12
Article Cryoelectron Microscopy Maps of Human Papillomavirus 16 Reveal L2 Densities and Heparin Binding Site Highlights d 4.3-A ˚ resolution 3D reconstructions based on gold- standard FSC d Classification of HPV overcomes heterogeneity that limits the resolution d L1 protein was built and used to reveal L2 minor protein d Heparin binding elicits conformational changes to L2 Authors Jian Guan, Stephanie M. Bywaters, Sarah A. Brendle, ..., James F. Conway, Neil D. Christensen, Susan Hafenstein Correspondence [email protected] 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, 253–263 February 7, 2017 ª 2016 Published by Elsevier Ltd. http://dx.doi.org/10.1016/j.str.2016.12.001

Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

Article

Cryoelectron Microscopy

Maps of HumanPapillomavirus 16 Reveal L2 Densities and HeparinBinding Site

Highlights

d 4.3-A resolution 3D reconstructions based on gold-

standard FSC

d Classification of HPV overcomes heterogeneity that limits the

resolution

d L1 protein was built and used to reveal L2 minor protein

d Heparin binding elicits conformational changes to L2

Guan et al., 2017, Structure 25, 253–263February 7, 2017 ª 2016 Published by Elsevier Ltd.http://dx.doi.org/10.1016/j.str.2016.12.001

Authors

Jian Guan, Stephanie M. Bywaters,

Sarah A. Brendle, ..., James F. Conway,

Neil D. Christensen, Susan Hafenstein

[email protected]

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

Page 2: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

Structure

Article

Cryoelectron Microscopy Maps of HumanPapillomavirus 16 Reveal L2 Densitiesand Heparin Binding SiteJian 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, USA2Department of Pathology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA3Department of Structural Biology, University of Pittsburgh School of Medicine, 3501 5th Avenue, Pittsburgh, PA 15260, USA4Lead Contact

*Correspondence: [email protected]://dx.doi.org/10.1016/j.str.2016.12.001

SUMMARY

Human papillomavirus (HPV) is a significant healthburden and leading cause of virus-induced cancers.The current commercial vaccines are genotype spe-cific and provide little therapeutic benefit to patientswith existing HPV infections. Host entry mechanismsrepresent an excellent target for alternative thera-peutics, but HPV receptor use, the details of cellattachment, and host entry are inadequately under-stood. Here we present near-atomic resolution struc-tures of the HPV16 capsid and HPV16 in complexwith heparin, both determined from cryoelectronmicrographs collected with direct electron detectiontechnology. The structures clarify details of capsidarchitecture for the first time, including variation inL1 major capsid protein conformation and putativelocation of L2 minor protein. Heparin binds spe-cifically around the capsid icosahedral vertices andmay recapitulate the earliest stage of infection,providing a framework for continuing biochemical,genetic, and biophysical studies.

INTRODUCTION

Human papillomavirus 16 (HPV16) is the most prevalent onco-

genic genotype in HPV-associated anogenital and oral cancers.

Having co-evolved with the human host for more than a million

years, HPV develops chronic infections by activating the cell

cycle as epithelial cells differentiate in order to create a replica-

tion competent environment that allows viral genome expression

and packaging into infectious particles (Doorbar et al., 2015).

Since the first reported association with cancer, extensive HPV

research has focused on the prevention of cervical cancer,

including vaccine development to prevent virus infection (Malik

et al., 2014; Nicol et al., 2015; Toh et al., 2015). However, the

current commercial vaccines are genotype specific and provide

little therapeutic benefit to patients with existing HPV infections

Structure 25

(Dickson et al., 2015; Gilmer, 2015; McKee et al., 2015). Target-

ing host entry mechanisms may be a successful alternative

approach because virus entry is a key step of HPV infection

regardless of genotype. However, receptor use for HPV, the de-

tails of cell attachment, and capsid conformational changes

required for host entry are poorly understood.

HPV is epitheliotropic and its replication is tightly associated

with the terminal differentiation of basal cells into keratinocytes.

This restricted tropism makes production and purification of

high-titer virus preparations for research problematic. Therefore,

alternative HPV production methods have been developed

within the HPV community for molecular biology and structural

studies. Virus-like particles can be composed of only the major

structural protein (L1) or of the major and minor capsid proteins

(L1/L2) and are not infectious since they are devoid of viral

genome (Hernandez et al., 2012; Kirnbauer et al., 1992, 1993).

Pseudovirus is composed of both structural proteins, which

are expressed along with a plasmid so that a copy of the plasmid

DNA can be packaged as a mock genome (Buck et al., 2005a,

2005b). Quasiviruses assemble a structurally complete capsid

and are infectious since they package a copy of the cottontail

rabbit papillomavirus genome (CRPV) (Christensen, 2005). All

of these types of HPV capsids preserve the main attributes of

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

have been used for the HPV16 work presented here.

Structures of sub-viral particles have been solved by X-ray

crystallography, including the HPV16 L1 pentamer and a T = 1

particle (Chen et al., 2000; Dasgupta et al., 2011); however, the

complete HPV T = 7d capsid has only been visualized by cryoe-

lectron microscopy (cryo-EM) reconstructions (Baker et al.,

1991; Buck et al., 2008; Cardone et al., 2014; Guan et al.,

2015a; Lee et al., 2014). Currently, the highest-resolution struc-

ture of the intact T = 7d HPV16 capsid is a 9-A resolution cryo-

EM structure, fromwhich a pseudoatomic model was developed

using the major structural protein, L1 (Cardone et al., 2014). The

55–60 nm diameter HPV capsid is composed of 360 copies of

L1 arranged into 72 capsomers, each composed of five copies

of L1. Twelve capsomers lay on the icosahedral 5-fold vertices

and are referred to as pentavalent capsomers, whereas the

, 253–263, February 7, 2017 ª 2016 Published by Elsevier Ltd. 253

Page 3: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

remaining 60 capsomers are each surrounded by six other

capsomers and referred to as hexavalent capsomers. An exten-

sion of the L1 C terminus, or C-terminal arm, links capsomers

together and provides interactions leading to the formation of

disulfide bonds between Cys175 and Cys428 from two neigh-

boring L1 proteins (Buck et al., 2005a, 2005b; Cardone et al.,

2014; Sapp et al., 1998). Although the invading arm structure

of bovine papilloma structure is known (Wolf et al., 2010), the

stabilizing C-terminal arm structure has not been solved yet

for HPV.

Besides the L1 major capsid protein, there is an uncertain

number of L2 minor structural proteins that co-assemble with

L1 to form infectious virus capsids. The L2 minor protein has a

number of functions including encapsidation of DNA, involve-

ment in the conformational changes of the capsid during cell

entry, disruption of the endosomal membrane, and subcellular

trafficking of the incoming viral genome (Bronnimann et al.,

2013; Buck et al., 2008; Raff et al., 2013). Specifically, the N-ter-

minal residues of L2 (1–88) are exposed on the surface of the

capsid although the anti-L2 antibody, RG-1 recognition of L2

(17–36) is dependent on maturation status or cell attachment

(Day et al., 2008a, 2008b; Finnen et al., 2003; Lowe et al.,

2008; Okun et al., 2001). An N-terminal L2 motif (RXXR, furin

cleavage site) is conserved across HPV genotypes and thus

has promise as a new target for vaccine development (Bronni-

mann et al., 2013; Conway et al., 2011; Roden et al., 2001). A

previous structural study using pseudovirus maps (23 and

25 A) interpreted small densities beneath each capsomer as

belonging to L2 (Buck et al., 2008). Based on analysis of the

late gene products, L2 protein has been predicted to bind within

the center of the L1 capsomer with the antigenic region facing

outward (Lowe et al., 2008). Stoichiometric studies have indi-

cated that there are about 12–36 L2 molecules per HPV capsid

(Okun et al., 2001); however, other works have suggested the

amount of L2 per capsid is variable, and may include up to 72

L2 molecules per capsid, perhaps one L2 at each capsomer

(Buck et al., 2008; Doorbar and Gallimore, 1987; Trus et al.,

1997). Naturally produced virions average approximately 24–36

L2 molecules per capsid (Bronnimann et al., 2013; Buck et al.,

2008; Campos andOzbun, 2009). Despite these variable reports,

there is general acceptance that between 12 and 72 L2 proteins

per capsid are incorporated, each into a central internal cavity of

the capsomer via L2 C-terminal interactions.

During host entry and endocytosis, HPV conformational

changes are triggered by binding to a cell receptor (Abban and

Meneses, 2010; Kines et al., 2009; Selinka et al., 2003). Cell

surface heparan sulfates (HSs) serve as a primary attachment

receptor (Buck et al., 2006; Rommel et al., 2005; Schowalter

et al., 2011; Shafti-Keramat et al., 2003) and it is not the protein

backbone but the glycan moieties, specifically the sulfation

pattern, that are required for virus infection (Cerqueira et al.,

2013). HS proteoglycans are expressed and secreted by nearly

all mammalian cells, and are located on both the cell surface

and extracellular matrix (Bernfield et al., 1992; Buck et al.,

2006; Schowalter et al., 2011; Selinka et al., 2003). However,

because HS polysaccharides are expensive and difficult to

obtain, heparin has been used in lieu of HS in most biological

and structure studies (Cerqueira et al., 2013; Selinka et al.,

2007). Both heparin and HS binding have been shown to induce

254 Structure 25, 253–263, February 7, 2017

conformational changes of virus during entry (Florin et al., 2012;

Knappe et al., 2007; Levy et al., 2009). Co-crystallization of a

truncated L1 capsomer together with heparin oligosaccharides

(8 and 10 monosaccharide units) suggested four distinct heparin

binding sites that mapped to the L1 protein (Dasgupta et al.,

2011). Three of these HS binding sites were verified by subse-

quent mutational analysis (Dasgupta et al., 2011; Richards

et al., 2013). Nonetheless, the molecular mechanisms regarding

the binding of heparin during entry remain fragmented and not

completely understood.

With recent technological advances in cryo-EM image detec-

tion and the introduction of new software classification schemes,

we can now investigate HPV at near-atomic resolution. Here, we

present the near-atomic resolution structures of HPV16 alone

andHPV16 interactingwith heparinmolecules at 4-A resolutions.

Three-dimensional (3D) classification of the data revealed that

HPV capsids are heterogeneous in nature but contain sub-pop-

ulations of capsids with consistent diameters. The classification

of particles improved the high-resolution maps, which facilitated

building de novo the L1 protein (residues 3–485), revealing the

structure of the C-terminal arm. Solving the L1 asymmetric unit

allowed us to calculate an L1-only capsid that was used to iden-

tify the location of L2 densities in our maps. Binding of heparin

induced no conformational changes in the capsid that include

local changes in L1, but there are alterations to putative L2 den-

sities. Besides the identification of the heparin binding site in the

context of the intact capsid, the changes induced by heparin

binding provide the first glimpse of an HPV entry intermediate.

We propose an entry mechanism model that may include isola-

tion of specific 5-fold capsomers due to potential interaction

between heparin and L2 protein.

RESULTS

Near-Atomic Resolution Maps Were Reconstructed bySelecting Homogeneous Sub-populationsHPV16 capsids and capsids incubated with heparin in excess

were vitrified for cryo-EM data collection using new direct elec-

tron detector technology. The drift-corrected micrographs of

HPV and HPV-heparin displayed bumpy spherical shapes

with evident capsomer protrusions (Figures 1A and 1B). Cap-

sids contained internal density corresponding to packaged

DNA and appeared to have diameters of about 60 nm. Howev-

er, 3D classification using REgularized LIkelihood OptimizatioN

(RELION) showed that the populations of HPV and HPV-hepa-

rin capsids were heterogeneous in size (Figures S1 and S2).

Both capsid types included a large group of approximately

50% of the total number of particles that had the same diam-

eter of 569–570 A. Final reconstructions using AUTO3DEM

with the gold-standard approach used 50% of the HPV parti-

cles and 50% of the HPV-heparin complex particles to recon-

struct a homogeneous population in each case to near-atomic

resolution (Figures 1C and 1D). The 4.3-A resolution of the

maps was estimated where the Fourier shell correlation (FSC)

dropped below 0.143 (Figure 2) (Table 1). Central sections

through the density maps illustrate the modest quality of the re-

constructions (Figures 1E and 1F). At this resolution, we can

see strand separation, the secondary structures of a helices

and b strands (Figures 3AI, 3AII, 3BI, and 3BII), and side chains

Page 4: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

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).

Figure 1. Cryo-EM Reconstruction of HPV16 and HPV16-Heparin

(A and B) Representative regions of cryo-EM micrographs of the HPV16

particle capsids (A) and HPV16 capsids complexed with heparin (B).

(C and D) The surface rendered 3D maps of HPV16 (C) and capsid-heparin

complex (D) were radially colored according to the distance from the center

(color key in angstroms) of the capsid and surface rendered at 1s. The heparin

difference density (black, difference map rendered at 4.2 s) was found at

positions surrounding only the 5-fold vertices of the capsid.

(E and F) The central sections through the cryo-EM density maps show the

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

appearing at the 12 o’clock position.

(G) The zoom-in view of the HPV16-heparin complex shows the detailed fea-

tures of bound heparin molecules. The 5-fold symmetric axis was represented

by black pentagon.

of some residues can be identified (Figures 3A1–3A3 and

3B1–3B3).

Building L1 into the Cryo-EM Density Solved theStructure of the C-Terminal Invading Arm and Revealedthe Location of L2Strong 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 L1molecules 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

outside of the capsomer to the shoulder, ending at the outside

center of the capsomer (Figure 4B). Corresponding densities in

the pentavalent capsomer were significantly smaller than the

densities in hexavalent capsomers, both at the surface and the

internal part at the base.

Distinct Structural DifferencesWere Observed betweenPentavalent and Hexavalent CapsomersOur near-atomic structure of the HPV16 capsid supplied more

detailed information on the capsid surface topology than the

maps we produced before (Guan et al., 2015a, 2015b; Lee

et al., 2014). Besides the entire C-terminal arm, surface loops

could be distinguished separately, especially the BC and EF

Structure 25, 253–263, February 7, 2017 255

Page 5: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

Table 1. Cryo-EM Image Reconstruction Data

HPV HPV-Heparin

No. of micrographs 9,137 6,175

Defocus level range (mm) 1.04–5.19 0.78–4.85

No. of particles selected from

micrograph

115,112 102,829

No. of particles selected for

reconstruction

57,556 51,422

Final resolution 4.3 4.3

loops that were seen as merged densities in the previous lower-

resolution maps (Figure 4A). To further visualize the differences

between L1 conformations in the two different environments,

the newly built structures of the pentavalent and hexavalent

capsomers were superimposed (Figure 5A). The conformational

differences were obvious on both surface loops and C-ter-

minal arms, with a root-mean-square deviation (RMSD) value

of 4.77 A between the entire sets of L1 atoms of the two cap-

somers. Since the C-terminal arm conformations were notice-

ably different, the RMSD was recalculated with this part of the

structure truncated, and found to be 1.49 A, indicating sig-

nificantly different loop structures throughout. Specifically the

hexavalent C-terminal arm extended further away from the

hexavalent capsomer, whereas the pentavalent arms were

arranged closer in a curved arc at the site of interaction with

the neighboring hexavalent capsomer. The individual BC, DE,

FG, HI, and EF loops are in decidedly different conformations

throughout; however, overall the hexavalent capsomers formed

amore open crown-like structure comparedwith the pentavalent

capsomers.

Differences between the pentavalent and hexavalent cap-

somers were also apparent in the local resolution map (Figures

5B and 5C). The pentavalent capsomer surface had lower local

resolution compared with the hexavalent capsomer, especially

near the center, suggesting the pentavalent capsomer loops

had more flexibility. Overall the very center and the outer edges

of the capsomers had lower resolution than the mid-regions of

the capsomers. Specifically, the BC and EF loops, which were

Figure 3. Secondary Structure and Residue Side Chain of the L1 Prote

Secondary structure of a helix and b sheet (blue) in the L1 protein was shown

HPV-heparin complex (BI) and (BII). The side chain of the residues were also iden

HPV16 map; and (B1) 311.Tyr, (B2) 447.Trp, (B3) 467.Lys in the HPV16-heparin m

256 Structure 25, 253–263, February 7, 2017

located at the tip of the star-shaped capsomer, had the lowest

resolution (6.5 A) in both pentavalent and hexavalent capsomers.

The HPV-Heparin Complex Showed that HeparinReceptors Only Bound at the 5-fold Vertex of theHPV16 VirusSignificant extra densities were found at each 5-fold vertex of the

HPV-heparin complex map compared with the HPV map. This

putative heparin density was comparable with the magnitude

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,

there was noisy discontinuous density stretching out and away

from the bound heparin (Figure 1E). The mixture of polysaccha-

ride chain lengths (10–30) in the heparin incubated with the virus

in our study likely caused the lesser densities extending away

from the bound heparin, which corresponded to longer polysac-

charide chains of bound heparin that were averaged with shorter

chains during the reconstruction process.

Heparin Binding Triggers Conformational Changes of L2Protein without Altering the Capsid DiameterNo significant difference in diameter between HPV16 and

HPV16-heparin complex maps was seen in the radial density

plots (Figure 6A), indicating that heparin binding did not trigger

a change in the overall size of the capsid. The structure of L1

was built into the HPV-heparin complex map to make an asym-

metric unit and an L1-only capsid map was calculated as

described above for the capsid. The mass axis of L1 proteins

in pentavalent and hexavalent capsomers in the HPV and the

HPV-heparin maps were calculated (Figure 6B) to track both

rotational and translational movements of L1 proteins. L1 pro-

teins rotated no more than 0.4� and shifted less than 0.1 A (Fig-

ure S3). Thus, no significant movement of L1 protein was found

after binding with heparin.

To check the surface loop rearrangement of the L1 pro-

tein, the HPV and HPV-heparin asymmetric unit structures

were superimposed. An RMSD of 0.76 A suggested slight

changes may have occurred after heparin binding. Super-

imposing the HPV hexavalent and pentavalent capsomers on

in Were Identified in the Maps

in the corresponding density in the map (green) of HPV16 (AI) and (AII) and

tified in each of maps. For example, (A1) 311.Tyr, (A2) 447.Trp, (A3) 467.Lys in

ap.

Page 6: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

Figure 4. The Molecule Building of L1 Protein and Putative L2

Densities in the Map

(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

densities are continuous and clearly identified in the map.

(B) Difference density calculated from subtracting the L1-only capsid map

from the HPV16 experimental map (gray) showed difference densities (red) on

top view to show exposed densities on the surfaces of both pentavalent and

hexavalent capsomers, with wormlike shapes extending between capsid floor

and center of the capsomer.

(C) Cross-section view to show the L2 densities inside the capsomers.

the corresponding HPV-heparin capsomers resulted in RMSD

values of 0.51 and 1.12 A, respectively (Figure 6C). There was

no significant loop rearrangement among all HI, EF, DE, FG,

and BC loops (Figure 6C), and no significant change in the diam-

eter of the capsid.

By subtracting the calculated HPV-heparin L1-only map from

HPV-heparin experimental map, we found a difference density

that had significant conformational changes compared with the

difference densities from the HPV map (Figures 4B, 4C, 6D, 6E,

and S4). After binding with heparin, difference densities in the

HPV-heparincomplexmap includedputativeL2density thatmap-

ped tobothpentavalent andhexavalent 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 andPentavalent CapsomersThe 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,

oneEF loop, andoneC-terminal arm from twoneighboringL1pro-

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, and59.Lys from theBC loop, andC-terminal arm residues

of 428.Cys, 429.Gln, 431.His, 432.Thr, and 433.Pro (Figure 7B).

DISCUSSION

Variations in the size of the HPV capsid have been attributed pre-

viously to progressive stages of maturation of the capsid through

formation of disulfide linkages conferred by the C-terminal arm

using the pseudovirus model. Similarly, our 3D classification of

the quasi-HPV capsids used in our study (Experimental Proced-

ures) showed different-diameter capsids existing with similar

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

may be linked to survival outside of the host in a hostile environ-

ment and may represent an evolving adaptability. For instance,

the range in diameter may be linked to stability, providing classes

of more (or less) stable particles. Once assembled into capsids,

the heterogeneous characteristic of HPV during and after the

maturation process may provide an advantage ensuring the sur-

vival of an existing sub-population, within the mixed population

of capsids.

Structure 25, 253–263, February 7, 2017 257

Page 7: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

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.

Compared with the well-understood nature and structure

of the L1 major protein, the location and number of L2 minor

proteins per capsid have remained controversial, despite the

essential requirement for genome encapsidation and subcellular

trafficking. During the past 25 years, extensive biochemical and

structural studies have attempted to lift the veil shrouding L2

protein incorporation. Using the L1-only calculated map, we

identified non-L1 density in our experimental map and suggest

that difference density can be attributed to L2. L2 protein was

observed to intertwine with L1 protein at the base part of the

capsomer and extend along the side wall of the capsomer in

the canyon to the top surface until reaching the upper center

of the capsomer. This observation was consistent with a previ-

ous structural result that showed small L2 protein densities at

the lower center of each capsomer (Buck et al., 2008). With the

higher resolution that is now attainable, our map reveals more

about the location and features of the L2 protein. Because of

the quasi-equivalent 5-fold symmetry characteristics of the hex-

avalent capsomer, the features of putative L2 densities were

different between pentavalent and hexavalent capsomers. The

smaller potential density of L2 at the surface and inner parts of

pentavalent capsomer suggested a flexible interaction of L1

and L2 proteins in the pentavalent capsomer, which were aver-

aged out during the reconstruction process. Consistent with a

previous model, L1 residues 261–269 map near the L2 density

that drapes over the outside of the capsomers, suggesting

possible interactions (Lowe et al., 2008). The variation of L2 dis-

tribution among capsomers suggested random L2 incorporation

during capsid formation, which may be related to the long virion

maturation time and the high heterogeneity of the population.

Among the 72 capsomers comprising the intact icosahe-

dral capsid, L2 molecules may be incorporated asymmetrically

and randomly distributed in both pentavalent and hexavalent

capsomers. Alternatively, L2 proteins may be located on each

of the 12 pentavalent capsomers with the other additional L2

copies randomly distributed in hexavalent positions. Finally,

258 Structure 25, 253–263, February 7, 2017

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.

An anisotropic distribution of L2 protein in the capsid implies

that a unique vertex may exist, which may function during host

cell binding and infection. However, due to the magnitude of

these difference densities they cannot be definitively appor-

tioned or assigned to L2 molecules.

The L2N terminus has been shown previously to be accessible

at the surface of the capsomer (Conway et al., 2011; Lowe et al.,

2008), and such a location is suggested in our map (Figure 3B).

The incorporated L2 protein may start from the middle center

of the capsomer, extending to the surface along the side wall

to reach the capsid floor with most of the molecule including

the L2 C terminus intertwining with the L1 protein and interacting

with the genome at the base of the capsomer. At the current res-

olution we cannot solve the molecular structure of L2 peptide

within the density (Figure 4) or define the orientation. However,

the morphological features and conformational changes of the

putative L2 protein densities might be used to guide future

studies to contribute to understanding the mechanisms of entry

and infection.

Variations in the micro-environment of the T = 7d icosahedral

capsid (Guan et al., 2015a, 2015b; Lee et al., 2014; Xia et al.,

2016; Zhao et al., 2014) underlie functional differences between

pentavalent and hexavalent capsomers. Specifically, the U4

antibody binds preferentially at the icosahedral 5-fold (Guan

et al., 2015a), which is consistent with the previous studieswhere

U4 antibody prohibited the virus from binding cells (Day et al.,

2007; Deschuyteneer et al., 2010). Considering the functional dif-

ferences between pentavalent and hexavalent L1 environments,

preferential heparin binding, and the established importance of

L2 function during entry, we conclude that the 5-fold vertex

can be considered the functional site for binding host cells during

entry. This model suggests L2 incorporation must include the

vertex pentamers.

Previous work has shown that HS binding triggered conforma-

tional changes to the capsid, which resulted in reduced affinity to

the primary HS attachment receptor and more exposure of

Page 8: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

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.

L2 protein for furin cleavage (Day et al., 2008a, 2008b; Selinka

et al., 2003). The HPV-heparin complex captured in our study

showed no significant conformational changes of the L1 protein,

but suggested major L2 alteration. Post-attachment alterations

to the HPV capsid have been reported previously (Guan et al.,

2015a) and may correspond to the heparin-triggered conforma-

tional changes we report here that include small L1 protein

movements at the apical surface of the capsomers. The surface

loops on hexavalent capsomer had no significant changes

after binding heparin (Figure 6C), which coincides with the re-

tained ability of antibody HPV16.V5 to neutralize well after cell

attachment (Guan et al., 2015a; Lee et al., 2014). Compared

with HPV16.V5, the redisposition and more exposure of L2 den-

sities on the surface may affect other antibody binding, for

example, H33.J3, which displayed improved neutralizing

efficiency to cell-bound virions (Selinka et al., 2003). The lack

of global conformational changes of L1 protein after binding

heparin and the presence of possible L2 density near the

bound heparin molecule suggested that the L2 protein was likely

participating in the binding with heparin (Figures 6D and 6E). If

heparin binds to both L1 and L2 proteins, perhaps the L1 interac-

tions facilitate the orientation and the L2 binding with heparin

provides the contacts that triggered major conformational

St

changes. Themovement 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;

however, further work remains to reveal the L2 structure and

subsequent changes.

Of the three heparin binding sites identified previously, the site

occupied by heparin in our density map overlapped with two res-

idues of site 3 (57.Asn, 59.Lys, 442.Lys, 443.Lys), whereas no

traces of bound heparin were found at the top of the capsomers

involving the FG and HI loops previously identified as sites 1 and

2 (Richards et al., 2013), and as the primary binding sites. How-

ever, considerable differences exist among these three experi-

ments including the form of HPV capsid (L1 only pentamers,

pseudovirus, or quasivirus) or the type of heparin (heparin oligo-

saccharide 8–10, heparin oligomers, or heparin 10–30) used. The

seemingly disparate results suggest several possibilities. First,

due to the variations in the reagents, the heparin used in these

experiments was only capable of interacting at site 3 on our

quasivirus capsids due to steric limitations of binding. Certainly,

it seems likely from our structure that heparin binding to site 3

would interfere with binding at site 2. Second, a stepwise

sequence of events is conceivable. For example, heparin bound

HPV capsids at site 1 during the 1-hr incubation; the bound hep-

arin initiated the conformational changes revealed in the maps;

whereupon heparin dissociated from site 1 and then bound

site 3 as revealed in the HPV-heparin complex structure.

ructure 25, 253–263, February 7, 2017 259

Page 9: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

Figure 7. Epitope Analysis of Heparin Based on the Model and the

4.7-A Maps

The heparin binding site described by the model. (A) The fitted heparin

molecule (purple, yellow, and red) and L1 protein (blue) were used to locate the

contacts between heparin and L1. (B) The residues on the L1 protein are

indicated at their position with sequence number, residue name, and side

chain shown in red. The residues comprising the binding site identified here are

listed in the table along with their position on the L1 capsomer.

Until now, no structural tools to detect HPV conformational

changes during entry have been described. By using near-

atomic reconstruction maps, local conformational changes

were seen for the first time. Since heparin molecules were only

found at the pentavalent capsomer, triggering the conforma-

tional changes to expose more L2 densities in the canyon, these

changes suggested that HPV infection is initiated through a

pentavalent capsomer as a unique 5-fold vertex that may func-

tion during later stages of entry that include release of the DNA

genome tethered to L2. In support of this model is the possibility

that L2 might be incorporated into the HPV capsid to form a

unique vertex. Further studies will be needed to resolve these

questions.

EXPERIMENTAL PROCEDURES

Preparation of HPV16 Quasiviruses

HPV16 quasivirus containing L1 and L2 proteins and encapsidating a CRPV

genome having the SV40 origin of replication was prepared as described

previously (Brendle et al., 2010; Mejia et al., 2006; Pyeon et al., 2005). In

brief, HPV16 sheLL plasmid (kindly provided by John Schiller, NIH) was trans-

260 Structure 25, 253–263, February 7, 2017

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�Cand concentrated to 0.8mg/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 1.04–5.19

and 0.78–4.85 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

the process by splitting the dataset into two independently refined halves

(Tang et al., 2007; Yan et al., 2007). The refinements for sharpening high-

resolution information were performed using EMBFACTOR (Fernandez

et al., 2008; Rosenthal and Henderson, 2003). Reported resolutions are esti-

mated according to the gold-standard FSC = 0.143 criterion (Scheres and

Chen, 2012).

For heterogeneity investigation, 3D classifications and refinements were

performed using RELION software (Scheres, 2012). A windowing operation

was applied to the particles in Fourier space to downsize the particle images

to yield a final box size of 260 pixels2 (corresponding to a pixel size of 2.3 A).

The final pixel size was chosen after optimization of the balance between

the Nyquist frequency limit and the memory requirements for computations.

Model Building

The atomic model of the HPV16 L1 capsomer with the C-terminal arm trun-

cated (PDB: 3OAE; residues 20–403, 438–474) was fitted as a rigid body

into the cryo-EM map using UCSF Chimera (Dasgupta et al., 2011; Pettersen

et al., 2004). Placement of the pseudoatomic model of an asymmetric unit of

HPV16 capsid (PDB: 3J6R; residues 9–486) was used as a guideline during

building (Cardone et al., 2014). Model building proceeded de novo to place

the missing C-terminal arm residues 404–437 and the N and C termini through

COOT (Emsley et al., 2010). Specifically, 17 residues were built into density at

the N-terminal end and 11 residues at the C-terminal end. Each L1 molecule in

the asymmetric unit was built independently using COOT (Emsley et al., 2010).

Icosahedral symmetry was applied to the refined asymmetric unit model to

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,

kernel width �4.8 A, Gaussian smoothing kernel, and scaling factor of 1.

The capsid atomic model was then further refined iteratively using the Phenix

Real Space Refinement program (Afonine et al., 2012). At each iteration, the

best model was visually inspected in COOT and adjusted manually accord-

ingly to the best fit into the density and guided by torsion restraints, planar

Page 10: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

peptide restraints, and Ramachandran restrains. The quality of the final model

was evaluated by MolProbity (http://molprobity.biochem.duke.edu/) (Chen

et al., 2010) (Table S1). The same building and refinement procedures were

performed for the HPV-heparin complex map. Difference maps were calcu-

lated using SITUS (Wriggers, 2010; Wriggers et al., 1999).

Coordinates for the disaccharide unit of heparin isolated from (PDB: 3OAE)

were used as the basis for the model building (Dasgupta et al., 2011). The hep-

arin polysaccharide model was built manually using disaccharide units fitted

one-by-one into the difference map made by subtracting HPV16 from the

HPV16-heparin complex map. The result of initial fitting was refined using

the HPV16-heprin complex map. The final result included ten disaccharide

units of heparin built in the density with a correlation coefficient of 83%accord-

ing to Chimera (Pettersen et al., 2004). Contacts between the fitted heparin and

L1 protein structures were identified using Chimera with the criteria for van der

Waals overlap distances set at �0.4 and 0.0 A. Clashes between atoms were

defined by any overlap of 0.6 A or more.

ACCESSION NUMBERS

Cryo-EM maps for the HPV16 and HPV16-heparin complexes have been

deposited in the EM database (www.emdatabank.org/) with accession

numbers EMD: 6620 and 6619. Coordinates for the atomic model of the

asymmetric unit of HPV16 L1 proteins and the complex with the heparin

molecule have been deposited in the PDB under accession codes PDB:

5KEP and 5KEQ.

SUPPLEMENTAL INFORMATION

Supplemental Information includes four figures and one table and can be

found with this article online at http://dx.doi.org/10.1016/j.str.2016.12.001.

AUTHOR CONTRIBUTIONS

J.G., S.M.B., and S.A.B. performed the research; R.E.A. performed micro-

scopy and collected the cryo-EM data; A.M.M. and J.F.C. collected the

cryo-EM data; J.G. processed and analyzed the cryo-EM data; J.G., N.D.C.,

J.F.C., and S.H. wrote the paper.

ACKNOWLEDGMENTS

This work was supported in part by the J. Gittlen Memorial Golf Tournament

and the Pennsylvania Department of Health CURE funds. Research reported

in this publication was also supported by the Office of the Director, NIH, under

award numbers S10OD019995 and S10OD011986. The content is solely the

responsibility of the authors and does not necessarily represent the official

views of the NIH.

Received: June 8, 2016

Revised: October 7, 2016

Accepted: December 12, 2016

Published: January 5, 2017

REFERENCES

Abban, C.Y., and Meneses, P.I. (2010). Usage of heparan sulfate, integrins,

and FAK in HPV16 infection. Virology 403, 1–16.

Afonine, P.V., Grosse-Kunstleve, R.W., Echols, N., Headd, J.J., Moriarty,

N.W., Mustyakimov, M., Terwilliger, T.C., Urzhumtsev, A., Zwart, P.H., and

Adams, P.D. (2012). Towards automated crystallographic structure refinement

with phenix.refine. Acta Crystallogr. D Biol. Crystallogr. 68, 352–367.

Baker, T.S., Newcomb, W.W., Olson, N.H., Cowsert, L.M., Olson, C., and

Brown, J.C. (1991). Structures of bovine and human papillomaviruses.

Analysis by cryoelectron microscopy and three-dimensional image recon-

struction. Biophys. J. 60, 1445–1456.

Bernfield, M., Kokenyesi, R., Kato, M., Hinkes,M.T., Spring, J., Gallo, R.L., and

Lose, E.J. (1992). Biology of the syndecans: a family of transmembrane hep-

aran sulfate proteoglycans. Annu. Rev. Cell Biol. 8, 365–393.

Brendle, S.A., Culp, T.D., Broutian, T.R., and Christensen, N.D. (2010). Binding

and neutralization characteristics of a panel of monoclonal antibodies to

human papillomavirus 58. J. Gen. Virol. 91, 1834–1839.

Bronnimann, M.P., Chapman, J.A., Park, C.K., and Campos, S.K. (2013). A

transmembrane domain and GxxxG motifs within L2 are essential for papillo-

mavirus infection. J. Virol. 87, 464–473.

Buck, C.B., Pastrana, D.V., Lowy, D.R., and Schiller, J.T. (2005a). Generation

of HPV pseudovirions using transfection and their use in neutralization assays.

Methods Mol. Med. 119, 445–462.

Buck, C.B., Thompson, C.D., Pang, Y.-Y.S., Lowy, D.R., and Schiller, J.T.

(2005b). Maturation of papillomavirus capsids. J. Virol. 79, 2839–2846.

Buck, C.B., Thompson, C.D., Roberts, J.N., M€uller, M., Lowy, D.R., and

Schiller, J.T. (2006). Carrageenan is a potent inhibitor of papillomavirus infec-

tion. PLoS Pathog. 2, e69.

Buck, C.B., Cheng, N., Thompson, C.D., Lowy, D.R., Steven, A.C., Schiller,

J.T., and Trus, B.L. (2008). Arrangement of L2within the papillomavirus capsid.

J. Virol. 82, 5190–5197.

Campos, S.K., and Ozbun, M.A. (2009). Two highly conserved cysteine resi-

dues in HPV16 L2 form an intramolecular disulfide bond and are critical for

infectivity in human keratinocytes. PLoS One 4, e4463.

Cardone, G., Moyer, A.L., Cheng, N., Thompson, C.D., Dvoretzky, I., Lowy,

D.R., Schiller, J.T., Steven, A.C., Buck, C.B., and Trus, B.L. (2014).

Maturation of the human papillomavirus 16 capsid. MBio 5, e01104–e01114.

Cerqueira, C., Liu, Y., K€uhling, L., Chai, W., Hafezi, W., van Kuppevelt, T.H.,

K€uhn, J.E., Feizi, T., and Schelhaas, M. (2013). Heparin increases the infectivity

of human papillomavirus type 16 independent of cell surface proteoglycans

and induces L1 epitope exposure: interactions of HPV-16 with GAGs. Cell

Microbiol. 15, 1818–1836.

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

papillomavirus 16. Mol. Cell 5, 557–567.

Chen, V.B., Arendall, W.B., Headd, J.J., Keedy, D.A., Immormino, R.M.,

Kapral, G.J., Murray, L.W., Richardson, J.S., and Richardson, D.C. (2010).

MolProbity: all-atom structure validation for macromolecular crystallography.

Acta Crystallogr. D Biol. Crystallogr. 66, 12–21.

Christensen, N.D. (2005). Cottontail rabbit papillomavirus (CRPV) model

system to test antiviral and immunotherapeutic strategies. Antivir. Chem.

Chemother. 16, 355–362.

Conway, M.J., Cruz, L., Alam, S., Christensen, N.D., and Meyers, C. (2011).

Cross-neutralization potential of native human papillomavirus N-terminal L2

epitopes Torbjorn Ramqvist (ed.). PLoS One 6, e16405.

Dasgupta, J., Bienkowska-Haba, M., Ortega, M.E., Patel, H.D., Bodevin, S.,

Spillmann, D., Bishop, B., Sapp, M., and Chen, X.S. (2011). Structural basis

of oligosaccharide receptor recognition by human papillomavirus. J. Biol.

Chem. 286, 2617–2624.

Day, P.M., Thompson, C.D., Buck, C.B., Pang, Y.-Y.S., Lowy, D.R., and

Schiller, J.T. (2007). Neutralization of human papillomavirus with monoclonal

antibodies reveals different mechanisms of inhibition. J. Virol. 81, 8784–8792.

Day, P.M., Gambhira, R., Roden, R.B.S., Lowy, D.R., and Schiller, J.T. (2008a).

Mechanisms of human papillomavirus type 16 neutralization by l2 cross-

neutralizing and l1 type-specific antibodies. J. Virol. 82, 4638–4646.

Day, P.M., Lowy, D.R., and Schiller, J.T. (2008b). Heparan sulfate-independent

cell binding and infection with furin-precleaved papillomavirus capsids.

J. Virol. 82, 12565–12568.

Deschuyteneer, M., Elouahabi, A., Plainchamp, D., Plisnier, M., et al. (2010).

Molecular and structural characterization of the L1 virus-like particles that

are used as vaccine antigens in CervarixTM, the AS04-adjuvanted HPV-16

and -18 cervical cancer vaccine. Hum. Vaccin. 6, 407–419.

Dickson, E.L., Vogel, R.I., Luo, X., and Downs, L.S. (2015). Recent trends in

type-specific HPV infection rates in the United States. Epidemiol. Infect.

143, 1042–1047.

Doorbar, J., and Gallimore, P.H. (1987). Identification of proteins encoded by

the L1 and L2 open reading frames of human papillomavirus 1a. J. Virol. 61,

2793–2799.

Structure 25, 253–263, February 7, 2017 261

Page 11: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

Doorbar, J., Egawa, N., Griffin, H., Kranjec, C., andMurakami, I. (2015). Human

papillomavirus molecular biology and disease association: human papilloma-

virus. Rev. Med. Virol. 25, 2–23.

Emsley, P., Lohkamp, B., Scott, W.G., and Cowtan, K. (2010). Features and

development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501.

Fernandez, J.J., Luque, D., Caston, J.R., and Carrascosa, J.L. (2008).

Sharpening high resolution information in single particle electron cryomicro-

scopy. J. Struct. Biol. 164, 170–175.

Finnen, R.L., Erickson, K.D., Chen, X.S., and Garcea, R.L. (2003). Interactions

between papillomavirus L1 and L2 capsid proteins. J. Virol. 77, 4818–4826.

Florin, L., Sapp, M., and Spoden, G.A. (2012). Host-cell factors involved in

papillomavirus entry. Med. Microbiol. Immunol. 201, 437–448.

Gilmer, L.S. (2015). Human papillomavirus vaccine update. Prim. Care

42, 17–32.

Guan, J., Bywaters, S.M., Brendle, S.A., Lee, H., Lee, H., Ashley, R.E.,

Makhov, A.M., Conway, J.F., Christensen, N.D., and Hafenstein, S. (2015a).

Structural comparison of four different antibodies interacting with human

papillomavirus 16 and mechanisms of neutralization. Virology 483, 253–263.

Guan, J., Bywaters, S.M., Brendle, S.A., Lee, H., Ashley, R.E., Christensen,

N.D., and Hafenstein, S. (2015b). The U4 antibody epitope on human papillo-

mavirus 16 identified by cryo-electronmicroscopy S. Perlman (ed.). J. Virol. 89,

12108–12117.

Hernandez, B.Y., Ton, T., Shvetsov, Y.B., Goodman, M.T., and Zhu, X. (2012).

Human papillomavirus (HPV) L1 and L1-L2 virus-like particle-based multiplex

assays for measurement of HPV virion antibodies. Clin. Vaccin. Immunol. 19,

1348–1352.

Kines, R.C., Thompson, C.D., Lowy, D.R., Schiller, J.T., and Day, P.M. (2009).

The initial steps leading to papillomavirus infection occur on the basement

membrane prior to cell surface binding. Proc. Natl. Acad. Sci. USA 106,

20458–20463.

Kirnbauer, R., Booy, F., Cheng, N., Lowy, D.R., and Schiller, J.T. (1992).

Papillomavirus L1 major capsid protein self-assembles into virus-like particles

that are highly immunogenic. Proc. Natl. Acad. Sci. USA 89, 12180–12184.

Kirnbauer, R., Taub, J., Greenstone, H., Roden, R., D€urst, M., Gissmann, L.,

Lowy, D.R., and Schiller, J.T. (1993). Efficient self-assembly of human papillo-

mavirus type 16 L1 and L1-L2 into virus-like particles. J. Virol. 67, 6929–6936.

Knappe, M., Bodevin, S., Selinka, H.-C., Spillmann, D., Streeck, R.E., Chen,

X.S., Lindahl, U., and Sapp, M. (2007). Surface-exposed amino acid residues

of HPV16 L1 protein mediating interaction with cell surface heparan sulfate.

J. Biol. Chem. 282, 27913–27922.

Lee, H., Brendle, S.A., Bywaters, S.M., Guan, J., Ashley, R.E., Yoder, J.D.,

Makhov, A.M., Conway, J.F., Christensen, N.D., et al. (2014). A CryoEM study

identifies the complete H16.V5 epitope and reveals global conformational

changes initiated by binding of the neutralizing antibody fragment. J. Virol.

89, 1428–1438.

Levy, H.C., Bowman, V.D., Govindasamy, L., McKenna, R., Nash, K.,

Warrington, K., Chen, W., Muzyczka, N., Yan, X., Baker, T.S., et al. (2009).

Heparin binding induces conformational changes in Adeno-associated virus

serotype 2. J. Struct. Biol. 165, 146–156.

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-

tion correction enable near-atomic-resolution single-particle cryo-EM. Nat.

Methods 10, 584–590.

Lowe, J., Panda, D., Rose, S., Jensen, T., Hughes, W.A., Tso, F.Y., and

Angeletti, P.C. (2008). Evolutionary and structural analyses of alpha-papillo-

mavirus capsid proteins yields novel insights into L2 structure and interaction

with L1. Virol. J. 5, 150.

Malik, H., Khan, F.H., and Ahsan, H. (2014). Human papillomavirus: current

status and issues of vaccination. Arch. Virol. 159, 199–205.

McKee, S.J., Bergot, A.-S., and Leggatt, G.R. (2015). Recent progress in

vaccination against human papillomavirus-mediated cervical cancer: HPV

vaccination. Rev. Med. Virol. 25, 54–71.

Mejia, A.F., Culp, T.D., Cladel, N.M., Balogh, K.K., Buck, C.B., and

Christensen, N.D. (2006). Preclinical model to test human papillomavirus virus

262 Structure 25, 253–263, February 7, 2017

(HPV) capsid vaccines in vivo using infectious HPV/cottontail rabbit papilloma-

virus chimeric papillomavirus particles. J. Virol. 80, 12393–12397.

Nicol, A.F., de Andrade, C.V., Russomano, F.B., Rodrigues, L.S.L., Oliveira,

N.S., Provance, D.W., and Nuovo, G.J. (2015). HPV vaccines: their pathol-

ogy-based discovery, benefits, and adverse effects. Ann. Diagn. Pathol. 19,

418–422.

Okun, M.M., Day, P.M., Greenstone, H.L., Booy, F.P., Castle, P.E., FitzGerald,

P.C., Kr€uger Kjaer, S., Lowy, D.R., and Schiller, J.T. (2001). L1 Interaction do-

mains of papillomavirus L2 necessary for viral genome encapsidation. J. Virol.

75, 4332–4342.

Pastrana, D.V., Buck, C.B., Pang, Y.-Y.S., Thompson, C.D., Castle, P.E.,

FitzGerald, P.C., Kr€uger Kjaer, S., Lowy, D.R., and Schiller, J.T. (2004).

Reactivity of human sera in a sensitive, high-throughput pseudovirus-based

papillomavirus neutralization assay for HPV16 and HPV18. Virology 321,

205–216.

Pettersen, E.F., Goddard, T.D., Huang, C.C., Couch, G.S., Greenblatt, D.M.,

Meng, E.C., and Ferrin, T.E. (2004). UCSF chimera: a visualization system

for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612.

Pyeon, D., Lambert, P.F., and Ahlquist, P. (2005). Production of infectious

human papillomavirus independently of viral replication and epithelial cell

differentiation. Proc. Natl. Acad. Sci. USA 102, 9311–9316.

Raff, A.B., Woodham, A.W., Raff, L.M., Skeate, J.G., Yan, L., Da Silva,

D.M., Schelhaas, M., and Kast, W.M. (2013). The evolving field of human

papillomavirus receptor research: a review of binding and entry. J. Virol.

87, 6062–6072.

Richards, K.F., Bienkowska-Haba, M., Dasgupta, J., Chen, X.S., and Sapp,

M. (2013). Multiple heparan sulfate binding site engagements are required

for the infectious entry of human papillomavirus type 16. J. Virol. 87,

11426–11437.

Roden, R.B., Day, P.M., Bronzo, B.K., Yutzy, W.H., Yang, Y., Lowy, D.R., and

Schiller, J.T. (2001). Positively charged termini of the L2 minor capsid protein

are necessary for papillomavirus infection. J. Virol. 75, 10493–10497.

Rohou, A., and Grigorieff, N. (2015). CTFFIND4: fast and accurate defocus

estimation from electron micrographs. J. Struct. Biol. 192, 216–221.

Rommel, O., Dillner, J., Fligge, C., Bergsdorf, C., Wang, X., Selinka, H.C., and

Sapp, M. (2005). Heparan sulfate proteoglycans interact exclusively with con-

formationally intact HPV L1 assemblies: basis for a virus-like particle ELISA.

J. Med. Virol. 75, 114–121.

Rosenthal, P.B., and Henderson, R. (2003). Optimal determination of particle

orientation, absolute hand, and contrast loss in single-particle electron cryo-

microscopy. J. Mol. Biol. 333, 721–745.

Sapp, M., Fligge, C., Petzak, I., Harris, J.R., and Streeck, R.E. (1998).

Papillomavirus assembly requires trimerization of the major capsid protein

by disulfides between two highly conserved cysteines. J. Virol. 72, 6186–6189.

Scheres, S.H.W. (2012). RELION: implementation of a Bayesian approach to

cryo-EM structure determination. J. Struct. Biol. 180, 519–530.

Scheres, S.H.W., and Chen, S. (2012). Prevention of overfitting in cryo-EM

structure determination. Nat. Methods 9, 853–854.

Schowalter, R.M., Pastrana, D.V., and Buck, C.B. (2011). Glycosaminoglycans

and sialylated glycans sequentially facilitate Merkel cell polyomavirus infec-

tious entry. PLoS Pathog. 7, e1002161.

Selinka, H.-C., Giroglou, T., Nowak, T., Christensen, N.D., and Sapp, M.

(2003). Further evidence that papillomavirus capsids exist in two distinct

conformations. J. Virol. 77, 12961–12967.

Selinka, H.-C., Florin, L., Patel, H.D., Freitag, K., Schmidtke, M.,Makarov, V.A.,

and Sapp, M. (2007). Inhibition of transfer to secondary receptors by heparan

sulfate-binding drug or antibody induces noninfectious uptake of human papil-

lomavirus. J. Virol. 81, 10970–10980.

Shafti-Keramat, S., Handisurya, A., Kriehuber, E., Meneguzzi, G.,

Slupetzky, K., and Kirnbauer, R. (2003). Different heparan sulfate proteogly-

cans serve as cellular receptors for human papillomaviruses. J. Virol. 77,

13125–13135.

Page 12: Cryoelectron Microscopy Maps of Human Papillomavirus 16 …php.scripts.psu.edu/dept/hafenstein/pdfs/1-s2.0-S... · 2019. 10. 15. · Structure Article Cryoelectron Microscopy Maps

Tang, G., Peng, L., Baldwin, P.R., Mann, D.S., Jiang, W., Rees, I., and Ludtke,

S.J. (2007). EMAN2: an extensible image processing suite for electron micro-

scopy. J. Struct. Biol. 157, 38–46.

Toh, Z.Q., Licciardi, P.V., Fong, J., Garland, S.M., Tabrizi, S.N., Russell, F.M.,

and Mulholland, E.K. (2015). Reduced dose human papillomavirus vaccina-

tion: an update of the current state-of-the-art. Vaccine 33, 5042–5050.

Trus, B.L., Roden, R.B., Greenstone, H.L., Vrhel, M., Schiller, J.T., and Booy,

F.P. (1997). Novel structural features of bovine papillomavirus capsid revealed

by a three-dimensional reconstruction to 9 A resolution. Nat. Struct. Biol. 4,

413–420.

Wolf, M., Garcea, R.L., Grigorieff, N., and Harrison, S.C. (2010). Subunit inter-

actions in bovine papillomavirus. Proc. Natl. Acad. Sci. USA 107, 6298–6303.

Wriggers, W. (2010). Using Situs for the integration of multi-resolution struc-

tures. Biophys. Rev. 2 (1), 21–27.

Wriggers, W., Milligan, R.A., andMcCammon, J.A. (1999). Situs: a package for

docking crystal structures into low-resolution maps from electron microscopy.

J. Struct. Biol. 125, 185–195.

Xia, L., Xian, Y., Wang, D., Chen, Y., Huang, X., Bi, X., Yu, H., Fu, Z., Liu, X., Li,

S., et al. (2016). A human monoclonal antibody against HPV16 recognizes an

immunodominant and neutralizing epitope partially overlapping with that of

H16.V5. Sci. Rep. 6, 19042.

Yan, X., Sinkovits, R.S., and Baker, T.S. (2007). AUTO3DEM—an automated

and high throughput program for image reconstruction of icosahedral parti-

cles. J. Struct. Biol. 157, 73–82.

Zhao, Q., Potter, C.S., Carragher, B., Lander, G., Sworen, J., Towne, V.,

Abraham, D., Duncan, P., Washabaugh, M.W., and Sitrin, R.D. (2014).

Characterization of virus-like particles in GARDASIL� by cryo transmission

electron microscopy. Hum. Vaccin. Immunother. 10, 734–739.

Structure 25, 253–263, February 7, 2017 263