Slow Delivery Immunization Enhances HIV Neutralizing Antibody and Germinal Center Responses via Modulation of Immunodominance

Kimberly M Cirelli, Diane G Carnathan, Bartek Nogal, Jacob T Martin, Oscar L Rodriguez, Amit A Upadhyay, Chiamaka A Enemuo, Etse H Gebru, Yury Choe, Federico Viviano, Catherine Nakao, Matthias G Pauthner, Samantha Reiss, Christopher A Cottrell, Melissa L Smith, Raiza Bastidas, William Gibson, Amber N Wolabaugh, Mariane B Melo, Benjamin Cossette, Venkatesh Kumar, Nirav B Patel, Talar Tokatlian, Sergey Menis, Daniel W Kulp, Dennis R Burton, Ben Murrell, William R Schief, Steven E Bosinger, Andrew B Ward, Corey T Watson, Guido Silvestri, Darrell J Irvine, Shane Crotty, Kimberly M Cirelli, Diane G Carnathan, Bartek Nogal, Jacob T Martin, Oscar L Rodriguez, Amit A Upadhyay, Chiamaka A Enemuo, Etse H Gebru, Yury Choe, Federico Viviano, Catherine Nakao, Matthias G Pauthner, Samantha Reiss, Christopher A Cottrell, Melissa L Smith, Raiza Bastidas, William Gibson, Amber N Wolabaugh, Mariane B Melo, Benjamin Cossette, Venkatesh Kumar, Nirav B Patel, Talar Tokatlian, Sergey Menis, Daniel W Kulp, Dennis R Burton, Ben Murrell, William R Schief, Steven E Bosinger, Andrew B Ward, Corey T Watson, Guido Silvestri, Darrell J Irvine, Shane Crotty

Abstract

Conventional immunization strategies will likely be insufficient for the development of a broadly neutralizing antibody (bnAb) vaccine for HIV or other difficult pathogens because of the immunological hurdles posed, including B cell immunodominance and germinal center (GC) quantity and quality. We found that two independent methods of slow delivery immunization of rhesus monkeys (RMs) resulted in more robust T follicular helper (TFH) cell responses and GC B cells with improved Env-binding, tracked by longitudinal fine needle aspirates. Improved GCs correlated with the development of >20-fold higher titers of autologous nAbs. Using a new RM genomic immunoglobulin locus reference, we identified differential IgV gene use between immunization modalities. Ab mapping demonstrated targeting of immunodominant non-neutralizing epitopes by conventional bolus-immunized animals, whereas slow delivery-immunized animals targeted a more diverse set of epitopes. Thus, alternative immunization strategies can enhance nAb development by altering GCs and modulating the immunodominance of non-neutralizing epitopes.

Keywords: FNA; GC- T(FH); HIV vaccine; affinity maturation; immune complexes; memory B cells; non-human primates; rhesus macaque genome; somatic hypermutation.

Conflict of interest statement

DECLARATION OF INTERESTS

The authors declare no competing interests.

Copyright © 2019 Elsevier Inc. All rights reserved.

Figures

Figure 1.. Sustained delivery immunization enhances B…
Figure 1.. Sustained delivery immunization enhances BGC cell responses.
(A) Immunization and sampling schedule of first immunization. Bolus Group 2 (Gp2), 2w osmotic pump (OP), and 4w OP RMs were immunized and sampled at the same time. Bolus Gp1 were immunized and sampled separately. Bolus Gp1&2 data have been pooled. (B) Representative BGC cell flow cytometry, gated on viable CD20+ B cells pre- and post-immunization. See Fig S1 for full gating strategy. (C) BGC cell frequencies over time. Black circles, pooled bolus Gp1&2; grey circles, bolus Gp2. (D) Cumulative BGC cell responses (AUC of C) to immunization within individual LNs at weeks 1, 3–7 [AUC]. (E) Representative flow cytometry of Env trimer-specific B cells pre- and post-immunization. (F) Env-specific B cell frequencies over time. (G) Cumulative Env trimer-specific B cells (AUC of F) within each LN. (H) Representative flow cytometry Env trimer-specific BGC cells pre- and post-immunization. (I) Env trimer-specific BGC cell frequencies over time. (J) Cumulative Env trimer-specific BGC cells (AUC of I) within each LN. (K) Env trimer-specific BGC cells over time. (L) Cumulative Env trimer-specific BGC cells (AUC of K) within each LN. (M) Frequencies of high-affinity Env trimer-specific BGC cells over time. (N) Cumulative high-affinity Env trimer-specific BGC cells (AUC of M) within each LN. (O) High-affinity Env trimer-specific BMem cells over time. (P) Cumulative high-affinity Env trimer-specific BMem cell responses within individual LNs. (Q) Representative flow cytometry of GC-TFH cells, gated on CD4+ T cells. See Fig S2 for full gating strategy. (R) GC-TFH cells over time. (S) Cumulative GC-TFH cell response between w1 plus w3-6 [AUC]. Mean ± SEM are graphed. Statistical significance tested using unpaired, two-tailed Mann-Whitney U tests. *p≤0.05, **p≤0.01. ***p≤0.001, ****p≤0.0001 See also Figures S1–2 and Data S1.
Figure 2.. Germinal center responses following 2…
Figure 2.. Germinal center responses following 2nd Env trimer immunization.
(A) Immunization and sampling schedule of 2nd immunization. Groups were immunized and sampled contemporaneously. (B) Frequencies of total BGC cells over time. (C) Env trimer-specific B cell frequencies over time. (D-E) Env trimer-specific BGC cells over time. (F) High-affinity Env trimer-specific BGC cells over time. (G) High-affinity Env trimer-specific BMem cells over time. (H) GC-TFH cell frequencies after 2nd immunization. (I) Representative flow cytometry of Env-specific CD4+ T cells from LNs. Cells were left unstimulated or stimulated with a peptide pool spanning Olio6CD4ko. (J) Env-specific CD4+ T cells at w12 (bolus) or w14 (OP). (K) Representative flow cytometry of GC-TFH, mantle (m)TFH and non-TFH cell subsets. (L) Flow cytometry of AIMOB assay (OX40+4-1BB+), gated on GC-TFH, mTFH or non-TFH cells. (M) Quantification of Env-specific CD4+ T cells by subset. Mean ± SEM are graphed. Statistical significance tested using unpaired, two-tailed Mann-Whitney U test. *p≤ 0.05, **p≤0.01, ***p≤0.00. See also Figure S2.
Figure 3.. Sustained delivery immunization induces higher…
Figure 3.. Sustained delivery immunization induces higher nAb titers
(A) BG505 Env trimer IgG binding endpoint titers over time. (B) His IgG binding endpoint titers over time. (C) BG505 (autologous) nAb titers over time. (D) Peak BG505 nAb titers after two immunizations. (E) Neutralization breadth on a 12-virus panel. GMT and ± geometric SD. Statistical significance was tested using unpaired, two-tailed Mann-Whitney U test. *p≤0.05. See also Figure S2.
Figure 4.. Immunoglobulin gene germline annotations using…
Figure 4.. Immunoglobulin gene germline annotations using long-read genomic DNA sequencing
(A) Locus and assembly summaries for the RM Ig loci. (B) A representative region where PacBio primary contigs resolved gaps in the current RM reference genome. PacBio reads span these gaps (inset). (C) Overview of V gene allelic variant discovery process. Reads overlapping annotations on primary contigs were assessed for the presence of SNPs, which were used to partition reads for allele-specific assemblies. (D) SNPs (e.g., green and red) within or near genes (red boxes) were used to partition reads to each respective haplotype, allowing for the identification of heterozygous (pink) and homozygous (grey) gene segments. (E) Primary and alternate contig alleles. (F) Variable (V) genes from PacBio assembly that were present in IMGT or NCBI V gene repositories. (G) V gene counts from PacBio primary contig assemblies, compared to human gene loci. (H) Quantification of Env-specific B cells lineages from individual LNs. (I) Phylogenetic analysis of a lineage found in both LNs in one animal. Blue, left LN; Red, right LN. Dot size represents number of reads with that sequence. (J) Lineages shared between R and L LNs within an animal. (K) Mutation frequencies in IGHV, IGLV, or IGKV. Violin plots; Dash = mean. Mean ± SEM; statistical significance in H and J tested using unpaired, two-tailed Mann-Whitney U test. Significance in K tested using Student’s t-test. *p≤ 0.05, **p≤0.01, ***p≤0.001 See also Figure S3 and Tables S1–2.
Figure 5.. Slow delivery immunization shifts immunodominance.
Figure 5.. Slow delivery immunization shifts immunodominance.
(A) IGHV or (B) IGLV use by antigen-specific B cells within a LN. Each data point is single LN. Statistical significance tested using multiple t-tests with FDR = 5%; ****q+ lineage. Blue, left LN; Red, right LN. Dot size represents number of reads with that sequence. (D) The base of Env is hidden on the virion surface. Soluble trimer allows access of the base to B cells. Glycans restrict access to the main Env trimer surface. (E) Binding curves of mAbs isolated from w7 to BG505 Env trimer. (F) Cross-competition ELISA assay. Data is representative of two experiments, each performed in duplicate. (G) 3D EM reconstruction of BDA1 Fab (blue) in complex with BG505 Env trimer. (H) Binding curves of BDA1 and related mutants to BG505 Env trimer. BDA12 has a single w12 mutation in L-CDR3. BDA13 contains this mutation and three additional w12 mutations in L-CDR2. Data is representative of two experiments, each performed in duplicate. (I) Composite 3D reconstruction of Env trimer bound to Fabs isolated from all animals as determined by polyclonal EM analysis. Numbers of individuals with Fab that binds region are listed. Base (purple), glycan hole-I (light blue), C3/V5 (dark blue), fusion peptide (orange), V1/V3 apex (green). Apex specific Fab is transparent to represent rarity. Mean ± SEM. See also Figures S4–6 and Table S3.
Figure 6.. Dose escalating immunization strategy results…
Figure 6.. Dose escalating immunization strategy results in higher nAb titers.
(A) Immunization and sampling schedule. Groups were immunized and sampled contemporaneously. (B) Total BGC frequencies over time. Data from bolus Gp2 (Fig 1) are included in these analyses (grey circles). (C) Cumulative BGC cell response to the first immunization, calculated between w3-7 [AUC]. (D) Env trimer-specific BGC cells frequencies over time. (E) Cumulative Env trimer-specific BGC cell responses to one immunization. (F) Total GC-TFH cell frequencies over time. (G) Cumulative GC-TFH responses to one immunization (AUC of F). (H) Env-specific CD4+ responses after one immunization. (I) Ratio of Env+ BGC to Env-specific GC-TFH cells at w5, calculated as Env+ BGC (% B cells)/ Env-specific GC-TFH (% CD4+). (J) BG505 Env trimer binding IgG titers over time. (K) Autologous BG505 nAb titers over time. (L) Peak BG505 nAb titers after three immunizations. (M) Composite 3D reconstruction of Env trimer bound to Fabs isolated from all animals after two immunizations. 3D EM reconstructions from individual animals can be seen in Figure S8A. (N) Correlation between peak GC-TFH and BGC cell % during 1st immunization from both studies. (O) Correlation between Env+ BGC cells (% B cells) and peak neutralization titers. Env+ BGC cell values are from w7 or peak frequencies during 1st immunization. Peak nAb titers are after 2nd immunization. Serological data represent GMT ± geometric SD. Cell-frequency data represent mean ± SEM. Statistical significance was tested using unpaired, two-tailed Mann-Whitney U tests. *p

Figure 7.. Slow delivery immunization results in…

Figure 7.. Slow delivery immunization results in enhanced antigen retention in LNs.

(A) Quantitation of…

Figure 7.. Slow delivery immunization results in enhanced antigen retention in LNs.
(A) Quantitation of fluorescent Env in draining inguinal and iliac and non-draining axillary LNs. Mean ± SEM; statistical significance was tested using two-way ANOVA. *adjusted p FH cells and activation and recruitment of a diverse set of B cells. Greater GC-TFH help supports a wider repertoire of B cells, which is more likely to contain nAb precursors, later in the response (w3–7). Antigen delivered via conventional bolus immunization can be subject to degradative processes and nonnative forms of antigen can be presented by FDCs late in the response, while OPs protect the antigen prior to release. Immune complex (IC) formation is enhanced by slow delivery immunization. See also Figure S8 and Movie S1.
All figures (7)
Figure 7.. Slow delivery immunization results in…
Figure 7.. Slow delivery immunization results in enhanced antigen retention in LNs.
(A) Quantitation of fluorescent Env in draining inguinal and iliac and non-draining axillary LNs. Mean ± SEM; statistical significance was tested using two-way ANOVA. *adjusted p FH cells and activation and recruitment of a diverse set of B cells. Greater GC-TFH help supports a wider repertoire of B cells, which is more likely to contain nAb precursors, later in the response (w3–7). Antigen delivered via conventional bolus immunization can be subject to degradative processes and nonnative forms of antigen can be presented by FDCs late in the response, while OPs protect the antigen prior to release. Immune complex (IC) formation is enhanced by slow delivery immunization. See also Figure S8 and Movie S1.

References

    1. Abbott RK, Lee JH, Menis S, Skog P, Rossi M, Ota T, Kulp DW, Bhullar D, Kalyuzhniy O, Havenar-Daughton C, et al. (2018). Precursor Frequency and Affinity Determine B Cell Competitive Fitness in Germinal Centers, Tested with Germline-Targeting HIV Vaccine Immunogens. Immunity 48, 133–146.e136.
    1. Alkan C, Sajjadian S, and Eichler EE (2011). Limitations of next-generation genome sequence assembly. Nat. Methods 8, 61–65.
    1. Andrew S (2010). FastQC: A quality control tool for high throughput sequence data. 2010.
    1. Andrews SF, Graham BS, Mascola JR, and McDermott AB (2018). Is It Possible to Develop a “Universal” Influenza Virus Vaccine? Immunogenetic Considerations Underlying B-Cell Biology in he Development of a Pan-Subtype Influenza A Vaccine Targeting the Hemagglutinin Stem. Cold Spring Harb Perspect Biol 10, a029413.
    1. Angeletti D, and Yewdell JW (2018). Understanding and Manipulating Viral Immunity: Antibody Immunodominance Enters Center Stage. Trends Immunol 39, 549–561.
    1. Angeletti D, Gibbs JS, Angel M, Kosik I, Hickman HD, Frank GM, Das SR, Wheatley AK, Prabhakaran M, Leggat DJ, et al. (2017). Defining B cell immunodominance to viruses. Nat. Immunol 18, 456–463.
    1. Bianchi M, Turner HL, Nogal B, Cottrell CA, Oyen D, Pauthner M, Bastidas R, Nedellec R, McCoy LE, Wilson IA, et al. (2018). Electron-Microscopy-Based Epitope Mapping Defines Specificities of Polyclonal Antibodies Elicited during HIV-1 BG505 Envelope Trimer Immunization. Immunity 49, 288–300.e288.
    1. Bolger AM, Lohse M, and Usadel B (2014). Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120.
    1. Burton DR, and Hangartner L (2016). Broadly Neutralizing Antibodies to HIV and Their Role in Vaccine Design. Annu. Rev. Immunol 34, 635–659.
    1. Chaisson MJ, and Tesler G (2012). Mapping single molecule sequencing reads using basic local alignment with successive refinement (BLASR): application and theory. BMC Bioinformatics 13, 238.
    1. Chin C-S, Peluso P, Sedlazeck FJ, Nattestad M, Concepcion GT, Clum A, Dunn C, O’Malley R, Figueroa-Balderas R, Morales-Cruz A, et al. (2016). Phased diploid genome assembly with single-molecule real-time sequencing. Nat. Methods 13, 1050–1054.
    1. Chowdhury A, Del Rio PME, Tharp GK, Trible RP, Amara RR, Chahroudi A, Reyes-Teran G, Bosinger SE, and Silvestri G (2015). Decreased T Follicular Regulatory Cell/T Follicular Helper Cell (TFH) in Simian Immunodeficiency Virus-Infected Rhesus Macaques May Contribute to Accumulation of TFH in Chronic Infection. J. Immunol 195, 3237–3247.
    1. Cirelli KM, and Crotty S (2017). Germinal center enhancement by extended antigen availability. Curr. Opin. Immunol 47, 64–69.
    1. Corcoran MM, Phad GE, Vázquez Bernat N, Stahl-Hennig C, Sumida N, Persson MAA, Martin M, and Karlsson Hedestam GB (2016). Production of individualized V gene databases reveals high levels of immunoglobulin genetic diversity. Nat Commun 7, 13642.
    1. Crotty S (2014). T follicular helper cell differentiation, function, and roles in disease. Immunity 41, 529–542.
    1. Dan JM, Lindestam Arlehamn CS, Weiskopf D, da Silva Antunes R, Havenar-Daughton C, Reiss SM, Brigger M, Bothwell M, Sette A, and Crotty S (2016). A Cytokine-Independent Approach To Identify Antigen-Specific Human Germinal Center T Follicular Helper Cells and Rare Antigen-Specific CD4+ T Cells in Blood. J. Immunol 197, 983–993.
    1. deCamp A, Hraber P, Bailer RT, Seaman MS, Ochsenbauer C, Kappes J, Gottardo R, Edlefsen P, Self S, Tang H, et al. (2014). Global Panel of HIV-1 Env Reference Strains for Standardized Assessments of Vaccine-Elicited Neutralizing Antibodies. J. Virol 88, 2489–2507.
    1. DeMuth PC, Garcia-Beltran WF, Ai-Ling ML, Hammond PT, and Irvine DJ (2013). Composite dissolving microneedles for coordinated control of antigen and adjuvant delivery kinetics in transcutaneous vaccination. Adv Funct Mater 23, 161–172.
    1. DeMuth PC, Min Y, Irvine DJ, and Hammond PT (2014). Implantable silk composite microneedles for programmable vaccine release kinetics and enhanced immunogenicity in transcutaneous immunization. Adv Healthc Mater 3, 47–58.
    1. Edgar RC (2004). MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res 32, 1792–1797.
    1. Ehrenmann F, and Lefranc M-P (2011). IMGT/DomainGapAlign: IMGT standardized analysis of mino acid sequences of variable, constant, and groove domains (IG, TR, MH, IgSF, MhSF). Cold Spring Harb Protoc 2011, 737–749.
    1. Ehrenmann F, Kaas Q, and Lefranc M-P (2010). IMGT/3DstructureADB and IMGT/DomainGapAlign: a database and a tool for immunoglobulins or antibodies, T cell receptors, MHC, IgSF and MhcSF. Nucleic Acids Res 38, D301–D307.
    1. Feng Y, Tran K, Bale S, Kumar S, Guenaga J, Wilson R, de Val N, Arendt H, DeStefano J, Ward AB, et al. (2016). Thermostability of Well-Ordered HIV Spikes Correlates with the Elicitation of Autologous Tier 2 Neutralizing Antibodies. PLoS Pathog 12, e1005767.
    1. Francica JR, Sheng Z, Zhang Z, Nishimura Y, Shingai M, Ramesh A, Keele BF, Schmidt SD, Flynn BJ, Darko S, et al. (2015). Analysis of immunoglobulin transcripts and hypermutation following SHIV(AD8) infection and protein-plus-adjuvant immunization. Nat Commun 6, 6565.
    1. Fu L, Niu B, Zhu Z, Wu S, and Li W (2012). CDAHIT: accelerated for clustering the next-generation sequencing data. Bioinformatics 28, 3150–3152.
    1. Gibbs RA, Rogers J, Katze MG, Bumgarner R, Weinstock GM, Mardis ER, Remington KA, Strausberg RL, Venter JC, Wilson RK, et al. (2007). Evolutionary and biomedical insights from the rhesus macaque genome. Science 316, 222–234.
    1. Gitlin AD, Mayer CT, Oliveira TY, Shulman Z, Jones MJK, Koren A, and Nussenzweig MC (2015). HUMORAL IMMUNITY. T cell help controls the speed of the cell cycle in germinal center B cells. Science 349, 643–646.
    1. Gitlin AD, Shulman Z, and Nussenzweig MC (2014). Clonal selection in the germinal centre by regulated proliferation and hypermutation. Nature 509, 637–640.
    1. Grabherr MG, Haas BJ, Yassour M, Levin JZ, Thompson DA, Amit I, Adiconis X, Fan L, Raychowdhury R, Zeng Q, et al. (2011). Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat. Biotechnol 29, 644–652.
    1. Gupta NT, Vander Heiden JA, Uduman M, Gadala-Maria D, Yaari G, and Kleinstein SH (2015). Change-O: a toolkit for analyzing large-scale B cell immunoglobulin repertoire sequencing data. Bioinformatics 31, 3356–3358.
    1. Havenar-Daughton C, Carnathan DG, Torrents de la Peña A, Pauthner M, Briney B, Reiss SM, Wood JS, Kaushik K, van Gils MJ, Rosales SL, et al. (2016a). Direct Probing of Germinal Center Responses Reveals Immunological Features and Bottlenecks for Neutralizing Antibody Responses to HIV Env Trimer. Cell Rep 17, 2195–2209.
    1. Havenar-Daughton C, Lee JH, and Crotty S (2017). Tfh cells and HIV bnAbs, an immunodominance model of the HIV neutralizing antibody generation problem. Immunological Reviews 275, 49–61.
    1. Havenar-Daughton C, Reiss SM, Carnathan DG, Wu JE, Kendric K, Torrents de la Peña A, Kasturi SP, Dan JM, Bothwell M, Sanders RW, et al. (2016b). Cytokine-Independent Detection of Antigen-Specific Germinal Center T Follicular Helper Cells in Immunized Nonhuman Primates Using a Live Cell Activation-Induced Marker Technique. J. Immunol 197, 994–1002.
    1. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, et al. (2012). Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N. Engl. J. Med 366, 1275–1286.
    1. Hogenesch H (2002). Mechanisms of stimulation of the immune response by aluminum adjuvants. Vaccine 20, S34–S39.
    1. Hogenesch H (2012). Mechanism of immunopotentiation and safety of aluminum adjuvants. Front Immunol 3, 406.
    1. Hu JK, Crampton JC, Cupo A, Ketas T, van Gils MJ, Sliepen K, de Taeye SW, Sok D, Ozorowski G, Deresa I, et al. (2015). Murine Antibody Responses to Cleaved Soluble HIV-1 Envelope Trimers Are Highly Restricted in Specificity. J. Virol 89, 10383–10398.
    1. Huang J, Kang BH, Ishida E, Zhou T, Griesman T, Sheng Z, Wu F, Doria-Rose NA, Zhang B, McKee K, et al. (2016). Identification of a CD4-Binding-Site Antibody to HIV that Evolved Near-Pan Neutralization Breadth. Immunity 45, 1108–1121.
    1. Hutchison S, Benson RA, Gibson VB, Pollock AH, Garside P, and Brewer JM (2012). Antigen depot is not required for alum adjuvanticity. Faseb J 26, 1272–1279.
    1. Julien J-P, Cupo A, Sok D, Stanfield RL, Lyumkis D, Deller MC, Klasse PJ, Burton DR, Sanders RW, Moore JP, et al. (2013). Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science 342, 1477–1483.
    1. Katoh K, and Standley DM (2013). MAFFT Multiple Sequence Alignment Software Version 7: Improvements in Performance and Usability. Molecular Biology and Evolution 30, 772–780.
    1. Kent WJ (2002). BLAT--the BLAST-like alignment tool. Genome Res 12, 656–664.
    1. Klasse PJ, Ketas TJ, Cottrell CA, Ozorowski G, Debnath G, Camara D, Francomano E, Pugach P, Ringe RP, LaBranche CC, et al. (2018). Epitopes for neutralizing antibodies induced by HIV-1 envelope glycoprotein BG505 SOSIP trimers in rabbits and macaques. PLoS Pathog 14, e1006913.
    1. Klein F, Mouquet H, Dosenovic P, Scheid JF, Scharf L, and Nussenzweig MC (2013). Antibodies in HIV-1 vaccine development and therapy. Science 341, 1199–1204.
    1. Kong R, Xu K, Zhou T, Acharya P, Lemmin T, Liu K, Ozorowski G, Soto C, Taft JD, Bailer RT, et al. (2016). Fusion peptide of HIV-1 as a site of vulnerability to neutralizing antibody. Science 352, 828–833.
    1. Kubota SI, Takahashi K, Nishida J, Morishita Y, Ehata S, Tainaka K, Miyazono K, and Ueda HR (2017). Whole-Body Profiling of Cancer Metastasis with Single-Cell Resolution. Cell Rep 20, 236–250.
    1. Kulp DW, Steichen JM, Pauthner M, Hu X, Schiffner T, Liguori A, Cottrell CA, Havenar-Daughton C, Ozorowski G, Georgeson E, et al. (2017). Structure-based design of native-like HIV-1 envelope trimers to silence non-neutralizing epitopes and eliminate CD4 binding. Nat Commun 8, 1655.
    1. Kumar V, Vollbrecht T, Chernyshev M, Mohan S, Hanst B, Bavafa N, Lorenzo A, Ketteringham R, Eren K, Golden M, et al. (2018). Long-read amplicon denoising. 1–10.
    1. Kuraoka M, Schmidt AG, Nojima T, Feng F, Watanabe A, Kitamura D, Harrison SC, Kepler TB, and Kelsoe G (2016). Complex Antigens Drive Permissive Clonal Selection in Germinal Centers. Immunity 44, 542–552.
    1. Lander GC, Stagg SM, Voss NR, Cheng A, Fellmann D, Pulokas J, Yoshioka C, Irving C, Mulder A, Lau P-W, et al. (2009). Appion: an integrated, database-driven pipeline to facilitate EM image processing. J. Struct. Biol 166, 95–102.
    1. Langmead B, and Salzberg SL (2012). Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359.
    1. Lefranc MP, and Lefranc G (2001). The immunoglobulin factsbook.
    1. Locci M, Havenar-Daughton C, Landais E, Wu J, Kroenke MA, Arlehamn CL, Su LF, Cubas R, Davis MM, Sette A, et al. (2013). Human circulating PD-1+CXCR3-CXCR5+ memory Tfh cells are highly functional and correlate with broadly neutralizing HIV antibody responses. Immunity 39, 758–769.
    1. Lyumkis D, Julien J-P, de Val N, Cupo A, Potter CS, Klasse PJ, Burton DR, Sanders RW, Moore JP, Carragher B, et al. (2013). Cryo-EM structure of a fully glycosylated soluble cleaved HIV-1 envelope trimer. Science 342, 1484–1490.
    1. Mascola JR, Snyder SW, Weislow OS, Belay SM, Belshe RB, Schwartz DH, Clements ML, Dolin R, Graham BS, Gorse GJ, et al. (1996). Immunization with envelope subunit vaccine products elicits neutralizing antibodies against laboratory-adapted but not primary isolates of human immunodeficiency virus type 1. The National Institute of Allergy and Infectious Diseases AIDS Vaccine Evaluation Group. J. Infect. Dis 173, 340–348.
    1. Mesin L, Ersching J, and Victora GD (2016). Germinal Center B Cell Dynamics. Immunity 45, 471–482.
    1. Montefiori DC, Roederer M, Morris L, and Seaman MS (2018). Neutralization tiers of HIV-1. Curr Opin HIV AIDS 13, 128–136.
    1. Moody MA, Pedroza-Pacheco I, Vandergrift NA, Chui C, Lloyd KE, Parks R, Soderberg KA, Ogbe AT, Cohen MS, Liao H-X, et al. (2016). Immune perturbations in HIV-1-infected individuals who make broadly neutralizing antibodies. Science Immunology 1, aag0851–aag0851.
    1. Nakane T, Kimanius D, Lindahl E, and Scheres SH (2018). Characterisation of molecular motions in cryo-EM single-particle data by multi-body refinement in RELION. Elife 7, 1485.
    1. Nishimura Y, and Martin MA (2017). Of Mice, Macaques, and Men: Broadly Neutralizing Antibody Immunotherapy for HIV-1. Cell Host Microbe 22, 207–216.
    1. Noe SM, Green MA, Hogenesch H, and Hem SL (2010). Mechanism of immunopotentiation by aluminum-containing adjuvants elucidated by the relationship between antigen retention at the inoculation site and the immune response. Vaccine 28, 3588–3594.
    1. Pauthner M, Havenar-Daughton C, Sok D, Nkolola JP, Bastidas R, Boopathy AV, Carnathan DG, Chandrashekar A, Cirelli KM, Cottrell CA, et al. (2017). Elicitation of Robust Tier 2 Neutralizing Antibody Responses in Nonhuman Primates by HIV Envelope Trimer Immunization Using Optimized Approaches. Immunity 46, 1073–1088.e1076.
    1. Petrovas C, Yamamoto T, Gerner MY, Boswell KL, Wloka K, Smith EC, Ambrozak DR, Sandler NG, Timmer KJ, Sun X, et al. (2012). CD4 T follicular helper cell dynamics during SIV infection. J. Clin. Invest 122, 3281–3294.
    1. Picelli S, Faridani OR, Björklund AK, Winberg G, Sagasser S, and Sandberg R (2014). Full-length RNA-seq from single cells using Smart-seq2. Nature Protocols 9, 171–181.
    1. Plotkin SA (2010). Correlates of protection induced by vaccination. Clin. Vaccine Immunol 17, 1055–1065.
    1. Potter CS, Chu H, Frey B, Green C, Kisseberth N, Madden TJ, Miller KL, Nahrstedt K, Pulokas J, Reilein A, et al. (1999). Leginon: a system for fully automated acquisition of 1000 electron micrographs a day. Ultramicroscopy 77, 153–161.
    1. Price MN, Dehal PS, and Arkin AP (2010). FastTree 2--approximately maximum-likelihood trees for large alignments. PLoS ONE 5, e9490.
    1. Ramesh A, Darko S, Hua A, Overman G, Ransier A, Francica JR, Trama A, Tomaras GD, Haynes BF, Douek DC, et al. (2017). Structure and Diversity of the Rhesus Macaque Immunoglobulin Loci through Multiple De Novo Genome Assemblies. Front Immunol 8, 220–19.
    1. Reiss S, Baxter AE, Cirelli KM, Dan JM, Morou A, Daigneault A, Brassard N, Silvestri G, Routy J-P, Havenar-Daughton C, et al. (2017). Comparative analysis of activation induced marker (AIM) assays for sensitive identification of antigen-specific CD4 T cells. PLoS ONE 12, e0186998.
    1. Renier N, Adams EL, Kirst C, Wu Z, Azevedo R, Kohl J, Autry AE, Kadiri L, Umadevi Venkataraju K, Zhou Y, et al. (2016). Mapping of Brain Activity by Automated Volume Analysis of Immediate Early Genes. Cell 165, 1789–1802.
    1. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, Paris R, Premsri N, Namwat C, de Souza M, Adams E, et al. (2009). Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N. Engl. J. Med 361, 2209–2220.
    1. Richman DD, Wrin T, Little SJ, and Petropoulos CJ (2003). Rapid evolution of the neutralizing antibody response to HIV type 1 infection. Proceedings of the National Academy of Sciences 100, 4144–4149.
    1. Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, and Mesirov JP (2011). Integrative genomics viewer. Nat. Biotechnol 29, 24–26.
    1. Sanders RW, Derking R, Cupo A, Julien J-P, Yasmeen A, de Val N, Kim HJ, Blattner C, la Peña, de AT, Korzun J, et al. (2013). 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 9, e1003618.
    1. Sanders RW, van Gils MJ, Derking R, Sok D, Ketas TJ, Burger JA, Ozorowski G, Cupo A, Simonich C, Goo L, et al. (2015). HIV-1 VACCINES. HIV-1 neutralizing antibodies induced by native-like envelope trimers. Science 349, aac4223–aac4223.
    1. Scheres SHW (2012). RELION: implementation of a Bayesian approach to cryo-EM structure determination. J. Struct. Biol 180, 519–530.
    1. Schwickert TA, Victora GD, Fooksman DR, Kamphorst AO, Mugnier MR, Gitlin AD, Dustin ML, and Nussenzweig MC (2011). A dynamic T cell-limited checkpoint regulates affinity-dependent B cell entry into the germinal center. J. Exp. Med 208, 1243–1252.
    1. Shi Y, Hogenesch H, and Hem SL (2001). Change in the degree of adsorption of proteins by aluminum-containing adjuvants following exposure to interstitial fluid: freshly prepared and aged model vaccines. Vaccine 20, 80–85.
    1. Sorzano COS, Bilbao-Castro JR, Shkolnisky Y, Alcorlo M, Melero R, Caffarena-Fernández G, Li M, Xu G, Marabini R, and Carazo JM (2010). A clustering approach to multireference alignment of single-particle projections in electron microscopy. J. Struct. Biol 171, 197–206.
    1. Stewart-Jones GBE, Soto C, Lemmin T, Chuang G-Y, Druz A, Kong R, Thomas PV, Wagh K, Zhou T, Behrens A-J, et al. (2016). Trimeric HIV-1-Env Structures Define Glycan Shields from Clades A, B, and G. Cell 165, 813–826.
    1. Sundling C, Li Y, Huynh N, Poulsen C, Wilson R, O’Dell S, Feng Y, Mascola JR, Wyatt RT, and Karlsson Hedestam GB (2012). High-resolution definition of vaccine-elicited B cell responses against the HIV primary receptor binding site. Sci Transl Med 4, 142ra96–142ra96.
    1. Tam HH, Melo MB, Kang M, Pelet JM, Ruda VM, Foley MH, Hu JK, Kumari S, Crampton J, Baldeon AD, et al. (2016). Sustained antigen availability during germinal center initiation enhances antibody responses to vaccination. Proc. Natl. Acad. Sci. U.S.a 113, 201606050–E201606648.
    1. Tange O (2011). Gnu parallel-the command-line power tool.
    1. Tas JMJ, Mesin L, Pasqual G, Targ S, Jacobsen JT, Mano YM, Chen CS, Weill J-C, Reynaud C-A, Browne EP, et al. (2016). Visualizing antibody affinity maturation in germinal centers. Science 351, 1048–1054.
    1. Thorvaldsdóttir H, Robinson JT, and Mesirov JP (2013). Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinformatics 14, 178–192.
    1. Turner JS, Benet ZL, and Grigorova IL (2017). Antigen Acquisition Enables Newly Arriving B Cells To Enter Ongoing Immunization-Induced Germinal Centers. J. Immunol 199, 1301–1307.
    1. Upadhyay AA, Kauffman RC, Wolabaugh AN, Cho A, Patel NB, Reiss SM, Havenar-Daughton C, Dawoud RA, Tharp GK, Sanz I, et al. (2018). BALDR: a computational pipeline for paired heavy and light chain immunoglobulin reconstruction in single-cell RNA-seq data. Genome Med 10, 20.
    1. Vander Heiden JA, Yaari G, Uduman M, Stern JNH, O’Connor KC, Hafler DA, Vigneault F, and Kleinstein SH (2014). pRESTO: a toolkit for processing high-throughput sequencing raw reads of lymphocyte receptor repertoires. Bioinformatics 30, 1930–1932.
    1. Victora GD, and Wilson PC (2015). Germinal center selection and the antibody response to influenza. Cell 163, 545–548.
    1. Victora GD, Schwickert TA, Fooksman DR, Kamphorst AO, Meyer-Hermann M, Dustin ML, and Nussenzweig MC (2010). Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter. Cell 143, 592–605.
    1. Voss NR, Yoshioka CK, Radermacher M, Potter CS, and Carragher B (2009). DoG Picker and TiltPicker: software tools to facilitate particle selection in single particle electron microscopy. J. Struct. Biol 166, 205–213.
    1. Watson CT, and Breden F (2012). The immunoglobulin heavy chain locus: genetic variation, missing data, and implications for human disease. Genes Immun 13, 363–373.
    1. Watson CT, Glanville J, and Marasco WA (2017). The Individual and Population Genetics of Antibody Immunity. Trends Immunol 38, 459–470.
    1. Wei X, Decker JM, Wang S, Hui H, Kappes JC, Wu X, Salazar-Gonzalez JF, Salazar MG, Kilby JM, Saag MS, et al. (2003). Antibody neutralization and escape by HIV-1. Nature 422, 307–312.
    1. Weissburg RP, Berman PW, Cleland JL, Eastman D, Farina F, Frie S, Lim A, Mordenti J, Peterson MR, Yim K, et al. (1995). Characterization of the MN gp120 HIV-1 Vaccine: Antigen Binding to Alum. Pharmaceutical Research 12, 1439–1446.
    1. West AP, Scharf L, Scheid JF, Klein F, Bjorkman PJ, and Nussenzweig MC (2014). Structural insights on the role of antibodies in HIV-1 vaccine and therapy. Cell 156, 633–648.
    1. Yamamoto T, Lynch RM, Gautam R, Matus-Nicodemos R, Schmidt SD, Boswell KL, Darko S, Wong P, Sheng Z, Petrovas C, et al. (2015). Quality and quantity of TFH cells are critical for broad antibody development in SHIVAD8 infection. Sci Transl Med 7, 298ra120–298ra120.
    1. Ye J, Ma N, Madden TL, and Ostell JM (2013). IgBLAST: an immunoglobulin variable domain sequence analysis tool. Nucleic Acids Res 41, W34–W40.
    1. Yeh C-H, Nojima T, Kuraoka M, and Kelsoe G (2018). Germinal center entry not selection of B cells is controlled by peptide-MHCII complex density. Nat Commun 9, 928.
    1. Zhou T, Doria-Rose NA, Cheng C, Stewart-Jones GBE, Chuang G-Y, Chambers M, Druz A, Geng H, McKee K, Kwon YD, et al. (2017). Quantification of the Impact of the HIV-1-Glycan Shield on Antibody Elicitation. Cell Rep 19, 719–732.

Source: PubMed

3
Suscribir