HIV Envelope Glycoform Heterogeneity and Localized Diversity Govern the Initiation and Maturation of a V2 Apex Broadly Neutralizing Antibody Lineage

Elise Landais, Ben Murrell, Bryan Briney, Sasha Murrell, Kimmo Rantalainen, Zachary T Berndsen, Alejandra Ramos, Lalinda Wickramasinghe, Melissa Laird Smith, Kemal Eren, Natalia de Val, Mengyu Wu, Audrey Cappelletti, Jeffrey Umotoy, Yolanda Lie, Terri Wrin, Paul Algate, Po-Ying Chan-Hui, Etienne Karita, IAVI Protocol C Investigators, IAVI African HIV Research Network, Andrew B Ward, Ian A Wilson, Dennis R Burton, Davey Smith, Sergei L Kosakovsky Pond, Pascal Poignard, Elise Landais, Ben Murrell, Bryan Briney, Sasha Murrell, Kimmo Rantalainen, Zachary T Berndsen, Alejandra Ramos, Lalinda Wickramasinghe, Melissa Laird Smith, Kemal Eren, Natalia de Val, Mengyu Wu, Audrey Cappelletti, Jeffrey Umotoy, Yolanda Lie, Terri Wrin, Paul Algate, Po-Ying Chan-Hui, Etienne Karita, IAVI Protocol C Investigators, IAVI African HIV Research Network, Andrew B Ward, Ian A Wilson, Dennis R Burton, Davey Smith, Sergei L Kosakovsky Pond, Pascal Poignard

Abstract

Understanding how broadly neutralizing antibodies (bnAbs) to HIV envelope (Env) develop during natural infection can help guide the rational design of an HIV vaccine. Here, we described a bnAb lineage targeting the Env V2 apex and the Ab-Env co-evolution that led to development of neutralization breadth. The lineage Abs bore an anionic heavy chain complementarity-determining region 3 (CDRH3) of 25 amino acids, among the shortest known for this class of Abs, and achieved breadth with only 10% nucleotide somatic hypermutation and no insertions or deletions. The data suggested a role for Env glycoform heterogeneity in the activation of the lineage germline B cell. Finally, we showed that localized diversity at key V2 epitope residues drove bnAb maturation toward breadth, mirroring the Env evolution pattern described for another donor who developed V2-apex targeting bnAbs. Overall, these findings suggest potential strategies for vaccine approaches based on germline-targeting and serial immunogen design.

Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1. Functional Screening Identifies a New…
Figure 1. Functional Screening Identifies a New V2-Apex Specific Broadly Neutralizing Antibody Lineage
(A) Longitudinal plasma samples from donor PC64 were tested for neutralization against heterologous pseudoviruses. The percent of viruses neutralized (> 50% inhibition of infectivity at the lowest plasma dilution, 1:50) from a cross-clade (A, B, C) 37-virus panel is shown as shaded blue bars. Neutralization Inhibitory Dilution 50 (ID50, plasma dilution giving 50% inhibition of infection) of PC64 longitudinal plasma samples against 92TH021 WT (black circles) and N160K mutant (red circles) pseudoviruses are plotted. The evolution of the viral load (green circles) in the plasma is also plotted. The time points at which PCT64 antibodies were isolated andenv sequenced and/or cloned from plasma are indicated by corresponding symbols. The number (N) of PCT64 antibodies isolated and their range of somatic hypermutation frequency (%SHM) is also indicated. See also Table S1. (B) Evolutionary distance between the PCT64 mAbs is illustrated as a phylogeny for both heavy and light chain nucleotide sequences. (C) Mapping of the PCT64 epitope. Fold decrease in neutralization IC50 of individual JR-CSF Ala mutants by PCT64-35G as compared to WT is color-coded as indicated and presented on the BG505 SOSIP.664 trimer structure (Julien et al., 2013) based on HxB2 numbering and alignments. Data are representative of at least two independent experiments. See also Table S3. (D) PCT64 mAbs characteristics. Heavy and light chain V- and J-gene nucleotide (nt) somatic mutation percentages, neutralization breadth (% virus neutralized at IC50<50 μg/mL) and potency (GeoMean IC50 in μg/mL) on a 37- and 109-virus panel are tabulated and color-coded as indicated. See also Tables S1, S5, and S6. Data are representative of at least two independent experiments. (E) Comparison of PC64 plasma neutralization breadth and potency on the 109-virus panel subdivided by subtype, with that of a theoretical combination of PCT64 mAbs, color-coded as indicated. See also Tables S2, S4, S7, and Figure S1A.
Figure 2. Longitudinal Next-Generation Sequencing of the…
Figure 2. Longitudinal Next-Generation Sequencing of the Memory B Cell Repertoire Reveals the Phylogeny of the PCT64 bnAb Lineage
PC64 IgG libraries prepared from total PBMCs were amplified with IgG-specific primers for all human VH gene families. (A) Frequency of PCT64 bnAbs at each time point, plotted as a percentage of total PCT64 lineage sequences from all time points. Plasma neutralization score (see Landais et al., 2016) from a heterologous 37-virus panel is plotted as a dashed line. (B) Somatic hypermutation frequency was calculated for each time point either as divergence (number of amino acid changes compared to LMCA; top panel) or as sum of the evolutionary distance (middle panel). Data are presented as whisker plots showing mean, 95% upper and lower quartiles, SD, and outliers. The evolutionary distance normalized by divergence using mean values is shown in the bottom panel. (C) Longitudinal phylogeny of PCT64 HC sequences (colored by mpi). The PCT64 mAb sequences are named in black, and star symbols represent autologous (black) and heterologous (red) neutralization. See also Figure S2.
Figure 3. Crystal Structure of PCT64-35B bnAb…
Figure 3. Crystal Structure of PCT64-35B bnAb Fab Shows an Extended PGT145-like CDRH3 Conformation
(A) PCT64-35B Fab crystal structure with CDRH3 side chains as sticks. Heavy chain in beige, CDRH3 in orange, and light chain in purple. (B) Overall organization of the variable region of PCT64-35B Fab; secondary structure rendering of CDRs and framework (FR) regions. (C) Superposition of PCT64-35B Fab variable region (blue) and PGT145 variable region (gray), showing differential CDRH3 orientations. (D) Logograms of CDRH2 and CDRH3 residues (Kabat numbering) for PCT64 mAbs segregated in two groups based on acquisition of heterologous breadth. See also Figures S1B and S3.
Figure 4. Longitudinal Full Length Env Next…
Figure 4. Longitudinal Full Length Env Next Generation Sequencing Allows a Comprehensive Analysis of Env Evolution and Escape from PCT64 Abs
HIV subtype A env phylogeny from donor PC64, colored by sample date, estimated by maximum likelihood from full-length envPacBio high-quality consensus sequences (HQCSs) (with bubbles representing sample proportion) (http://test.datamonkey.org/flea-demo/PC64_kinetics/) and clonal Sanger sequences (Monogram Biosciences Lab Corp) (indicated by dashed lines). Data are representative of one experiment. See also Figure S4.
Figure 5. Analysis of Env Escape from…
Figure 5. Analysis of Env Escape from PCT64 Abs Reveals that Localized Diversity at Key V2 Epitope Residues Drove bnAb Maturation toward Breadth
(A) Left panel: V1/V2 amino acid sequences of PC64 Env cloned at various time points, aligned vertically. The number of clones is indicated and amino-acid identity is color-coded. Middle panel: Logograms of Env C-strand residues 160–171 for each time point. Net charge of the 160–171 peptide for each clone is color-coded in shades of blue (from 0 to 1). Right panel: Autologous neutralization of each pseudotyped Env clone by PCT64 mAbs (grouped and colored by isolation time point). The neutralization IC50 (μg/mL) is tabulated and color-colored as indicated in the legend. The development of the PCT64 bnAb lineage and V1V2-directed heterologous plasma neutralization are indicated on the right. (B) Evolution of PCT64 mAb neutralization against autologous pseudotyped virus mutants representing various Env immunotypes at residues 160, 166, 167, 169, 181 at several time points. Residues mutated from WT are shaded in pink. The neutralization IC50 (μg/mL) is tabulated and colored as indicated. Gray cells represent the absence of neutralization. (C) Frequency of PC64 Env immunotypes (combination of residues 160, 166, 167, 169, 181) at several time points, with viral load in plasma (black). Neutralization potency of PCT64 mAbs presented in (B) against these immunotypes is shown as shades of blue, green, orange, and red for PCT64 mAbs isolated at 13, 18, 24, and 35 mpi, respectively. Antibody icons are colored by mpi, with the frequency of PCT64 HC sequences found in the periphery (Figure 2A) shown by icon size. GL: Germline. Development of V2 apex-directed heterologous plasma neutralization (Figure 1A) is indicated in black above. Data are representative of at least two independent experiments. See also Figures S5 and S6.
Figure 6. Env Glycoform Heterogeneity Played a…
Figure 6. Env Glycoform Heterogeneity Played a Role in Elicitation of the PCT64 bnAb Lineage
(A) Autologous neutralization of the MRCA pseudotyped virus by titrated amounts of PCT64 mAbs. (B) Autologous neutralization of PC64 Env clones by PCT64-infGL and -LMCA mAbs. (C) Autologous neutralization of MRCA pseudotyped virus by titrated amounts of WT and mutant PCT64 mAbs. PCT64-LMCA (top) and PCT64-13C (bottom) heavy chain mutants paired with PCT64-infGL WT, PCT64-infGL-CDRL3 mutant(+SAR) or PCT64-13C light chains. (D) Top and side views of negative-stain EM 3D reconstructions (see Figure S7C) of PC64 unliganded (left) or PGV04-bound (right) SOSIPs produced in 293-F cells. Atomic models of BG505 (gp120 in blue, gp41 in yellow: PBD 5CEZ) in complex with PGV04 (red) (PDB: 3J5M) and with Fabs removed were docked into EM density maps. (E) Cryo-EM reconstruction of PC64-M4c054 SOSIP with autologous PCT64-13C Fab. Side and top view of the EM density map showing PCT64-13C Fab bound to the V1/V2 apex. The final resolution is ~13Å (Figure S7D). (F) BLI curves of indicated antibodies immobilized on anti-human IgG Fc sensors and indicated PC64 SOSIP trimers (1 μM) in solution as analytes. PC64 SOSIP trimers were produced in the presence of furin in 293F (top panels; normal glycan processing), or 293S (bottom panels;GnTI−/− with no hybrid/complex glycan processing leading to enrichment in Man5GlcNAc glycans). Data are representative of at least two independent experiments. See also Figure S7.

References

    1. Adams PD, Afonine PV, Bunkóczi G, Chen VB, Davis IW, Echols N, Headd JJ, Hung LW, Kapral GJ, Grosse-Kunstleve RW, et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr D Biol Crystallogr. 2010;66:213–221.
    1. Andrabi R, Voss JE, Liang CH, Briney B, McCoy LE, Wu CY, Wong CH, Poignard P, Burton DR. Identification of common features in prototype broadly neutralizing antibodies to HIV envelope V2 apex to facilitate vaccine design. Immunity. 2015;43:959–973.
    1. Behrens AJ, Vasiljevic S, Pritchard LK, Harvey DJ, Andev RS, Krumm SA, Struwe WB, Cupo A, Kumar A, Zitzmann N, et al. Composition and antigenic effects of individual glycan sites of a trimeric hiv-1 envelope glycoprotein. Cell Rep. 2016;14:2695–2706.
    1. Bhiman JN, Anthony C, Doria-Rose NA, Karimanzira O, Schramm CA, Khoza T, Kitchin D, Botha G, Gorman J, Garrett NJ, et al. Viral variants that initiate and drive maturation of V1V2-directed HIV-1 broadly neutralizing antibodies. Nat Med. 2015;21:1332–1336.
    1. Bonsignori M, Montefiori DC, Wu X, Chen X, Hwang KK, Tsao CY, Kozink DM, Parks RJ, Tomaras GD, Crump JA, et al. Two distinct broadly neutralizing antibody specificities of different clonal lineages in a single HIV-1-infected donor: implications for vaccine design. J Virol. 2012;86:4688–4692.
    1. Bonsignori M, Zhou T, Sheng Z, Chen L, Gao F, Joyce MG, Ozorowski G, Chuang GY, Schramm CA, Wiehe K, et al. NISC Comparative Sequencing Program. Maturation pathway from germline to broad hiv-1 neutralizer of a CD4-mimic antibody. Cell. 2016;165:449–463.
    1. Bonsignori M, Kreider EF, Fera D, Meyerhoff RR, Bradley T, Wiehe K, Alam SM, Aussedat B, Walkowicz WE, Hwang K-K, et al. Staged induction of HIV-1 glycan-dependent broadly neutralizing antibodies. Sci Transl Med. 2017;9:eaai7514.
    1. Briney BS, Willis JR, Crowe JE., Jr Human peripheral blood antibodies with long HCDR3s are established primarily at original recombination using a limited subset of germline genes. PLoS ONE. 2012;7:e36750.
    1. Briney B, Le K, Zhu J, Burton DR. Clonify: unseeded antibody lineage assignment from next-generation sequencing data. Sci Rep. 2016a;6:23901.
    1. Briney B, Sok D, Jardine JG, Kulp DW, Skog P, Menis S, Jacak R, Kalyuzhniy O, de Val N, Sesterhenn F, et al. Tailored immunogens direct affinity maturation toward hiv neutralizing antibodies. Cell. 2016b;166:1459–1470. e11.
    1. Burton DR, Hangartner L. Broadly neutralizing antibodies to HIV and their role in vaccine design. Annu Rev Immunol. 2016;34:635–659.
    1. Burton DR, Ahmed R, Barouch DH, Butera ST, Crotty S, Godzik A, Kaufmann DE, McElrath MJ, Nussenzweig MC, Pulendran B, et al. A blueprint for HIV vaccine discovery. Cell Host Microbe. 2012;12:396–407.
    1. Cale EM, Gorman J, Radakovich NA, Crooks ET, Osawa K, Tong T, Li J, Nagarajan R, Ozorowski G, Ambrozak DR, et al. Virus-like particles identify an HIV v1v2 apex-binding neutralizing antibody that lacks a protruding loop. Immunity. 2017;46:777–791. e10.
    1. Cao L, Diedrich JK, Kulp DW, Pauthner M, He L, Park SR, Sok D, Su CY, Delahunty CM, Menis S, et al. Global site-specific N-glycosylation analysis of HIV envelope glycoprotein. Nat Commun. 2017;8:14954.
    1. Doores KJ, Burton DR. Variable loop glycan dependency of the broad and potent HIV-1-neutralizing antibodies PG9 and PG16. J Virol. 2010;84:10510–10521.
    1. Doria-Rose NA, Klein RM, Daniels MG, O’Dell S, Nason M, Lapedes A, Bhattacharya T, Migueles SA, Wyatt RT, Korber BT, et al. Breadth of human immunodeficiency virus-specific neutralizing activity in sera: clustering analysis and association with clinical variables. J Virol. 2010;84:1631–1636.
    1. Doria-Rose NA, Schramm CA, Gorman J, Moore PL, Bhiman JN, DeKosky BJ, Ernandes MJ, Georgiev IS, Kim HJ, Pancera M, et al. NISC Comparative Sequencing Program. Developmental pathway for potent V1V2-directed HIV-neutralizing antibodies. Nature. 2014;509:55–62.
    1. Doria-Rose NA, Bhiman JN, Roark RS, Schramm CA, Gorman J, Chuang GY, Pancera M, Cale EM, Ernandes MJ, Louder MK, et al. New member of the V1V2-directed CAP256-VRC26 lineage that shows increased breadth and exceptional potency. J Virol. 2015;90:76–91.
    1. Doria-Rose NA, Altae-Tran HR, Roark RS, Schmidt SD, Sutton MS, Louder MK, Chuang GY, Bailer RT, Cortez V, Kong R, et al. Mapping polyclonal HIV-1 Antibody responses via next-generation neutralization fingerprinting. PLoS Pathog. 2017;13:e1006148.
    1. Dosenovic P, von Boehmer L, Escolano A, Jardine J, Freund NT, Gitlin AD, McGuire AT, Kulp DW, Oliveira T, Scharf L, et al. Immunization for HIV-1 broadly neutralizing antibodies in human Ig knockin mice. Cell. 2015;161:1505–1515.
    1. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010;26:2460–2461.
    1. Emsley P, Cowtan K. Coot: model-building tools for molecular graphics. Acta Crystallogr D Biol Crystallogr. 2004;60:2126–2132.
    1. Escolano A, Steichen JM, Dosenovic P, Kulp DW, Golijanin J, Sok D, Freund NT, Gitlin AD, Oliveira T, Araki T, et al. Sequential immunization elicits broadly neutralizing anti-hiv-1 antibodies in Ig knockin mice. Cell. 2016;166:1445–1458. e12.
    1. Fauci AS, Marston HD. Ending AIDS–is an HIV vaccine necessary? N Engl J Med. 2014;370:495–498.
    1. Gao F, Bonsignori M, Liao HX, Kumar A, Xia SM, Lu X, Cai F, Hwang KK, Song H, Zhou T, et al. Cooperation of B cell lineages in induction of HIV-1-broadly neutralizing antibodies. Cell. 2014;158:481–491.
    1. Gorman J, Soto C, Yang MM, Davenport TM, Guttman M, Bailer RT, Chambers M, Chuang GY, DeKosky BJ, Doria-Rose NA, et al. NISC Comparative Sequencing Program. Structures of HIV-1 Env V1V2 with broadly neutralizing antibodies reveal commonalities that enable vaccine design. Nat Struct Mol Biol. 2016;23:81–90.
    1. Gray ES, Moore PL, Choge IA, Decker JM, Bibollet-Ruche F, Li H, Leseka N, Treurnicht F, Mlisana K, Shaw GM, et al. CAPRISA 002 Study Team. Neutralizing antibody responses in acute human immunodeficiency virus type 1 subtype C infection. J Virol. 2007;81:6187–6196.
    1. Guenaga J, Dubrovskaya V, de Val N, Sharma SK, Carrette B, Ward AB, Wyatt RT. Structure-guided redesign increases the propensity of hiv env to generate highly stable soluble trimers. J Virol. 2015;90:2806–2817.
    1. Hraber P, Seaman MS, Bailer RT, Mascola JR, Montefiori DC, Korber BT. Prevalence of broadly neutralizing antibody responses during chronic HIV-1 infection. AIDS. 2014;28:163–169.
    1. Huerta-Cepas J, Dopazo J, Gabaldón T. ETE: a Python environment for tree exploration. BMC Bioinformatics. 2010;11:24.
    1. Jardine JG, Kulp DW, Havenar-Daughton C, Sarkar A, Briney B, Sok D, Sesterhenn F, Ereño-Orbea J, Kalyuzhniy O, Deresa I, et al. HIV-1 broadly neutralizing antibody precursor B cells revealed by germline-targeting immunogen. Science. 2016;351:1458–1463.
    1. Julien JP, Cupo A, Sok D, Stanfield RL, Lyumkis D, Deller MC, Klasse PJ, Burton DR, Sanders RW, Moore JP, et al. Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science. 2013;342:1477–1483.
    1. Karplus PA, Diederichs K. Linking crystallographic model and data quality. Science. 2012;336:1030–1033.
    1. Katoh K, Standley DM. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol. 2013;30:772–780.
    1. Katoh K, Kuma K, Toh H, Miyata T. MAFFT version 5: improvement in accuracy of multiple sequence alignment. Nucleic Acids Res. 2005;33:511–518.
    1. Kimanius D, Forsberg BO, Scheres SH, Lindahl E. Accelerated cryo-EM structure determination with parallelisation using GPUs in RELION-2. eLife. 2016;5:e18722.
    1. Klein F, Diskin R, Scheid JF, Gaebler C, Mouquet H, Georgiev IS, Pancera M, Zhou T, Incesu RB, Fu BZ, et al. Somatic mutations of the immunoglobulin framework are generally required for broad and potent HIV-1 neutralization. Cell. 2013;153:126–138.
    1. Kong R, Xu K, Zhou T, Acharya P, Lemmin T, Liu K, Ozorowski G, Soto C, Taft JD, Bailer RT, et al. Fusion peptide of HIV-1 as a site of vulnerability to neutralizing antibody. Science. 2016;352:828–833.
    1. Laird Smith M, Murrell B, Eren K, Ignacio C, Landais E, Weaver S, Phung P, Ludka C, Hepler L, Caballero G, et al. The IAVI Protocol C Investigators & The IAVI African HIV Research Network. Rapid sequencing of complete env genes from primary HIV-1 samples. Virus Evol. 2016;2:vew018.
    1. Landais E, Huang X, Havenar-Daughton C, Murrell B, Price MA, Wickramasinghe L, Ramos A, Bian CB, Simek M, Allen S, et al. Broadly neutralizing antibody responses in a large longitudinal sub-Saharan HIV primary infection cohort. PLoS Pathog. 2016;12:e1005369.
    1. Lee JH, de Val N, Lyumkis D, Ward AB. Model building and refinement of a natively glycosylated HIV-1 Env protein by high-resolution cryoelectron microscopy. Structure. 2015;23:1943–1951.
    1. Liao HX, Lynch R, Zhou T, Gao F, Alam SM, Boyd SD, Fire AZ, Roskin KM, Schramm CA, Zhang Z, et al. NISC Comparative Sequencing Program. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature. 2013;496:469–476.
    1. MacLeod DT, Choi NM, Briney B, Garces F, Ver LS, Landais E, Murrell B, Wrin T, Kilembe W, Liang CH, et al. IAVI Protocol C Investigators & The IAVI African HIV Research Network. Early antibody lineage diversification and independent limb maturation lead to broad HIV-1 neutralization targeting the Env high-mannose patch. Immunity. 2016;44:1215–1226.
    1. Masella AP, Bartram AK, Truszkowski JM, Brown DG, Neufeld JD. PANDAseq: paired-end assembler for illumina sequences. BMC Bioinformatics. 2012;13:31.
    1. McCoy AJ, Grosse-Kunstleve RW, Adams PD, Winn MD, Storoni LC, Read RJ. Phaser crystallographic software. J Appl Cryst. 2007;40:658–674.
    1. McLellan JS, Pancera M, Carrico C, Gorman J, Julien JP, Khayat R, Louder R, Pejchal R, Sastry M, Dai K, et al. Structure of HIV-1 gp120 V1/V2 domain with broadly neutralizing antibody PG9. Nature. 2011;480:336–343.
    1. Otwinowski Z, Minor W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 1997;276:307–326.
    1. Pan Z, Liu Z, Cheng H, Wang Y, Gao T, Ullah S, Ren J, Xue Y. Systematic analysis of the in situ crosstalk of tyrosine modifications reveals no additional natural selection on multiply modified residues. Sci Rep. 2014;4:7331.
    1. Pancera M, McLellan JS, Wu X, Zhu J, Changela A, Schmidt SD, Yang Y, Zhou T, Phogat S, Mascola JR, Kwong PD. Crystal structure of PG16 and chimeric dissection with somatically related PG9: structure-function analysis of two quaternary-specific antibodies that effectively neutralize HIV-1. J Virol. 2010;84:8098–8110.
    1. Panico M, Bouché L, Binet D, O’Connor MJ, Rahman D, Pang PC, Canis K, North SJ, Desrosiers RC, Chertova E, et al. Mapping the complete glycoproteome of virion-derived HIV-1 gp120 provides insights into broadly neutralizing antibody binding. Sci Rep. 2016;6:32956.
    1. Pejchal R, Walker LM, Stanfield RL, Phogat SK, Koff WC, Poignard P, Burton DR, Wilson IA. Structure and function of broadly reactive antibody PG16 reveal an H3 subdomain that mediates potent neutralization of HIV-1. Proc Natl Acad Sci USA. 2010;107:11483–11488.
    1. Pejchal R, Doores KJ, Walker LM, Khayat R, Huang PS, Wang SK, Stanfield RL, Julien JP, Ramos A, Crispin M, et al. A potent and broad neutralizing antibody recognizes and penetrates the HIV glycan shield. Science. 2011;334:1097–1103.
    1. Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, Ferrin TE. UCSF Chimera–a visualization system for exploratory research and analysis. J Comput Chem. 2004;25:1605–1612.
    1. Potter CS, Chu H, Frey B, Green C, Kisseberth N, Madden TJ, Miller KL, Nahrstedt K, Pulokas J, Reilein A, et al. Leginon: a system for fully automated acquisition of 1000 electron micrographs a day. Ultramicroscopy. 1999;77:153–161.
    1. Price MN, Dehal PS, Arkin AP. FastTree 2–approximately maximum-likelihood trees for large alignments. PLoS ONE. 2010;5:e9490.
    1. Rusert P, Kouyos RD, Kadelka C, Ebner H, Schanz M, Huber M, Braun DL, Hozé N, Scherrer A, Magnus C, et al. Swiss HIV Cohort Study. Determinants of HIV-1 broadly neutralizing antibody induction. Nat Med. 2016;22:1260–1267.
    1. Sanders RW, Derking R, Cupo A, Julien JP, Yasmeen A, de Val N, Kim HJ, Blattner C, de la Peña AT, Korzun J, 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. Schweighardt B, Liu Y, Huang W, Chappey C, Lie YS, Petropoulos CJ, Wrin T. Development of an HIV-1 reference panel of subtype B envelope clones isolated from the plasma of recently infected individuals. J Acquir Immune Defic Syndr. 2007;46:1–11.
    1. Seaman MS, Janes H, Hawkins N, Grandpre LE, Devoy C, Giri A, Coffey RT, Harris L, Wood B, Daniels MG, et al. Tiered categorization of a diverse panel of HIV-1 Env pseudoviruses for assessment of neutralizing antibodies. J Virol. 2010;84:1439–1452.
    1. Sheng Z, Schramm CA, Connors M, Morris L, Mascola JR, Kwong PD, Shapiro L. Effects of darwinian selection and mutability on rate of broadly neutralizing antibody evolution during HIV-1 infection. PLoS Comput Biol. 2016;12:e1004940.
    1. Simek MD, Rida W, Priddy FH, Pung P, Carrow E, Laufer DS, Lehrman JK, Boaz M, Tarragona-Fiol T, Miiro G, et al. Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. J Virol. 2009;83:7337–7348.
    1. Sok D, Laserson U, Laserson J, Liu Y, Vigneault F, Julien JP, Briney B, Ramos A, Saye KF, Le K, et al. The effects of somatic hypermutation on neutralization and binding in the PGT121 family of broadly neutralizing HIV antibodies. PLoS Pathog. 2013;9:e1003754.
    1. Sok D, van Gils MJ, Pauthner M, Julien JP, Saye-Francisco KL, Hsueh J, Briney B, Lee JH, Le KM, Lee PS, et al. Recombinant HIV envelope trimer selects for quaternary-dependent antibodies targeting the trimer apex. Proc Natl Acad Sci USA. 2014;111:17624–17629.
    1. Steichen JM, Kulp DW, Tokatlian T, Escolano A, Dosenovic P, Stanfield RL, McCoy LE, Ozorowski G, Hu X, Kalyuzhniy O, et al. HIV vaccine design to target germline precursors of glycan-dependent broadly neutralizing antibodies. Immunity. 2016;45:483–496.
    1. Tian M, Cheng C, Chen X, Duan H, Cheng HL, Dao M, Sheng Z, Kimble M, Wang L, Lin S, et al. Induction of HIV neutralizing antibody lineages in mice with diverse precursor repertoires. Cell. 2016;166:1471–1484. e18.
    1. van Gils MJ, Euler Z, Schweighardt B, Wrin T, Schuitemaker H. Prevalence of cross-reactive HIV-1-neutralizing activity in HIV-1-infected patients with rapid or slow disease progression. AIDS. 2009;23:2405–2414.
    1. Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P, Goss JL, Wrin T, Simek MD, Fling S, Mitcham JL, et al. Protocol G Principal Investigators. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science. 2009;326:285–289.
    1. Walker LM, Huber M, Doores KJ, Falkowska E, Pejchal R, Julien JP, Wang SK, Ramos A, Chan-Hui PY, Moyle M, et al. Protocol G Principal Investigators. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature. 2011;477:466–470.
    1. Wardemann H, Yurasov S, Schaefer A, Young JW, Meffre E, Nussenzweig MC. Predominant autoantibody production by early human B cell precursors. Science. 2003;301:1374–1377.
    1. Zhang K. Gctf: Real-time CTF determination and correction. J Struct Biol. 2016;193:1–12.
    1. Zheng SQ, Palovcak E, Armache JP, Verba KA, Cheng Y, Agard DA. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat Methods. 2017;14:331–332.

Source: PubMed

3
Subskrybuj