Вы находитесь на странице: 1из 12

Article

Cryoelectron Microscopy Maps of Human


Papillomavirus 16 Reveal L2 Densities and Heparin
Binding Site
Highlights Authors
d 4.3-A resolution 3D reconstructions based on gold- Jian Guan, Stephanie M. Bywaters,
standard FSC Sarah A. Brendle, ..., James F. Conway,
Neil D. Christensen, Susan Hafenstein
d Classification of HPV overcomes heterogeneity that limits the
resolution Correspondence
d L1 protein was built and used to reveal L2 minor protein shafenstein@hmc.psu.edu

d Heparin binding elicits conformational changes to L2 In Brief


Guan et al. solved near-atomic resolution
cryo-EM maps of HPV16 and HPV16-
heparin, building the L1 protein structure
including C-terminal arm. The structural
analysis revealed the heterogeneity of
HPV16, the location and features of L2
protein, the heparin binding site, and
suggested L2 conformational changes
during cell entry.

Accession Numbers
5KEP
5KEQ

Guan et al., 2017, Structure 25, 253263


February 7, 2017 2016 Published by Elsevier Ltd.
http://dx.doi.org/10.1016/j.str.2016.12.001
Structure

Article

Cryoelectron Microscopy Maps of Human


Papillomavirus 16 Reveal L2 Densities
and Heparin Binding Site
Jian Guan,1 Stephanie M. Bywaters,2 Sarah A. Brendle,2 Robert E. Ashley,1 Alexander M. Makhov,3 James F. Conway,3
Neil D. Christensen,2 and Susan Hafenstein1,4,*
1Division of Infectious Diseases, Department of Medicine, Penn State College of Medicine, The Pennsylvania State University College of

Medicine, Mail Code H036, 500 University Drive, P.O. Box 850, Hershey, PA 17033-0850, USA
2Department of Pathology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
3Department of Structural Biology, University of Pittsburgh School of Medicine, 3501 5th Avenue, Pittsburgh, PA 15260, USA
4Lead Contact

*Correspondence: shafenstein@hmc.psu.edu
http://dx.doi.org/10.1016/j.str.2016.12.001

SUMMARY (Dickson et al., 2015; Gilmer, 2015; McKee et al., 2015). Target-
ing host entry mechanisms may be a successful alternative
Human papillomavirus (HPV) is a significant health approach because virus entry is a key step of HPV infection
burden and leading cause of virus-induced cancers. regardless of genotype. However, receptor use for HPV, the de-
The current commercial vaccines are genotype spe- tails of cell attachment, and capsid conformational changes
cific and provide little therapeutic benefit to patients required for host entry are poorly understood.
with existing HPV infections. Host entry mechanisms HPV is epitheliotropic and its replication is tightly associated
with the terminal differentiation of basal cells into keratinocytes.
represent an excellent target for alternative thera-
This restricted tropism makes production and purification of
peutics, but HPV receptor use, the details of cell
high-titer virus preparations for research problematic. Therefore,
attachment, and host entry are inadequately under- alternative HPV production methods have been developed
stood. Here we present near-atomic resolution struc- within the HPV community for molecular biology and structural
tures of the HPV16 capsid and HPV16 in complex studies. Virus-like particles can be composed of only the major
with heparin, both determined from cryoelectron structural protein (L1) or of the major and minor capsid proteins
micrographs collected with direct electron detection (L1/L2) and are not infectious since they are devoid of viral
technology. The structures clarify details of capsid genome (Hernandez et al., 2012; Kirnbauer et al., 1992, 1993).
architecture for the first time, including variation in Pseudovirus is composed of both structural proteins, which
L1 major capsid protein conformation and putative are expressed along with a plasmid so that a copy of the plasmid
location of L2 minor protein. Heparin binds spe- DNA can be packaged as a mock genome (Buck et al., 2005a,
2005b). Quasiviruses assemble a structurally complete capsid
cifically around the capsid icosahedral vertices and
and are infectious since they package a copy of the cottontail
may recapitulate the earliest stage of infection,
rabbit papillomavirus genome (CRPV) (Christensen, 2005). All
providing a framework for continuing biochemical, of these types of HPV capsids preserve the main attributes of
genetic, and biophysical studies. the native capsid structure and have been used successfully
for vaccine development and for studies of antigenicity, receptor
usage, entry mechanisms, and capsid structure. Quasiviruses
INTRODUCTION have been used for the HPV16 work presented here.
Structures of sub-viral particles have been solved by X-ray
Human papillomavirus 16 (HPV16) is the most prevalent onco- crystallography, including the HPV16 L1 pentamer and a T = 1
genic genotype in HPV-associated anogenital and oral cancers. particle (Chen et al., 2000; Dasgupta et al., 2011); however, the
Having co-evolved with the human host for more than a million complete HPV T = 7d capsid has only been visualized by cryoe-
years, HPV develops chronic infections by activating the cell lectron microscopy (cryo-EM) reconstructions (Baker et al.,
cycle as epithelial cells differentiate in order to create a replica- 1991; Buck et al., 2008; Cardone et al., 2014; Guan et al.,
tion competent environment that allows viral genome expression 2015a; Lee et al., 2014). Currently, the highest-resolution struc-
and packaging into infectious particles (Doorbar et al., 2015). ture of the intact T = 7d HPV16 capsid is a 9-A resolution cryo-
Since the first reported association with cancer, extensive HPV EM structure, from which a pseudoatomic model was developed
research has focused on the prevention of cervical cancer, using the major structural protein, L1 (Cardone et al., 2014). The
including vaccine development to prevent virus infection (Malik 5560 nm diameter HPV capsid is composed of 360 copies of
et al., 2014; Nicol et al., 2015; Toh et al., 2015). However, the L1 arranged into 72 capsomers, each composed of five copies
current commercial vaccines are genotype specific and provide of L1. Twelve capsomers lay on the icosahedral 5-fold vertices
little therapeutic benefit to patients with existing HPV infections and are referred to as pentavalent capsomers, whereas the

Structure 25, 253263, February 7, 2017 2016 Published by Elsevier Ltd. 253
remaining 60 capsomers are each surrounded by six other conformational changes of virus during entry (Florin et al., 2012;
capsomers and referred to as hexavalent capsomers. An exten- Knappe et al., 2007; Levy et al., 2009). Co-crystallization of a
sion of the L1 C terminus, or C-terminal arm, links capsomers truncated L1 capsomer together with heparin oligosaccharides
together and provides interactions leading to the formation of (8 and 10 monosaccharide units) suggested four distinct heparin
disulfide bonds between Cys175 and Cys428 from two neigh- binding sites that mapped to the L1 protein (Dasgupta et al.,
boring L1 proteins (Buck et al., 2005a, 2005b; Cardone et al., 2011). Three of these HS binding sites were verified by subse-
2014; Sapp et al., 1998). Although the invading arm structure quent mutational analysis (Dasgupta et al., 2011; Richards
of bovine papilloma structure is known (Wolf et al., 2010), the et al., 2013). Nonetheless, the molecular mechanisms regarding
stabilizing C-terminal arm structure has not been solved yet the binding of heparin during entry remain fragmented and not
for HPV. completely understood.
Besides the L1 major capsid protein, there is an uncertain With recent technological advances in cryo-EM image detec-
number of L2 minor structural proteins that co-assemble with tion and the introduction of new software classification schemes,
L1 to form infectious virus capsids. The L2 minor protein has a we can now investigate HPV at near-atomic resolution. Here, we
number of functions including encapsidation of DNA, involve- present the near-atomic resolution structures of HPV16 alone
ment in the conformational changes of the capsid during cell and HPV16 interacting with heparin molecules at 4-A resolutions.
entry, disruption of the endosomal membrane, and subcellular Three-dimensional (3D) classification of the data revealed that
trafficking of the incoming viral genome (Bronnimann et al., HPV capsids are heterogeneous in nature but contain sub-pop-
2013; Buck et al., 2008; Raff et al., 2013). Specifically, the N-ter- ulations of capsids with consistent diameters. The classification
minal residues of L2 (188) are exposed on the surface of the of particles improved the high-resolution maps, which facilitated
capsid although the anti-L2 antibody, RG-1 recognition of L2 building de novo the L1 protein (residues 3485), revealing the
(1736) is dependent on maturation status or cell attachment structure of the C-terminal arm. Solving the L1 asymmetric unit
(Day et al., 2008a, 2008b; Finnen et al., 2003; Lowe et al., allowed us to calculate an L1-only capsid that was used to iden-
2008; Okun et al., 2001). An N-terminal L2 motif (RXXR, furin tify the location of L2 densities in our maps. Binding of heparin
cleavage site) is conserved across HPV genotypes and thus induced no conformational changes in the capsid that include
has promise as a new target for vaccine development (Bronni- local changes in L1, but there are alterations to putative L2 den-
mann et al., 2013; Conway et al., 2011; Roden et al., 2001). A sities. Besides the identification of the heparin binding site in the
previous structural study using pseudovirus maps (23 and context of the intact capsid, the changes induced by heparin
25 A) interpreted small densities beneath each capsomer as binding provide the first glimpse of an HPV entry intermediate.
belonging to L2 (Buck et al., 2008). Based on analysis of the We propose an entry mechanism model that may include isola-
late gene products, L2 protein has been predicted to bind within tion of specific 5-fold capsomers due to potential interaction
the center of the L1 capsomer with the antigenic region facing between heparin and L2 protein.
outward (Lowe et al., 2008). Stoichiometric studies have indi-
cated that there are about 1236 L2 molecules per HPV capsid RESULTS
(Okun et al., 2001); however, other works have suggested the
amount of L2 per capsid is variable, and may include up to 72 Near-Atomic Resolution Maps Were Reconstructed by
L2 molecules per capsid, perhaps one L2 at each capsomer Selecting Homogeneous Sub-populations
(Buck et al., 2008; Doorbar and Gallimore, 1987; Trus et al., HPV16 capsids and capsids incubated with heparin in excess
1997). Naturally produced virions average approximately 2436 were vitrified for cryo-EM data collection using new direct elec-
L2 molecules per capsid (Bronnimann et al., 2013; Buck et al., tron detector technology. The drift-corrected micrographs of
2008; Campos and Ozbun, 2009). Despite these variable reports, HPV and HPV-heparin displayed bumpy spherical shapes
there is general acceptance that between 12 and 72 L2 proteins with evident capsomer protrusions (Figures 1A and 1B). Cap-
per capsid are incorporated, each into a central internal cavity of sids contained internal density corresponding to packaged
the capsomer via L2 C-terminal interactions. DNA and appeared to have diameters of about 60 nm. Howev-
During host entry and endocytosis, HPV conformational er, 3D classification using REgularized LIkelihood OptimizatioN
changes are triggered by binding to a cell receptor (Abban and (RELION) showed that the populations of HPV and HPV-hepa-
Meneses, 2010; Kines et al., 2009; Selinka et al., 2003). Cell rin capsids were heterogeneous in size (Figures S1 and S2).
surface heparan sulfates (HSs) serve as a primary attachment Both capsid types included a large group of approximately
receptor (Buck et al., 2006; Rommel et al., 2005; Schowalter 50% of the total number of particles that had the same diam-
et al., 2011; Shafti-Keramat et al., 2003) and it is not the protein eter of 569570 A. Final reconstructions using AUTO3DEM
backbone but the glycan moieties, specifically the sulfation with the gold-standard approach used 50% of the HPV parti-
pattern, that are required for virus infection (Cerqueira et al., cles and 50% of the HPV-heparin complex particles to recon-
2013). HS proteoglycans are expressed and secreted by nearly struct a homogeneous population in each case to near-atomic
all mammalian cells, and are located on both the cell surface resolution (Figures 1C and 1D). The 4.3-A resolution of the
and extracellular matrix (Bernfield et al., 1992; Buck et al., maps was estimated where the Fourier shell correlation (FSC)
2006; Schowalter et al., 2011; Selinka et al., 2003). However, dropped below 0.143 (Figure 2) (Table 1). Central sections
because HS polysaccharides are expensive and difficult to through the density maps illustrate the modest quality of the re-
obtain, heparin has been used in lieu of HS in most biological constructions (Figures 1E and 1F). At this resolution, we can
and structure studies (Cerqueira et al., 2013; Selinka et al., see strand separation, the secondary structures of a helices
2007). Both heparin and HS binding have been shown to induce and b strands (Figures 3AI, 3AII, 3BI, and 3BII), and side chains

254 Structure 25, 253263, February 7, 2017


Figure 2. Plot Showing the FSC versus Spatial Frequency of the
Icosahedrally Averaged Reconstructions for HPV16 and HPV16-
Heparin Complex
The resolution of the reconstructions was assessed where the FSC curve
crossed a correlation value of 0.143 (dashed line).

of some residues can be identified (Figures 3A13A3 and


3B13B3).

Building L1 into the Cryo-EM Density Solved the


Structure of the C-Terminal Invading Arm and Revealed
the Location of L2
Strong continuous density facilitated the modeling of the L1
protein structure from residues 3 to 485, which included the
C-terminal arm that links capsomers. The structure building
was initiated with a combination of the previously solved pseu-
doatomic L1 model (PDB: 3J6R) and the crystal structure of
the truncated L1 (PDB: 3OAE). During the iterative modeling
and refinement, six full-length L1 molecules were built separately
to make an asymmetric unit of the capsid (Figure 4A). After
applying icosahedral symmetry to this asymmetric unit an L1-
only density map was generated (see Experimental Procedures).
A difference map was calculated by subtracting the L1-only
density from the L1/L2-containing HPV16 experimental map. Dif-
ference density mapped to both pentavalent and hexavalent
capsomers (Figures 4B and 4C). Putative L2 density began at
the inside base of the capsomer (Figure 4C), rose along the
Figure 1. Cryo-EM Reconstruction of HPV16 and HPV16-Heparin
outside of the capsomer to the shoulder, ending at the outside
(A and B) Representative regions of cryo-EM micrographs of the HPV16
particle capsids (A) and HPV16 capsids complexed with heparin (B).
center of the capsomer (Figure 4B). Corresponding densities in
(C and D) The surface rendered 3D maps of HPV16 (C) and capsid-heparin the pentavalent capsomer were significantly smaller than the
complex (D) were radially colored according to the distance from the center densities in hexavalent capsomers, both at the surface and the
(color key in angstroms) of the capsid and surface rendered at 1s. The heparin internal part at the base.
difference density (black, difference map rendered at 4.2 s) was found at
positions surrounding only the 5-fold vertices of the capsid. Distinct Structural Differences Were Observed between
(E and F) The central sections through the cryo-EM density maps show the
Pentavalent and Hexavalent Capsomers
quality of the reconstructions. Capsids were cut vertically through the 2-, 3-,
and 5-fold icosahedral symmetry axes (black lines), with the central 2-fold axis Our near-atomic structure of the HPV16 capsid supplied more
appearing at the 12 oclock position. detailed information on the capsid surface topology than the
(G) The zoom-in view of the HPV16-heparin complex shows the detailed fea- maps we produced before (Guan et al., 2015a, 2015b; Lee
tures of bound heparin molecules. The 5-fold symmetric axis was represented et al., 2014). Besides the entire C-terminal arm, surface loops
by black pentagon. could be distinguished separately, especially the BC and EF

Structure 25, 253263, February 7, 2017 255


Table 1. Cryo-EM Image Reconstruction Data located at the tip of the star-shaped capsomer, had the lowest
resolution (6.5 A) in both pentavalent and hexavalent capsomers.
HPV HPV-Heparin
No. of micrographs 9,137 6,175
The HPV-Heparin Complex Showed that Heparin
Defocus level range (mm) 1.045.19 0.784.85 Receptors Only Bound at the 5-fold Vertex of the
No. of particles selected from 115,112 102,829 HPV16 Virus
micrograph Significant extra densities were found at each 5-fold vertex of the
No. of particles selected for 57,556 51,422 HPV-heparin complex map compared with the HPV map. This
reconstruction putative heparin density was comparable with the magnitude
Final resolution 4.3 4.3 of the average capsid density, which suggested nearly full occu-
pancy of bound heparin. The regions of strongest density were
located between neighboring pairs of BC and EF loops; however,
loops that were seen as merged densities in the previous lower- there was noisy discontinuous density stretching out and away
resolution maps (Figure 4A). To further visualize the differences from the bound heparin (Figure 1E). The mixture of polysaccha-
between L1 conformations in the two different environments, ride chain lengths (1030) in the heparin incubated with the virus
the newly built structures of the pentavalent and hexavalent in our study likely caused the lesser densities extending away
capsomers were superimposed (Figure 5A). The conformational from the bound heparin, which corresponded to longer polysac-
differences were obvious on both surface loops and C-ter- charide chains of bound heparin that were averaged with shorter
minal arms, with a root-mean-square deviation (RMSD) value chains during the reconstruction process.
of 4.77 A between the entire sets of L1 atoms of the two cap-
somers. Since the C-terminal arm conformations were notice- Heparin Binding Triggers Conformational Changes of L2
ably different, the RMSD was recalculated with this part of the Protein without Altering the Capsid Diameter
structure truncated, and found to be 1.49 A, indicating sig- No significant difference in diameter between HPV16 and
nificantly different loop structures throughout. Specifically the HPV16-heparin complex maps was seen in the radial density
hexavalent C-terminal arm extended further away from the plots (Figure 6A), indicating that heparin binding did not trigger
hexavalent capsomer, whereas the pentavalent arms were a change in the overall size of the capsid. The structure of L1
arranged closer in a curved arc at the site of interaction with was built into the HPV-heparin complex map to make an asym-
the neighboring hexavalent capsomer. The individual BC, DE, metric unit and an L1-only capsid map was calculated as
FG, HI, and EF loops are in decidedly different conformations described above for the capsid. The mass axis of L1 proteins
throughout; however, overall the hexavalent capsomers formed in pentavalent and hexavalent capsomers in the HPV and the
a more open crown-like structure compared with the pentavalent HPV-heparin maps were calculated (Figure 6B) to track both
capsomers. rotational and translational movements of L1 proteins. L1 pro-
Differences between the pentavalent and hexavalent cap- teins rotated no more than 0.4 and shifted less than 0.1 A (Fig-
somers were also apparent in the local resolution map (Figures ure S3). Thus, no significant movement of L1 protein was found
5B and 5C). The pentavalent capsomer surface had lower local after binding with heparin.
resolution compared with the hexavalent capsomer, especially To check the surface loop rearrangement of the L1 pro-
near the center, suggesting the pentavalent capsomer loops tein, the HPV and HPV-heparin asymmetric unit structures
had more flexibility. Overall the very center and the outer edges were superimposed. An RMSD of 0.76 A suggested slight
of the capsomers had lower resolution than the mid-regions of changes may have occurred after heparin binding. Super-
the capsomers. Specifically, the BC and EF loops, which were imposing the HPV hexavalent and pentavalent capsomers on

Figure 3. Secondary Structure and Residue Side Chain of the L1 Protein Were Identified in the Maps
Secondary structure of a helix and b sheet (blue) in the L1 protein was shown in the corresponding density in the map (green) of HPV16 (AI) and (AII) and
HPV-heparin complex (BI) and (BII). The side chain of the residues were also identified in each of maps. For example, (A1) 311.Tyr, (A2) 447.Trp, (A3) 467.Lys in
HPV16 map; and (B1) 311.Tyr, (B2) 447.Trp, (B3) 467.Lys in the HPV16-heparin map.

256 Structure 25, 253263, February 7, 2017


difference densities from the HPV map (Figures 4B, 4C, 6D, 6E,
and S4). After binding with heparin, difference densities in the
HPV-heparin complex map included putative L2 density that map-
ped to both pentavalent and hexavalent capsomers. Compared to
virus alone, there was redisposition within the HPV-heparin cap-
somer and more putative L2 densities on the top surface of the
capsomer and inside the capsomers, especially for the pentava-
lent capsomers. These changes suggest that heparin binding
initiates conformational changes that may result in additional L2
residues becoming exposed at the capsid surface and the side
wall of capsomers, which is consistent with previous studies.
The HPV-heparin local resolution map was compared with the
HPV map to show that areas of poorer local resolution mapped
to the centers of the capsomers in the complex map (Figure 5C).
However, resolution at the rim of the BC and EF loops on penta-
valent capsomers improved slightly after binding with heparin,
possibly due to a stabilizing effect (Figure 5C, indicated by
arrow). Overall, both the pentavalent and hexavalent capsomers
showed regions of poorer resolution after heparin binding, sug-
gesting local flexible movements.

The Receptor Binding Site Is between Hexavalent and


Pentavalent Capsomers
The structures of multiple disaccharide units of heparin were
manually fitted one-by-one into the difference map calculated
by subtracting HPV from the HPV16-heparin reconstruction. The
fitting result for ten units was then refined in the HPV16-heparin
complex map. The resultant heparin model and the L1 protein
model were then used to describe the binding site (Figure 7A).
The HPV-heparin interaction appears to be conferred by sulfated
polysaccharide residues and six different side chains from two
contributing L1 proteins. Specifically, in the model two BC loops,
one EF loop, and one C-terminal arm from two neighboring L1 pro-
teins participated in the binding. One hexavalent L1 protein
contributed the 59.Lys of the BC loop and 176.Thr and 177.Asn
of the EF loop for interaction. All the other binding residues in the
model were from one pentavalent L1 that contributed a 57.Asn,
58.Asn, and 59.Lys from the BC loop, and C-terminal arm residues
of 428.Cys, 429.Gln, 431.His, 432.Thr, and 433.Pro (Figure 7B).
Figure 4. The Molecule Building of L1 Protein and Putative L2
Densities in the Map
DISCUSSION
(A) The six copies of L1 (blue) were built into the HPV16 map density (gray) to
complete the asymmetric unit. The pentavalent and hexavalent capsomers are
indicated by a black pentagon and hexagon, respectively. The C-terminal arm Variations in the size of the HPV capsid have been attributed pre-
densities are continuous and clearly identified in the map. viously to progressive stages of maturation of the capsid through
(B) Difference density calculated from subtracting the L1-only capsid map formation of disulfide linkages conferred by the C-terminal arm
from the HPV16 experimental map (gray) showed difference densities (red) on using the pseudovirus model. Similarly, our 3D classification of
top view to show exposed densities on the surfaces of both pentavalent and
the quasi-HPV capsids used in our study (Experimental Proced-
hexavalent capsomers, with wormlike shapes extending between capsid floor
ures) showed different-diameter capsids existing with similar
and center of the capsomer.
(C) Cross-section view to show the L2 densities inside the capsomers. surface features (Figure S1). This size variation in capsids may
relate to the CRPV genome or maturation status. Variation also
suggests that heterogeneity is a natural attribute of HPV, which
the corresponding HPV-heparin capsomers resulted in RMSD may be linked to survival outside of the host in a hostile environ-
values of 0.51 and 1.12 A, respectively (Figure 6C). There was ment and may represent an evolving adaptability. For instance,
no significant loop rearrangement among all HI, EF, DE, FG, the range in diameter may be linked to stability, providing classes
and BC loops (Figure 6C), and no significant change in the diam- of more (or less) stable particles. Once assembled into capsids,
eter of the capsid. the heterogeneous characteristic of HPV during and after the
By subtracting the calculated HPV-heparin L1-only map from maturation process may provide an advantage ensuring the sur-
HPV-heparin experimental map, we found a difference density vival of an existing sub-population, within the mixed population
that had significant conformational changes compared with the of capsids.

Structure 25, 253263, February 7, 2017 257


Figure 5. Molecule Model Comparison and
Local Resolution of Pentavalent and Hexa-
valent Capsomers
(A) The L1 molecular structures of pentavalent
(red) and hexavalent (blue) capsomers are super-
imposed to show the structural differences,
especially between the C-terminal arms (blue ar-
row), with an RMSD value of 4.77 A between the
atom sets in two capsomers.
(B and C) The zoomed view of a pentavalent and
hexavalent capsomer of each local resolution map
of HPV16 (B) and HPV16-heparin (C). Resolution of
the map (color key in angstroms) shows that inner
and outermost densities have the lowest resolution
and, presumably, the most flexibility. (C) The res-
olution of the BC and EF loop densities (arrow)
improves after binding with heparin.

the third possibility for L2 incorporation


into the capsid is that all copies of L2
may cluster in a unique locus that includes
a pentavalent capsomer and some of the
surrounding five hexavalent capsomers.
Compared with the well-understood nature and structure An anisotropic distribution of L2 protein in the capsid implies
of the L1 major protein, the location and number of L2 minor that a unique vertex may exist, which may function during host
proteins per capsid have remained controversial, despite the cell binding and infection. However, due to the magnitude of
essential requirement for genome encapsidation and subcellular these difference densities they cannot be definitively appor-
trafficking. During the past 25 years, extensive biochemical and tioned or assigned to L2 molecules.
structural studies have attempted to lift the veil shrouding L2 The L2 N terminus has been shown previously to be accessible
protein incorporation. Using the L1-only calculated map, we at the surface of the capsomer (Conway et al., 2011; Lowe et al.,
identified non-L1 density in our experimental map and suggest 2008), and such a location is suggested in our map (Figure 3B).
that difference density can be attributed to L2. L2 protein was The incorporated L2 protein may start from the middle center
observed to intertwine with L1 protein at the base part of the of the capsomer, extending to the surface along the side wall
capsomer and extend along the side wall of the capsomer in to reach the capsid floor with most of the molecule including
the canyon to the top surface until reaching the upper center the L2 C terminus intertwining with the L1 protein and interacting
of the capsomer. This observation was consistent with a previ- with the genome at the base of the capsomer. At the current res-
ous structural result that showed small L2 protein densities at olution we cannot solve the molecular structure of L2 peptide
the lower center of each capsomer (Buck et al., 2008). With the within the density (Figure 4) or define the orientation. However,
higher resolution that is now attainable, our map reveals more the morphological features and conformational changes of the
about the location and features of the L2 protein. Because of putative L2 protein densities might be used to guide future
the quasi-equivalent 5-fold symmetry characteristics of the hex- studies to contribute to understanding the mechanisms of entry
avalent capsomer, the features of putative L2 densities were and infection.
different between pentavalent and hexavalent capsomers. The Variations in the micro-environment of the T = 7d icosahedral
smaller potential density of L2 at the surface and inner parts of capsid (Guan et al., 2015a, 2015b; Lee et al., 2014; Xia et al.,
pentavalent capsomer suggested a flexible interaction of L1 2016; Zhao et al., 2014) underlie functional differences between
and L2 proteins in the pentavalent capsomer, which were aver- pentavalent and hexavalent capsomers. Specifically, the U4
aged out during the reconstruction process. Consistent with a antibody binds preferentially at the icosahedral 5-fold (Guan
previous model, L1 residues 261269 map near the L2 density et al., 2015a), which is consistent with the previous studies where
that drapes over the outside of the capsomers, suggesting U4 antibody prohibited the virus from binding cells (Day et al.,
possible interactions (Lowe et al., 2008). The variation of L2 dis- 2007; Deschuyteneer et al., 2010). Considering the functional dif-
tribution among capsomers suggested random L2 incorporation ferences between pentavalent and hexavalent L1 environments,
during capsid formation, which may be related to the long virion preferential heparin binding, and the established importance of
maturation time and the high heterogeneity of the population. L2 function during entry, we conclude that the 5-fold vertex
Among the 72 capsomers comprising the intact icosahe- can be considered the functional site for binding host cells during
dral capsid, L2 molecules may be incorporated asymmetrically entry. This model suggests L2 incorporation must include the
and randomly distributed in both pentavalent and hexavalent vertex pentamers.
capsomers. Alternatively, L2 proteins may be located on each Previous work has shown that HS binding triggered conforma-
of the 12 pentavalent capsomers with the other additional L2 tional changes to the capsid, which resulted in reduced affinity to
copies randomly distributed in hexavalent positions. Finally, the primary HS attachment receptor and more exposure of

258 Structure 25, 253263, February 7, 2017


Figure 6. Conformational Changes of HPV
Capsid Triggered by Heparin
(A) Radial density plots of HPV16 and HPV16-
heparin.
(B) The mass axis of L1 protein in pentavalent and
hexavalent capsomers for HPV (red) and HPV-HS
(blue) maps.
(C) The conformational changes of L1 surface
loops induced by heparin binding are indicated by
superimposing the pentavalent and hexavalent
capsomer models from HPV16 (red) and HPV16-
heparin (blue). The movement of FG, HI, BC, DE,
and EF loops were indicated. The RMSD value of
pentavalent and hexavalent capsomers before and
after heparin binding were measured as 1.12 and
0.51 A, respectively.
(D) L2 densities (blue) calculated by subtracting the
L1 only capsid model from HPV16-heparin map
was shown from top, whereas the heparin den-
sities were highlight in black.
(E) The cross-section view L2 densities (blue)
together with heparin (black) to show the densities
inside the capsomer. Pentavalent and hexavalent
capsomers were indicated by black pentagon and
hexagon.

changes. The movement of L2 after hepa-


rin binding also includes conformational
changes of L2 at the side wall of the cap-
somer near the canyon (Figures 6D, 6E,
and S4). These changes may indicate
that the binding sites for furin cleavage,
cyclophilin B, or other factors of the L2
protein are located at the side wall of the
capsomer, and the release of L2 and the
genome may be from the canyon during
the latter dis-assembly of the capsid;
L2 protein for furin cleavage (Day et al., 2008a, 2008b; Selinka however, further work remains to reveal the L2 structure and
et al., 2003). The HPV-heparin complex captured in our study subsequent changes.
showed no significant conformational changes of the L1 protein, Of the three heparin binding sites identified previously, the site
but suggested major L2 alteration. Post-attachment alterations occupied by heparin in our density map overlapped with two res-
to the HPV capsid have been reported previously (Guan et al., idues of site 3 (57.Asn, 59.Lys, 442.Lys, 443.Lys), whereas no
2015a) and may correspond to the heparin-triggered conforma- traces of bound heparin were found at the top of the capsomers
tional changes we report here that include small L1 protein involving the FG and HI loops previously identified as sites 1 and
movements at the apical surface of the capsomers. The surface 2 (Richards et al., 2013), and as the primary binding sites. How-
loops on hexavalent capsomer had no significant changes ever, considerable differences exist among these three experi-
after binding heparin (Figure 6C), which coincides with the re- ments including the form of HPV capsid (L1 only pentamers,
tained ability of antibody HPV16.V5 to neutralize well after cell pseudovirus, or quasivirus) or the type of heparin (heparin oligo-
attachment (Guan et al., 2015a; Lee et al., 2014). Compared saccharide 810, heparin oligomers, or heparin 1030) used. The
with HPV16.V5, the redisposition and more exposure of L2 den- seemingly disparate results suggest several possibilities. First,
sities on the surface may affect other antibody binding, for due to the variations in the reagents, the heparin used in these
example, H33.J3, which displayed improved neutralizing experiments was only capable of interacting at site 3 on our
efficiency to cell-bound virions (Selinka et al., 2003). The lack quasivirus capsids due to steric limitations of binding. Certainly,
of global conformational changes of L1 protein after binding it seems likely from our structure that heparin binding to site 3
heparin and the presence of possible L2 density near the would interfere with binding at site 2. Second, a stepwise
bound heparin molecule suggested that the L2 protein was likely sequence of events is conceivable. For example, heparin bound
participating in the binding with heparin (Figures 6D and 6E). If HPV capsids at site 1 during the 1-hr incubation; the bound hep-
heparin binds to both L1 and L2 proteins, perhaps the L1 interac- arin initiated the conformational changes revealed in the maps;
tions facilitate the orientation and the L2 binding with heparin whereupon heparin dissociated from site 1 and then bound
provides the contacts that triggered major conformational site 3 as revealed in the HPV-heparin complex structure.

Structure 25, 253263, February 7, 2017 259


fected together with linear CRPV/SV40ori DNA into 293TT cells and prepared
as described previously (Buck et al., 2005a, 2005b; Pastrana et al., 2004).
HPV16 was allowed to mature overnight and then pelleted by centrifugation.
The centrifuged pellet was resuspended in 1 M NaCl and 0.2 M Tris (pH 7.4).
After CsCl gradient purification, the lower band was collected, concentrated,
and dialyzed against PBS, as described previously (Guan et al., 2015a). The
concentrated HPV16 particles were applied to Formavar-coated copper grids,
stained with 2% phosphotungstic acid, and analyzed for integrity and concen-
tration on a JEOL JEM-1400 electron microscope.

Cryo-EM
HPV16 was incubated with a 5-fold excess of heparin molecules (Sigma-
Aldrich, catalog no. H1027) according to an average mass of 16 kDa per mole-
cule and 360 binding sites predicted per capsid. HPV-heparin was incubated
for 1 hr at 37 C and concentrated to 0.8 mg/mL in PBS buffer. An aliquot of 3 mL
of virus-Fab complex was vitrified on QUANTIFOIL R2/1 holey carbon support
grids (Quantifoil) that were vitrified by plunge-freezing into liquid ethane using a
Cryoplunge 3 (Gatan). Low-dose micrographs were recorded using an FEI
Polara G2 microscope operating at 300 kV. Images were collected under the
software control of the EPU program using an FEI Falcon 2 direct electron
detector with a nominal magnification of 93,0003 yielding a calibrated pixel
size at the sample of 1.15 A. A similar procedure was followed to prepare
and collect data for untreated HPV particles. Defocus ranges of 1.045.19
and 0.784.85 mm (Table 1) were measured respectively for micrographs of
HPV16 and HPV16-heparin, respectively. RELION, AUTO3DEM, and EMAN2
program suites were used for image processing and 3D reconstructions
(Scheres, 2012; Tang et al., 2007; Yan et al., 2007).

Image Processing
Whole-frame alignment was carried out using MotionCorr to account for
stage drift (Li et al., 2013). The microscope contrast transfer function pa-
rameters were estimated for each micrograph using ctffind4 (Rohou and
Grigorieff, 2015). Semiautomatic particle selection was performed using
EMAN2 e2boxer.py to obtain the particle coordinates, followed by particle
extraction, linearization, normalization, apodization, and 3D reconstruction
of the images using AUTO3DEM 4.05 following the gold standard of initiating
Figure 7. Epitope Analysis of Heparin Based on the Model and the the process by splitting the dataset into two independently refined halves
4.7-A Maps (Tang et al., 2007; Yan et al., 2007). The refinements for sharpening high-
The heparin binding site described by the model. (A) The fitted heparin resolution information were performed using EMBFACTOR (Fernandez
molecule (purple, yellow, and red) and L1 protein (blue) were used to locate the et al., 2008; Rosenthal and Henderson, 2003). Reported resolutions are esti-
contacts between heparin and L1. (B) The residues on the L1 protein are mated according to the gold-standard FSC = 0.143 criterion (Scheres and
indicated at their position with sequence number, residue name, and side Chen, 2012).
chain shown in red. The residues comprising the binding site identified here are For heterogeneity investigation, 3D classifications and refinements were
listed in the table along with their position on the L1 capsomer. performed using RELION software (Scheres, 2012). A windowing operation
was applied to the particles in Fourier space to downsize the particle images
Until now, no structural tools to detect HPV conformational to yield a final box size of 260 pixels2 (corresponding to a pixel size of 2.3 A).
changes during entry have been described. By using near- The final pixel size was chosen after optimization of the balance between
atomic reconstruction maps, local conformational changes the Nyquist frequency limit and the memory requirements for computations.
were seen for the first time. Since heparin molecules were only
Model Building
found at the pentavalent capsomer, triggering the conforma-
The atomic model of the HPV16 L1 capsomer with the C-terminal arm trun-
tional changes to expose more L2 densities in the canyon, these cated (PDB: 3OAE; residues 20403, 438474) was fitted as a rigid body
changes suggested that HPV infection is initiated through a into the cryo-EM map using UCSF Chimera (Dasgupta et al., 2011; Pettersen
pentavalent capsomer as a unique 5-fold vertex that may func- et al., 2004). Placement of the pseudoatomic model of an asymmetric unit of
tion during later stages of entry that include release of the DNA HPV16 capsid (PDB: 3J6R; residues 9486) was used as a guideline during
genome tethered to L2. In support of this model is the possibility building (Cardone et al., 2014). Model building proceeded de novo to place
the missing C-terminal arm residues 404437 and the N and C termini through
that L2 might be incorporated into the HPV capsid to form a
COOT (Emsley et al., 2010). Specifically, 17 residues were built into density at
unique vertex. Further studies will be needed to resolve these the N-terminal end and 11 residues at the C-terminal end. Each L1 molecule in
questions. the asymmetric unit was built independently using COOT (Emsley et al., 2010).
Icosahedral symmetry was applied to the refined asymmetric unit model to
EXPERIMENTAL PROCEDURES generate the whole capsid through SITUS (Wriggers, 2010; Wriggers et al.,
1999), with the parameters of mass-weight of the atom, pixel size 1.147 A,
Preparation of HPV16 Quasiviruses kernel width 4.8 A, Gaussian smoothing kernel, and scaling factor of 1.
HPV16 quasivirus containing L1 and L2 proteins and encapsidating a CRPV The capsid atomic model was then further refined iteratively using the Phenix
genome having the SV40 origin of replication was prepared as described Real Space Refinement program (Afonine et al., 2012). At each iteration, the
previously (Brendle et al., 2010; Mejia et al., 2006; Pyeon et al., 2005). In best model was visually inspected in COOT and adjusted manually accord-
brief, HPV16 sheLL plasmid (kindly provided by John Schiller, NIH) was trans- ingly to the best fit into the density and guided by torsion restraints, planar

260 Structure 25, 253263, February 7, 2017


peptide restraints, and Ramachandran restrains. The quality of the final model Brendle, S.A., Culp, T.D., Broutian, T.R., and Christensen, N.D. (2010). Binding
was evaluated by MolProbity (http://molprobity.biochem.duke.edu/) (Chen and neutralization characteristics of a panel of monoclonal antibodies to
et al., 2010) (Table S1). The same building and refinement procedures were human papillomavirus 58. J. Gen. Virol. 91, 18341839.
performed for the HPV-heparin complex map. Difference maps were calcu- Bronnimann, M.P., Chapman, J.A., Park, C.K., and Campos, S.K. (2013). A
lated using SITUS (Wriggers, 2010; Wriggers et al., 1999). transmembrane domain and GxxxG motifs within L2 are essential for papillo-
Coordinates for the disaccharide unit of heparin isolated from (PDB: 3OAE) mavirus infection. J. Virol. 87, 464473.
were used as the basis for the model building (Dasgupta et al., 2011). The hep-
Buck, C.B., Pastrana, D.V., Lowy, D.R., and Schiller, J.T. (2005a). Generation
arin polysaccharide model was built manually using disaccharide units fitted
of HPV pseudovirions using transfection and their use in neutralization assays.
one-by-one into the difference map made by subtracting HPV16 from the
Methods Mol. Med. 119, 445462.
HPV16-heparin complex map. The result of initial fitting was refined using
the HPV16-heprin complex map. The final result included ten disaccharide Buck, C.B., Thompson, C.D., Pang, Y.-Y.S., Lowy, D.R., and Schiller, J.T.
units of heparin built in the density with a correlation coefficient of 83% accord- (2005b). Maturation of papillomavirus capsids. J. Virol. 79, 28392846.
ing to Chimera (Pettersen et al., 2004). Contacts between the fitted heparin and Buck, C.B., Thompson, C.D., Roberts, J.N., Mu ller, M., Lowy, D.R., and
L1 protein structures were identified using Chimera with the criteria for van der Schiller, J.T. (2006). Carrageenan is a potent inhibitor of papillomavirus infec-
Waals overlap distances set at 0.4 and 0.0 A. Clashes between atoms were tion. PLoS Pathog. 2, e69.
defined by any overlap of 0.6 A or more. Buck, C.B., Cheng, N., Thompson, C.D., Lowy, D.R., Steven, A.C., Schiller,
J.T., and Trus, B.L. (2008). Arrangement of L2 within the papillomavirus capsid.
ACCESSION NUMBERS J. Virol. 82, 51905197.
Campos, S.K., and Ozbun, M.A. (2009). Two highly conserved cysteine resi-
Cryo-EM maps for the HPV16 and HPV16-heparin complexes have been dues in HPV16 L2 form an intramolecular disulfide bond and are critical for
deposited in the EM database (www.emdatabank.org/) with accession infectivity in human keratinocytes. PLoS One 4, e4463.
numbers EMD: 6620 and 6619. Coordinates for the atomic model of the Cardone, G., Moyer, A.L., Cheng, N., Thompson, C.D., Dvoretzky, I., Lowy,
asymmetric unit of HPV16 L1 proteins and the complex with the heparin D.R., Schiller, J.T., Steven, A.C., Buck, C.B., and Trus, B.L. (2014).
molecule have been deposited in the PDB under accession codes PDB: Maturation of the human papillomavirus 16 capsid. MBio 5, e01104e01114.
5KEP and 5KEQ.
Cerqueira, C., Liu, Y., Ku hling, L., Chai, W., Hafezi, W., van Kuppevelt, T.H.,
hn, J.E., Feizi, T., and Schelhaas, M. (2013). Heparin increases the infectivity
Ku
SUPPLEMENTAL INFORMATION of human papillomavirus type 16 independent of cell surface proteoglycans
and induces L1 epitope exposure: interactions of HPV-16 with GAGs. Cell
Supplemental Information includes four figures and one table and can be Microbiol. 15, 18181836.
found with this article online at http://dx.doi.org/10.1016/j.str.2016.12.001.
Chen, X.S., Garcea, R.L., Goldberg, I., Casini, G., and Harrison, S.C. (2000).
Structure of small virus-like particles assembled from the L1 protein of human
AUTHOR CONTRIBUTIONS
papillomavirus 16. Mol. Cell 5, 557567.

J.G., S.M.B., and S.A.B. performed the research; R.E.A. performed micro- Chen, V.B., Arendall, W.B., Headd, J.J., Keedy, D.A., Immormino, R.M.,
scopy and collected the cryo-EM data; A.M.M. and J.F.C. collected the Kapral, G.J., Murray, L.W., Richardson, J.S., and Richardson, D.C. (2010).
cryo-EM data; J.G. processed and analyzed the cryo-EM data; J.G., N.D.C., MolProbity: all-atom structure validation for macromolecular crystallography.
J.F.C., and S.H. wrote the paper. Acta Crystallogr. D Biol. Crystallogr. 66, 1221.
Christensen, N.D. (2005). Cottontail rabbit papillomavirus (CRPV) model
ACKNOWLEDGMENTS system to test antiviral and immunotherapeutic strategies. Antivir. Chem.
Chemother. 16, 355362.
This work was supported in part by the J. Gittlen Memorial Golf Tournament Conway, M.J., Cruz, L., Alam, S., Christensen, N.D., and Meyers, C. (2011).
and the Pennsylvania Department of Health CURE funds. Research reported Cross-neutralization potential of native human papillomavirus N-terminal L2
in this publication was also supported by the Office of the Director, NIH, under epitopes Torbjorn Ramqvist (ed.). PLoS One 6, e16405.
award numbers S10OD019995 and S10OD011986. The content is solely the Dasgupta, J., Bienkowska-Haba, M., Ortega, M.E., Patel, H.D., Bodevin, S.,
responsibility of the authors and does not necessarily represent the official Spillmann, D., Bishop, B., Sapp, M., and Chen, X.S. (2011). Structural basis
views of the NIH. of oligosaccharide receptor recognition by human papillomavirus. J. Biol.
Chem. 286, 26172624.
Received: June 8, 2016
Day, P.M., Thompson, C.D., Buck, C.B., Pang, Y.-Y.S., Lowy, D.R., and
Revised: October 7, 2016
Schiller, J.T. (2007). Neutralization of human papillomavirus with monoclonal
Accepted: December 12, 2016
antibodies reveals different mechanisms of inhibition. J. Virol. 81, 87848792.
Published: January 5, 2017
Day, P.M., Gambhira, R., Roden, R.B.S., Lowy, D.R., and Schiller, J.T. (2008a).
REFERENCES Mechanisms of human papillomavirus type 16 neutralization by l2 cross-
neutralizing and l1 type-specific antibodies. J. Virol. 82, 46384646.
Abban, C.Y., and Meneses, P.I. (2010). Usage of heparan sulfate, integrins, Day, P.M., Lowy, D.R., and Schiller, J.T. (2008b). Heparan sulfate-independent
and FAK in HPV16 infection. Virology 403, 116. cell binding and infection with furin-precleaved papillomavirus capsids.
Afonine, P.V., Grosse-Kunstleve, R.W., Echols, N., Headd, J.J., Moriarty, J. Virol. 82, 1256512568.
N.W., Mustyakimov, M., Terwilliger, T.C., Urzhumtsev, A., Zwart, P.H., and Deschuyteneer, M., Elouahabi, A., Plainchamp, D., Plisnier, M., et al. (2010).
Adams, P.D. (2012). Towards automated crystallographic structure refinement Molecular and structural characterization of the L1 virus-like particles that
with phenix.refine. Acta Crystallogr. D Biol. Crystallogr. 68, 352367. are used as vaccine antigens in CervarixTM, the AS04-adjuvanted HPV-16
Baker, T.S., Newcomb, W.W., Olson, N.H., Cowsert, L.M., Olson, C., and and -18 cervical cancer vaccine. Hum. Vaccin. 6, 407419.
Brown, J.C. (1991). Structures of bovine and human papillomaviruses. Dickson, E.L., Vogel, R.I., Luo, X., and Downs, L.S. (2015). Recent trends in
Analysis by cryoelectron microscopy and three-dimensional image recon- type-specific HPV infection rates in the United States. Epidemiol. Infect.
struction. Biophys. J. 60, 14451456. 143, 10421047.
Bernfield, M., Kokenyesi, R., Kato, M., Hinkes, M.T., Spring, J., Gallo, R.L., and Doorbar, J., and Gallimore, P.H. (1987). Identification of proteins encoded by
Lose, E.J. (1992). Biology of the syndecans: a family of transmembrane hep- the L1 and L2 open reading frames of human papillomavirus 1a. J. Virol. 61,
aran sulfate proteoglycans. Annu. Rev. Cell Biol. 8, 365393. 27932799.

Structure 25, 253263, February 7, 2017 261


Doorbar, J., Egawa, N., Griffin, H., Kranjec, C., and Murakami, I. (2015). Human (HPV) capsid vaccines in vivo using infectious HPV/cottontail rabbit papilloma-
papillomavirus molecular biology and disease association: human papilloma- virus chimeric papillomavirus particles. J. Virol. 80, 1239312397.
virus. Rev. Med. Virol. 25, 223. Nicol, A.F., de Andrade, C.V., Russomano, F.B., Rodrigues, L.S.L., Oliveira,
Emsley, P., Lohkamp, B., Scott, W.G., and Cowtan, K. (2010). Features and N.S., Provance, D.W., and Nuovo, G.J. (2015). HPV vaccines: their pathol-
development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486501. ogy-based discovery, benefits, and adverse effects. Ann. Diagn. Pathol. 19,
Fernandez, J.J., Luque, D., Caston, J.R., and Carrascosa, J.L. (2008). 418422.
Sharpening high resolution information in single particle electron cryomicro- Okun, M.M., Day, P.M., Greenstone, H.L., Booy, F.P., Castle, P.E., FitzGerald,
scopy. J. Struct. Biol. 164, 170175. ger Kjaer, S., Lowy, D.R., and Schiller, J.T. (2001). L1 Interaction do-
P.C., Kru
Finnen, R.L., Erickson, K.D., Chen, X.S., and Garcea, R.L. (2003). Interactions mains of papillomavirus L2 necessary for viral genome encapsidation. J. Virol.
between papillomavirus L1 and L2 capsid proteins. J. Virol. 77, 48184826. 75, 43324342.
Florin, L., Sapp, M., and Spoden, G.A. (2012). Host-cell factors involved in Pastrana, D.V., Buck, C.B., Pang, Y.-Y.S., Thompson, C.D., Castle, P.E.,
papillomavirus entry. Med. Microbiol. Immunol. 201, 437448. ger Kjaer, S., Lowy, D.R., and Schiller, J.T. (2004).
FitzGerald, P.C., Kru
Gilmer, L.S. (2015). Human papillomavirus vaccine update. Prim. Care Reactivity of human sera in a sensitive, high-throughput pseudovirus-based
42, 1732. papillomavirus neutralization assay for HPV16 and HPV18. Virology 321,
Guan, J., Bywaters, S.M., Brendle, S.A., Lee, H., Lee, H., Ashley, R.E., 205216.
Makhov, A.M., Conway, J.F., Christensen, N.D., and Hafenstein, S. (2015a). Pettersen, E.F., Goddard, T.D., Huang, C.C., Couch, G.S., Greenblatt, D.M.,
Structural comparison of four different antibodies interacting with human Meng, E.C., and Ferrin, T.E. (2004). UCSF chimera: a visualization system
papillomavirus 16 and mechanisms of neutralization. Virology 483, 253263. for exploratory research and analysis. J. Comput. Chem. 25, 16051612.
Guan, J., Bywaters, S.M., Brendle, S.A., Lee, H., Ashley, R.E., Christensen, Pyeon, D., Lambert, P.F., and Ahlquist, P. (2005). Production of infectious
N.D., and Hafenstein, S. (2015b). The U4 antibody epitope on human papillo- human papillomavirus independently of viral replication and epithelial cell
mavirus 16 identified by cryo-electron microscopy S. Perlman (ed.). J. Virol. 89, differentiation. Proc. Natl. Acad. Sci. USA 102, 93119316.
1210812117.
Raff, A.B., Woodham, A.W., Raff, L.M., Skeate, J.G., Yan, L., Da Silva,
Hernandez, B.Y., Ton, T., Shvetsov, Y.B., Goodman, M.T., and Zhu, X. (2012). D.M., Schelhaas, M., and Kast, W.M. (2013). The evolving field of human
Human papillomavirus (HPV) L1 and L1-L2 virus-like particle-based multiplex papillomavirus receptor research: a review of binding and entry. J. Virol.
assays for measurement of HPV virion antibodies. Clin. Vaccin. Immunol. 19, 87, 60626072.
13481352.
Richards, K.F., Bienkowska-Haba, M., Dasgupta, J., Chen, X.S., and Sapp,
Kines, R.C., Thompson, C.D., Lowy, D.R., Schiller, J.T., and Day, P.M. (2009).
M. (2013). Multiple heparan sulfate binding site engagements are required
The initial steps leading to papillomavirus infection occur on the basement
for the infectious entry of human papillomavirus type 16. J. Virol. 87,
membrane prior to cell surface binding. Proc. Natl. Acad. Sci. USA 106,
1142611437.
2045820463.
Roden, R.B., Day, P.M., Bronzo, B.K., Yutzy, W.H., Yang, Y., Lowy, D.R., and
Kirnbauer, R., Booy, F., Cheng, N., Lowy, D.R., and Schiller, J.T. (1992).
Schiller, J.T. (2001). Positively charged termini of the L2 minor capsid protein
Papillomavirus L1 major capsid protein self-assembles into virus-like particles
are necessary for papillomavirus infection. J. Virol. 75, 1049310497.
that are highly immunogenic. Proc. Natl. Acad. Sci. USA 89, 1218012184.
Kirnbauer, R., Taub, J., Greenstone, H., Roden, R., Du rst, M., Gissmann, L., Rohou, A., and Grigorieff, N. (2015). CTFFIND4: fast and accurate defocus
Lowy, D.R., and Schiller, J.T. (1993). Efficient self-assembly of human papillo- estimation from electron micrographs. J. Struct. Biol. 192, 216221.
mavirus type 16 L1 and L1-L2 into virus-like particles. J. Virol. 67, 69296936. Rommel, O., Dillner, J., Fligge, C., Bergsdorf, C., Wang, X., Selinka, H.C., and
Knappe, M., Bodevin, S., Selinka, H.-C., Spillmann, D., Streeck, R.E., Chen, Sapp, M. (2005). Heparan sulfate proteoglycans interact exclusively with con-
X.S., Lindahl, U., and Sapp, M. (2007). Surface-exposed amino acid residues formationally intact HPV L1 assemblies: basis for a virus-like particle ELISA.
of HPV16 L1 protein mediating interaction with cell surface heparan sulfate. J. Med. Virol. 75, 114121.
J. Biol. Chem. 282, 2791327922. Rosenthal, P.B., and Henderson, R. (2003). Optimal determination of particle
Lee, H., Brendle, S.A., Bywaters, S.M., Guan, J., Ashley, R.E., Yoder, J.D., orientation, absolute hand, and contrast loss in single-particle electron cryo-
Makhov, A.M., Conway, J.F., Christensen, N.D., et al. (2014). A CryoEM study microscopy. J. Mol. Biol. 333, 721745.
identifies the complete H16.V5 epitope and reveals global conformational Sapp, M., Fligge, C., Petzak, I., Harris, J.R., and Streeck, R.E. (1998).
changes initiated by binding of the neutralizing antibody fragment. J. Virol. Papillomavirus assembly requires trimerization of the major capsid protein
89, 14281438. by disulfides between two highly conserved cysteines. J. Virol. 72, 61866189.
Levy, H.C., Bowman, V.D., Govindasamy, L., McKenna, R., Nash, K.,
Scheres, S.H.W. (2012). RELION: implementation of a Bayesian approach to
Warrington, K., Chen, W., Muzyczka, N., Yan, X., Baker, T.S., et al. (2009).
cryo-EM structure determination. J. Struct. Biol. 180, 519530.
Heparin binding induces conformational changes in Adeno-associated virus
serotype 2. J. Struct. Biol. 165, 146156. Scheres, S.H.W., and Chen, S. (2012). Prevention of overfitting in cryo-EM
structure determination. Nat. Methods 9, 853854.
Li, X., Mooney, P., Zheng, S., Booth, C.R., Braunfeld, M.B., Gubbens, S.,
Agard, D.A., and Cheng, Y. (2013). Electron counting and beam-induced mo- Schowalter, R.M., Pastrana, D.V., and Buck, C.B. (2011). Glycosaminoglycans
tion correction enable near-atomic-resolution single-particle cryo-EM. Nat. and sialylated glycans sequentially facilitate Merkel cell polyomavirus infec-
Methods 10, 584590. tious entry. PLoS Pathog. 7, e1002161.
Lowe, J., Panda, D., Rose, S., Jensen, T., Hughes, W.A., Tso, F.Y., and Selinka, H.-C., Giroglou, T., Nowak, T., Christensen, N.D., and Sapp, M.
Angeletti, P.C. (2008). Evolutionary and structural analyses of alpha-papillo- (2003). Further evidence that papillomavirus capsids exist in two distinct
mavirus capsid proteins yields novel insights into L2 structure and interaction conformations. J. Virol. 77, 1296112967.
with L1. Virol. J. 5, 150. Selinka, H.-C., Florin, L., Patel, H.D., Freitag, K., Schmidtke, M., Makarov, V.A.,
Malik, H., Khan, F.H., and Ahsan, H. (2014). Human papillomavirus: current and Sapp, M. (2007). Inhibition of transfer to secondary receptors by heparan
status and issues of vaccination. Arch. Virol. 159, 199205. sulfate-binding drug or antibody induces noninfectious uptake of human papil-
McKee, S.J., Bergot, A.-S., and Leggatt, G.R. (2015). Recent progress in lomavirus. J. Virol. 81, 1097010980.
vaccination against human papillomavirus-mediated cervical cancer: HPV Shafti-Keramat, S., Handisurya, A., Kriehuber, E., Meneguzzi, G.,
vaccination. Rev. Med. Virol. 25, 5471. Slupetzky, K., and Kirnbauer, R. (2003). Different heparan sulfate proteogly-
Mejia, A.F., Culp, T.D., Cladel, N.M., Balogh, K.K., Buck, C.B., and cans serve as cellular receptors for human papillomaviruses. J. Virol. 77,
Christensen, N.D. (2006). Preclinical model to test human papillomavirus virus 1312513135.

262 Structure 25, 253263, February 7, 2017


Tang, G., Peng, L., Baldwin, P.R., Mann, D.S., Jiang, W., Rees, I., and Ludtke, Wriggers, W., Milligan, R.A., and McCammon, J.A. (1999). Situs: a package for
S.J. (2007). EMAN2: an extensible image processing suite for electron micro- docking crystal structures into low-resolution maps from electron microscopy.
scopy. J. Struct. Biol. 157, 3846. J. Struct. Biol. 125, 185195.
Toh, Z.Q., Licciardi, P.V., Fong, J., Garland, S.M., Tabrizi, S.N., Russell, F.M., Xia, L., Xian, Y., Wang, D., Chen, Y., Huang, X., Bi, X., Yu, H., Fu, Z., Liu, X., Li,
and Mulholland, E.K. (2015). Reduced dose human papillomavirus vaccina- S., et al. (2016). A human monoclonal antibody against HPV16 recognizes an
tion: an update of the current state-of-the-art. Vaccine 33, 50425050. immunodominant and neutralizing epitope partially overlapping with that of
Trus, B.L., Roden, R.B., Greenstone, H.L., Vrhel, M., Schiller, J.T., and Booy, H16.V5. Sci. Rep. 6, 19042.
F.P. (1997). Novel structural features of bovine papillomavirus capsid revealed Yan, X., Sinkovits, R.S., and Baker, T.S. (2007). AUTO3DEMan automated
by a three-dimensional reconstruction to 9 A resolution. Nat. Struct. Biol. 4, and high throughput program for image reconstruction of icosahedral parti-
413420. cles. J. Struct. Biol. 157, 7382.
Wolf, M., Garcea, R.L., Grigorieff, N., and Harrison, S.C. (2010). Subunit inter- Zhao, Q., Potter, C.S., Carragher, B., Lander, G., Sworen, J., Towne, V.,
actions in bovine papillomavirus. Proc. Natl. Acad. Sci. USA 107, 62986303. Abraham, D., Duncan, P., Washabaugh, M.W., and Sitrin, R.D. (2014).
Wriggers, W. (2010). Using Situs for the integration of multi-resolution struc- Characterization of virus-like particles in GARDASIL by cryo transmission
tures. Biophys. Rev. 2 (1), 2127. electron microscopy. Hum. Vaccin. Immunother. 10, 734739.

Structure 25, 253263, February 7, 2017 263

Вам также может понравиться