences. This probably results because these hnRNPs are RNA editing is widespread in the mitochondria of proto-
bound by the abnormally high concentration of this RNA zoans and plants and also in chloroplasts. In the mitochon-
sequence in the.nuclei of neurons in such patients. The ab- dria of certain pathogenic trypanosomes, more than half the
normal concentrations of these hnRNP proteins are thought sequence of some mRNAs is altered from the sequence of the
to lead to alterations in the rate of splicing of different alter- corresponding primary transcripts. Additions and deletions
native splice sites in multiple pre-mRNAs normally regulated of specific numbers of U's fo llows temp lates provided by
by these hnRNP proteins. base-paired short "guide" RNAs. These RNAs are encoded
by thousands of mini-mitochondrial DNA circles catenated
to many fewer large mitochondrial DNA molecules. The rea-
Expression of Dscam lsoforms in Drosophila Retinal Neurons son for this baroque mechanism for encoding mitochondrial
The most extreme example of regulated alternative RNA proteins in such protOzoans is not clear. But this system does
processing yet uncovered occurs in expression of the Dscam represent a potential target for drugs to inhibit the complex
gene in Drosophila. Mutations in this gene interfere with the processing enzymes essential to the microbe that do not exist
normal synaptic connections made between axons and den- in the cells of their human or other vertebrate hosts.
drites during fly development. Analysis of the Dscam gene In higher eukaryotes, RNA editing is much rarer, and
showed that it contains 95 alternatively spliced exons that thus far, only single-base changes have been observed. Such
could be spliced to generate 38,016 possible isoforms! Re- minor editing, however, turns out to have significant func-
cent results have shown that Drosophila mutants with aver- tional consequences in some cases. An important example of
sion of the gene that can be spliced in only about 22,000 RNA editing in mamma ls involves the apoB gene. This gene
different ways have specific defects in connectivity between encodes two alternative forms of a c;erum protein central to
neurons. These results indicate that expression of most of the uptake and transport of cholesterol. Consequently, it is
the possible Dscam isoforms through regulated RNA splic- important in the pathogen ic processes that lead to athero-
ing helps to specify the tens of millions of different specific sclerosis, the arterial disease that is the major cause of death
synapti<.. connections between neurons in the Drosophila in the developed world. The apoB gene expresses both the
brain. In other words, the correct w iring of neurons in the serum protein apolipoprotein B-100 (apoB-100) in hepato-
brain requires regulated RNA splicing. cytes, the major cell type in the liver, and apoB-48, expressed

364 CHAPTERs Post-transcriptional Gene Control


in intestinal epithelial cells. The ~240-kDa apoB-48 corre- The nuclear envelope is a double membrane that separates
sponds to theN-terminal region of the ~500-kDa apoB-100. the nucleus from the cytoplasm (see Figure 9-32). Like the
As we detail in Chapter 10, both apoB proteins are compo- plasma membrane surrounding cells, each nuclear mem-
nents of large lipoprotein complexes that transport lipids in brane consists of a water-impermeable phospholipid bilayer
the serum. However, only low-density lipoprotein (LDL) and multiple associated proteins. mRNPs and other macro-
complexes, which contain apoB-100 on their surface, deliver molecules including tRNAs and ribosomal subunits traverse
cholesterol to body tissues by binding to the LDL receptor the nuclear envelope through nuclear pores. This section will
present on all cells. focus on the export of mRNPs through the nuclear pore and
The cell-type-specific expression of tht> two forms of apoB the mechanisms that allow some lt:vcl of regulation of this
results from editing of apoB pre-mRNA so as to change the step. Transport of other cargoes across the nuclear pore is
nucleotide at position 6666 in the sequence from a C to a U. discussed in Chapter 13.
This alteration, which occurs only in intestinal cells, converts
a CAA codon for glutamine to a UAA stop codon, leading to
Macromolecules Exit and Enter the Nucleus
synthesis of the shorter apoB-48 (Figure 8-19). Studies with
the partially purified enzyme that performs the post-tran- Through Nuclear Pore Complexes
scriptional deamination of C 6666 to U shows that it can recog- Nuclear pore complexes (NPCs) arc large, symmetrical struc-
nize and edit an RNA as short as 26 nucleotides with the tures composed of multiple copies of approximately 30 dif-
sequence surrounding C 6666 in the a{JOB primary transcript. ferent proteins called nucleoporins. Embedded in the nuclear
envelope, NPCs are cylindrical in shape with a diameter of
- 30 nm (Figure 8-20a). A special class of nucleoporins called
FG-nucleoporins line the central channel through the NPC.
KEY CONCEPTS of Section 8.2 FG-nucleoporins form a semi-permeable barrier that allows
small molecules to diffuse freely, but restricts the passage of
Regulation of Pre-mRNA Processing larger molecules. The FG-nucleoporin globular domain an-
Because of alternative splicing of primary transcripts, the chors the protein in the NPC scaffold. From these anchors,
use of alternative promoters, and cleavage at different poly(A) long random-coil stretches extend into the channel. These ex-
sites, different mRNAs may be expressed from the same gene tensions are composed of hydrophilic amino acid sequences
in different cell types or at different developmental stages (sec punctuated by hydrophobic PG-repeats, short sequences rich
Figure 6-3 and Figure 8-16). in hydrophobic phenylalanine (F) and glycine (G). These FG-
Alternative splicing can be regulated by RNA-binding pro- rcpeat domains form a cloud of polypeptide chains in the
teins that bind to specific sequences near regulated splice central channel and extending into the nucleoplasm and cyto-
plasm, effectively limiting the free diffusion of macromole-
. sites. Splicing repressors may sterically block the binding of
cules across the channel. Water, ions, metabolites, and small
splicing factors to specific sites in pre-mRNAs or inhibit their
function. Splicing activators enhance splicing by interacting globular proteins up to - 40-60 kDa can diffuse through the
with splicing factors, thus promoting their association with a cloud of FG-repeat domains. However, the FG-domains in
regulated splice.site. The RNA sequences bound by splicing the central channel form a barrier restricting the diffusion of
repressors are called intronic or exonic splicing silencers, de- larger macromolecules between the cytoplasm and nucleus.
pending on their location in an intron or exon. RNA se- Proteins and RNPs larger than ~40-60 kDa must be se-
quences bound by splicing activators arc called intronic or lectively transported across the nuclear envelope with the as-
exonic splicing enhancers. sistance of soluble transporter proteins that bind them and
also interact reversibly with the FG-repeats of FG-nucleoporins.
In RNA editing the nucleotide sequence of a pre-mRNA is As a consequence of these reversible interactions with FG-
a ltered in the nucleus. In vertebrates, this process is fairly domains, the transporter and its stably bound cargo can be
rare and entails deamina~ion of a single base in the mRNA passed from FG-domain to FG-domain, allowing them both
sequence, resulting in a change in the amino acid specified by to diffuse down a concentration gradient from the nucleus to
the corresponding codon and production of a functionally the cytoplasm.
different protein (see Figure 8-19). mRNPs are transported through the NPC by the mRNP
exporter, a heterodimer consisting of a large subunit, called
nuclear export factor 1 (NXF1 ), and a small subunit, nuclear
export transporter 1 (NXT1) (Figure 8-20b) . NXFl binds
8.3 Transport of mRNA Across nuclear mRNPs through associations with both RNA and
other proteins in the mRNP complex. One of the most im-
the Nuclear Envelope
portant of these is REF (RNA export factor), a component
Fully processed mRNAs in the nucleus remain bound by of the exon-junction complexes discussed earlier, which is
hnRNP proteins in complexes referred to as nuclear mRNPs. bound approximately 20 nucleotides 5' to each cxon-exon
Before an mRNA can be translated into its encoded protein, junction (see Figure 8-21 ). The NXF L/NXT1 mRNP exporter
it must be exported out of the nucleus into the cytoplasm. also associates with SR proteins bound to cxonic splicing

8.3 Transport of mRNA Across the Nuclear Envelope 365


(a) Cytosol (b) Cytosol
FG Nucleoporins ~mRNP
Direction of t
Central
channel
Cytoplasmic
fi laments
tra nsport I
Outer nuclear
Outer nuclea r
Core scaffold membrane membrane ""'
/
Outer ri ng

REF, ot her
'\Inner nuclear
adapter membrane
Lumen Luminal Nuclear
proteins
ring basket

Nucleus Nucleus

FIGURE 8 -20 Model of transporter passage through an NPC. in pu rple. The FG-domains of FG-nucleop'orins (tan worms) have an
(a) Diagram of NPC structure. The nuclear envelope lipid bilayers are extended, random-coil conformation that forms a molecular cloud of
represented in green. Transmembrane proteins represented in red continuously moving random-coil polypeptide. (b) Nuclear transport-
form a luminal ring around which the nuclear envelope membrane folds ers (NXF 1/NXT1) have hydrophobic regions on their surface that bind
to form the inner and outer nuclear membranes. Transmembrane reversibly to the FG-domains in the FG-nucleoporins. As a conse-
domains of these proteins are connected to globular domains on the quence, they can penetrate the molecular cloud in the NPC central
inside of the pore to which the other nucleoporins bind, forming the channel and diffuse in and out of the nucleus. [Adapted from D. Grunwald,
core scaffold. The globular domains of the FG-nudeoporins are shown R. H. Singer, and M. Rout, 2011 , Nature 475:333.)

enhancers. Thus SR proteins associated with exons function Protein filaments extend from the core scaffold into the
to direct both the splicing of pre-mRNAs and the export of nucleoplasm forming a "nuclear basket " (see Figure 8-20a).
fully processed mRNAs through NPCs to the cytoplasm. Protein filaments also extend into the cytoplasm. T hese fila-
mRNPs are probably bound along their length by multiple ments assist in mRNP export. Gle2, an adapter protein that
NXFl/NXTl mRNP exporters, which interact with the FG- reversibly binds both NXFl and a nucleoporin in the nuclear
domains of FG-nucleoporins to faci litate export of mRNPs basket, brings nuclear mRNPs to the pore in preparation for
through the NPC central channel (Figure 8-20b). export. A nucleoporin in the cytoplasmic filaments of the NPC

CBC

Nucleus

Cytoplasm

FIGURE 8-21 Remodeling of mRNPs during


nuclear export. Some mRNP proteins (rectan-
gles) dissociate from nuclear mRNP complexes
before expurllhrough an NPC. Some (ovals) are ciF4E
exported through the NPC associated with the
mRNP but dissociate in the cytoplasm and are
shuttled back into the nucleus through an NPC.
In the cytoplasm, translation initiation factor
eiF4E replaces CBC bound to the 5' cap and
PABPI replaces PABPII.

366 CHAPTER 8 Post-transcriptional Gene Control


binds an RNA helicase (Dbp5) that functions in the dissocia- plasm, where they dissociate. As a result, the direction of
tion of NXFl/NXTl and other hnRNP proteins from the mRNP export may be driven by simple diffusion down a
mRNP as it reaches the cytoplasm. concentration gradient of the transport-competent mRNP
In a process called mRNP remodeling, the proteins associ- exporter-mRNP complex across the NPC, from high in the
ated with an mRNA in the nuclear mRNP complex are ex- nucleus to low in the cytoplasm.
changed for a different set of proteins as the mRNP is
transported through the NPC (Figure 8-21). Some nuclear Nuclear Export of Balbiani Ring mRNPs The larval salivary
mRNP proteins dissociate early in transport, remaining in the glands of the insect Chironomous tentans provide a good
nucleus to bind to newly synthesized nascent pre-mRNA. model system for electron mil:roscopy studies of the forma-
Other nuclear mRNP proteins remain with the mRNP com- tion of hnRNPs and their export through NPCs. In these
plex as it traverses the pore and do not dissociate from the larvae, genes in large chromosomal puffs called Balbiani
mRNP until the complex reaches the cytoplasm. Proteins in rings are abundantly transcribed into nascent pre-mRNAs
this category include the NXFl/NXTl mRNP exporter, cap- that associate with hnRNP proteins and are processed into
binding complex (CBC) bound to the 5 ' cap, and PABPII coiled mRNPs with a final mRNA length of -75 kb (Figure
bound to the poly(A) tail. They dissociate from the mRNP on 8-23a, b). These giant mRNAs encode large glue proteins
the cytoplasmic side of the NPC through the action of the that adhere the developing larvae to a leaf. After processing
Dbp5 RNA helicase that associates with cytoplasmic NPC fila- of the pre-mRNA in Balbiani ring hnRNPs, the resulting
ments, as discussed above. These proteins are then imported mRNPs move through nuclear pores to the cytoplasm. Elec-
back into the nucleus as discussed for other nuclear proteins in tron micrographs of sections of these cells show mRNPs that
Chapter 13, where they can function in the export of another appear to uncoil during their passage through nuclear pores
mRNP. In the cytoplasm, the cap-binding translation initiation and then bind to ribosomes as they enter the cytoplasm. This
factor eiF4E replaces CBC bound to the 5' cap of nuclear uncoiling is probably a consequence of the remodeling of
mRNPs (Figure 4-24). ln vertebrates, the nuclear poly(A)- mRNPs as the result of phosphorylation of mRNP proteins
binding protein PABPII is replaced with the cytoplasmic by cytoplasmic kinases and the action of the RNA helicase
poly(A)-binding protein PABPI (so named because it was dis- associated with NPC cytoplasmic filament~, as discussed in
covered before PABPII). Only a single PABP is found in bud- the previous section. The observation that mRNPs become
ding yeast, in both the nucleus and the cytoplasm. associated with ribosomes during transport indicates that
the 5' end leads the way through the nuclear pore complex.
Yeast SR Protein Studies of S. cerevisiae indicate that the di- Detailed electron microscopic studies of the transport of Bal-
rection of mRNP export from the nucleus into the cytoplasm biani ring mRNPs through nuclear pore complexes led to the
is controlled by phosphorylation and dephosphorylation of model depicted in Figure 8-23c.
mRNP adapter proteins such as REF that assist in the binding
of the NXFl/NXTl exporter to mRNPs. In one case, a yeast
SR protein (Npl3) functions as an adapter protein that pro-
Pre-mRNAs in Spliceosomes Are Not
motes the binding of the yeast mRNP exporter (Figure 8-22).
The SR protein initially binds to nascent pre-mRNA in its Exported from the Nucleus
phosphorylated form. When 3' cleavage and polyadenylation It is critical that only fully processed mature mRNAs be ex-
are completed, the adapter protein is dephosphorylated by a ported from the nucleus because translation of incompletely
specific nuclear protein phosphatase essential for mRNP ex- processed pre-mRNAs containing introns would produce de-
port. Only the dephosphorylated adapter protein can bind the fective proteins that might interfere with the functioning of
mRNP exporter, thereby coupling mRNP export to correct the cell. To prevent this, pre-mRNAs associated with snRNPs
polyadenylation. This is one form of mRNA "quality con- in spliceosomes usually are prevented from being transported
trol." If the nascent mRNf is not correctly processed, it is not to the cytoplasm.
recognized by the phosphatase that dephosphorylates Npl3. In one type of experiment demonstrating this restriction,
Consequently, it is not bound by the mRNA exporter and not a gene encoding a pre-mRNA with a single intron that nor-
exported from the nucleus. Instead it is degraded by exo- mally is spliced out was mutated to introduce deviations
somes, the multiprotein complexes that degrade unprotected from the consensus splice-site sequences. Mutation of either
RNAs in the nucleus and cytoplasm (see Figure 8-1). the 5' or the 3' invariant splice-site bases at the ends of the
Following export to the cytoplasm, the Npl3 SR protein inrron resulted in pre-mRNAs that were bound by snRNPs
is phosphorylated by a specific cytoplasmic protein kinase. to form spliceosomes; however, RNA splicing was blocked,
This causes it to dissociate from the mRNP, along with the and the pre-mRNA was retained in the nucleus. In contrast,
mRNP exporLtr. In this way, dephosphorylation of adapter mutation of both the 5' and 3' splice sites in the same pre-
mRNP proteins in the nucleus once RNA processing is com- mRNA resulted in export of the unspliced pre-mRNA, al-
plete and their phosphorylation and resulting dissociation in though less efficiently than for the spliced mRNA. When
the cytoplasm results in a higher concentration of mRNP both splice sites were mutated, the pre-mRNAs were not ef-
exporter-mRNP complexes in the nucleus, where they form, ficiently bound by snRNPs, and, consequently, their export
and a lower concentration of these complexes in the cyto- was not blocked.

8.3 Transport of mRNA Across the Nuclear Envelope 367


RNA pol II

RNA pol II RNA pol II

II
Nucleoplsam

.
NPC

Cytoplasm

lmportin
AAAAAAA
II AAAAAAA
~NXF1/NXT1
NXF1/N~
II

e a NXF1/NXT1

FIGURE 8 -22 Reversible phosphorylation and direction of mRNP Skyl phosphorylates Npl3 in the cytoplasm, causing lit dissociation of
nuclear export. Step 0 :The yeast SR protein Npl3 binds nascent the mRNP exporter and phosphorylated Npl3, probably through the
pre-mRNAs in its phosphorylated form. Step fJ: When polyadenylation action of an RNA helicase associated with NPC cytoplasmic filaments.
has occurred successfully, the Glc? nuclear phosphatase essential for mThe mRNA transporter and phosphorylated Npl3 are transported
mRNP export dephosphorylates Npl3, promoting the binding of the back into the nucleus through NPCs. 6 Transported mRNA is available
yeast mRNP exporter, NXFl /NXTl . Step 11:The mRNP exporter allows for translation in the cytoplasm. [From E. lzaurralde, 2004, Nat. Struct. Mol.
diffusion of the mRNP complex through the central channel of the Bioi. 11 :210-212. See W. Gilbert and C. Guthrie, 2004, Mol. Ce/113:201-212.]
nuclear pore complex (NPC). Step m: The cytoplasmic protein kinase

Recent studies in yeast have shown that a nuclear protein HIV Rev Protein Regulates the Transport
that associates with a nucleoporin in the NPC nuclear basket of Unspliced Viral mRNAs
is required to retain pre-mRNAs associated with snRNPs in
the nucleus. If either this protein or the nucleoporin to which As discussed earlier, transport of mRNPs containing mature,
it binds is deleted, unspliced prc-mRNAs are exported. functional mRNAs from the nucleus to the cytoplasm entails
a complex mechanism that is crucial to gene expression (sec
Many cases of thalassemia, an inherited disease that re- Figures 8-21, 8-22, and 8-23). Regulation of this transport
sults in abnormally low levels of globin proteins, are due theoretically could provide another means of gene control,
to mutations in globin-gene splice sites that decrease the effi- although it appears to be relatively rare. Indeed, the only
ciency of splicing but do not prevent association of the pre- known examples of regulated mRNA export occur during
mRNA with snRNPs. The resulting unspliced globin pre-mRNAs the cellular response to conditions (e.g., heat shock) that
are retained in reticulocyte nuclei and are rapidly degraded. cause protein denaturation or during viral infection when

368 CHAPTER 8 Post-transcriptional Gene Control


(b) FIGURE 8 -23 Formation of heterogeneous ribonucleoprotein
particles (hnRNPs) and export of mRNPs from t he nucleus.
(a) Model of a single chromatin transcription loop and assembly of
Balbiani ring (BR) mRNP in Chironomous ten tans. Nascent RNA transcripts
produced from the template DNA rapidly associate with proteins,
forming hnRNPs. The gradual increase in size of the hnRNPs reflects

\ the increasing length of RNA transcripts at greater distances from the


transcription start site. The model was reconstructed from electron
micrographs of serial thin sections of salivary gland cells. (b) Schematic
diagram of the biogenesis of hnRNPs. Following processing of the
pre-mRNA, the resulting ribonucleoprotein particle is referred to as
an mRNP. (c) Model for the transport of BR mRNPs through the nuclear
pore complex (NPC) based on electron microscopic studies. Note that
the curved mRNPs appear to uncoil as they pass through nuclear pores.
As the mRNA enters the cytoplasm, it rapidly associates with ribosomes,
indicating that the 5' end passes through the NPC first. [Part (a) from
C. Erricson et al., 1989, Cell 56:631 ; courtesy of B. Daneholt. Parts (b) and
(c) adapted from B. Daneholt, 1997, Ce//88:585. See also B. Daneholt, 2001,
Template mRNP Proc. Nat'/. Acad. Sci. USA 98:7012.)
DNA

m RNA

virus-induced alterations in nuclear transport maximize viral have some mechanism for overcoming this block, permitting
replication. Here we describe the regulation of mRNP ex- export of the longer viral mRNAs. Some retroviruses have
port mediated by a protein encoded by human immunodefi- evolved a sequence called the collstitutive transport element
ciency virus (HIV). (CTE), which binds to the NXFl/NXTl mRNP exporter with
A retrovirus, HJV integrates a DNA copy of its RNA ge- high affinity, thereby permitting export of unspliced retroviral
nome into the host-cell DNA (see Figure 4 -49). The integrated Rl"'A into the cytoplasm. HJV solved the problem differently.
viral DNA, or provirus, contains a single transcription unit, Studies with H IV mutants showed that transport of un-
which is transcribed into a. single primary transcript by cell ular spliced 9-k b and singly spliced 4 -kb viral mRNAs from the
RNA polymerase II. The HIV transcript can be spliced in alter- nucleus to the cytoplasm requires the virus-encoded Rev pro-
native ways to yield three classes of mRNAs: a 9-kb unspliced tein. Subsequent biochemical experiments demonstrated that
mRNA; ~4-kb mRNAs formed by removal of one intron; and Rev binds to a specific Rev-response element (RRE) present in
~2-kb mRNAs formed by removal of two or more introns HJV RNA. In cells infected with HJV mutants lacking the RRE,
(Figure 8-24). After their synthesis in the host-cell nucleus, all unspliced and singly spliced viral mRNAs remain in the nu -
three classes of HJV mRNAs are transported to the cytoplasm cleus, demonstrating that the RRE is required for Rev-mediated
and translated into viral proteins; some of the 9-kb unspliced stimulation of nuclear export. Early in an infection, before
RNA is used as the viral genome in progeny virions that bud any Rev protein is synthesized, only the multiply spliced 2-kb
from the cell surface. mRNAs can be exported. One of these 2-kb mRNAs encodes
Since the 9-kb and 4-kb HIV mRNAs contain splice sites, Rev, which contains a leucine-rich nuclear export signal that
they can be viewed as incompletely spliced mRNAs. As dis- interacts with transporter Exportin 1. As discussed in Chapter
cussed earlier, association of such incompletely spliced mRNAs 13, translation and nuclear import of Rev results in export of
with snRNPs in spliceosomes normally b locks their export the larger unspliced and singly spliced HIV mRNAs through
from the nucleus. Thus HJV, as well as other retroviruses, must the nuclear pore complex.

8.3 Transport of mRNA Across the Nuclear Envelope 369


HIV provirus

~~----------------~~--~c==
/

NUCLEAR mRNAs
l RRE

Transcription, splicing
CYTOPLASMIC mRNAs
9-kb +Rev
('
Unspliced 9 kb

4-kb ~'
Singly spliced 4 kb

2-kb ~
-Rev 0:>.::;;;;;;;;;;;;:;~ _____., 0 Rev protein
Multiply spliced ' 2 kb Translation

Nucleoplasm Cytopl asm

FIGURE 8-24 Transport of HIV mRNAs from the nucleus to the RNA species are translated into different viral proteins. Rev protein,
cytoplasm. The HIV genome, which contains several coding regions, is encoded by a 2-kb mRNA, interact s with the Rev-response element
transcribed into a single 9-kb primary transcript. Several -4-kb mRNAs (RRE) in the unspliced and singly spliced mRNAs, stimulating their
result from alternative splicing out of any one of several introns transport to the cytoplasm. [Adapted from B. R. Cullen and M. H. Malim, 1991,
(dashed lines) and several -2-kb mRNAs from splicing out oftwo or Trends Biochem. Sci. 16:346.]
more alternative introns. After transport to the cytoplasm, the various

scription elongation in the promoter proximal region are the


KEY CONCEPTS of Section 8.3 initial mechanisms for controlling the expression of genes in
the gene expression pathway of DNA~RNA~protein. In
Transport of mRNA Across the Nuclear Envelope
preceding sections of this chapter, we learned that the expres-
Most mRNPs are exported from the nucleus by a het- sion of protein isoforms is controlled by regulating alternative
erodimeric mRNP exporter that interacts with FG-rcpcats of RNA splicing and cleavage and polyadenylation at alternative
FG-nucleoporins (see Figure 8-20) . The direction of trans- poly(A) sites. Although nuclear export of fully and correctly
port (nucleus~cytoplasm) may result from dissociation of processed mRNPs to the cytoplasm is rarely regulated, the
the exporter-mRNP complex in the cytoplasm by phosphor- export of improperly processed or aberrantly remodeled pre-
ylation of mRNP proteins by cytoplasmic kinases and the mRNPs is prevented, and such abnormal transcripts are de-
action of an RNA helicase associated with cytoplasmic fila- graded by the exosome. However, retroviruses, including
ments of the nuclear pore complexes. HIV, have evolved mechanisms that permit pre-mRNAs that
The mRNP exporter binds to most mRNAs cooperatively retain splice sites to be exported and t ranslated.
v.ith SR proteins bound to exons and with REF associated with In this section we consider other mechanisms of post-
the exon-junction complexes that bind to mRNAs following transcriptional control that contribute to regulating the ex-
RNA splicing, and also binds to additional mRNP proteins. pression of some genes. Most of these mechanisms operate
in the cytoplasm, controlling the stability or loca lization of
Pre-mRNAs bound by a spliceosome normally arc not ex-
mRNA or its translation into protein. We begin by discuss-
ported from the nucleus, ensuring that only fully processed,
ing two recently discovered and related mechanisms of gene
functional mRNAs reach the cytoplasm for translation.
control that provide powerful new techniques for manipulat-
ing the expression of specific genes for experimental and
therapeutic purposes. These mechanisms are controlled by
short, - 22-nuclcotide, single-stranded RNAs called micro
8.4 Cytoplasmic Mechanisms RNAs (miRNAs) and short interfering (siRNAs). Both base-
pair with specific target mRNAs, either inhibiting their
of Post-transcriptional Control translation (miRNAs) or causing their degradation (siRNAs).
Before proceeding, let's quickly review the steps in gene ex- Humans express -500 miRNAs. Most of these are expressed
pression at which control is exerted. We saw in the previous in specific cell types at particular times during embryogenesis
chapter that regulation of transcription initiation and tran- and after birth. Many miRNAs can target more than one

370 CHAPTER 8 Post-transcriptional Gene Control

.
(a) miRNA ~translation inhibition (b) siRNA ~RNA cleavage

,o ~ OH I 1 I I ;1'-~""'l ~..,.~,..,1
:"T"IT"""j ;~~
II"'TI"T"Inii'"T'I'I"1111"'TI'T'l:-;
I I I L.LI I I I I I I I I I I I I LLr.
Target RNA Target RNA t
uc
C A
C A
5' -UCCCUGAGA GUGUGA 3' ~-UAGGUAGUUUCAUGUUGUUGGG-3'
11111 1 11 I I II I 1111111111111 111111111
3' -UCCAGGGACUCAACCAACACUCAA- 5' 3' - CUUAUCCGUCAAAGUACAACAACCUUCU- 5'

lin-4 miRNA and lin-14 mRNA (C.elegans) miR-196a and HOXBB mRNA (H. sapiens)

5' -UGUUAGCUGGAUGAAAACTT- 3' 5' -UCGGACCAGGCUUCAUUCCCC_ 3'


11111111 111111111 1111111111111 1 11111
3' -GCCACAAUCGAAACACUUUUGAAGGC- 5' 3' - UUAGGCCUGGUCCGAAGUAGGGUUAGU- 5'

CXCR4 m iRNA and target mRNA (H. sapiens) miR-166 and PHAVOLUTA mRNA (A. thaliana)

FIGURE 8-25 Base pairing with target RNAs distinguishes miRNA mRNA. (b) siRNA hybridizes perfectly with its target mRNA, causing
and siRNA. (a) miRNAs hybridize imperfectly with their target mRNAs, cleavage of the mANA at the position indicated by the red arrow,
repressing translation of the mRNA. Nucleotides 2 to 7 of an miRNA triggering its rapid degradation. [Adapted from P. D. Zamore and B. Haley,
(highlighted blue) are the most critical for targeting it to a specific 2005, Science 3 09:151 9.]

mRNA. Consequently, these newly discovered mechanisms that they regulate need not be perfect (Figure 8-25). In fact,
contribute significantly to the regulation of gene expression. considerable experimentation with synthetic miRNAs has
siRNAs, involved in the process called RNA interference, are shown that complementarity between the six or seven 5 ' nucle-
also an important cellular defense against viral infection and otides of an miRNA and its target mRNA 3' untranslated
excessive transposition by transposons. region are most critical for target mRNA selection.
Most miRNAs are processed from RNA polymerase II tran-
scripts of several hundred to thousands of nucleotides in length
Micro RNAs Repress Translation
called pri (for primary transcript)-miRNAs (figure 8-26). Pri-
of Specific mRNAs miRNAs can contain the sequence of one or more miRNAs.
Micro RNAs (miRNAs) were fim discovered during analysis miRNAs are also processed out of some excised introns and
of mutations in the lin-4 and let-7 genes of the nematode C. from 3' untranslated regions of some pre-mRNAs. Within
elegans, which influence development of the organism, Clon- these long transcripts are sequences that fold into hairpin struc-
ing and analysis 'o f wild-type lin-4 and let-7 revealed that they tures of -70 nucleotides in length with imperfect base pairing
encode not protein products but rather RNAs only 21 and 22 in the stem. A nuclear RNase specific for double-stranded
nucleotides long, respectively. The RNAs hybridize to the 3' RNA called Drosha acts with a nuclear double-stranded RNA-
untranslated regions of specific target mRNAs. For example, binding protein called DGCR8 in humans (Pasha in Dro-
the lin-4 miRNA, which is expressed early in embryogenesis, sophila) and cleaves the hairpin region out of the long precursor
hybridizes to the 3' untranslatcd regions of both the lin-14 RNA, generating a pre-miRNA. Pre-miRNAs are recognized
and lin-28 mRNAs in rhe.cytoplasm, thereby repressing their and bound by a specific nuclear transporter, Exportin5, which
translation by a mechanism discussed below, Expression of interacts with the FG-domains of nucleoporins, allowing the
lin-4 miRNA ceases later in development, allowing translation complex to diffuse through the inner channel of the nuclear
of newly synthesized lin-14 and lin-28 mRNAs at that time. pore complex, as discussed above (see Figure 8-20, and Chap-
Expression of let-7 miRNA occurs at comparable times dur- ter 13). Once in the cytoplasm, a cytoplasmic double-stranded
ing embryogenesis of all bilaterally symmetric animals. RNA-specific RNase called Dicer acts with a cytoplasmic
miRNA regulation of translation appears to be widespread double-stranded RNA-binding protein called TRBP in
in a ll multicellular plants and animals. In the past few years, humans (for Tar binding protein; called Loquacious in Dro-
small RNAs of 20- 26 nucleotides have been isolated, cloned, sophila) to further process the pre-miRNA into a double-
and sequenced from various tissues ot multiple model organ- stranded miRNA. The double-stranded miRNA is approximately
isms. Recent estimates suggest the expression of one-third of two turns of an A-form RNA helix in length, with strands
all human genes is regulated by -500 human miRNAs iso- 21-23 nucleotides long and two unpaired 3'-nucleotides at
lated from various tissues. The potential for regulation of mul- each end. Finally, one of the two strands is selected for assem-
tiple mRNAs by one miRNA is great because base pairing bly into a mature RNA-induced silencing complex (RISC)
between the miRNA and the sequence in the 3' ends of mRNAs containing a single-stranded macure miRNA bound by a

8.4 Cytoplasmic Mechanisms of Post-transcriptional Control 37 1


multidomain Argonaute protein, a member of a protein
family with a recognizable conserved sequence. Several Ar-
miR-1-1 gene gonaute proteins are expressed in some organisms, espe-
..F'~J.'"'ff~' cially plants, and are found in distinct RISC complexes with
different functions.
The miRNA-RISC complexes associate with target
mRNPs by base pairing between the Argonaute-bound ma-
ture miRNA and complementary regions in the 3' untrans-
lated regions (3' UTRs) of target mRNAs (see Figure 8-25).
Inhibition of target mRNA translation requires the binding
of two or more RISC complexes to distinct complementary
regions in the target mRNA 3' UTR. It has been suggested
that this may allow combinatorial regulation of mRNA

~
translation by separately regulating the transcription of two or
Drosha more different pri-miRNAs, which are processed to miRNAs
Pasha that are required in combination tO suppress the translation
of a specific target mRNA.
pre-miR-1-1
The binding of several RISC complexes to an mRNA in-
LO Nucleus hibits translation initiation by a mechanism currently being
c
e analyzed. Binding of RISC complexes causes the bound
0
c. mRNPs to associate with dense cytOplasmic domains many
X
TRBP w Cytoplasm times the size of a ribosome called cytoplasmic RNA-processing
Dicer bodies, or simply P bodies. P bodies, which will be described
in greater detail below, are sites of RNA degradation that
TRBP Dicer '\
contain no ribosomes or translation factors, potentially ex-
"'JGAAcc
. s -p CAUACUUCUUUAUAUGCCCAUA U plaining the inhibition of translation. The association with P
pre-m1R-1-1 1111111111111 11 IIII I G
3' -AUGUAUGAAGAAAUGUA G GGUAUC C bodies may also explain why expression of an miRNA often
----~ GAAU decreases the stability of a targeted mRNA.

5
l
pCAUACUUCUUUAUAUGCCCAUA- 3'
As mentioned earlier, approximately 500 different human
miRNAs have been observed, many of them expressed only
miR-1-1 111111111111111 Ill in specific cell types. Determining the function of these miRNAs
3'-AUGUAUGAAGAAAUGUA G GGUp- s
is currently a highly active area of research. In one example,

l a specific miRNA called miR-133 is induced when myoblasts


differentiate into muscle cells. miR-133 suppresses the trans-
Mature miR-1-1 lation of PTB, a regulatory splicing factor that functions sim-
bound to an ilarly to Sxl in Drosophila (see Figure 8-16). PTB binds to the
Argonaute RISC
protein 3' splice-site region in the pre-mRNAs of many genes, leading
to exon skipping or use of alternative 3 ' splice sites. When
miR-133 is expressed in differentiating myoblasts, the PTB
concentration falls without a significant decrease in the con-
FIGURE 826 miRNA processing. This diagram shows transcription centration of PTB mRNA. As a result, alternative isoforms of
and processing of the miR-1-1 miRNA. The primary miRNA transcript multiple proteins important for muscle-cell function are ex-
(pri-miRNA) is transcribed by RNA polymerase II. The nuclear double- pressed in the differentiated cells.
stranded RNA-specific endoribonuclease Drosha, with its partner
Other examples of miRNA regulation in various organ-
double-stranded RNA-binding protein DGCRS (Pasha in Drosophila),
isms are being discovered at a rapid pace. Knocking out the
make the initial cleavages in the pri-miRNA, generating a - 70-nucle-
otide pre-miRNA that is exported to the cytoplasm by nuclear dicer gene eliminates the generation of miRNA in mammals.
transporter ExportinS. The pre-miRNA is further processed in the This causes embryonic death early in mouse development.
cytoplasm to a double-stranded miRNA with a two-base single- However, when dicer is knocked out only in limb primordia,
stranded 3' end by Dicer in conjunction with the double-stranded the influence of miRNA on the development of the nones-
RNA-binding protein TRBP (loquatious in Drosophila). Finally, one of sential limbs can be observed (Figure 8-27). Although all
the two strands i~ incorporated into an RISC complex, where it is bound major cell types differentiate and the fundamental aspects of
by an Argonaute protein. [Adapted from P. D. Zamore and B. Haley, 2005, limb patterning are maintained, development is abnormal-
Science 309:1 519.] demonstrating the importance of miRNAs in regulating
the proper level of translation of multiple mRNAs. Of the
-500 human miRNAs, 53 appear to be unique to primates.
It seems likely that new miRNAs arose readily during evolution
by the duplication of a pri-miRNA gene followed by mutation

372 CHAPTER 8 Post-transcriptional Gene Control


Wild type Dicer mut ant cases, the mature short single-stranded RNA, either mature
siRNA or mature miRNA, is assembled into RISC complexes
in which the short RNAs are bound by an Argonaute pro-
tein. What distinguishes a RISC complex containing an
siRNA from one containing an miRNA is that the siRNA
base-pairs extensively with its target RNA and induces its
cleavage, whereas a RISC complex associated with an
miRNA recognizes its target through imperfect base-pairing
and results in inhibition of translation.
The Argonaute protein is responsible for cleavage of tar-
EXPERIMENTAL FIGURE 8 27 miRNA function in limb get RNA; one domain of the Argonaute protein is homolo-
development. Micrographs comparing normal (left) and Dicer
gous to RNase H enzymes that degrade the RNA of an
knockout (right) limbs of embryonic development day-13 mouse
RNA-DNA hybrid (see Figure 6-14). When the 5' end of the
embryos immunostained for the GdS protein, a marker of joint
short RNA of a RISC complex base-pairs precisely with a
formation. Dicer is knocked out in developing mouse embryos by
conditional expression of Cre to induce deletion of the Dicer gene only
target mRNA over a distance of one turn of an RNA helix
in these cells (see Figure 5-42). [From B. D. Harfe et al., 2005, Proc. Nor'/. Acad.
(10-12 base pairs), th is domain of Argonaute cleaves the
Sci. USA 102:10896.] phosphodiesrer bond of the target RNA across from nucle-
otides 10 and 11 of the siRNA (see Figure 8-25 ). The cleaved
RNAs arc released and subsequently degraded by cytoplas-
mic exosomes and 5' exoribonucleases. If base pairing is not
of bases encoding the mature miRNA. miRNAs are particu- perfect, the Argonaute domain does not cleave or release the
larly abundant in plants-more than 1.5 million distinct target mRNA. Instead, if several miRNA-RISC complexes
miRNAs have been characterized in Arabidopsis thaliana! associate with a target mRNA, its translation is tnhibired
and the mRNA becomes associated with P bodies, where, as
mentioned earlier, it is probably degraded by a different and
RNA Interference Induces Degradation
slower mechanism than the degradation pathway initiated
of Precisely Complementary mRNAs by RISC cleavage of a perfectly complementary target RNA.
RNA interference (RNAi) was discovered unexpectedly dur- When double-stranded RNA is introduced into the cyto-
ing attempts to experimentally manipulate the expression of plasm of eukaryotic cells, it enters the pathway for assembly
specific genes. Researchers tried to inhibit the expression of a of siRNAs into a RISC complex because it is recognized by
gene in C. elegans by microinjecting a single-stranded, com- the cytoplasmic Dicer enzyme and TRBP double-stranded
plementary RNA that would hybridize to the encoded mRNA RNA-binding protein that process pre-miRNAs (see Figure
and prevent its translation, a method called antisense inhibi- 8-26). This process of RNA interference is believed to be an
tion. But in control experiments, perfectly base-paired double- ancient cellular defense against certain viruses and mobile
stranded RNA a few hundred base pairs long was much more genetic elements in both plants and animals. Plants with mu-
effective at inhibiting expression of the gene than the anti- tations in the genes encoding Dicer and RISC proteins ex-
sense strand a lone (see Figure 5-45). Similar inhibition of hibit increased sensitivity to infection by RNA viruses and
gene expression by an introduced double-stranded RNA soon increased movement of transposons within their genomes.
was observed in plants. In each case, the double-stranded The double-stranded RNA intermediates generated during
RNA induced degradation of all cellular RNAs containing a replication of RNA viruses are thought to be recognized by
sequence that was exactly the same as one strand of the double- the Dicer ribonuclease, inducing an RNAi response that ulti-
stranded RNA. Because of the specificity of RNA interference mately degrades viral mRNAs. During transposition, trans-
in targeting mRNAs for destruction, it has become a power- posons are inserted into cellular genes in a random orientation,
ful experimental tool for studying gene function. and their transcription from different promoters produces
Subsequent biochemical studies with extracts of Dro- complementary RKAs that can hybrid ize with each other, initi-
sophila embryos showed that a long double-stranded RNA ating the RNAi system that then interferes with the expression
that mediates interference is initially processed into a double- of transposon proteins required for additional transpositions.
stranded short interfering RNA (siRNA). The strands in In plants and C. elegans the RNAi response can be in-
siRNA contain 21-23 nucleotides hybridized to each other duced in all cells of the organism by introduction of double-
so that the two bases at the 3' end of each strand are single- stranded RNA into just a few cell s. Such organism-wide
stranded. Further studies revealed that the cytoplasmic double- induction requires production of a protein that is homolo-
stranded RNA-specific ribonuclease that cleaves long gous to the RNA replicases of RNA viruses. It has been
double-stranded RNA into siRNAs is the same Dicer en- revealed that double-stranded siRNAs arc replicated and then
zyme involved in processing pre-miRNAs after their nuclear transferred to other cells in these organisms. In plants, transfer
export to the cytoplasm (see Figure 8-26). This discovery led of siRNAs might occur through plasmodesmata, the cytoplas-
to the realization that RNA interference and miRNA-medi- mic connections between plant cells that traverse the cell walls
ated translational repression are related processes. In both between them (see Figure 20-38 ). Organism-wide induction

8.4 Cytoplasmic Mechanisms of Post-transcriptional Control 373


of RNA interference does not occur in Drosophila or mam- mRNAs stored in oocytes are not translated efficiently. At
mals, presumably because their genomes do not encode the appropriate time during oocyte maturation or after ferti-
RNA replicase homologs. lization of an egg cell, usually in response to an external sig-
In mammalian cells, the introduction of long RNA-RNA nal, approximately 150 A residues are added to the short
duplex molecules into the cytoplasm results in the generalized poly(A) tails on these mRNAs in the cytoplasm, stimulating
inhibition of protein synthesis via the PKR pathway, discussed their translation.
further below. This greatly limits the use of long double- Studies with mRNAs stored in Xenopus oocytes have
stranded RNAs to experimentally induce an RNAi response helped elucidate the mechanism of this type of translational
against a specific targeted mRNA. furtunacdy, researchers control. Experiments in which short-tailed mRNAs are in-
discovered that one strand of double-stranded siRNAs 21-23 jected into oocytes have shown that two sequences in their 3'
nucleotides in length with two-base 3' single-stranded regions UTR are required for their polyadenylation in the cytoplasm:
leads to the generation of mature siRNA RISC complexes the AAUAAA poly(A) signal that is also required for the nu- .
without inducing the generalized inhibition of protein synthe- clear polyadenylation of pre-mRNAs, and one or more cop-
sis. This has allowed researchers to use synthetic double- ies of an upstream U-rich cytoplasmic polyadenylation
stranded siRNAs to "knock down" the expression of specific element (CPE). This regulatory element is bound by a highly
genes in human cells as well as in other mammals. This conserved CPE-binding protein (CPEB) that contains an
method of siRNA knockdown is now widely used in studies RRM domain and a zinc-finger domain.
of diverse processes, including the RNAi pathway itself. According to the current model, in the absence of a stimu-
latory signal, CPEB bound to the U-rich CPE interacts with the
protein Maskin, which in turn binds to eiF4E associated with
Cytoplasmic Polyadenylation Promotes
the mRNA 5' cap (Figure 8-28, left). As a result, eiF4E cannot
Translation of Some mRNAs interact with other initiation factors and the 40S ribosomal
In addition to repression of translation by miRNAs, other subunit, so translation initiation is blocked. During oocyte
protein-mediated translational controls help regulate expres- maturation, a specific CPEB serine is phosphorylated, causing
sion of some genes. Regulatory sequences, or elements, in Maskin to dissociate from the complex. This allows cytoplas-
mRNAs that interact with specific proteins to control transla- mic forms of the cleavage and polyadenylation specificity fac-
tion generally are present in the untranslated region (UTR) at tor (CPSF) and poly(A) polymerase to bind to the mRNA
the 3' or 5' end of an mRNA. Here we discuss a type of pro- cooperatively with CPEB. Once the poly(A) polymerase cata-
tein-mediated translational control involving 3' regulatory ele- lyzes the addition of A residues, PABPI can bind to the length-
ments. A different mechanism involving RNA-binding proteins ened poly(A) tail, leading to the stabilized interaction of all the
that interact with 5' regulatory elements is discussed later. factors needed to initiate translation (Figure 8-28, right; see
Translation of many eukaryotic mRNAs is regulated by also Figure 4-24). In the case of Xenopus oocyte maturation,
sequence-specific RNA-binding proteins that bind coopera- the protein kinase that phosphorylates CPEB is activated in
tively to neighb<:>ring sites in 3' UTRs . This allows them to response ro the hormone progesterone. Thus timing of the
function in a combinatorial manner, similar to the cooperative translation of stored mRNAs encoding proteins needed for
binding of transcription factors to regulatory sites in an en- oocyte maturation is regulated by this external signal.
hancer or promoter region. In most cases studied, translation Considerable evidence indicates that a similar mechanism
is repressed by protein binding to 3' regulatory elements and of translational control plays a role in learning and memory.
regulation results from derepression at the appropriate time or In the central nervous system, the axons from a thousand or
place in a cell or developing embryo. The mechanism of such so neurons can make connections (synapses) with the den-
repression is best understood for mRNAs that must undergo drites of a single postsynaptic neuron (Figure 22-23) . When
cytoplasmic polyadenylat1on before they can be translated. one of these axons is stimulated, the postsynaptic neuron "re-
Cytoplasmic polyadenylation is a critical aspect of gene members" which one of these thousands of synapses was
expression in the early embryo of animals. The egg cells stimulated. The next time that synapse is stimulated, the
(oocytes) of multicellular animals contain many mRNAs, en- strength of the response triggered in the postsynaptic cell dif- .
coding numerous different proteins that are not translated fers from the first time. This change in response has been
until after the egg is fertilized by a sperm cell. Some of these shown to result largely from the translational activation of
"stored" mRNAs have a short poly(A) tail, consisting of mRNAs stored in the region of the synapse, leading to the
only -20-40 A residues, to which just a few molecules of local synthesis of new proteins that increase the size and alter
cytoplasmic poly(A)-binding protein (PABPI) can bind. As the neurophysiological characteristics of the synapse. The
discussed in Chapter 4, multi pit> PA RPI molecules hound to finding that CPEB is present in neuronal dendrites has led to
the long poly(A) tail of an mRNA interact with the eiF4G the proposal that cytoplasmic polyadenylation stimulates
initiation factor, thereby stabilizing the interaction of the translation of specific mRNAs in dendrites, much as it does
mRNA 5' cap with eiF4E, which is required for translation in oocytes. In this case, presumably, synaptic activity (rather
initiation (see Figure 4-24). Because this stabilization cannot than a hormone) is the signal that induces phosphorylation of
occur with mRNAs that have short poly(A) tails, such CPEB and subsequent activation of translation.

374 CHAPTERs Post-transcriptional Gene Control


Translationally dormant Translationally active

1 - - - - - :UUUUAU AAUAAA-A
I I I I

mANA I I
CPE Poly(A)
signal
FIGURE 8-28 Model for control of cytoplasmic polyadenylation specificity factor (CPSF) then binds to the poly(A) site, interacting with
and translation initiation. (Left) In immature oocytes, mRNAs both bound CPEB and the cytoplasmic form of poly(A) polymerase
containing the U-rich cytoplasmic polyadenylation element (CPE) have (PAP). After the poly(A) tail is lengthened, multiple copies of the
short poly(A) tails. CPE-binding protein (CPEB) mediates repression of cytoplasmic poly(A)-binding protein I (PABPI) can bind to it and
interact with eiF4G, which functions with other initiation factors to
... translation through the interactions depicted, which prevent assembly
of an initiation complex at the 5' end ofthe mRNA. (Right) Hormone bind the 405 ribosome subunit and initiate translation. [Adapted from
stimulation of oocytes activates a protein kinase that phosphorylates R. Mendez and J. D. Richter, 2001, Nature Rev. Mol. Cell Bioi. 2:521 .]
CPEB, causing it to release Maskin. The cleavage and polyadenylation

Cytoplasmic mRNAs are degraded by one of the three


Degradation of mRNAs in the Cytoplasm
pathways shown in Figure 8-29. For mQst mRNAs, the
Occurs by Several Mechanisms deadenylation-dependent pathway is followed: the length of
The concentration of an mRNA is a function of both its rate the poly(A) tail gradually decreases with time through the
of synthesis and its rate of degradation. For this reason, if action of a deadenylating nuclease. When it is shortened suf-
two genes are transcribed at the same rate, the steady-state ficiently, PABPI molecules can no longer bind and stabilize
concentration of the corresponding mRNA that is more sta- the interaction of the 5' cap and translation initiation factors
ble will be higher than the concentration of the other. The (see Figure 4-24). The exposed cap then is removed by a de-
stability of an mRNA also determines how rapidly synthesis capping enzyme (Dcp1/Dcp2 in 5. cerevisiae), and the un -
. of the encoded protein can be shut down . For a stable protected mRNA is degraded by a 5'~3' exonuclease (Xrnl
mRNA, synthesis of the encoded protein persists long after in 5. cerevisiae). Removal of the poly(A) tail also makes
transcription of the gene is repressed. Most bacterial mRNAs mRNAs susceptible to degradation by cytoplasmic exosomes
are unstable, decaying exponentially with a typical half-life containing 3'~5' exonucleases. The 5'~3' exonucleases
of a few minute~. For this reason, a bacterial cell can rapidly predominate in yeast, and the 3' ~5 ' exosome predominates
adjust the synthesis of proteins to accommodate changes in in mammalian cells. The decapping enzymes and 5'~3' exo-
the cellular environment. Most cells in multicellular organ- nuclease are concentrated in the P bodies, regions of the cy-
isms, on the other hand, exist in a fairly constant environ- toplasm of unusually high density.
ment and carry out a specific set of functions over periods of Some mRNAs are degraded primarily by a deadenylation-
days to months or even the lifetime of the organism (nerve independent decapping pathway (see Figure 8-29). This is be-
cells, for example). Accoroingly, most mRNAs of higher eu- cause certain sequences at the 5' end of an mRNA seem to
karyotes have half-lives of many hours. make the cap sensitive to the decapping enzyme. For these
However, some proteins in eukaryotic cells are required mRNAs, the rate at which they are decapped controls the rate
only for short periods and must be expressed in bursts. For at which they are degraded because once the 5' cap is removed,
example, as discussed in the chapter introduction, certain sig- the RNA is rapidly hydrolyzed by the 5' ~3' exonuclease.
naling molecules called cytokines, which are involved in the The rate of mRNA deadenylation varies inversely with the
immune response of mammals, are synthesized and secreted frequency of translation initiation for an mRNA: the higher the
in short bursts (see Chapter 23). Similarly, many of the tran- frequency of initiation, the slower the rate of deadenylation.
scription factors that regulate the onset of the S phase of the This relation probably is due to the reciprocal interactions be-
cell cycle, such as c-Fos and c-Jun, are synthesized for brief tween translation initiation factors bound at the 5' cap and
periods only (Chapter 19 ). Expression of such proteins oc- PABPI bound to the poly(A) tail. For an mRNA that is trans-
curs in short bursts because transcription of their genes can lated at a high rate, initiation factors are bound to the cap much
be rapidly turned on and off, and their mRNAs have unusu- of the time, stabilizing the binding of PABPI and thereby pro-
ally short half-lives, on the order of 30 minutes or less. tecting the poly(A) tail from the deadenylation exonuclease.

8.4 Cytoplasmic Mechanisms of Post-transcriptional Control 375


Decapping pathway Deadenylation-dependent Endonucleolytic
(deadenylation-independent) pathways pathway

I~wo
- - - - - - AAAAAA - - - - - - AAAAAA - - - - - - AAAAAA

1
1a
- -----A
Poly{A) shortenmg

-----~- AAAAAA
Endonucleolytic
cleavage

Decapping / ""- . Exo~~c:e~lyt1c /


/ ~ decay / _

- - - - - - AAAAAA ------ A
5->3 / Exosome
Exonucleolytic

c decay

A
FIGURE 8 -29 Pathways for degradation of eukaryotic mRNAs. may either (1) be decapped and degraded by a 5' -t3' exonuclease or
In the deadenylation-dependent (middle) pathways, the poly(A) tail is (2) be degraded by a 3 '-t5' exonuclease in cytoplasmic exosomes.
progressively shortened by a deadenylase (orange) until it reaches a Some mRNAs (right) are cleaved internally by an endonuclease and the
length of 20 or fewer A residues, at which point the interaction with fragment s degraded by an exosome. Oth;r mRNAs (left) are decapped
PABPI is destabilized, leading to weakened interactions between the before they are deadenylated and then degraded by a 5' -t3' exonucle
5' cap and translation-initiation factors. The deadenylated mRNA then ase. [Adapted from M. Tucker and R. Parker, 2000, Ann. Rev. Biochem. 69:571.]

Many short-lived mRNAs in mammalian cells contain mul- translational modifications such as phosphorylation. Such
tiple, sometimes overlapping copies of the sequence AUUUA in mechanisms affect the translation rate of mpst mRNAs and
their 3' untranslated region. Specific RNA-binding proteins hence the overall rate of cellular protein synthesis.
have been found that both bind to these 3' AU-rich sequences
and also interact with a deadenylating enzyme and with the TOR Pathway The TOR pathway was discovered through
exosome. This causes rapid deadenylation and subsequent research into the mechanism of action of rapamycin, an an-
3'-t5' degradation of these mRNAs. In this mechanism, the tibiotic produced by a strain of Streptomyces bacteria, which
rate of mRNA degradation is uncoupled from the frequency of is useful fo r suppressing t he immune response in organ
translation. Thus mRNAs containing the AUUUA sequence can transplant patients . The target of rapamycin (TOR) was
be translated at high frequency yet also be degraded rapidly, identified by isolating yeast m utants resistant to rapa mycin
allowing the encoded proteins to be expressed in short bursts. inhibition of cell growth. TOR is a large (-2400 amino acid
As shown in Figure 8-29, some mRNAs are degraded by residue) protein kinase that regulates several cellular pro-
an endonucleolytic pathway that docs not involve decapping cesses in yeast cells in response to nutritional status. In mul-
or significant deadenylation. One example of this type of path- ticellular eukaryotes, metazoan TOR (mTOR) also responds
way is the RNAi pathway discussed above (see Figure 8-25). to multiple signals from cell-surface-signaling proteins to co-
Each siRNA-RISC complex can degrade thousands of targeted ordinate cell growth with developmental programs as well as
RNA molecules. The fragments generated by internal cleavage nutritional status.
then are degraded by exonucleases. Current understanding of the mTOR pathway is sum-
marized in Figure 8-30. Active mTOR stimulates the overall
P Bodies As mentioned above, P bodies are sires of transla-
rate of protein synthesis by phosphorylating two critical pro-
tional repression of mRNAs bound by miRNA-RISC com-
teins that regulate translation directly. mTOR also activates
plexes. They arc also the major sites of mRNA degradation in
t ranscription factors that contr o l exp ression of ribosomal
the cytoplasm. These dense regions of cytoplasm contain the
components, tRNAs, and tra nslation fac tors, furthe r activat-
dccapping enzyme (Dcp 1/Dcp2 in yeast), activators of decap-
ing protein synthesis and cell growth.
ping (Dhh, Patl, Lsml-7 in yeast), the major 5'-t3' exonucle-
Recall that the first step in translation of a eukaryotic
ase (Xrnl), as well as densely associated mRNAs. P bodies arc
mRNA is binding of the ei F4 initiation complex to t he 5 ' cap
dynamic structures that grow and shrink in size depending on
via its eiF4E cap-bind ing subunit (see Figure 4-24). The con-
the rate at which mR.t"iPs associate with them, the rate at which
centration of active elF4E is regulated by a small family of
mRNAs are degraded, and the rate at which mRNPs exit P
homologous elf4E-binding proteins (4E-BPs) that inhibit
bodies and reenter the pool of translated mRNPs. mRNAs
Lhe iutera<.:tion of e!F4E with mRNA 5' caps. 4-BPs are di-
whose translation is inhibited by imperfect base-pa iring of
rect targets of mTOR. When phosphorylated by mTOR, 4E-
miRNAs (Figure 8-25) are major components of P-bodies.
BPs release elF4E, stimulating translation initiation. mTOR
also phosphorylates and activates another protein kinase
Protein Synthesis Can Be Globally Regulated (56K) that phosphorylates the small ribosomal subun it pro-
Like proteins involved in other processes, translation initia- tein S6 and probably additional substrates, leading to a fur-
tion factors and ribosomal proteins can be regulated by post- ther increase in the rate of protein synthesis.

376 CHAPTER 8 Post-transcriptional Gene Control


Growth factor
receptor

Nutrients

Cytoplasm

n utrients

~~/\
Protein
synthesis
Rib osom e Pol Ill M acroautophagy
bio g enesis transcript ion

FIGURE 8-30 mTOR pathway. mTOR is an active protein mTOR protein kinase activity. Low nutrient concentration also
kinase when bound by a complex of Rheb and an associated GTP regulates Rheb GTPase activity, by a mechanism that does not require
(lower /eft).ln contrast, mTOR is inactive when bound by a complex TSClfTSC2. Active mTOR phosphorylates 4E-BP, causing it to release
of Rheb associated with GDP (lower right). When active, the TSCl fTSC2 eiF4E, stimulating translation initiation. It also phosphorylates and
Rheb-GTPase activating protein (Rheb-GAP) causes hydrolysis of Rheb- activates 56 kinase (S6K), which in turn phosphorylates ribosomal
bound GTP to GDP, thereby inactivating mTOR. The TSClfTSC2 proteins, stimulating translation. Activated mTOR also activates
Rheb-GAP is activated (arrows) by phosphorylation by AMP kinase transcription factors for RNA polymerases I, II, and Ill, leading to
(AMPK) when cellular energy charge is low and by other cellular stress synthesis and assembly of ribosomes, tRNAs, and translation factors.
responses. Signal-transduction pathways activated by cell-surface In the absence of mTOR activity, all of these processes are inhibited. In
growth factor receptors lead to phosphorylation of inactivating sites contrast, activated mTOR inhibits macroautophagy, which is stimulated
on TSClfTSC2, inhibiting its GAP activity. Consequently, they leave a in cells with inactive mTOR. [Adapted from S. Wullschleger et al., 2006, Cell
higher fraction of cellular Rheb in the GTP conformation that activates 124:471.]

Translation of a specific subset of mRNAs that have a string translation factor genes. Finally, mTOR stimulates process-
of pyrimidines in their 5' untranslated regions (called TOP ing of the rRNA precursor (Section 8.5). As a consequence of
mRNAs for tract of oligopyrimidine) is stimulated particularly phosphorylation of these several mTOR substrates, the syn-
strongly by mTOR. The T0P mRNAs encode ribosomal pro- thesis and assembly of ribosomes as well as the synthesis of
teins and translation elongation factors. mTOR also activates translation factors and tRNAs are greatly increased. Alterna-
the RNA polymerase I transcription factor TIF-lA, stimulating tively, when mTOR kinase activity is inhibited, these sub-
transcription of the large rRNA precursor (see Figure 7-52). strates become dephosphorylated, greatly decreasing the rate
mTOR activates transcription by RNA polymerase III as of protein synthesis and the production of ribosomes, transla-
well, by phosphorylating and thereby activating protein ki- tion factors, and tRNAs, thus halting cell growth.
nases that phosphorylate MAFl, a protein inhibitor of RNA mTOR activity is regulated by a monomeric small G pro-
polymerase III transcription. MAFl phosphorylation causes it tein in the Ras protein family called Rheb. Like other small G
to be exported from the nucleus, relieving repression of RNA proteins, Rheb is in its :=~ctive conformation when it is bound
polymerase III transcription. When mTOR activity falls , to GTP. RhebGTP binds the mTOR complex, stimulating
MAFl in the cytoplasm is rapidly dephosphorylated and im- mTOR kinase activity, probably by inducing a conformation
ported into the nucleus where it represses transcription by change in its kinase domain. Rheb is in turn regulated by a
RNA polymerase III. heterodimer composed of subunits TSCl and TSC2, named
In addition, mTOR activates two RNA polymerase II ac- for their involvement in the medical syndrome tuberous scle-
tivators that stimulate transcription of ribosomal protein and rosis complex, as discussed below. In the active conformation,

8.4 Cytoplasmic Mechanisms of Post-transcriptional Control 377


the TSClfTSC2 heterodimer functions as a GTPase activat- High mTOR protein kinase activity in tumors correlates
ing protein for Rheb (Rheb-GAP), causing hydrolysis of the with a poor clinical prognosis. Consequently, mTOR inhibi-
Rheb-bound GTP to GDP. This converts Rheb to its GOP- tors are currently in clinical trials to test their effectiveness for
bound conformation, which binds to the mTOR complex treating cancers in conjunction with other modes of therapy.
and inhibits its kinase activity. Finally, the activity of the Rapamycin and other structurally related mTOR inhibitors
TSClfTSC2 Rheb-GAP is regulated by several inputs, allow- are potent suppressors of the immune response because they
ing the cell to integrate different cellular signaling pathways to inhibit activation and replication ofT lymphocytes in response
control the overall rate of protein synthesis. Signaling from cell- to foreign antigens (Chapter 23). Several viruses encode pro-
surface growth factor receptors leads to phosphorylaLiun of teins that activate mTOR early after viral infection. The result-
TSClfTSC2 at inhibitory sites, causing an increase in RhebGTP ing stimulation of translation has an obvious selective advantage
and activation of mTOR kinase activity. This type of regula- for these cellular parasites.
tion through cell-surface receptors links the control of cell
growth to developmental processes controlled by cell-cell eiF2 Kinases eiF2 kinases also regulate the global rate of cel-
interactions. lular protein synthesis. Figure 4-24 summarizes the steps in
mTOR activity also is regulated in response to nutritional translation initiation. Translation initiation factor e!F2
status. When energy from nutrients is not sufficient for cell brings the charged initiator tRNA to the small ribosome sub-
growth, the resulting fall in the ratio of ATP to AMP concen- unit P site. e!F2 is a trimeric G protein and consequently
trations is detected by AMP kinase (AMPK). The activated exists in either a GTP-bound or a GDP-bound conforma-
AMP kinase phosphorylates TSC1fTSC2 at activating sites, tion. Only the GTP-bound form of elf2 is able to bind the
stimulating its Rheb-GAP activity and consequently inhibit- charged initiator tRNA and associate with the small ribo-
ing mTOR kinase activity and the global rate of translation. somal subunit. The small subunit with bound initiation fac-
Hypox1a and other cellular stresses also activate the TSCll tors and charged initiator tRNA then interacts with the eiF4
TSC2 Rheb-GAP. Finally, the concentration of nutrients in complex bound to the 5' cap of an mRNA via its eiF4E sub-
the extracellular space also regulates Rheb, by an unknown unit. The small ribosomal subunit then "scans down the
mechanism that does not require the TSClfTSC2 complex. mRNA in the 3' direction until it reaches an AUG initiation
In addition to regulating the global rate of cellular pro- codon that can base-pair with the initiator tRNA in its P site.
tein synthesis and the production of ribosomes, tRNAs, and When this occurs, the GTP bound by eiF2 is hydrolyzed to
' '
translation factors, mTOR regulates at least one other pro- GDP and the resulting elF2 GDP complex is released. GTP
cess involved in the response to low levels of nutrients: mac- hydrolysis results in an irreversible "proofreading" step that
roautophagy (or simply autophagy). Starved cells degrade prepares the small ribosomal subunit to associate with the
cytoplasmic constituents, including whole organelles, to sup- large subunit only when an initiator tRNA is properly bound
ply energy and precursors for essential cellular processes. in the P site and is properly base-paired with the AUG start
During this process a large, double-membrane structure en- codon. Before ciF2 can participate in another round of ini-
gulfs a reg10n ~f cytoplasm to form an autophagosome, tiation, its bound GDP must be replaced with a GTP. This
which then fuses with a lysosome where the entrapped pro- process is catalyzed by the translation initiation factor eiF2B,
teins, lipids, and other macromolecules are degraded, com- a guanine nucleotide exchange factor (GEF) specific for eiF2.
pleting the process of macroautophagy. Activated mTOR A mechanism for inhibiting general protein synthesis in
inhibits macroautophagy in growing cells when nutrients are stressed cells involves phosphorylation of the eiF2 o: subunit
plentiful. Macroautophagy is stimulated when mTOR activ- at a specific serine. Phosphorylation at this site does not in-
ity falls in nutrient-deprived cells. terfere with ciF2 function in protein synthesis directly.
Rather, phosphorylated eiF2 has very high affinity for the
Genes encoding components of the mTOR pathway elF2 guanine nucleotide exchange factor, eiF2B, which can-
are mutated in many human cancers, resulting in cell not release the phosphorylated eiF2 and consequently is
growth in the absence of normal growth signals. TSCl and blocked from catalyzing GTP exchange of additional elf2
TSC2 (see Figure 8-30) were initially identified because one factors. Since there is an excess of eiF2 over eiF2B, phos-
or the other of the proteins is mutant in a rare human genetic phorylation of a fraction of elF2 results in inhibition of all
syndrome: tuberous sclerosis complex. Patients with this dis- the cellular eiF2B. The remaining ciF2 accumulates in its
order develop benign tumors in multiple tissues. The disease GDP-bound form, which cannot participate in protein syn-
results because inactivation of either TSC1 or TSC2 elimi- thesis, thereby inhibiting nearly all cellular protein synthesis.
nates the Rheb-GAP activity of the TSC1!TSC2 heterodi- However, some mRNAs have 5' regions that allow transla-
mer, resulting in an abnormally high and unregulated level tion initiation at the low elF2-GTP concentration that rc
of RhebGTP and the resulting high, unregulated activity of suits from elf2 phosphorylation. These mRNAs include
mTOR. Mutations in components of cell-surface receptor those for chaperone proteins that function to refold cellular
signal-transduction pathways that lead to inhibition ofTSCl/ proteins denatured as the result of cellular stress, additional
TSC2 Rheb-GAP activity are also common in human tumors proteins that help the cell to cope with stress, and transcrip-
and contribute to cell growth and replication in the absence tion factors that activate transcription of the genes encoding
of normal signals for growth and proliferation. these stress-induced proteins.

378 CHAPTER 8 Post-transcriptional Gene Control


Human cells contain four eiF2 kinases that phosphor- Control of intracellular iron concentration by the iron
ylate the same inhibitory eiF2a serine. Each of these is regu- response element-binding protein (JRE-BP) is an elegant ex-
lated by a different type of cellular stress, inhibiting protein ample of a single protein that regulates the translation of one
synthesis and allowing cells to divert the large fraction of mRNA and the degradation of another. Precise regulation
cellular resources usually devoted to protein synthesis in of cellular iron ion concentration is critical to the cell. Mul-
growing cells for use in responding to the stress. tiple enzymes and proteins contain Fe2+ as a cofactor, such as
The GCN2 (general control non-dercpressible 2) elf2- enzymes of the Krebs cycle (see Figure 12-1 0) and electron-
kinase is activated by binding uncharged tRNAs. The con- carrying proteins involved in the genera non of ATP by mito-
centration of unch::~rged tRNAs increases when cells are chonJria and chloroplasts (Chapter 12). On the other hand,
starved for amino acids, activating GCN2 eiF2-kinase activ- excess Fe2 generates free radicals that react with and dam-
ity and greatly inhibiting protein synthesis. age cellular macromolecules. When intracellular iron stores
PEK (pancreatic eiF2 kinase) is activated when proteins are low, a dual-control system operates to increase the level
translocated into the endoplasmic reticulum (ER) do not fold of cellular iron; when iron is in excess, the system operates
properly because of abnormalities in the ER lumen environ- ro prevent accumulation of toxic levels of free ions.
ment. Inducers include abnormal carbohydrate concentration, One component in this system is the regulation of the
because this inhibits the glycosylation of many ER proteins, production of ferritin, an intracellular iron-binding protem
and inactivating mutations in an ER chaperone required for that binds and stores excess cellular iron. The 5' untranslated
proper folding of many ER proteins (Chapters 13 and 14). region of ferritin mRNA contains iron-response elements
Heme-regulated inhibitor (HRI) is activated in develop- (IREs) that have a stem-loop structure. The IRE-binding pro-
ing red blood cells when the supply of the heme prosthetic tein (IRE-BP) recognizes five specific bases in the IRE loop
group is too low to accommodate the rate of globin protein and the duplex nature of the stem. At low iron concentra-
synthesis. This negative feedback loop lowers the rate of tions, IRE-BP is in an active conformation that binds to the
globin protein synthesis until it matches the rate of heme syn- IREs (Figure 8-Jla). The bound IRE-BP blocks the small ri-
thesis. HRI is also activated in other types of cells in response bosomal subunit from scanning for the AUG start codon (see
._ to oxidative stress or heat shock . Figure 4-24 ), thereby inhibiting translation initiation. The
Finally, protein kinase RNA activated (PKR) is activated resulting decrease in ferritin means less iron is complexed
by double-stranded RNAs longer than -30 base pairs. Under with the ferritin and therefore more is available to iron-
normal circumstances in mammalian cells, such double- requiring enzymes. At high iron concentrations, IRE-BP is in
stranded RNAs are produced only during a viral infection. an inactive conformation that does not bind to the 5' IREs,
Long regions of double-stranded RNA are generated in repli- so translation initiation can proceed. The newly synthesized
cation intermediates of RNA viruses or from hybridization of ferritin then binds free iron ions, preventing their accumula-
complementary regions of RNA transcribed from both tion to harmful levels.
strands of DNA virus genomes. Inhibition of protein synthe- The other part of this regulatory system controls the im-
sis prevents the production of progeny virions, protecting port of iron into cells. In vertebrates, ingested iron is carried
neighboring cells from infection. Interestingly, adenoviruses through the circulatory system bound to a protein called
evolved a defense against PKR: they express prodigious transferrin. After binding to the transferrin receptor (TfR) tn
amounts of an -1 60-nucleotide virus-associated (VA) RNA the plasma membrane, the transferrin-iron complex is
with long double-stranded hairpin regions. VA RNA is tran- brought into cells by receptor-mediated endocytosis (Chap-
scribed by RNA polymerase !II and exported from the nu- ter 14 ). The 3' untranslated region of TfR mRNA contains
cleus by Exportin5, the exportin for pre-miRNAs (see Figure IREs whose stems have AU-rich destabilizing sequences (Fig-
8-27). VA RNA binds to PKR with high affinity, inhibiting ure 8-Jlb). At high iron concentrations, when the IRE-BP is
its protein kinase activity and preventing the inhibition of in the inactive, nonbinding conformation, these AU-rich se-
protein synthesis observed in cells infected with a mutant quences promote degradation of TfR mRNA by the same
adenovirus from which the VA gene was deleted. mechanism that leads to rapid degradation of other short-
lived mRNAs, as described previously. The resulting de-
crease in production of the transferrin receptor quickly
Sequence-Specific RNA-Binding Proteins
reduces iron import, thus protecting the cell from excess
Control Specific mRNA Translation iron. At low iron concentrations, however, IRE-BP can bind
In contrast to global mRNA regulation, mechanisms have to the 3' IREs in TfR mRNA. The bound IRE-BP blocks
also evolved for controlling the translation of certain specific recognition of the destabilizing AU-rich sequences by the
mRNAs. This is usually done by sequence-specific RNA- proteins that would otherwise rapidly degrade lhc mRNAs.
binding proteins that bind to a particular sequence or RNA As a result, production of the transferrin receptor increases
structure in the mRNA. When binding is in the 5' untrans- and more iron is brought into the cell.
lated region (5' UTR) of an mRNA, the ribosome's ability to Other regulated RNA-binding proteins may also func-
scan to the first initiation codon is blocked, inhibiting trans- tion to control mRNA translation or degradation, much like
lation initiation. Binding in other regions can either promote the dual-acting IRE-BP. For example, a heme-sensitive RNA-
or inhibit mRNA degradation. binding protein controls translation of the mRNA encoding

8.4 Cytoplasmic Mechanisms of Post-transcriptional Control 379


(a) Ferritin mRNA their degradation by the exosome, and the general restriction
IREs coding reg1on COOH against nuclear export of incompletely spliced pre-mRNAs

~
that remain associated with a spliceosome.
High iron H2 "! Another mechanism called nonsense-mediated decay
5' ~B!!IIIl!II!!!D- An ~ ~ (NMD) causes degradation of mRNAs in wh ich o ne or more
exons have been incorrectly spliced. Such incorrect splicing
Inactive IRE-BP ~ Translated
often will alter the open reading frame of the mRNA 3' to the

Active IRE-BP Q 1 ferritin improper exon junction, resulting in introduction of an out-of-


frame missense mutation and an incorrect stop codon. For
nearly all properly spliced mRNAs, the stop codon is in the last
exon. The process of nonsense-mediated decay resu lts in the
Low iron rapid degradation of mRNAs with stop codons that occur be-
fore the last splice junction in the mRNA since in most cases,
5' L --llir!'mlnl!mlml- An -If--+ No translation
such mRNAs arise from errors in RNA splicing. However,
initiation

v
NMD can also result from a mutation creating a stop codon
(b) TfR mRNA IREs within a gene or a frame-shifting deletion or insertion. NMD

..::.:~~;.. gJ1
AU-rich region was initially discovered during the'study of patients with 13-
~ ,,,~~ thalasemia, who produce a low level of 13-globin protein asso-
ciated with a low level of 13-globin mRNA (Figure 8-32a, b).
5 A ---+ ',,.,.,
n 1"':,'- J 1 A search for possible molecular signals that might indi-
( .1( - ,

Inactive IRE-BP ~ I,.,,. --:._, cate the positions of splice junctions in a processed mRNA

01
Degraded led to the discovery of exon-junction complexes. As noted
mononucleotides
already, these complexes of several proteins. (including Y14,
Active IRE-BP
Magoh, eiF4IIIA, UPF2, UPF3, and REF), bind -20 nucle-
otides 5' to an exon-exon junction following RNA splicing

~ ~ ~
(Figure 8-32c), stimulate export of mRNPs from the nucleus
Low iron
by interacting with the mRNA exporter (see Figure 8-21 ).
5' .ii!ffiii.I.WQ An -If--+ Little Analysis of yeast mutants indicated that one of the proteins
degradation
in exon-junction complexes (UPF3) functions in nonsense-
FIGURE 8 -31 Iron-dependent regulation of mRNA translation mediated decay. In the cytoplasm, this component of exon-
and degradation. The iron response element-binding protein (IRE-BP) j unction complexes interacts with a protein (UPFl) and a
controls (a) translation of ferritin mRNA and (b) degradation of protein kinase (SMGl) that phosphorylates UPFl, causing
transferrin-receptor (TfR) mRNA. At low intracellular iron concentra-
the mRNA to associate with P bodies, repressing translation
tions IRE-BP binds to iron-response elements (IREs) in the 5' or 3'
of the mRNA. An additional protein (UPF2) associated with
untranslated regiori of these mRNAs. At high iron concentrations,
the mRNP complex binds a P-body associated deadenylase
IRE-BP undergoes a conformational change and cannot bind either
mRNA. The dual control by IRE-BP precisely regulates the level of free
that rapidly removes the poly(A) tail from an associated
iron ions within cells. See the text for discussion.
mRNA, leading to its rapid decapping and degrada tion by
the P-body associated 5' ~3' exonuclease (see Figure 8-29).
In the case of properly spliced mRNAs, the exon-junction
complexes associate with the nuclear cap-binding complex
aminolevulinate (ALA) synthase, a key enzyme in the synthe- (CBP80, CBP20) as the mRNP is transported through a nu-
sis of heme. Similarly, in vitro studies have shown that the clear pore complex, thereby protecting the mRNA from deg-
mRNA encoding the milk protein casein is stabilized by the radation. The exon-junction complexes arc thought to be
hormone prolactin and rapidly degraded in its absence. dislodged from the mRNA by passage of the first "pioneer"
ribosome to translate the mRNA. However, for mRNAs with
a stop codon before the final exon junction, one or more
Surveillance Mechanisms Prevent Translation
exon-junction complexes remain associated with the mRNA,
of Improperly Processed mRNAs resulting in nonsense-mediated decay (Figure 8-32).
Translation of an improperly processed mRNA could lead to
production of an abnormal protein that interferes with the Localization of mRNAs Permits Production
gene's normal function. This effect is equiva lent to that re
of Proteins at Specific Regions
suiting from dominant-negative mLJtations, discussed in
Chapter 5 (Figure 5-44). Several mechanisms collectively Within the Cytoplasm
termed mRNA surveillance help cells avoid the translation of Many cellular processes depend on localization of particular
improperly processed mRNA molecules. We have previously proteins to specific structures or regions of the cel l. In later
mentioned two such surveillance mechanisms: the recogni- chapters we examine how some proteins are transported after
tion of improperly processed pre-mRNAs in the nucleus and their synthesis to their proper cellular location. Alternatively,

380 CHAPTER 8 Post-transcriptional Gene Control


(a) (c) EJC
Exon-exon
CG deletion CBP80
Initiation of
. CBP20
p1oneer round m7 lip')
~-Globin of translation Gppp
genomic -20-24 nt Norm Ter
t t
DNA
AUG Poly(A)
site l
(b) CBP80 and
UPF1

.. ..
wt ~- ~0- transiently
globin thalasemia or weakly
interact PTC
- + - + Act D

+-
CBP80 and UPF1
promote SMG1-UPF1
binding to eRF1-eRF3
to promote SURF
complex formation

CBP80 and UPF1


promote SMG1-UPF1
binding to a
PTC-distal EJC

l
SMG1 CBP80
phosphorylates CBP20
m Gppp llip')-~--11!11-lllild ..I!II~AAAAA
l
7
UPF1
t t t
AUG PTC Norm Ter

Translational repression and mRNA decay

FIGURE 8-32 Discovery of nonsense-mediated mRNA decay (3-thalasemia had much less (3-globin mRNA than the patient with
(NMD). (a) Patients with (3-thalasemia express very low levels of a wild-type (3-g lobin gene (- Act D). The mutant (3-globin mRNA
(3-globin mRNA. A common cause of this syndrome is a single-base- decayed rapidly when transcription was inhibited ( + Act D), whereas
pair deletion in exon 1 or exon 2 of the (3-globin gene. Ribosomes the wild-type (3-globin mRNA remained stable. (c) Current model of
translating the mutant mRNA read out of frame following the deletion NMD. PTC. premature termination codon. Norm Term, the normal
and encounter a stop codon in the wrong reading frame before they termination codon. SURF, complex of protein kinase SMG1 , UPF1 , and
translate across the last exon junction in the mRNA. Consequently, translation termination factors eRF1 and eRF3. Formation of the SURF
they do not displace an exon-junction complex (EJC) from the mRNA. complex leads to phosphorylation of UPF1 and association of
Cytoplasmic proteins associate with the EJC and induce degradation phospho-UPF1 with a UPF2-UPF3 complex bound to any exon-exon
of the mRNA. (b) Bone marrow was obtained from a patient with a junction complexes that were not displaced from the mRNA by the
wild-type (3-globin gene and from a patient with (3-thalasemia. RNA first, pioneer ribosome to translate the message. This leads to the
was isolated from the bone marrow cells shortly after collection, or association ofthe PTe-containing mRNA with P-bodies, removal of the
30 min after incubation in media with Actinomycin D, a drug that poly(A) tail, and degradation of the mRNA. [Part (b) from L. E. Maquat et al.,
inhibits transcription. The amount of (3-globin RNA was measured 1981, Ce// 27:543. Part (c) adapted from J. Hwang et al., 2010, Mol. Ce//39:396.]
using the S1-nuclease protection method (arrow). The patient with

protein localization can be achieved by localization of mRNAs of 3000 mRNAs analyzed were localized to specific subcellular
to specific regions of the cytoplasm in which their encoded pro- regions, raising the possibility that this is a much more general
teins function. In most cases examined thus far, such mRNA phenomenon than previously appreciated.
localization is specified by sequences in the 3' untranslated re-
gion of the mRNA. A recent genomic-level study of mRNA Localization of mRNAs to the b ud in S. cerevisiae The most
localization in Drosophila embryos revealed that -70 percent thoroughly understood example o f mRNA localization occurs

8.4 Cytop lasmic Mechanisms of Post-tran sc riptional Control 381


FIGURE 8-33 Switching of mating type in haploid yeast cells. (a)
(a) Division by budding forms a larger mother cell (M) and smaller
~Bud
daughter cell (D), both of which have the same mating type as the
original cell (a in this example). The mother cell can switch mating type
during G1 of the next cell cycle and then divide again, producing two / \Division
cells ofthe opposite type (a in this example). Switching depends on
transcription of the HO gene, which occurs only in the absence of Ashl
protein. The smaller daughter cells, which produce Ash 1 protein,
MG) G) D
cannot switch; after growing in size through interphase, they dividP to
form a mother cell and daughter cell. (b) Model for restriction of
HO transcripti~shl r j
Switching /
mating-type switching to mother cells in 5. cerevisiae. Ashl protein
prevents a cell from transcribing the HO gene whose encoded protein
initiates the DNA rearrangement that results in mating-type switching
from a to ex or ex to a. Switching occurs only in the mother cell, after it
M0 G)D
separates from a newly budded daughter cell, because the Ashl /1
protein is present only in the daughter cell. The molecular basis for this
differential localization of Ash 1 is the one-way transport of ASH I mRNA
into the bud. A linking protein, She2, binds to specific 3'untranslated
88 M D
a

M
sequences in the ASH I mRNA and also binds to She3 protein. This
protein in turn binds to a myosin motor, Myo4, which moves along (b)
actin filaments into the bud. [SeeS. Koon and B. J. Schnapp, 2001, Curr.
Biology 11 :R166.]

Ash1
mRNA

>---c~~
in the budding yeastS. cerevisiae. As discussed in Chapter 7,
whether a haploid yeast cell exhibits the a or a mating type
is determined by whether a or a genes are present at the ex-
pressed MAT locus on chromosome III (see Figure 7-33). ~ She2
.... J9 ~~
Myo4 ""
The process that transfers a or a genes from the silent mating- Actin
type locus to the expressed MAT locus is initiated by a se-
quence-specific endonuclease ca lled HO. Transcription of
the HO gene is dependent on the SWI/SNF chromatin-
remodeling complex (see Chapter 7, Section 7.5). Daughter
yeast cells arising by budding from mother cells contain a
transcriptional repressor called Ash 1 (for Asymmetric syn-
thesis of HO) that prevents recruitment of the SWIISNF tion signal to which She2 binds, usually in their 3' UTR. The
complex to the HO gene, thereby preventing its transcrip- process can be visualized in live cells by the experiment
tion. The absence of Ashl from mother cells allows them to shown in Figure 8-34. RNAs can be fluorescently labeled by
transcribe the HO gene. As a consequence mother cells including in their sequence high-affinity binding sites for
switch their mating type, while daughter cells generated by RNA-binding proteins, such as bacteriophage proteins MS2
budding do not (Figure 8-33a). coat protein and bacteriophage }..N protein, that bind to dif-
Ashl protein accumulates only in daughter cells because ferent stem loops of specific sequence (Figure 8-34a). When
the mRNA encoding it is localized to daughter cells. The lo- such engineered mRNAs are expressed in budding yeast cells
calization process requires three proteins: She2 (for SWI- along with the bacteriophage proteins fused to proteins that
dependent HO expression), an RNA-binding protein that fluoresce different colors, the fusion proteins bind to these
binds specifically to a localization signal with a specific RNA specific RNA sequences, thereby labeling the RNAs that
structure in the ASHl mRNA; Myo4, a myosin motor pro- contain them with different colors. In the experiment shown
tein that moves cargos on actin filaments (see Chapter 17); in Figure 8-34b, ASHl mRNA was labeled by the binding of
and She3, which links She2 and therefore ASHl mRNA to green fluorescent protein fused to ;\N. Another mRNA local-
Myo4 (Figure 8-33b). ASHl mRNA is transcribed in the nu- ized to the bud by this system, the IST2 mRNA encoding a
cleus of the mother cell before mitosis. Movement of Myo4 component of the growing bud membrane, was labeled by
with its bound ASHl mRNA along actin filaments that ex- the binding of red fluorescent protein fused to M$2 coat
tend from the mother cell into the bud carries the ASHl protein. Video of a budding cell showed that the differently
mRNA into the growing bud before cell division. labeled ASHl and IST2 mRNAs accumulated in the same
At least 23 other mRNAs were found to be transported large cytoplasmic RNP particle containing multiple mRNAs
by the She2, She3, Myo4 ~ystem. All have an RNA localiza- in the mother cell cytoplasm, as can be seen from the merge

382 CHAPTER 8 Post-transcriptional Gene Control


(a) Binding sites for GFP-f..N

A
(b)

, ' , " , ' , .." , " '


ASH1 m RNA I
~ ' I
.. - ;

...
~

. M ' '..
:

,, .,
Binding sites for RFP-MS2
N
(/)
::E " " ' 't
" 'l " "
~ N

, ,.
1-
!a

... " ' , ".


, "
'.. ... ' '
AAAA
" "
I

'
t
1""1 ,.
I j I

') .~
I

,.

EXPERIMENTAL FIGURE 834 Transport of mRNP particles the right, GFP-A.N and RFP-MS2 were independently visualized by using
from a yeast mother cell into the bud. (a) Yeast cells were engineered millisecond alternating laser excitation of GFP and RFP. (b) Frames from
to express an ASH1 mRNA with binding sites for the bacteriophage A.N a video of fluorescing cells are shown. The nucleus next to the large
protein in its 5' untranslated region and an IST2 mRNA with binding vacuole in the mother cell near the center of the micrographs, as well
sites for bacteriophage MS2 coat protein in its 3' untranslated region. A as nuclei in neighboring cells, was observed by green and red
fusion of green fluorescent protein to A.N protein (GFP-A.N) and a fusion fluorescence as shown in the top and middle rows. A merge of the two
of red fluorescent protein to MS2 coat protein (RFP-MS2) also were images is shown in the bottom row, which also indicates the time
expressed in the same cells. In other experiments, these fluorescently elapsed between images. An RNP particle containing both the ASH 1
tagged sequence-specific RNA-binding proteins were shown to bind mRNA with A.N-binding sites and the IST2 mRNA with MS2-binding
to their own specific binding sites engineered into the ASH1 and IST2 sites was observed in the mother cell cytoplasm in the left column of
mRNAs, and not to each others' binding sites. Both fluorescently images (arrow). The particle increased in intensity between 0.00 and
tagged proteins also contained a nuclear localization signal so that the 46.80 seconds, indicating that more of these mRNAs joined the RNP
fluorescent proteins that were not bound to their high-affinity binding particle. The RNP particle was transported into the bud between 46.80
sites in these mRNAs were transported into nuclei through nuclear and 85.17 seconds and then became localized to the bud tip.
pore complexes (see Chapter 13). This was necessary to prevent high [From 5. Lange et al., 2008, Traffic 9:1256. See this paper to v1ew the v1deo.]
fluorescence from excess GFP-A.N and RFP-MS2 in the cytoplasm. At

of the green and red fluorescent signals. The RNP particle short poly(A) tails that do not allow translation initiation.
was then transported into the bud within about one minute. Once again, large RNP particles containing multiple mRNAs
bearing localization signals form in the cytoplasm near the
Localization of mRNAs to synapses in the mammalian ner- cell nucleus. In this case, the RNP particles arc transported
vous system As mentioned earlier, in neurons, localization down the axon to synapses by kinesin motor proteins that
of specific mRNAs at synapses far from the nucleus in the cell travel down microtubules extending the length of the axon
body plays an essential function in learning and memory (Fig- (see Chapter 18). Electrical activity at a given synapse may
ure 8-35). Like the localized mRNAs in yeast, these mRNAs
contain RNA localization signals in their 3' untranslated re-
gion. Some of these mRNAs are initially synthesized with EXPERIME TAL FIGURII:: 835 A specific neuronal mRNA
localizes to synapses. Sensory neurons from the sea slug Ap/ysia
californica were cultured with target motor neurons so that processes
from the sensory neurons formed synapses with processes from the
motor neurons. The micrograph at the left shows motor neuron
processes visualized with a blue fluorescent dye. GFP-VAMP (green)
was expressed in sensory neurons and marks the location of synapses
formed between sensory and motor neuron processes (arrows). The
micrograph on the right shows red fluorescence from in situ hybridiza-
tion of an antlsensonn mHNA probe. Sensorin is a neurotransmitter
expressed by the sensory neuron only; sensory neuron processes are
not otherwise visualized in this preparation, but they lie adjacent to the
motor neuron processes. The in situ hybridization results indicate that
sensorin mRNA is localized to synapses. [From V. Lyles, Y. Zhao, and K. C.
Martin, 2006, Neuron 49:323.]

8.4 Cytoplasmic Mechanisms of Post-transcriptional Control 383


then stimulate the mRNAs' polyadcnylation in the region
of the synapse, activating the translation of encoded proteins Many mRNAs are transported to specific subcellular loca-
that increase the size and alter the neurophysiological proper- tions by sequence-specific RNA-binding proteins that bind
tics of the one synapse whi le leaving unaffected the hundreds to localization sequences usually found in the 3' UTR. These
thousands of other synapses made by the neuron. RNA-binding proteins then associate directly or via interme-
diary proteins with motor proteins that carry large RNP
complexes w ith many mRNAs bearing the same localization
signal on actin or microtubule fibers to specific locations in
KEY CONCEPTS of Section 8.4 the cytoplasm.
Cytoplasmic Mechanisms of
Post-transcriptional Control
Translation can be repressed by micro RNAs (miRNAs),
which form imperfect hybrids with sequences in the 3' un-
translated region (UTR) of specific target mRNAs. 8.5 Processing of rRNA and tRNA
~ The related phenomenon of RNA interference, which Approximately 80 percent of the total RNA in rapidly grow-
probably evolved as an early defense system against viruses ing mammalian cells (e.g., cultured,HeLa cells) is rRNA, and
and transposons, leads to degradation of mRNAs that form 15 percent is tRNA; protein-coding mRNA thus constitutes
perfect hybrids with short interfer ing RNAs (siRNAs). only a small portion of the total RNA. The primary tran-
Both miRNAs and siRNAs contain 21-23 nucleotides, are scripts produced from most rRNA genes and from tRNA
generated from longer precursor molecules, and are bound genes, like pre-mRNAs, are extensively processed to yield
by an Argonaute protein and assembled into a multiprotein the mature, functional forms of these RNAs.
RNA-induced silencing complex (RISC) that either represses The ribosome is a highly evolved, complex structure (see
translation of target mRNAs or cleaves them (see figures Figure 4-23 ), optimized for its function in protein synthesis.
8-25 and 8-26). Ribosome synthesis requires t he function and coordination of
all three nuclear RNA polymerases. The 28S and 5.8S rRNAs
Cytoplasmic polyadenylation is required for translation of associated with the large ribosomal subunit and the single
mRNAs with a short poly( A) tail. Binding of a specific protein 185 rRNA of the small subunit are transcribed by RNA
to regu latory elements in their 3' UTRs represses translation
polymerase I. The 5S rRNA of the large subunit is transcribed
of these mRNAs. Phosphorylation of this RNA-binding pro- by RNA polymerase Ill, and the mRNAs encoding the ribo-
tein, induced by an external signal, leads to lengthening of somal proteins are transcribed by RNA polymerase II. In ad-
the 3' poly(A) tail and thus translation (see Figure 8-28). dition to the four rRNAs and -70 ribosomal proteins, at
Most mRNAs are degraded as the result of the gradual least 150 other RNAs and proteins interact transiently with
shortening of their poly(A) tail (deadenylation) followed by the two ribosomal subunits during their assembly through a
exosome-mediated 3' ~5 digestion, or removal of the 5' cap series of coordinated steps. Furthermore, multiple specific
and digestion by a 5'~3' exonuclease (see Figure 8-29). bases and riboses of the mature rRNAs are modified to opti-
Eukaryotic mRNAs encoding proteins that are expressed mize their function in protein synthesis. Although most of the
in short bursts generally have repeated copies of an AU-rich steps in ribosomal subunit synthesis and assembly occur in
sequence in their 3' UTR. Specific proteins that bind to these the nucleolus (a subcompartment of the nucleus not bounded
elements also interact with the deadenylating enzyme and by a membrane), some occur in the nucleoplasm during pas-
cytoplasmic exosomes, promoting rapid RNA degradation. sage from the nucleolus to nuclear pore complexes. A quality-
control step occurs before nuclear export so that only fully
Binding of various proteins to regulatory elements in the functional subunits are exported to the cytoplasm, where the
3' or 5' UTRs of mRNAs regulates the translation or degra- final steps of ribosome subunit maturation occur. tRNAs also
dation of many mRNAs in the cytoplasm. are processed from precursor primary transcripts in the nu-
Translation of ferritin mRNA and degradation of trans- cleus and modified extensively before they are exported to
ferrin receptor (TfR) mRNA are both regulated by the same the cytoplasm and used in protein synthesis. First we'll dis-
iron-sensitive RNA-binding protein. At low iron concentra- cuss the processing and modification of rRNA and the as-
tions, this protein is in a conformation that binds to specific sembly and nuclear export of ribosomes. Then we'll consider
elements in the mRNAs, inhibiting ferritin mRNA transla- the processing and modification of tRNAs.
tion or degradation of TfR mRN A (see Figure 8-31 ). This
dual control precisely regulates the iron level within cells.
Pre-rRNA Genes Function as Nucleolar
Nonsense-mediated decay and other mRNA surveillance Organizers and Are Similar in All Eukaryotes
mechanisms prevent the translation of improperly processed
mRNAs encoding abnormal proteins that might interfere The 28S and 5.8S rRNAs associated with the large (60S)
with functioning of the corresponding normal proteins. ribosomal subunit and the 18S rRNA associated with the
small (405) ribosomal subunit in higher eukaryores (a nd the

384 CHAPTER 8 Post-transcriptional Gene Control


..
I '
The synthesis and most of the processing of pre-rRNA
occurs in the nucleolus. When prc-rRNA genes initially were
identified in the nucleolus by in situ hybridization, it was not
known whether any other DNA was required to form the
nuc leolus. Subsequent experiments with transgenic Dro-
sophila strains demonstrated that a single complete pre-
Transcription rRNA transcription unit induces formation of a sma ll
unit
nucleolus. Thus a single pre-rRNA gene is sufficient to be a
nucleolar organizer, and all the OLher components of the ri-
bosome d iffuse to the newly formed prc-rRNA. The structure
of the nucleolus observed by light and electron microscopy
results from the processing of pre-RNA and the assembly of
ribosomal subunits.

Nont ranscribed
} spacer Small Nucleolar RNAs Assist
in Processing Pre-rRNAs
Ribosomal subunit assembly, maturation, and export to the
cytoplasm are best understood in the yeast S. cereviswe.
However, nearly al l the proteins and RNAs involved are
Transcription highly conserved in multicellular eukaryotes, where the fun-
unit damental aspects of ribosome biosynthesis are likely to be
the same. As for pre-mRNAs, nascent pre-rRNA transcripts
arc immediately bound by proteins, forming preribosomal
ribonucleoprotein particles (pre-rRNPs). For reasons not yet
known, cleavage of the pre-rRNA does not begin until tran-
EXPERIMENTAL FIGURE 836 Electron micrograph of scription of the pre-rRNA is nearly complete. In yeast, it
pre-rRNA transcription units from the nucleolus of a frog oocyte. takes approximately six minutes for a pre-rRNA to be tran-
Each "feather represents multiple pre-rRNA molecules associated with scribed. Once transcription is complete, the rRNA is cleaved,
protein in a pre-ribonucleoprotein complex (pre-rRNP) emerging from and bases and riboses are modified in about 10 seconds. In a
a transcription unit. Note the dense " knob" at the 5' end of each rapidly growing yeast cell, -40 pairs of ribosomal subumts
nascent pre-RNP thought to be a processome. Pre-rRNA transcription are synthesized, processed, and transported to the cytoplasm
units are arran ged in tandem, separated by nontranscribed spacer every second. This extremely high rate of ribosome synthesis
regions of nucleolar chromatin. [Courtesy of Y. Osheim and 0. J. Miller, Jr.]
despite the seemingly long period required to transc ribe a
pre-rRNA is possible because pre-rRNA genes are packed

functionally equivalent rRNAs in all other eukaryotes) are


185 5.85 285
encoded by a single type of p re-rRNA transcription unit. In
Human, - 13.7 kb
human cells, transcription by RNA polymerase I yields a 455 3'
5'
(-13.7 kb) primary transcript (pre-rRNA), which is processed
into the mature 285, L8S, and 5.85 rRNAs found in cytoplas- X. laevis (frog), -7.9 kb
mic ribosomes. The fourt11 rRNA, 55, is encoded separately
and transcribed outside the nucleolus. Sequencing of the D. melanogaster (fruit fly), -7.7 kb
DNA encoding the 455 pre-rRNA from many species showed
that this DNA shares several properties in all eukaryotes. First, S. cerevisiae (yeast), - 6.6 kb
the pre-rRNA genes are arranged in long tandem arrays sepa-
rated by nontranscribed spacer regions ranging in length from - Transcribed spacer
- 2 kb in frogs to -30 kb in humans (Figure 8-36). Second, the
- Region preserved in rRNA
genomic regions corresponding to the three mature rRNAs
are always arranged in the same 5'~3' order: 185, 5.85, and FIGURE 8-37 General structu re of eukaryotic pre-rRNA t ran-
285. T hird, in all eukaryotic cells (and even in bacteria), the scription units. The three coding regions (red) encode the 185, 5.85,
pre-rRNA gene codes for regions that are removed during and 285 rRNAs found in ribosomes of higher eukaryotes or their
processing and rapid ly degraded. These regions probably equivalents in other species. The order of these coding regions in the
contribute to proper folding of the rRNAs but are notre- genome is always 5 '~3'. Variations in the lengths of the transcribed
quired once the folding has occurred. The general structure of spacer regions (blue) account for the major difference in the lengths
pre-rRNAs is diagrammed in Figure 8-37. of pre-rRNA transcription units in different organisms.

8.5 Processing of rAN A and tRNA 385


with RNA polymerase I molecules transcribing the same involved in protein synthesis. The positions of the specific
gene simultaneously (see Figure 8-36) and because there are sites of 2' -0-methylation and pseudouridine formation are
I 00-200 such genes on chromosome XU, the yeast nucleolar determined by approximately 150 different small nucleolus-
organizer. restricted RNA species, called small nucleolar RNAs
The primary transcript of~ 7 kb is cut in a series of cleav- (snoRNAs), which hybridize transiently to pre-rRNA mole-
age and cxonuclcolytic steps that ultimately yield the mature cules. Like the snRNAs that function in prc-mRNA processing,
rRNAs found in nbosomes (Figure 8-38). During processing, snoRNAs associate with proteins, forming ribonucleopro-
pre-rRNA also is extensively modified, mostly by methyla- tein particles called snoRNPs. One class of more than 40
tion of the 2'-hyJroxyl group of specific riboses and conver- snoRNPs (contaming box C+D snoRNAs) positions a meth-
sion of specific uridine residues to pseudouridine. These yltransferase enzyme near methylation sites in the pre-
post-transcriptional modifications of rRNA are probably mRNA. The multiple different box C+ D snoRNAs direct
important for protein synthesis, because they are highly con- methylation at multiple sites through a similar mechanism.
served. Virtually all of these modifications occur in the most They share common sequence and structural features and
conserved core structure of the ribosome, which is directly are bound by a common set of proteins. One or two regions

Primary
transcript
,
5- - - -- - - -- - - -
~ 3'----+ e ,
.. Rat1
Co transcr~pt1onal l
..,,.'11~-rucleolvt.c cleavage

355 ------------~---------------

l
~vie
,_ C+D s~r- -
L.a.o..ArA

V CH3 CH 3

~ ~~
t ----~-----------------
1 ~ I
355
Exosome -t') ~
1 Cleav

335 J~----------------------
Xrn1 r >
Rat1 \.....>
~ Cleavag.e l

'\.IR., qJl
325 ------~--------

- - - - - - -- 275A2
Xrn1
Rat1
G e

Xrn1 ~

Cleavage on
cytoplasm
Rat1 ~ -1--------- 275A3
Exxuclease prccesson:. 1
27585 --:l 275BL ~:+------

Q :essong
vage
ress.ng
svage
Xrn1
75 5 - D 75L - t)
l Exosom~ l Exosom)

- + - +
185 5.855 255 or 5.85L 255
FIGURE 8-38 rRNA processing. Endoribonuc!eases that make rRNAs occurs following the initial cleavage at the 3 ' end, before the
internal cleavages are represented as scissors. Exoribonuc!eases that initial cleavage at the 5' end. Proteins and snoRNPs known to partici-
digest from one end, either 5' or 3', are shown as Pac-Men. Most pate in these steps are indicated. [From J. Venema and D. Tollervey, 1999,
2' -0-ribose methylation (CH 3) and generation of pseudouridines in the Ann. Rev. Genetics 33:261.)

386 CHAPTER 8 Post-transcriptional Gene Control


OH OH
Uridine

Box H Box ACA

OH OH
Pseudouridine

FIGURE 8 - 39 snoRNP-directed modification of pre-rRNA. (a) A in the stems. Pre-rRNA hybridizes to the single-stranded bulges,
snoRNA called box C +D snoRNA is involved in ribose 2'-0-methylation. demarcating a site of pseudouridylation. (c) Conversion from uridine
Sequences in this snoRNA hybridize to two different regions in the to pseudouridine directed by the box H + ACA snoRNAs of part (b ).
pre-rRNA, directing methylation at the indicated sites. (b) Box H + ACA [Part (a) from T. Kiss, 2001 , EMBOJ. 2 0:3617. Part (b) from U. T. Meier, 2005,
snoRNAs fold into two stem loops with internal single-stranded bulges Chromosoma 114:1 .]

of each of these snoRNAs are precisely complementary to Some snoRNAs are expressed from their own promoters
sites on the pre-rRNA and direct the methyltransferase to by RNA polymerase II or III. Remarkabl}, however, the
specific riboses in the hybrid region (Figure 8-39a). A second large majority of snoRNAs are processed from spliced-out
major class of snoRNPs (containing box H + ACA snoRNAs) intron s of genes encoding functional mRNAs for proteins
positions the enzyme that converts uridine to pseudouridine involved in ribosome synthesis or translation. Some snoRNAs
(Figure 8-39b). This conversion involves rotation of the py- are processed from imrons spliced from apparently nonfunc-
rimidine ring (Figure 8-39c). Bases on either side of the mod- tional mRNAs. The genes encoding these mRNAs seem to
ified uridine in the pre-rRNA base-pair with bases in the exist only to express snoRNAs from excised introns.
bulge of a stem in the H + ACA snoRNAs, leaving the modified Unlike 185, 5 .85, and 285 genes, 55 rRNA genes are
uridine bulged out of the helical double-manded region, like the transcribed by RNA polymerase Ill in the nucleoplasm out-
branch point A bulges out in pre-mRNA spliceosomal splicing side the nucleolus. With only minor additional processing to
(see Figure 8-1 0). Other modifications of pre-rRNA nucle- remove nucleotides at the 3' end, 55 rRNA diffuses to the
otides, such as adenine dimethylation, are carried out by spe- nucleolus, where it assembles with the pre-rRNA precursor
cific proteins without the assistance of guiding snoRNAs. and remains associated with the region that is cleaved into
The U3 snoRNA is assembled into a large snoRNP contain- the precursor of the large ribosomal subunit.
ing -72 proteins called the small subunit (SSU) processomc, Most of the ribosomal proteins of the small 40S ribosomal
which specifies cleavage at site A 0 , the initial cut near the 5' subunit associate with the nascent pre-rRNA during transcrip-
end of the pre-rRNA (see Figure 8-38). U3 snoRNA base-pairs tion (figure 8-40). Cleavage of the full-length pre-rRNA in the
with an upstream region of the pre-rl~.NA to specify the loca- 90S RNP precursor releases a pre-405 particle that requires
tion of the cleavage. The processome is thought to form the only a few more remodeling steps before it is transported to the
"5' knob" visible in electron micrographs of pre-rRNPs (see cytoplasm. Once the pre-405 particle leaves the nucleolus, it
Figure 8-36). Base pairing of other snoRNPs specify additional traverses the nucleoplasm quickly and is exported through nu-
cleavage reactions that remove transcribed spacer regions. The clear pore complexes (NPCs), as discussed below. final matu-
first cleavage to initiate processing of t he 5.85 and 255 rRNAs ration of t he small ribosomal subunit occurs in the cytoplasm:
of the large subunit is performed by RNase M RP, a complex exonucleol) tic processing of the 205 rRNA into mature small
of nine proteins with an RNA. Once cleaved from pre-rRNAs, subunit 185 rRNA by the cytoplasmic 5' ~3 ' exoribonuclease
these sequences arc degraded by the same exosome-associated Xrn 1 and the dimethylation of two adjacent adenines near the
3'~5' nuclear exonucleases that degrade introns spliced from 3' end of 185 rRNA by the cytoplasmic enzyme Diml.
pre-mRNAs. Nuclear 5'~3' exoribonucleases (Ratl; Xrnl) In contrast to the pre-405 particle, the precursor of the
also remove some regions of 5' spacer. large subunit requires considerably more remodeling through

8.5 Processing of rRNA and tRNA 387


Nucleolus Nucleoplasm Cytoplasm

Early Intermediate Late Mature

rONA

SSU processome

rRNA C) Helicases
RNA polymerase I 0 Intranuclear transport (Noc proteins)

0 () U3-associated factors 0 GTPases

U3-snoRNP o- AAA-type ATPase

0 rRNA processing/modification factors Export factors (Nmd3, Nxt1, Ran-GTP)

FIGURE 8-40 Ril?osomal subunit assembly. Ribosomal proteins subunits in the cytoplasm. Other factors that associate transiently with
and RNAs in the maturing small and large ribosomal subunits are the maturing subunits are depicted in different colors, as shown in the
depicted in blue, with a shape similar to the icons for the mature key. [From H. Tschochner and E. Hurt, 2003, Trends Cell Bio/.13:255.]

many more transient interactions with nonribosomal pro- occur in the nucleoplasm, during passage from the nucleolus
teins before it is sufficiently mature for export to the cyto- to nuclear pore complexes (see Figure 8-40). Much remains
plasm. Consequently, it takes a considerably longer period to be learned about the complex, fascinating, and essential
for the maturing 60S subunit to exit the nucleus (30 minutes remodeling processes that occur during formation of the ri-
compared to 5 minutes for export of the 40S subunit in cul- bosomal subunits.
tured human cells). Multiple presumptive RNA helicases and The large ribosomal subunit is one of the largest struc-
small G proteins are associated w ith the maturing pre-60S tures to pass through nuclear pore complexes. Maturation of
subunits. Some RNA helicases arc necessary to dislodge the the large subunit in the nucleoplasm leads to the generation
snoRNPs that base-pair perfectly with pre-rRNA over up to of binding sites for a nuclear export adapter called Nmd3 .
30 base pairs. Other RNA helicases may function in the dis- Nmd3 is bound by the nuclear transporter Exportinl (also
ruption of protein-RNA interactions. The requirement for so called Crml ). This is another quality-control step, because
many GTPases suggests that there are many quality-control only correctl y assembled subunits can bind Nmd3 and be ex-
checkpoints in the assembly and remodeling of the large sub- ported. The small ~ubunit of the mRNP exporter (NXT 1) also
unit RNP, where one step must be completed before a GTPase becomes associated with the nearly mature large ribosomal
is activated to allow the next step to proceed. Members of the subunit. These nuclear transporters interact with FG-domains
AAA ATPase family are also bound transiently. This class of of FG-nucleoporins. This mechanism allows penetration of
proteins is often involved in large molecular movements and the molecular "cloud" that fills most of the central channel of
may be required to fold the complex, large rRNA into the the NPC (sec Figure 8-20). Several specific nucleoporins
proper conformation. Some steps in 60S subunit maturation without FG-domains are also required for ribosomal subunit

388 CHAPTER 8 Post-transcriptional Gene Control


export and may have additional functions specific for this A whole raft of self-splicing sequences subsequently were
task. The dimensions of ribosomal subunits (- 25-30 nm in found in pre-rRNAs from other single-celled organisms, m
diameter) and the central channel of the NPC are compara- mitochondrial and chloroplast pre-rRNAs, in several pre-
ble, so passage may not require distortion of either the ribo- mRNAs from certain E. coli bacteriophages, and in some
somal subunit or the channel. Final maturation of the large bacterial tRNA primary transcripts. The self-splicing se-
subunit in the cytoplasm includes removal of these export quences in all these precursors, referred to as group I m-
factors. As for the export of most macromolecules from the trons, use guanosine as a cofactor and can fold by internal
nucleus, including tRNAs and pre-miRNAs (but not most base pairi ng to juxtapose closely the two exons that must be
mRNPs), ribosome ~uhunit export requires the function of a joined. As discussed earl ier, certain mitochondrial and chlo-
small G protein called Ran, as discussed in Chapter 13. roplast pre-mRNAs and tRNAs contain a second type of
self-splicing intron, designated group 11.
The splicing mechanisms used by group I introns, group
Self-Splicing Group llntrons Were the First
II introns, and spliceosomes are generally similar, involving
Examples of Catalytic RNA two transesterification reactions, which require no input of
During the 1970s, the pre-rRNA genes of the protozoan Tet- energy (Figure 8-4 1). Structural studies of the group [ intron
rahymena thermophda were discovered to contain an intron. from Tetrahymena pre-rRNA combined with mutational and
Careful searches failed to uncover even one pre-rRNA gene hiochemical experiments have revea led that the RNA folds
without the extra sequence, indicating that splicing is required into a precise three-dimensional structure that, like protein
to produce mature rRNA in t hese organisms. In 1982, in vitro enzymes, contains deep grooves for binding substrates and
studies showing that the pre-rRNA was spliced at the correct solvent-inaccessible regions that function in catalysis. The
sites in the absence of any protein provided the first indication group I intron functions like a metalloenzyme to precisely
that RNA can function as a catalyst, like enz}mes. orient the atoms that participate in the two transestcrification

Self-splicing introns Spliceosome-catalyzed splicing


of pre-mRNA
Group I Group II

a
Spliceosome

.
5'
P~p
G

3' 5'
r1 HO
P+------'p/
A

3'
~

3'

1 1 1
__Q_G_3'0H
~p
D-
-- o~P ..;

1 1 1
p~ C\ _Q
- P HO --- p HO 3'

FIGURE 8-41 Splicing mechanisms in group I and group II involving th~ 2'-hydroxyl groups of branch-s1te As in group II introns
self-splicing introns and spliceosome-catalyzed splicing of and pre-mRNA introns spliced in spliceosomes {see Figure 8-8). The
pre-mRNA. The intron is shown in gray, the exons to be joined in red. subsequent transesterification that links the 5' and 3' exons is similar in
In group I introns, a guanosine cofactor {G) that is not part of the RNA all three splicing mechanisms. Note that spliced-out group I introns are
chain associates with the active site. The 3'-hydroxyl group of this linear structures, unlike the branched intron products in the other two
guanosine participates in a transesterification reaction with the cases. (Adapted from P. A. Sharp, 1987, Science 235:769.]
phosphate at the 5' end of the intron; this reaction is analogous to that

8.5 Processing of rRNA and tRNA 389


reactions adjacent to catalytic Mg 2 ions. Considerable evi- RNase P polypeptide increases the rate of cleavage by the RNA,
dence now indicates that splicing by group II introns and by allowing it to proceed at physiological Mg2 + concentrations. A
snRNAs in the spliceosome also involves bound catalytic comparable RNase P functions in eukaryotes.
Mg2 -'- ions. In both the group I and II self-splicing introns and About 10 percent of the bases in pre-tRNAs are modified
probably in the spliceosome, RNA functions as a ribozyme, enzymatically during processing. Three classes of base modifi-
an RNA sequence with catalytic ability. cations occur (Figure 8-42): (1) U residues at the 3' end of
pre-tRNA are replaced with a CCA sequence. The CCA se-
quence is found at the 3' end of all tRNAs and is required for
Pre-tRNAs Undergo Extensive Modification
their charging by aminoacyl-tRNA synthetases during protein
in the Nucleus synthesis. This step in tRNA synthesis likely functions as a
Mature cytosolic tRNAs, which average 75-80 nucleotides in quality-control point, since only properly folded tRNAs arc
length, arc produced from larger precursors (pre-tRNAs) syn- recognized by the CCA addition enzyme. (2) Methyl and iso-
thesized by RNA polymerase I1I in the nucleoplasm. Mature pentenyl groups are added to the heterocyclic ring of purine
tRNAs also contain numerous modified bases that are not bases, and the 2' -OH groups in the ribose of specific residues
present in tRNA primary transcripts. Cleavage and base are methylated. (3) Specific uridincs are converted to dihy-
modification occur during processing of all pre-tRNAs; some drouridine, pseudouridine, or ribothymidine residues. The
pre-tRNAs also are spliced during processing. All of these functions of these base and ribose modifications are not well
processing and modification events occur in the nucleus. understood, but since they are highly conserved, they proba-
A 5' sequence of variable length that is absent from mature bly have a positive influence on protein synthesis.
tRNAs is present in all prc-tRNAs (Figure 8-42). This occurs As shown in Figure 8-42, the pre-tRNA expressed from the
because the 5' end of mature tRNAs is generated by an endo- yeast tyrosine tRNA (tRNAT>r) gene contains a 14-base intron
nucleolrtic cleavage specified by the tRNA three-dimensional that is not present in mature tRNATrr. Some other eukaryotic
structure rather than the start site of transcription. These extra tRNA genes and some archaeal tRNA gene!! also contain in-
5' nuclcotides are removed by ribonuclease P (RNase P), a ri- trans. The introns in nuclear pre-tRNAs arc shorter than those
bonucleoprotein endonuclease. Studies with E. coli RNase P in pre-mRNAs and lack the consensus splice-site sequences
indicate that at high Mg2 concentrations, the RNA compo- found in pre-mRNAs (see Figure 8-7). Pre-tRNA introns also
nent alone can recognize and cleave E. coli pre-tRNAs. The are clearly distinct from the much longer self-splicing group I

3' 3'
OH OH

Processing

Mature tRNATyr

FIGURE 8-42 Changes that occur during the processing of the stem loops are converted to characteristic modified bases (yellow).
tyrosine pre-tRNA. A 14-nucleotide intron (blue) in the anticodon Not all pre-tRNAs contain introns that are spliced out during process-
loop is removed by splicing. A 16-nucleotide sequence (green) at the ing, but they all undergo the other types of changes shown here.
5' end is cleaved by RNase P. U residues at the 3' end are replaced by D = dihydrouridine; ljJ = pseudouridine.
the CCA sequence (red) found in all mature tRNAs. Numerous bases in

390 CHAPTERs Post-transcriptional Gene Control


and group II intro ns found in chloroplast and mitochondrial 3 kDa 10 kDa 40 kDa 40 kDa 70 kDa 500 kDa 2000 kDa

pre-rRNAs. The mechanism of pre-tRNA splicing differs in


three fundamental ways from the mechanisms utilized by self-
splicing introns and spliceosomes (see Figure 8-41). First, splic-
ing of pre-tRNAs is catalyzed by proteins, not by RNAs.
Second, a pre-tRNA intron is excised in one step that entails
simultaneous cleavage at both ends of the intron. Finally, hy-
drolysis of GTP and ATP is required to join the two rRNA FIGURE 843 Nuclear bodies are different ially permeable t o
halves generated by cleavage on either side of the intron. molecules in the bulk nucleoplasm. Each pair of panels shows a single
After pre-tRNAs are processed in the nucleoplasm, the area through a living Xenopus oocyte nucleus that was previously
mature tRNAs are transported to the cytoplasm through nu- injected with fluorescent dextran of the indicated molecular mass
clear pore complexes by Exportin-t, as discussed previously. (3-2000 kDa). Each section ofthe upper panel is a confocal image in
In the cytoplasm, tRNAs are passed between aminoacyl- which the intensity of fluorescence is a measure of dextran concentra-
rRNA synthetases, elongation factors, and ribosomes during tion (i.e., darker areas show regions where dextran has been excluded).
protein synthesis (Chapter 4). Thus tRNAs generally are as- Each section of the lower panel is a differential interference contrast
sociated with proteins and spend tittle time free in the cell, as image of the same field. Open arrowheads indicate nucleoli, closed
is also the case for mRNAs and rRNAs. arrowheads Cajal bodies (CBs) with attached nuclear speckles which are
much larger in Xenopus oocytes than in most somatic cells. Dextrans of
low molecular mass (e.g., 3 kDa) almost completely penetrated CBs but
Nuclear Bodies Are Functionally Specialized were excluded more from nuclear speckles and nucleoli. Exclusion of
Nuclear Domains dextran increased with molecular mass. Bar= 10 f.l.m. [From
K. E. Handwerger et al., 2005, Mol. Bioi. Cell 16:202.]
High- resolution visualization of plant- and animal-cell nu-
clei by electron microscopy and subsequent staining with
fluorescently labeled antibod ies has revealed domains in nu-
clei in addition to chromosome territories and nucleoli. methyl groups to the 2'-hydroxyl groups of specific riboses.
These specialized nuclear domains, called nuclear bod ies, are T hese post-transcriptional modifications arc important for
not surrounded by membranes but are nonetheless regions the proper assembly and function of snRNPs in pre-mRNA
of high concentrations of specific proteins and RNAs that splicing. These modifications occur in Cajal bodies, where
form distinct, rough ly spherical structures within the nu- they are directed by a class of snoRNA-like guide RNA mol-
cleus. T he most prominent nuclear bodies are nucleoli, the ecules called scaRNAs (small Cajal body-associated RNAs ).
sites of ribosomal subunit synthesis and assembly discussed There is also evidence that the Cajal body is the site of reas-
earlier. Several other types of nuclear bodies also have been sembly of the U4/U6/U5 tri-snRNP complexes required for
described in structural studies. pre-mRNA splicing from the free U4, US, and U6 snRNPs
Experiments with fluorescenrly labeled nuclear proteins released during the removal of each intron (see Figure 8-11 ).
have shown that the nucleus is a highly dynamic environ- Since Cajal bodies also contain a high concentration of the
ment, with rapid diffusion of proteins through the nucleo- U7 snRNP involved in the specialized 3'-end processing of
plasm. Proteins associated with nuclear bodies are often also the major histone mRNAs, it is likely that this process also
observed at lower concentrations in the nucleoplasm outside occurs in Cajal bodies, as may the assembly of the telom-
the nuclear bodies, and fluorescence studies indicate that erase RNP.
they diffuse in and out of the nuclear bodies. Based on these
measurements of molecular mobility in living cells, nuclear Nuclear Speckles Nuclear speckles were observed, using
bodies can be mathematically modeled as the expected fluorescently labeled antibodies to snRNP proteins and other
steady state for diffusin~ proteins that interact with suffi- proteins involved in pre-mRNA splicing, as approximately
cient affinity to form self-organized regions of high concen- 25-50 irregular, amorphous structures 0.5-2 1-Lm in diame-
trations of specific proteins but with low enough affinity for ter that are distributed through the nucleoplasm of verte-
each other to be able to diffuse in and out of the structure. In brate cells. Since speckles are not located at sites of
electron micrographs these structures appear to be a hetero- co-transcriptional pre-mRNA splicing, which are associated
geneous, sponge! ike network of interacting components. We closely with chromatin, they are thought to be storage re-
discuss a few examples of nuclear bodies here. gions for snRNPs and proteins involved in pre-mRNA splic-
ing that are released into the nucleoplasm when required.
Cajal Bodies Cajal bodies are -0.2-11-Lm spherical structures
that have been observed in large nuclei for more than a cen- Promyelocytic leukemia (PML) Nuclear Bodies The PML
tury (Figure 8-43 ). Current research indicates that like nucle- gene was originally discovered when chromosomal translo-
o li, Cajal bodies are centers of RNP-complex assembly for cations within the gene were observed in the leukemic cells
spliceosomal snRNPs and other RNPs. Like rRNAs, snRNAs of patients with the rare disease promyelocytic leukemia
undergo specific modifications, such as the conversion of (PML). When antibodies specific for the PML protein were
specific uridine residues to pseudouridine and addition of used in immunofluorescence microscopy studies, the protein

8.5 Processing of rRNA and tRNA 391


was found to localize to -10-30 roughly spherical regions
0.3-1 1-1-m in diameter in the nuclei of mammalian cells. Synthesis and processing of pre-rRNA occur in the nucleo-
Multiple functions have been proposed for these PML nu- lus. The SS rRNA component of the large ribosomal subunit
clear bodies, but a consensus is emerging that they function is synthesized in the nucleoplasm by RNA polymerase III.
as sites for the assembly and modification of protein com- Approximately 150 snoRNAs, associated with proteins in
plexes involved in DNA repair and the induction of apopto- snoRNPs, base-pair with specific sites in pre-rRNA where
sis. For example, the important p53 tumor suppressor they direct ribose methylation, modification of uridine to
protein appears to be post-translationally modified by phos- pseudouridine, and cleavage at specific sites during rRNA
phorylation and acetylation in PMI nuclear bodies in re- processing in the nucleulu~.
sponse to DNA damage, increasing its ability to activate the
Group I and group II self-splicing introns and probably
expression of DNA-damage response genes. PML nuclear
snRNAs in spliceosomes all function as ribozymes, or cata-
bodies are also required for cellular defenses against DNA
lytically active RNA sequences, that carry out splicing by
viruses that are induced by interferons, proteins secreted b)
analogous transesterificarion reactions requiring bound Mg2
virus-infected cells and T-lymphocytes involved in the im-
ions (see Figure 8-41).
mune response (sec Chapter 23).
PML nuclear bodies arc also sites of protein post- Pre-tRNAs synthesized by RNA polymerase Jll in the nu-
translational modificatton through the addition of a small, cleoplasm are processed by remo":al of the 5' -end sequence,
ubiquitin-like protein called SUMOl (small ubiquitin-like addition of CCA to the 3' end, and modification of multiple
moiety- I), which can control the activity and subcellular lo- internal bases (see Figure 8-42).
calization of the modified protein. Many transcriptional acti- Some pre-tRNAs contain a short intron that is removed by
vators are mhibited when they are sumoylated, and mutation a protein-catalyzed mechanism distinct from the splicing of
of their sire of sumoylation increases their activity in stimu- pre-mRNA and self-splicing introns.
lating transcription. These observations indicate that PML
All species of RNA molecules are associ a red with proteins
nuclear bodies arc involved in a mechanism of transcrip-
in various types of ribonucleoprotein particles, both in the
tional repression that remains to be studied and thoroughly
nucleus and after export to the cytoplasm.
understood.
Nuclear bodies are functionally specialized regions in the nu-
Nucleolar Functions in Addition to Ribosomal Subunit Syn- cleus where interacting proteins form self-organized structures.
Many of these, like the nucleolus, are regions of assembly of
thesis The first nuclear bodies to be observed, the nucleoli,
RNP complexes.
.. .
may have specialized regions of substructure that arc dedi-
cated to functions other than ribosome biogenesis. There is
evidence that immature SRP ribonucleoprotein complexes
involved in protein secretion and ER membrane insertion
(Chapter 13) are assembled in nucleoli and then exported to
Perspectives for the Future
the cytoplasm, where their final maturation takes place. The
Cdc14 protein phosphatase that regulates processes in the In this and the previous chapter, we have seen that in eu-
final stages of mitosis is sequestered in nucleoli in yeast cells karyotic cells, mRNAs are synthesized and processed in the
until chromosomes have been properly segregated into the nucleus, transported through nuclear pore complexes to the
bud (Chapter 19). Also, a tumor suppressor protein called cytoplasm, and then, in some cases, transported to specific
ARF, which is involved in the regulation of the protein en- areas of the cytoplasm before being translated by ribosomes.
coded by the most frequently mutated gene in human can- Each of these fundamental processes is carried out by com-
cers, p5 3, is sequestered in nucleoli and released in response plex macromolecular machines composed of scores of pro-
to DNA damage (Chapter 24). In addition, heterochromatin teins and in many cases RNAs as well. The complexity of
often forms on the surface of nucleoli (Figure 6-33), suggest- these macromolecular machines ensures accuracy in finding
ing that proteins associated with nucleoli also participate in promoters and splice sites in the long length of DNA and
the formation of this repressing chromatin structure. RNA sequences and provides various avenues for regulating
synthesis of a polypeptide chain. Much remains to be learned
about the structure, operation, and regulation of such
KEY CONCEPTS of Section 8.5 complex machines as spliceosomes and rhe cleavage/poly-
adenylation apparatus.
Processing of rRNA and tRNA Recent examples of the regulation of pre-mRNA splicing
A large precursor pre-rRNA ( 13.7 kb in humans) synthe- raise the question of how extracellular signals might control
sized by RNA polymerase I undergoes cleavage, exonucleo- such events, especially in the nervous system of vertebrates.
lytic digestion, and base modifications to yield mature 28S, A case in point is the remarkable situation in the chick inner
185, and 5.8S rRNAs, which associate with ribosomal pro- car, where multiple isoforms of the Ca 2 + -activated K chan-
teins into ribosomal subunits. nel called S/o are produced by alternative RNA splicing.
Cell-cell interactions appear to inform cells of their position

392 CHAPTER 8 Post- transcriptional Gene Control


in the cochlea, leading to alternative splicing of Slo pre-mRNA.
The challenging task facing researchers is to discover how
Key Terms
such cell-cell interactions regulate the activity of RNA- 5' cap 349 poly(A) tail 358
processing facrors. alternative splicing 361 pre-mRNA 349
The mechanism of mRNP transport through nuclear pore pre-rRNA 385
cleavage/polyadenylation
complexes poses many intriguing questions. Future research
complex 358 ribozyme 390
will likely reveal additional activities of hnRNP and nuclear
mRNP proteins and clarify their mechanisms of action. For cross-exon recognition RNA editing 364
instance, there i<> a small gene family encoding proteins ho- complex 356 RNA-induced silencing
mologous to the large subunit of the mRNA exporter. What Dicer 371 complex (RISC) 3 71
are the functions of these related proteins? Do they partici- Drosha 371 RNA interference (RNAi) 373
pate in the transport of overlapping sets of mRNPs? Some exosome 359 RNA splicing 348
hnRNP proteins contain nuclear-retention signals that pre- FG-nucleoporins 365 short interfering RNAs
vent nuclear export when fused to hnRNP proteins with (siRNA) 346
group I introns 357
nuclear-export signals (NESs). How are these hnRNP proteins
selectively removed from processed mRNAs in the nucleus, group II introns 357 siRNA knockdown 374
allowing the mRNAs to be transported to the cytoplasm? iron-response element- small nuclear RNAs
The localization of certain mRNAs to specific subcellular binding protein (snRNAs) 352
locations is fundamental to the development of multicellular (IRE-BP) 379 small nucleolar RNAs
organisms. As we will discuss in Chapter 21, during develop- micro RNAs (miRNAs) 370 (snoRNAs) 386
ment an individual cell frequently divides into daughter cells mRNA surveillance 380 spliceosome 353
that function differently from each other. In the language of mRNP exporter 365 SR proteins 356
developmental biology, the two daughter cells are said to
nuclear pore complex
have different developmental fates. In many cases, this dif-
(NPC) 365
ference in developmental fate results from the localization of
an mRNA to one region of the cell before mitosis so that
after cell division, it is present in one daughter cell and not
the other. Much exciting work remains to be done to fully
Review the Concepts
understand the molecular mechanisms controlling mRNA
localization that are critical for the normal development of 1. Describe three types of post-transcriptional regulation of
multicellular organisms. protein-coding genes.
Some of the most exciting and unanticipated discoveries 2. True or False? The CTD is responsible for mR.t'\IA-processing
. in molecular cell biology in recent years have concerned the steps that are specific for mRNA, and not other forms of RNA.
existence and function of miRNAs and the process of RNA Explain why you chose true or false.
interference. RNA interference (RNAi) provides molecular
3. There are a number of conserved sequences found in an
cell biologists with a powerful method for studying gene
mRNA which dictate where splicing occurs. Where are these
function. The discovery of -500 miRNAs in humans and
sequences found relative to the exon/intron junctions? What
other organisms suggests that multiple significant examples
is the significance of these sequences in the splicing process?
of translational control by this mechanism await characteri -
One of these important regions is the branch point A found
zation. Recent studies in S. pombe and plants link similar
in the intron. What is the role of the branch pmnt A in the
short nuclear R.NAs to the control of DNA methylation and
splicing process, and can this be accomplished with the OH
the formation of heterochromatin. Will similar processes
group on either the 2' or the 3' carbon?
control gene expression through the assembly of heterochro-
matin in humans and other animals? What other regulatory 4. What is the difference between hnRNAs, snRNAs,
processes might be directed by other kinds of small RNAs? miRNAs, siRNAs, and snoRNAs?
Since control by these mechanisms depends on base pairing 5. What are the mechanistic similarities between group II
between miRNAs and target mRNAs or genes, genomic and intron self-splicing and spliceosomal splicing? What is the
bioinformatic methods will probably suggest genes that may evidence that there may be an evolutionary relationship be-
be controlled by these mechanisms. What other processes in tween the rwo?
addition to translation control, mRNA degradation, and 6. You obtain the sequence of a gene containing 10 exons, 9
heterochromatin assembly might be controlled by miRNAs? introns, ;Jnd a 3' UTR containing a polyadenylation consen-
These are just a few of the fascinating questions concern- sus sequence. The fifth intron also contains a polyadenyla-
ing RNA processing, post-transcriptional control, and nu- tion site. To test whether both polyadenylation sites are
clear transport that will challenge molecular cell biologists in used, you isolate mRNA and find a longer transcript from
the coming decades. The astounding discoveries of entirely muscle tissue and a shorter transcript from all other tissues.
unanticipated mechanisms of gene control by miRNAs re- Speculate about the mechanism involved in the production
mind us that many more surprises are likely in the future. of these different transcripts.

Review the Concepts 393


7. RNA editing is a common process occurring in the mito- expresses a Pst-Miu fragment of the LAT gene (see diagram in
chondria of trypanosomes and plants, in chloroplasts, and in part b). The percentage of these transfected cells that then un-
rare cases in higher eukaryotes. What is RNA editing, and derwent drug-induced cell death was compared to that of con-
what benefit does it demonstrate in the documented example trol cells. The experiment was repeated in cells in which Dicer
of apoB in humans? expression was knocked down using Dicer siRNA. The data
8. A1> DNA is found in the nucleus, transcription is a nuclear- obtained are shown in the graph below. What conclusions can
localized process. Ribosomes responsible for protein synthe- be drawn from these data? Why did the scientists who con-
sis are found in the cytoplasm. Why is hnRNP trafficking to ducted this study examine the effects of silencing Dicer?
the cytoplasm restncted to the nuclear pore complexes? How
do the rG-repeats of the nuclear pore complexes act as a
specificity barrier in nuclear transport? Cells transfected with
(/)
60 control expression vector
9. A protein complex in the nucleus is responsible for trans- (/)en
=o
Q)- Cells transfected with LA T
porting mRNA molecules into the cytoplasm. Describe the (.)0.
expression vector
-0 o
a.
proteins that form this exporter. What two protein groups Q) 0)
are likely behind the mechanism involved in the directional ClCl
0) c
40 0 Cells transfected with LA T
expression vector and
movement of the mRNP and exporter into the cytosol. c:Q)C)
o expressing control siRNA
(.) ~
~ Q)
10. RNA knockdown has become a powerful tool in the arse- C>-o
a..c 0 Cells transfected with LA T
nal of methods ro deregulate gene expression. Briefly describe ::J
20 expression vector and
expressing Dicer siRNA
how gene expression can be knocked down. What effect would
introducing siRNAs to TSC I have on human cells?
11. Speculate about why plants deficient in Dicer activity b. Cells were transfected with an expression vector ex-
show increased sensitivity to infection by RNA viruses. pressing the Pst-Miu fragment of the LA T gene from which
12. mRNA sta bility is a key regulator of protein levels in a the region between the two Sty restriction sites was deleted
cell. Briefly describe the three mRNA degradation pathways. (DSty; diagram below). When these cells were induced to
A yea~t cell has a mutation in the DCPl gene, resulting in undergo apoptosis, they died at the same rate as did non-
decreased uncapping acti\ity. Would you expect to see a transfected cells. In additional studies, cells were transfected
change in the P bodies found in this mutant cell? with an expression vector expressing the Sty-Sty region of
13. mRNA localization now appears to be a common phe- the LA T gene. These cells exhibited the same resistance to
nomenon. What benefit docs mRNA localization have for a apoptosis as did cells transfected with the Pst-Mlu fragment.
cell? What is the evidence that some mRNAs are directed to What can be deduced from these findings about the region of
accumulate in specific subcellular location s? the LAT gene required to protect cells from apoptosis?

Pst/Miu fragment
I<
Analyze the Data
~Region deleted in t1Sty
~1ost humans are infected with herpes simplex virus-1 (HSV-1),
Pst Mlu
the causative agent of cold sores. The HSV-1 genome com-
prises about I 00 genes, most of which arc expressed in in-
fected host cells at the site of oral sores. The infectious
process involves replication of viral DNA, transcription and
translanon of viral genes, assembly of new viral particles,
and death of the host cell as the viral progeny are released. 5' - GTGGCGGCCCGGCCCGGGGCCCCGG G :CAAGGGGCCCCGGCCCGGGGCCCCAc- 3'
Unlike most other types of viruses, herpesvirus also has a Stem (5' arm) LOOP Stem (3' arm) I
latent phase, 10 which the virus remains hidden in neurons.
These latently infected neurons are the source of active infec- c. RNA encoded within the Sty-Sty region is predicted to
tions, causing cold sores when latency is overcome. form a stem loop (see diagram in part b). Northern blot anal-
Interestingly, only a single viral transcript is expressed ysis was performed on total-cell RNA isolated from control
during latency. This transcript, LA T (latency-associated cells (mock ), cells infected with wild-type HSV-1, cells in-
transcript), does not encode a protein, and neurons mfected fected with an HSV-1 deletion mutant from which these-
with mutant HSV - I lacking the LA T gene undergo cell death quence between the two Sty site:, in the LA T gene was deleted
by apoprosis at a rate twice that of cells infected with wild- (DSty), and cells infected with a rescued DSry virus into which
type HSV-1. To determine if LAT functions to block apop- the deleted region was re-inserted into the viral genome
tosis by encoding a miRKA, the following studies were done (StyR). The probe used for the Northern blot was the labeled
(see Gupta et al., 2006, Nature 442:82-85). 3' stem region of the LAT RNA in the Sty-Sty region, as dia-
a. A cell line was transfected (a process in which foreign grammed in part (b). The RNAs recognized by this probe
DNA is inserted into a cell) with an expression vector that were either - S 5 nucleotidcs or 20 nucleotides, as shown in

394 CHAPTER 8 Post-transcriptional Gene Control


.
the Northern blot below. Why were two different-sized RNAs ~loore, ~1. J., and N.J. Proudfoot. 2009. Pre-mR~ .-\ proccssmg
detected? When a second probe was used that was the labeled reache> back to transcription and ahead to translation. Ce// 136:
5' stem region of the RNA sequence shown in part (b), only 688-700.
Pawlicki, .J. ~!.,and J. A. SteltZ. 20 I 0. Nuclear networking
the -55-nucleotide RNA was detected. What can you deduce
fashions pre-messenger RNA and pnmary m1croRN \transcripts for
about the processing of RNA expressed from the LAT gene? function. Trends Cell Bioi. 20:52-61.
What enzyme likely produced the -55-nucleotide RNA? In Perales, R ., and D. Bentley. 2009. "Corranscriptionality": the
what part of t he cell ? What enzyme likely produced the 20- transcnption elongation complex as a nexu> for nuclear transac-
nucleotide RNA? In what part of the cell? tions. Mol. Cell. 36:178-191.
Pyl", A. ~1. 20 I 0. The rcrnar} ,truLture oi group 11 introns:
unplications for biological function and evolution. Crit. Ret.
Control wt HSV-1 ll Sty Biochem. Mol. Brol. 45:21 S-232.
Sharp, P. A. 2005. The discovery of split genes a nd RNA
u;
Q) splicing. Trends Bwchem. Sci. 30:2"9-281.
-o 60
g Valadkhan, S. 20 I 0. Role of the snR~As in sphceosomal aLnve
Q) 50 s1te. RNA Bioi. 7:345-353.
u:;,
c::
Wahl, :vi. C...., C. l.. Will, and R. Lt1hrmann. 2009. The spliceo-
'!t ~ome: design principles of a dynamic RNP machine. Cell136:701-718.
Q)
N
u; Regulation of Pre-mRNA Processing
<(
z Black, D. I. 2003. ~l ec hamsms of alternative pre-mR]';"A
a: 20
splicing. Amz. Rev. Bwchem. 72:291-336.
Chen, M., and J. l.. Manley. 2009. ).lechanisms of alternanve
>plicing regulation: insights from molecular and genomics approaches.
d. T GF-13 mR NA encodes a protein, t ransforming N,1t. Ret. Mol. Cell Bioi. 10:741-754.
growth factor 13, that inhibits cell growth and induces apop- l.icaralos1, D. D., and R. B. Darnell. 2010. RNA processing and
tosis. The 3' unrranslated region (3' UTR) of TGF-13 mRNA irs regulation: global insights IntO biologicalnerworks. Nat. Rez.
can form an imperfect duplex with miRNA encoded by the Genet. 11:7 5-87.
~laniatis, T., and B. Tasic_ 2002. Alternative prc-mR!\A splicing
5' stem region of the LA T Sty-Sty domain (miR-LAT), as
and proteome expansion 111 metazoans. Nature 418:236-243.
shown below. In what way might the expression levels of Raponi, ~!.,and D. Baralle. 20 I 0. Alternative splicing: good
TGF-13 differ in cell s infected with wild-type HSV-1 com- and bad effec t~ of translatlonall} silent subsmutions. FEBS .f.
pared to uninfected cells? What can you infer about latent 2 77:836-840.
HSV- 1 infections from these stud ies? \X1ang, E. T., et al. 2008. Alternative 1soform regulation in
human tissue transcriptomes. Nature 456:470-476.
Zhong, X. Y., et al. 2009. SR protems 111 \'Crtlcal inregration of
c gene n.pression from transcription to RNA processmg to transla-
miR-LAT 1\
C C : G ~ G ~ C ~ ~ ~ C _ C ' ~ 5' tion. Curr. Opm. Genet. Dev. 19:424-436.
I I I I I I I I I I I I I I I I I I I
5' C- G-C- C- C-C- G-G G-C-C-C-G-G-C-C-C-C-A Transport of mRNA Across the Nuclear Envelope
I I TGF-[3-3' UTR
Cole, C. N., and J. .J. Scarcel11. 2006. Tra nsport of messenger
CAG
Rl\'A from the nucleus ro the cytoplasm. Curr. Opm. Cell Bioi.
18:299-306.
Grunwald, D., R. H. Singer, and M. Rout. 2011. Nuclear
References export dynamiCS of R~A -protem complexes. Nature 4 7~ :333-341.
Iglesias, N., and F. Stutz. 2008. Regulation of mRNP dynamics
Processing of Eukaryotic Pre-,mRNA along the export pathwa}. FEBS /.ett . 582:198.,-1 996.
Bergkessel, M., G. M. Wilmes, and C. Guthrie. 2009. SnapShot: Kata hira, J., andY. Yoneda. 2009. Roles of the TREX complex
formation of mRNPs. Ce/1136:"94. m nuclear export of mR~A. RNA Bwl. 6:149-1 52.
Blanc, V., and N. 0. Davidson. 2010. APOBFC-1-mediated Rodriguez-Navarro, S., and E. Hurr. 20 I I. Lmkmg gene
RNA editing. Wiley InterdisCifJ. Ret. Syst. 810/. 'vfed. 2: 'i94-602. regulation to mRNA production and export. Curr. Opin. Cell Bwl.
de Almeida, S. F., and M. Carm o-rc)Tlseca. 2008 . The CTD role 23:302-309.
in cocra nscriptional Ri','A processing and suneillance. H: BS Lett. Stewart, 1\1. 20 I 0. Nuclear export of mRNA. Trends Biochem.
582: 1971-19.,6. So. 35:609-617.
Gu, M., and C. D. Lima. 2005. Processmg the mes>age: Wente, S. R., and M. P. Rout. 20 I 0. The nuclear pore complex
structural msights mto capping and decappmg mR~A. Curr. Ot1ilr. and nuclear transport. Cold Sprmg Har/1. Perspect. Bioi.
Struct. Bioi. 15:99- 106. 2( 10):a000562.
Hocine, S., R. I L Singer, and D. C..runwald. 2010. R~A process-
ing and export. Cold Spring Harb. Perspect. 810/. 2 ( 12):a000752. Cytoplasmic Mechanisms of Po st-transcriptional Control
Housele),]., and D. Tolleney. 2009. The many pathways of Ambros, V. 2004. The functions of animal microR::-..As. N,lfure
RNA degradation. Ce/1136:763-776. 431:350-355.
La mbowitz, A. \1., and S. Zimmer!}. 2004. ~1obile group II Buchan, J. R., and R. Parke r. 2009. Fukaryonc stres~ ~ranulcs:
mtrons. Amw. Reu. Genet. 38:1-35. the im and outs of translation. Mol. Ce/136:932-94 L.

References 395
Carthew, R. W., and F. J. Sontheimcr. 2009. Origins and Stmpson, L., et al. 2004. Mitochondrial proteins and complexes
mechamsms of miRNA~ and stRNAs. Cell 136:642-655. 111Letshmania and Trypanosoma involved 111 U-insertlon/deletion
Doma, \1. K., and R. Parker. 2007. RNA quality control tn RNA editing. RNA 10:159-170.
eukaryote~. Ce/1131:660-668. Siomi, M.. C., et al. 2011. PTWI-interacting small RNAs: the
Eulalio, A., I. Behm-Ansmant, and E. Izaurralde. 2007. P bodies: vanguard of genome defence. Nat. Rev. Mol. Cell Bzol. 12:246-258.
at the crossroads of post-transcriptional pathways. Nat. Rev. Mol. Willis, I. M., and R. D. Moir. 2007. Integration of nutritional and
Cell Bzol. 8:9-22. stress signaling pathways b) Mafl. Trends Biochem. SCI. 32:51-53.
Fabian, M. R., N. Sonenberg, and W. Filipowicz. 2010. Wullschleger, S., R. Loewith, and M. N. Hall. 2006. TOR
Regulation of mRNA translation and stability by microRNAs. signaling in growth and metabolism. Cell 124:471-484.
Annu. Rev. B10chem. 79:351-3 79. Zhang, H., J. M. Mamar, and A. Z. Fire. 2011. 'lnc-miRs':
Ghildiyal, .\1., and P. D. Zamore. 2009. Small silencing RNAs: funcnonal intron-interrupted miRNA genes. Genes Dev. 25:1589-
an expanding universe. Nat. Rev. Genet. 10:94-108. 1594. A. Z. Fire's Nobel Pnze lecture can be viewed at http://
Groppo, R., and j. D. Richter. 2009. Translational control from nohclprize.org/nobel_prizes/medicine/laureatcs/2006/announcement.
head to tail. Curr. Opi11. Cell Bioi. 21:444-451. html
Hirokawa, !':. 2006. mRNA transport in dendmes: RNA Zoncu, R., A. Efeyan, and D. M. Sahatmi. 201 I. mTOR: from
granules, motors, and tracks.]. Neuroscz. 26:7139-7142. growth signal integration to cancer, diabetes and ageing. Nat. Rev.
Huntzinger, E., and E. Izaurralde. 2011. Gene silencing by Mol. Cell Bioi. 12:21-35.
microRNAs: contributions of translational repression and mRNA
decay. Nat. Rev. Genet. 12:99-110. Processing of rRNA and tRNA
Jobson, R. W., andY. L. Qiu. 2008. Did RNA edtring in plant Evans, D., S.M. \1arquez, and ::-.l. R. Pace. 2006. RNase P: interface
organellar genomes originate under natural selecnon or through of the RNA and protein worlds. Trends Biochem. Sci. 31:333-341.
genetic drift? Bioi. Direct. 3:43. Farica, A., and D. Tollervey. 2002. Making ribosomes. Curr.
Kidner, C. A., and R. A. Marrienssen. 2005. The developmental Opm. Cell Bzol. 14:313-318.
role of microRNA in plants. Curr. Opm. Plant Bioi. 8:38-44. Hage. A. E., and D. Tollervey. 2004. A surfeit of factors: why ts
Leung, A. K., and P. A. Sharp. 2010. MicroRNA functions 111 nbosome assembly so much more complicated in eukaryores than
stress responses. Mol. Cell. 40:205-2 15. bacteria? RNA Bioi. 1:10-15.
Lodish, H. F., et al. 2008. Micromanagement of the immune Hamma, T., and A. R. Ferrc-D'Amarc. 2010. The box H/ACA
system by mtcroR;'\/As. Nat. Rev. lmmzmol. 8:120-130. ribonucleoprotein complex: mterplay of RNA and protem structures 111
Maquat, L. E., W. Y. Tarn, and 0. lsken. 2010. The pioneer post-transcripnonal RNA modification.]. Bzol. Chem. 285:805-809.
round of translation: features and functions. Cell 142:368-374. Handwcrger, K. E., and J. G. Gall. 2006. Subnuclear organelles:
Marrin, K. C., and A. Ephrussi. 2009. mRNA localization: gene new insights into form and function. Trends Cell Bzol. 16:19-26.
expression in the spatial dimension. Ce/1136:719-730. Kressler, D., E. Hurt, and J. Bassler. 2010. Drinng rihosomc
Mello, C. C., and D. Conte Jr. 2004. Revealing the world of assembly. Bioclnm. Biophys. Acta. 1803:673-683.
RNA Interference. Nature 431:338-342. C. C. :V1ello\ Nobel Pme Liang, B., and H. Li. 201 1. Structures of ribonucleoprotein
lecture can he viewed at http://nobelprize.org/nobel_prizcs/medicme/ particle modification enzymes. Q. Rev. Biophys. 44:95-122.
laureates/2006/announcement.html Marvin, M. C., and D. R. Engelke. 2009. RNase P: increased
Michels, A. A. 2011. MAF1: a new target of mTORCl. versatility through protein complexity? RNA Bioi. 6:40-42.
Biochem. Soc. TraJIS. 39:487-491. Ntzami, Z., S. Deryusheva, and J. G. Gall. 2010. The Cajal
Mihaylova, M. M., and R. j. Shaw. 2011. The AMPK signalling body and histone locus body. Cold Spnng Harb. Perspect. Bioi.
pathway coordinates cell growth, autophagy and metabolism. 2(7):a000653.
Nature Cell Bioi. 13:1016-1023. Phizicky, E. M., and A. K. Hopper. 2010. tRK'A biology charges
Parker, R., and H. Song. 2004. The enzymes and control of to the front. Genes Dev. 24:1832-1860.
eukaryotic mRNA turnover. Nat. Struct. Mol. Bzol. 11:121-127. Schmid, M., and T. H. Jemen. 2008. The exosome: a multipur-
Richter, J. D., and N. Sonenberg. 2005. Regulation of cap- pose RNA-decay machine. Trends Biochem. Sci. 33:501-510.
dependent translation by eiF4E inhibitory proteins. Nature Smhley, \1. R., and S. A. Strobel. 2006. RNA splicing: group I
433:4..,--480. mtron crystal structures reveal the basts of splice site ~election and
Ruvkun, G. B. 2004. The tiny RNA world. Haruey Lect. 99:1-21. metal ton catalysis. Curr. Opm. Struct. Rio/. 16:319-326.
Shaw, R. J. 2008. mTOR signaling: RAG GTPases transmit the Tschochner, H., and E. Hurr. 2003. Pre-ribosomes on the road
amino acid signal. Trends Riochem. Sci. 33:565-568. from the nucleolus to the cytoplasm. Trends Cell Bioi. 13:255-26 ."'!.

396 CHAPTER 8 Post-transcriptional Gene Control

Вам также может понравиться