Crystal structure, conformational fixation and entry-related interactions of mature ligand-free HIV-1 Env

Young Do Kwon, Marie Pancera, Priyamvada Acharya, Ivelin S Georgiev, Emma T Crooks, Jason Gorman, M Gordon Joyce, Miklos Guttman, Xiaochu Ma, Sandeep Narpala, Cinque Soto, Daniel S Terry, Yongping Yang, Tongqing Zhou, Goran Ahlsen, Robert T Bailer, Michael Chambers, Gwo-Yu Chuang, Nicole A Doria-Rose, Aliaksandr Druz, Mark A Hallen, Adam Harned, Tatsiana Kirys, Mark K Louder, Sijy O'Dell, Gilad Ofek, Keiko Osawa, Madhu Prabhakaran, Mallika Sastry, Guillaume B E Stewart-Jones, Jonathan Stuckey, Paul V Thomas, Tishina Tittley, Constance Williams, Baoshan Zhang, Hong Zhao, Zhou Zhou, Bruce R Donald, Lawrence K Lee, Susan Zolla-Pazner, Ulrich Baxa, Arne Schön, Ernesto Freire, Lawrence Shapiro, Kelly K Lee, James Arthos, James B Munro, Scott C Blanchard, Walther Mothes, James M Binley, Adrian B McDermott, John R Mascola, Peter D Kwong, Young Do Kwon, Marie Pancera, Priyamvada Acharya, Ivelin S Georgiev, Emma T Crooks, Jason Gorman, M Gordon Joyce, Miklos Guttman, Xiaochu Ma, Sandeep Narpala, Cinque Soto, Daniel S Terry, Yongping Yang, Tongqing Zhou, Goran Ahlsen, Robert T Bailer, Michael Chambers, Gwo-Yu Chuang, Nicole A Doria-Rose, Aliaksandr Druz, Mark A Hallen, Adam Harned, Tatsiana Kirys, Mark K Louder, Sijy O'Dell, Gilad Ofek, Keiko Osawa, Madhu Prabhakaran, Mallika Sastry, Guillaume B E Stewart-Jones, Jonathan Stuckey, Paul V Thomas, Tishina Tittley, Constance Williams, Baoshan Zhang, Hong Zhao, Zhou Zhou, Bruce R Donald, Lawrence K Lee, Susan Zolla-Pazner, Ulrich Baxa, Arne Schön, Ernesto Freire, Lawrence Shapiro, Kelly K Lee, James Arthos, James B Munro, Scott C Blanchard, Walther Mothes, James M Binley, Adrian B McDermott, John R Mascola, Peter D Kwong

Abstract

As the sole viral antigen on the HIV-1-virion surface, trimeric Env is a focus of vaccine efforts. Here we present the structure of the ligand-free HIV-1-Env trimer, fix its conformation and determine its receptor interactions. Epitope analyses revealed trimeric ligand-free Env to be structurally compatible with broadly neutralizing antibodies but not poorly neutralizing ones. We coupled these compatibility considerations with binding antigenicity to engineer conformationally fixed Envs, including a 201C 433C (DS) variant specifically recognized by broadly neutralizing antibodies. DS-Env retained nanomolar affinity for the CD4 receptor, with which it formed an asymmetric intermediate: a closed trimer bound by a single CD4 without the typical antigenic hallmarks of CD4 induction. Antigenicity-guided structural design can thus be used both to delineate mechanism and to fix conformation, with DS-Env trimers in virus-like-particle and soluble formats providing a new generation of vaccine antigens.

Conflict of interest statement

Competing financial interests The authors declare no competing financial interests.

Figures

Figure 1
Figure 1
Crystal structure of ligand-free HIV-1-Env trimer, and conformational changes related to individual subunit structures and virus entry. (a) The left gp120-gp41 protomer of the ligand-free BG505 SOSIP.664 trimer is shown in B-factor putty representation, the right protomer in Cα-backbone representation colored by average Cα distance between ligand-free Env and previously determined Env subunit structures, and the third protomer in grey cartoon representation. (b) Plot of pre-fusion Env B-factors versus Cα-subunit movement (see Supplementary Table 1 for a full listing of correlations and P-values for residue-level properties). (c) Correlations and P-values for pre-fusion B-factors of type 1 fusion machines versus pre-fusion to post-fusion movement of fusion subunit (see Supplementary Fig. 3 for subunit pictorials and correlation graphs). P-values in (b) and (c) were obtained by two-tailed student’s t test.
Figure 2
Figure 2
Ligand-free HIV-1-Env trimer is structurally compatible with epitopes of broadly neutralizing, but not ineffective, antibodies. (a) Superposition of ligand-free and antibody-bound HIV-1-Env structures. (Left) Ligand-free gp120 core monomer shown in ribbon representation, with regions of less (or greater) than 2 Å RMSD upon antibody binding shown in green (or magenta), and representative antibody-bound structures in gray. (Middle and right) Ligand-free and antibody-bound HIV-1-Env trimers. In the right panel, antibodies PGT122 and 35022 are shown in gray semitransparent surface, with rear protomer removed for clarity. RMSD values are reported in Supplementary Table 7. (b) Breadth-potency of broadly neutralizing (green) and ineffective (magenta) antibodies on a diverse 170 HIV-1-isolate panel. (c) Structural compatibility of ligand-free trimer by antibody epitope: an appropriate structural compatibility. The ligand-free Env structure is displayed as Cα-ribbon, with antibody epitope residues colored green (structurally compatible) or magenta (incompatible), and grey for non-epitope regions. RMSD (solid fill) and volume overlap (striped fill) with the respective antibody-Env complexes shown as a bar graph, with two linear scales split at RMSD and antibody-antigen volume overlap cutoffs of 2 and 500 Å3, respectively; bars below the respective cutoffs are colored green, and magenta otherwise. Antibody labels are colored green if the epitope is structurally compatible, magenta if incompatible, and gray if not present in the structure. (d) Ligand-free trimer structural compatibility versus antibody breadth. Volume overlap (left), RMSD (middle) and Antigenic Structural Compatibility (ASC) score (right) are graphed versus antibody breadth on a diverse 170 HIV-1-isolate panel; pictorial representations shown in Supplemental Fig. 5. P-values for Spearman correlations provided (n = 14).
Figure 3
Figure 3
Structural compatibility-guided negative selection and an appropriate target antigenicity. (a) Size-exclusion chromatography profile of crystallization-grade SOSIP before negative selection (left panel), and SDS-PAGE analyses of negative selection: first with antibody 447-52D, next with a cocktail of V3 antibodies, and third by CD4 (right panels). NR: non-reducing conditions, R: reducing conditions. (b) V3-epitope exposure on BG505 SOSIP.664 quantified by SPR on a panel of V3 antibodies and displayed as percent of total V3 epitope upon CD4 triggering. *V3 epitopes for antibodies 39F and 19b have not been structurally defined. (c) V3 antigenicity for all structurally defined antibodies in b mapped onto epitope atoms in the structure of the ligand-free Env trimer. (d) Appropriate target antigenicity. Binding antigenicity is displayed on the ligand-free Env trimer with epitope atoms with tight trimer binding to broadly neutralizing antibodies in green and to ineffective antibodies in magenta.
Figure 4
Figure 4
CD4-induced changes in antigenicity and conformational fixation of ligand-free HIV-1 Env. (a) BG505 SOSIP.664 ligand-free structural compatibility versus binding antigenicity, in the absence (left) and presence (right) of CD4 as measured by MSD. Antibodies are displayed in green (broadly neutralizing) and magenta (ineffective); the average binding of each is provided; and ineffective antibodies are labeled (CD4bs: CD4-binding site, CD4i: CD4-induced, non: non-neutralizing, V3: V3-loop directed). The change in binding to ineffective antibodies in the presence of CD4 is shown as a yellow dotted line. P-values for Spearman correlations are provided (n = 13). Binding levels for non-cognate antibodies lie within the gray shaded areas. (b) Conformational fixation of HIV-1-Env trimer. The central image depicts the ligand-free BG505 SOSIP.664 HIV-1-Env trimer, with two protomers shown in cartoon representation, one colored by domains (gp120 in wheat and gp41 in light pink), a second colored by RMSD distance between ligand-free trimer and subunit structures of CD4-bound gp120 and post-fusion gp41 (green if less than 2 Å, magenta if more than 2 Å). A third protomer is shown in gray. Insets: atomic-level details. *Residue 559 is disordered in the ligand-free structure. (c) Binding antigenicity of BG505 SOSIP.664 variants. Heat map showing binding of BG505 SOSIP.664 and variants that stabilized the ligand-free closed state to a panel of antibodies. (d) Ligand-free trimer structural compatibility versus BG505 SOSIP.664 201C-433C binding antigenicity in the absence (left) and presence (right) of CD4, with antibodies and average binding in green (broadly neutralizing) and magenta (ineffective) as in a. Elisa, Octet and Biacore data in Supplemental Data Set 2 and Supplementary Tables 5 and 6.
Figure 5
Figure 5
Atomic-level models and physical stability of ligand-free 201C-433C mutant (DS-SOSIP). (a) Atomic-level models of residues 201 and 433 in ligand-free pre-fusion closed state (grey) and CD4-bound state (yellow). Ribbon representations of the two structures are shown with residues that make-up the bridging sheet in the CD4-bound conformation colored orange, residues 201 and 433 in cyan spheres, and 201-433 Cα distance indicated. Variable loops are labeled. (Monomeric CD4-bound conformation modeled from PDB ID 2B4C, 3U4E and 3JWD). (b) Thermostability of the DS-SOSIP was assessed by differential scanning calorimetry. Raw data are shown in solid line (black for BG505 SOSIP.664 and red for DS-SOSIP), with corresponding curves from the fit shown in purple dashed lines. Tm values and error were obtained from the fit. (c) Physical stability of trimeric DS-SOSIP as determined by the quaternary-specific antibody VRC26.09 after 60 minutes of incubation at physical extremes or after 10 freeze-thaw cycles. Error bars are SEM of two technical replicates.
Figure 6
Figure 6
DS-SOSIP binds a single CD4 without the typical antigenic hallmarks of CD4 triggering. (a) Binding of soluble CD4 to SOSIP.664 and mutants measured by SPR with single-cycle kinetics. Values in parenthesis report standard errors from fit of the data to a 1:1 Langmuir binding model. Note that the level of binding for the P313W mutant is roughly 3-times higher than either wild-type SOSIP or 201C-433C. (b) Time course of CD4 activation of HIV-1 Env as measured by SPR. To initiate the time course, CD4 was mixed with HIV-1 Env at time point 0, and after incubation (time shown on x-axis), assessed by SPR for interaction with antibody (y-axis). (Left) Binding to antibody 17b, which recognizes a bridging sheet epitope; (right) binding to 3074, which recognizes a V3 epitope (right). (c) Sedimentation equilibrium analytical ultracentrifugation measurements of BG505 SOSIP.664 and 201C-433C variant in presence of excess 2-domain soluble CD4. Stochiometry for both 2-domain and 4-domain CD4 as well as residual calculation are provided in Supplementary Data Set 4.
Figure 7
Figure 7
An asymmetric intermediate in the HIV-1-entry pathway. (a) smFRET of JR-FL virions with and without 201C-433C substitution. Population FRET histograms are each paired with transition density plots, which display the relative density of observed transitions. Results for both ligand-free and dodecameric CD4 (sCD4D1D2-Igαtp), graphed in left and right pairs, respectively. The dominance of the low FRET state even in the presence of CD4 indicates DS-stabilized Env to remain in a closed state even when bound by CD4. (b) HIV-1 entry mechanism with conformation-blocking mutations, antigenicity, and interactions with functional ligands. A new mechanistic state, characterized by the binding of a single molecule of CD4 with no bridging sheet formation and reduced V3 exposure, is highlighted in red. Env density from refs. and .
Figure 8
Figure 8
A new generation of conformationally fixed HIV-1-Env trimeric immunogens. (a) HDX characterization of conformational mobility. Changes in exchange of hydrogen-deuterium relative to wild-type BG505 SOSIP.664 are displayed, with regions that become more ordered (blue) or disordered (red) shown on one lobe of the ligand-free trimer. Full HDX profiles are shown in Supplementary Fig. 7. (b) Antigenic characteristics of SOS virus-like particles (VLPs, comprising SOS mutant without IP) from strain JR-FL (modified with E168K to allow binding of V1V2-directed broadly neutralizing antibodies) and strain BG505 (modified with T332N to allow binding of 2G12 antibody). While broadly neutralizing antibody binding is maintained between parent and 201C-433C VLPs, the 201C-433C variant showed reduced ineffective antibody binding, especially in the presence of CD4. Broadly neutralizing antibodies binding are shown in green, ineffective in magenta and binding of antibodies in presence of 2 μg/ml of sCD4 in orange. Ineffective antibodies labeled (CD4bs: CD4-binding site, CD4i: CD4-induced, V3: V3-loop directed). (c) Information flow of antigenicity-guided immunogen design: from human antibody response through structure and antigenicity-guided design to conformationally fixed immunogens.

References

    1. Starcich BR, et al. Identification and characterization of conserved and variable regions in the envelope gene of HTLV-III/LAV, the retrovirus of AIDS. Cell. 1986;45:637–48.
    1. Wei X, et al. Antibody neutralization and escape by HIV-1. Nature. 2003;422:307–12.
    1. Chen L, et al. Structural basis of immune evasion at the site of CD4 attachment on HIV-1 gp120. Science. 2009;326:1123–7.
    1. Kwong PD, et al. HIV-1 evades antibody-mediated neutralization through conformational masking of receptor-binding sites. Nature. 2002;420:678–82.
    1. Fouts TR, Binley JM, Trkola A, Robinson JE, Moore JP. Neutralization of the human immunodeficiency virus type 1 primary isolate JR-FL by human monoclonal antibodies correlates with antibody binding to the oligomeric form of the envelope glycoprotein complex. J Virol. 1997;71:2779–85.
    1. Huang CC, et al. Structure of a V3-containing HIV-1 gp120 core. Science. 2005;310:1025–8.
    1. Weiss RA, et al. Neutralization of human T-lymphotropic virus type III by sera of AIDS and AIDS-risk patients. Nature. 1985;316:69–72.
    1. Bures R, et al. Immunization with recombinant canarypox vectors expressing membrane- anchored glycoprotein 120 followed by glycoprotein 160 boosting fails to generate antibodies that neutralize R5 primary isolates of human immunodeficiency virus type 1. AIDS Res Hum Retroviruses. 2000;16:2019–35.
    1. Flynn NM, et al. Placebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1 infection. J Infect Dis. 2005;191:654–65.
    1. Liu J, Bartesaghi A, Borgnia MJ, Sapiro G, Subramaniam S. Molecular architecture of native HIV-1 gp120 trimers. Nature. 2008;455:109–13.
    1. Bartesaghi A, Merk A, Borgnia MJ, Milne JL, Subramaniam S. Prefusion structure of trimeric HIV-1 envelope glycoprotein determined by cryo-electron microscopy. Nat Struct Mol Biol. 2013;20:1352–7.
    1. Julien JP, et al. Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science. 2013;342:1477–83.
    1. Lyumkis D, et al. Cryo-EM structure of a fully glycosylated soluble cleaved HIV-1 envelope trimer. Science. 2013;342:1484–90.
    1. Pancera M, et al. Structure and immune recognition of trimeric pre-fusion HIV-1 Env. Nature. 2014;514:455–61.
    1. Wu X, et al. Neutralization escape variants of human immunodeficiency virus type 1 are transmitted from mother to infant. J Virol. 2006;80:835–844.
    1. Binley JM, et al. A recombinant human immunodeficiency virus type 1 envelope glycoprotein complex stabilized by an intermolecular disulfide bond between the gp120 and gp41 subunits is an antigenic mimic of the trimeric virion- associated structure. J Virol. 2000;74:627–43.
    1. Sanders RW, et al. Stabilization of the soluble, cleaved, trimeric form of the envelope glycoprotein complex of human immunodeficiency virus type 1. J Virol. 2002;76:8875–89.
    1. Sanders RW, et al. A next-generation cleaved, soluble HIV-1 Env Trimer, BG505 SOSIP.664 gp140, expresses multiple epitopes for broadly neutralizing but not non-neutralizing antibodies. PLoS Pathog. 2013;9:e1003618.
    1. Kwon YD, et al. Unliganded HIV-1 gp120 core structures assume the CD4-bound conformation with regulation by quaternary interactions and variable loops. Proc Natl Acad Sci U S A. 2012;109:5663–8.
    1. Kwong PD, et al. Structure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibody. Nature. 1998;393:648–59.
    1. Zhou T, et al. Structural definition of a conserved neutralization epitope on HIV-1 gp120. Nature. 2007;445:732–7.
    1. Zhou T, et al. Structural Basis for Broad and Potent Neutralization of HIV-1 by Antibody VRC01. Science. 2010;329:811–817.
    1. Pejchal R, et al. A potent and broad neutralizing antibody recognizes and penetrates the HIV glycan shield. Science. 2011;334:1097–103.
    1. Scharf L, et al. Antibody 8ANC195 Reveals a Site of Broad Vulnerability on the HIV-1 Envelope Spike. Cell Rep. 2014;7:785–95.
    1. Wyatt R, Sodroski J. The HIV-1 envelope glycoproteins: fusogens, antigens, and immunogens. Science. 1998;280:1884–8.
    1. Chan DC, Fass D, Berger JM, Kim PS. Core structure of gp41 from the HIV envelope glycoprotein. Cell. 1997;89:263–73.
    1. Weissenhorn W, Dessen A, Harrison SC, Skehel JJ, Wiley DC. Atomic structure of the ectodomain from HIV-1 gp41. Nature. 1997;387:426–30.
    1. Munro JB, et al. Conformational dynamics of single HIV-1 envelope trimers on the surface of native virions. Science. 2014;346:759–63.
    1. Guttman M, et al. Antibody potency relates to the ability to recognize the closed, pre-fusion form of HIV Env. Nat Commun. 2015;6:6144.
    1. Jancarik JaKS-H. Sparse matrix sampling: a screening method for crystallization of proteins. J Appl Cryst. 1991;24:409–411.
    1. Majeed S, et al. Enhancing protein crystallization through precipitant synergy. Structure. 2003;11:1061–70.
    1. Adams PD, et al. Recent developments in the PHENIX software for automated crystallographic structure determination. J Synchrotron Radiat. 2004;11:53–5.
    1. Korber BTF, BT, Kuiken CL, Pillai SK, Sodroski JG. Numbering Positions in HIV Relative to HXB2CG. 1998.
    1. Burton DR, et al. Efficient neutralization of primary isolates of HIV-1 by a recombinant human monoclonal antibody. Science. 1994;266:1024–7.
    1. Huang J, et al. Broad and potent neutralization of HIV-1 by a human antibody that recognizes an intersubunit site on the envelope glycoprotein. Nature. 2014 In Press.
    1. Walker LM, et al. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature. 2011;477:466–70.
    1. Zolla-Pazner S, et al. The cross-clade neutralizing activity of a human monoclonal antibody is determined by the GPGR V3 motif of HIV type 1. AIDS Res Hum Retroviruses. 2004;20:1254–8.
    1. Liao HX, et al. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature. 2013;496:469–476.
    1. Guenaga J, et al. Well-ordered trimeric HIV-1 subtype B and C soluble spike mimetics generated by negative selection display native-like properties. PLoS Pathog. 2015;11:e1004570.
    1. Mbah HA, et al. Effect of soluble CD4 on exposure of epitopes on primary, intact, native human immunodeficiency virus type 1 virions of different genetic clades. J Virol. 2001;75:7785–8.
    1. Stanfield RL, Gorny MK, Williams C, Zolla-Pazner S, Wilson IA. Structural rationale for the broad neutralization of HIV-1 by human monoclonal antibody 447-52D. Structure (Camb) 2004;12:193–204.
    1. Jiang X, et al. Conserved structural elements in the V3 crown of HIV-1 gp120. Nat Struct Mol Biol. 2010;17:955–61.
    1. Thali M, et al. Characterization of conserved human immunodeficiency virus type 1 gp120 neutralization epitopes exposed upon gp120-CD4 binding. J Virol. 1993;67:3978–88.
    1. McLellan JS, et al. Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science. 2013;342:592–8.
    1. Wu X, et al. Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science. 2010;329:856–61.
    1. Doria-Rose NA, et al. Developmental pathway for potent V1V2-directed HIV-neutralizing antibodies. Nature. 2014;509:55–62.
    1. Posner MR, Cavacini LA, Emes CL, Power J, Byrn R. Neutralization of HIV-1 by F105, a human monoclonal antibody to the CD4 binding site of gp120. J Acquir Immune Defic Syndr. 1993;6:7–14.
    1. Arthos J, et al. Biochemical and biological characterization of a dodecameric CD4-Ig fusion protein: implications for therapeutic and vaccine strategies. J Biol Chem. 2002;277:11456–64.
    1. Daar ES, Ho DD. Relative resistance of primary HIV-1 isolates to neutralization by soluble CD4. Am J Med. 1991;90:22S–26S.
    1. Crooks ET, Tong T, Osawa K, Binley JM. Enzyme digests eliminate nonfunctional Env from HIV-1 particle surfaces, leaving native Env trimers intact and viral infectivity unaffected. J Virol. 2011;85:5825–39.
    1. Tong T, Crooks ET, Osawa K, Binley JM. HIV-1 virus-like particles bearing pure env trimers expose neutralizing epitopes but occlude nonneutralizing epitopes. J Virol. 2012;86:3574–87.
    1. Trkola A, et al. Human monoclonal antibody 2G12 defines a distinctive neutralization epitope on the gp120 glycoprotein of human immunodeficiency virus type 1. J Virol. 1996;70:1100–8.
    1. McLellan JS, et al. Structure of RSV fusion glycoprotein trimer bound to a prefusion-specific neutralizing antibody. Science. 2013;340:1113–7.
    1. McLellan JS, Yang Y, Graham BS, Kwong PD. Structure of respiratory syncytial virus fusion glycoprotein in the postfusion conformation reveals preservation of neutralizing epitopes. J Virol. 2011;85:7788–96.
    1. Swanson KA, et al. Structural basis for immunization with postfusion respiratory syncytial virus fusion F glycoprotein (RSV F) to elicit high neutralizing antibody titers. Proc Natl Acad Sci U S A. 2011;108:9619–24.
    1. Kovacs JM, et al. Stable, uncleaved HIV-1 envelope glycoprotein gp140 forms a tightly folded trimer with a native-like structure. Proc Natl Acad Sci U S A. 2014;111:18542–7.
    1. Pancera M, et al. Soluble mimetics of human immunodeficiency virus type 1 viral spikes produced by replacement of the native trimerization domain with a heterologous trimerization motif: characterization and ligand binding analysis. J Virol. 2005;79:9954–69.
    1. Moore JP. Native-like BG505 SOSIP.664 Trimers Induce Autologous Tier-2 NAbs against Complex Epitopes in Rabbits and Macaques. AIDS Res Hum Retroviruses. 2014;30:A67–A67.
    1. McLellan JS, et al. Structure of HIV-1 gp120 V1/V2 domain with broadly neutralizing antibody PG9. Nature. 2011;480:336–43.
    1. Tran EE, et al. Structural mechanism of trimeric HIV-1 envelope glycoprotein activation. PLoS Pathog. 2012;8:e1002797.
    1. Killikelly A, et al. Thermodynamic signatures of the antigen binding site of mAb 447-52D targeting the third variable region of HIV-1 gp120. Biochemistry. 2013;52:6249–57.
    1. Stanfield RL, Gorny MK, Zolla-Pazner S, Wilson IA. Crystal structures of human immunodeficiency virus type 1 (HIV-1) neutralizing antibody 2219 in complex with three different V3 peptides reveal a new binding mode for HIV-1 cross-reactivity. J Virol. 2006;80:6093–105.
    1. Gorny MK, et al. Human anti-V3 HIV-1 monoclonal antibodies encoded by the VH5-51/VL lambda genes define a conserved antigenic structure. PLoS One. 2011;6:e27780.
    1. Rini JM, et al. Crystal structure of a human immunodeficiency virus type 1 neutralizing antibody, 50.1, in complex with its V3 loop peptide antigen. Proc Natl Acad Sci U S A. 1993;90:6325–9.
    1. Otwinowski Z, Minor W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 1997;276:307–326.
    1. Emsley P, Cowtan K. Coot: model-building tools for molecular graphics. Acta Crystallogr D Biol Crystallogr. 2004;60:2126–32.
    1. Bernstein FC, et al. The Protein Data Bank: a computer-based archival file for macromolecular structures. J Mol Biol. 1977;112:535–42.
    1. Zhang Y, Skolnick J. TM-align: a protein structure alignment algorithm based on the TM-score. Nucleic Acids Res. 2005;33:2302–9.
    1. Pancera M, et al. Structure of HIV-1 gp120 with gp41-interactive region reveals layered envelope architecture and basis of conformational mobility. Proc Natl Acad Sci U S A. 2010;107:1166–71.
    1. Guttman M, et al. CD4-Induced Activation in a Soluble HIV-1 Env Trimer. Structure. 2014;22:974–84.
    1. Sanner MF, Olson AJ, Spehner JC. Reduced surface: an efficient way to compute molecular surfaces. Biopolymers. 1996;38:305–20.
    1. Hubbard SJaT, JM . “NACCESS”, Computer Program. Department of Biochemistry and Molecular Biology, University College London; 1993.
    1. Nicholls A, Sharp KA, Honig B. Protein folding and association: insights from the interfacial and thermodynamic properties of hydrocarbons. Proteins. 1991;11:281–96.
    1. Sarzotti-Kelsoe M, et al. Optimization and validation of the TZM-bl assay for standardized assessments of neutralizing antibodies against HIV-1. J Immunol Methods. 2014;409:131–46.
    1. Huang CC, et al. Structural basis of tyrosine sulfation and VH-gene usage in antibodies that recognize the HIV type 1 coreceptor-binding site on gp120. Proc Natl Acad Sci U S A. 2004;101:2706–11.
    1. Xiang SH, et al. Epitope mapping and characterization of a novel CD4-induced human monoclonal antibody capable of neutralizing primary HIV-1 strains. Virology. 2003;315:124–34.
    1. Huang J, et al. Broad and potent HIV-1 neutralization by a human antibody that binds the gp41-gp120 interface. Nature. 2014;515:138–42.
    1. Scheid JF, et al. Broad diversity of neutralizing antibodies isolated from memory B cells in HIV-infected individuals. Nature. 2009;458:636–40.
    1. Liao HX, et al. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature. 2013;496:469–76.
    1. Calarese DA, et al. Antibody domain exchange is an immunological solution to carbohydrate cluster recognition. Science. 2003;300:2065–71.
    1. Georgiev IS, et al. Delineating antibody recognition in polyclonal sera from patterns of HIV-1 isolate neutralization. Science. 2013;340:751–6.
    1. Doria-Rose NA, et al. Developmental pathway for potent V1V2-directed HIV-neutralizing antibodies. Nature. 2014;509:55–62.
    1. Seaman MS, et al. Tiered categorization of a diverse panel of HIV-1 Env pseudoviruses for assessment of neutralizing antibodies. J Virol. 2010;84:1439–52.
    1. Dreyfus C, et al. Highly conserved protective epitopes on influenza B viruses. Science. 2012;337:1343–8.
    1. Tharakaraman K, Subramanian V, Cain D, Sasisekharan V, Sasisekharan R. Broadly neutralizing influenza hemagglutinin stem-specific antibody CR8020 targets residues that are prone to escape due to host selection pressure. Cell Host Microbe. 2014;15:644–51.
    1. Palivizumab a humanized respiratory syncytial virus monoclonal antibody reduces hospitalization from respiratory syncytial virus infection in high-risk infants. The IMpact-RSV Study Group. Pediatrics. 1998;102:531–7.
    1. Mastronarde DN. Automated electron microscope tomography using robust prediction of specimen movements. J Struct Biol. 2005;152:36–51.
    1. Tang G, et al. EMAN2: an extensible image processing suite for electron microscopy. J Struct Biol. 2007;157:38–46.
    1. Shu Y, et al. Efficient protein boosting after plasmid DNA or recombinant adenovirus immunization with HIV-1 vaccine constructs. Vaccine. 2007;25:1398–408.
    1. Wei X, et al. Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy. Antimicrob Agents Chemother. 2002;46:1896–905.
    1. Tong T, Osawa K, Robinson JE, Crooks ET, Binley JM. Topological analysis of HIV-1 glycoproteins expressed in situ on virus surfaces reveals tighter packing but greater conformational flexibility than for soluble gp120. J Virol. 2013;87:9233–49.

Source: PubMed

3
Se inscrever