Memory B Cell Activation, Broad Anti-influenza Antibodies, and Bystander Activation Revealed by Single-Cell Transcriptomics

Felix Horns, Cornelia L Dekker, Stephen R Quake, Felix Horns, Cornelia L Dekker, Stephen R Quake

Abstract

Antibody memory protects humans from many diseases. Protective antibody memory responses require activation of transcriptional programs, cell proliferation, and production of antigen-specific antibodies, but how these aspects of the response are coordinated is poorly understood. We profile the molecular and cellular features of the antibody response to influenza vaccination by integrating single-cell transcriptomics, longitudinal antibody repertoire sequencing, and antibody binding measurements. Single-cell transcriptional profiling reveals a program of memory B cell activation characterized by CD11c and T-bet expression associated with clonal expansion and differentiation toward effector function. Vaccination elicits an antibody clone, which rapidly acquired broad high-affinity hemagglutinin binding during affinity maturation. Unexpectedly, many antibody clones elicited by vaccination do not bind vaccine, demonstrating non-specific activation of bystander antibodies by influenza vaccination. These results offer insight into how molecular recognition, transcriptional programs, and clonal proliferation are coordinated in the human B cell repertoire during memory recall.

Keywords: antibody repertoires; human antibody memory; infectious disease; influenza; single-cell transcriptomics; vaccination.

Conflict of interest statement

Declaration of Interests The authors declare no competing interests.

Copyright © 2019. Published by Elsevier Inc.

Figures

Figure 1.. Experimental Design and Workflow for…
Figure 1.. Experimental Design and Workflow for Studying B Cell Response to Influenza Vaccination Using Integrated Single-Cell and Antibody Repertoire Sequencing
(A) Study design. 2011–2012 seasonal trivalent influenza vaccine was administered, and peripheral blood samples were collected for analysis at the indicated days before and after vaccination. (B) Experiment workflow. Antibody heavy-chain repertoire sequencing (Rep-seq) was performed on samples from all time points. Single-cell transcriptional profiling and antibody heavy- and light-chain sequencing were performed on samples from d7 and d9 after vaccination.
Figure 2.. Clone Dynamics during B Cell…
Figure 2.. Clone Dynamics during B Cell Response to Influenza Vaccination
(A) Comparison of clonal abundance measurements across platforms, showing cells detected by single-cell sequencing and sequences detected by Rep-seq within each clone. Color indicates density of clones. ND, not detected. (B) Population dynamics of B cell clones. Each line represents a clone. Colored lines indicate vaccine-responsive clones (>50-fold expansion from D0 to D7 and >0.1% of repertoire at D7, as in Horns et al., 2019), according to the key in (C). Gray lines indicate non-vaccine-responsive clones. (C) Repertoire-wide distribution of the extent of clonal expansion after vaccination. Histogram shows the fold change from D0 to D7 of clones annotated as not vaccine responsive, which composed a substantial fraction of the repertoire at D7 (>0.1%) and had ≥1 cell detected by single-cell sequencing. Clones identified as vaccine responsive are indicated by markers (stars indicate vaccine binding, and circles indicate not vaccine binding). Dashed line shows the extent of expansion used as cutoff for identifying vaccine-responsive clones. Inf indicates undefined fold change arising because zero cells in the clone were detected at D0. D0, day of vaccination; D7, 7 days after vaccination; ND, not detected. See also Figure S1.
Figure 3.. Characterization of Gene Expression in…
Figure 3.. Characterization of Gene Expression in Single B Cells Isolated from Peripheral Blood after Influenza Vaccination
(A–C) Principal-component analysis and t-distributed stochastic neighbor embedding (tSNE) separates cells into distinct clusters. Each dot is a cell, colored by type or state revealed by gene expression profile (A), antibody isotype as revealed by single-cell antibody heavy-chain sequencing (B), or clonal population dynamics as revealed by Rep-seq (C). (D) Differential expression analysis identifies markers of distinct B cell states. Genes of immunological interest are labeled. (E–G) Gene expression distributions in distinct B cell states of established immune activation-related genes (E), transcription factors (F), and signaling receptors (G). See also Figure S2, Table S1, and Table S2.
Figure 4.. Binding of Influenza Vaccine-Responsive Antibodies…
Figure 4.. Binding of Influenza Vaccine-Responsive Antibodies to Vaccine
Binding of 21 monoclonal antibodies from 5 clones to the trivalent inactivated influenza vaccine from the 2011–2012 season was measured using enzyme-linked immunosorbent assay (ELISA), revealing that many vaccine-responsive antibodies do not bind vaccine. Ab, antibody; OD, optical density; hIgG1, human IgG1. See also Figure S3.
Figure 5.. Reconstructing Evolution of a Broadly…
Figure 5.. Reconstructing Evolution of a Broadly Binding High-Affinity Anti-influenza Antibody Clone
(A) Binding of antibodies from the L3 clone to a panel of influenza hemagglutinin (HA) variants was measured using ELISA. OD, optical density; hIgG1, human IgG1. (B) Evolutionary history of L3 depicted as a maximum-likelihood phylogeny based on heavy-chain sequence. Markers indicate antibodies detected by single-cell sequencing (N1–N7) or repertoire sequencing (R1–R7), or reconstructed ancestral sequences (germline and A1–A4). (C) Dissociation constants (KDs) of binding between L3 antibody variants and H1 (A/California/7/2009) and H3 (A/Perth/16/2009) hemagglutinin variants, as determined by biolayer interferometry. L3 antibodies include extant sequences (N1–N7), reconstructed ancestral sequences (germline and A1–A4), and engineered variants having the L3N6 sequences, but with heavy chain reverted to the inferred germline sequence (germline immunoglobulin heavy chain [IGH]), light chain reverted to the inferred germline sequence (germline immunoglobulin kappa chain [IGK]), or a light chain sequence substituted from a different clone (IGK swap). Jitter was added to germline and germline IGH to improve visualization of the data points. (D) Dissociation constants of binding between L3 antibodies compared with extent of somatic hypermutation. (E) Dissociation constants of binding between L3 antibodies and H1 variants from childhood (A/New Caledonia/20/1999) and adulthood (A/California/7/2009). Dashed line indicates equal KD for binding to both variants. Uncertainty of fitted parameters was smaller than the size of the markers used for plotting; therefore, error bars are not shown. See also Figure S4 and Figure S5.

References

    1. Altschul SF, Gish W, Miller W, Myers EW, and Lipman DJ (1990). Basic local alignment search tool. J. Mol. Biol 215, 403–410.
    1. Arneborn P, Biberfeld G, Forsgren M, and von Stedingk LV (1983). Specific and non-specific B cell activation in measles and varicella. Clin. Exp. Immunol 51, 165–172.
    1. Barnett BE, Staupe RP, Odorizzi PM, Palko O, Tomov VT, Mahan AE, Gunn B, Chen D, Paley MA, Alter G, et al. (2016). Cutting ddge: B cell-intrinsic T-bet expression is required to control chronic viral infection. J. Immunol 197, 1017–1022.
    1. Belshe RB, Newman FK, Cannon J, Duane C, Treanor J, Van Hoecke C, Howe BJ, and Dubin G (2004). Serum antibody responses after intradermal vaccination against influenza. N. Engl. J. Med 351, 2286–2294.
    1. Bernasconi NL, Traggiai E, and Lanzavecchia A (2002). Maintenance of serological memory by polyclonal activation of human memory B cells. Science 298, 2199–2202.
    1. Crotty S, Aubert RD, Glidewell J, and Ahmed R (2004). Tracking human antigen-specific memory B cells: a sensitive and generalized ELISPOT system. J. Immunol. Methods 286, 111–122.
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, and Gingeras TR (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21.
    1. Dreyfus C, Laursen NS, Kwaks T, Zuijdgeest D, Khayat R, Ekiert DC, Lee JH, Metlagel Z, Bujny MV, Jongeneelen M, et al. (2012). Highly conserved protective epitopes on influenza B viruses. Science 337, 1343–1348.
    1. Edgar RC (2004). MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 32, 1792–1797.
    1. Ester M, Kriegel H-P, Sander J, and Xu X (1996). A density-based algorithm for discovering clusters in large spatial databases with noise In KDD’96: Proceedings of the Second International Conference on Knowledge Discovery and Data Mining (AAAI Press; ), pp. 226–231.
    1. Gaucher D, Therrien R, Kettaf N, Angermann BR, Boucher G, Filali-Mouhim A, Moser JM, Mehta RS, Drake DR 3rd, Castro E, et al. (2008). Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J. Exp. Med 205, 3119–3131.
    1. Gupta NT, Adams KD, Briggs AW, Timberlake SC, Vigneault F, and Kleinstein SH (2017). Hierarchical clustering can identify B cell clones with high confidence in Ig repertoire sequencing data. J. Immunol 198, 2489–2499.
    1. Hao Y, O’Neill P, Naradikian MS, Scholz JL, and Cancro MP (2011). A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice. Blood 118, 1294–1304.
    1. Henn AD, Wu S, Qiu X, Ruda M, Stover M, Yang H, Liu Z, Welle SL, Holden-Wiltse J, Wu H, and Zand MS (2013). High-resolution temporal response patterns to influenza vaccine reveal a distinct human plasma cell gene signature. Sci. Rep 3, 2327.
    1. Horns F, Vollmers C, Croote D, Mackey SF, Swan GE, Dekker CL, Davis MM, and Quake SR (2016). Lineage tracing of human B cells reveals the in vivo landscape of human antibody class switching. eLife 5, e16578.
    1. Horns F, Vollmers C, Dekker CL, and Quake SR (2019). Signatures of selection in the human antibody repertoire: Selective sweeps, competing subclones, and neutral drift. Proc. Natl. Acad. Sci. USA 116, 1261–1266.
    1. Jash A, Wang Y, Weisel FJ, Scharer CD, Boss JM, Shlomchik MJ, and Bhattacharya D (2016). ZBTB32 restricts the duration of memory B cell recall responses. J. Immunol 197, 1159–1168.
    1. Jasiulewicz A, Lisowska KA, Pietruczuk K, Frąckowiak J, Fulop T, and Witkowski JM (2015). Homeostatic ‘bystander’ proliferation of human peripheral blood B cells in response to polyclonal T-cell stimulation in vitro. Int. Immunol 27, 579–588.
    1. Jiang N, He J, Weinstein JA, Penland L, Sasaki S, He X-S, Dekker CL, Zheng N-Y, Huang M, Sullivan M, et al. (2013). Lineage structure of the human antibody repertoire in response to influenza vaccination. Sci. Transl. Med 5, 171ra19.
    1. Juy D, Sterkers G, Gomez A, Zelizewski D, and Lévy J-P (1987). Polyclonal B-cell activation by influenza A/Texas virus-specific human T-cell clones. Ann. Inst. Pasteur Immunol 138, 371–382.
    1. Kallewaard NL, Corti D, Collins PJ, Neu U, McAuliffe JM, Benjamin E, Wachter-Rosati L, Palmer-Hill FJ, Yuan AQ, Walker PA, et al. (2016). Structure and function analysis of an antibody recognizing all influenza A sub-types. Cell 166, 596–608.
    1. Kluyver T, Ragan-Kelley B, Pérez F, Granger B, Bussonnier M, Frederic J, Kelley K, Hamrick J, Grout J, Corlay S, et al. (2016). Jupyter Notebooks—a publishing format for reproducible computational workflows In Positioning and Power in Academic Publishing: Players, Agents and Agendas. In Proceedings of the 20th International Conference on Electronic Publishing, Loizides F and Schmidt B, eds. (IOS Press; ), pp. 87–90.
    1. Lacotte S, Brun S, Muller S, and Dumortier H (2009). CXCR3, inflammation, and autoimmune diseases. Ann. N Y Acad. Sci 1173, 310–317.
    1. Lanzavecchia A, Parodi B, and Celada F (1983). Activation of human B lymphocytes: frequency of antigen-specific B cells triggered by alloreactive or by antigen-specific T cell clones. Eur. J. Immunol 13, 733–738.
    1. Larousserie F, Bardel E, Coulomb L’Herminé A, Canioni D, Brousse N, Kastelein RA, and Devergne O (2006). Variable expression of Epstein-Barr virus-induced gene 3 during normal B-cell differentiation and among B-cell lymphomas. J. Pathol 209, 360–368.
    1. Lee FE-H, Halliley JL, Walsh EE, Moscatiello AP, Kmush BL, Falsey AR, Randall TD, Kaminiski DA, Miller RK, and Sanz I (2011). Circulating human antibody-secreting cells during vaccinations and respiratory viral infections are characterized by high specificity and lack of bystander effect. J. Immunol 186, 5514–5521.
    1. Li S, Rouphael N, Duraisingham S, Romero-Steiner S, Presnell S, Davis C, Schmidt DS, Johnson SE, Milton A, Rajam G, et al. (2014). Molecular signatures of antibody responses derived from a systems biology study of five human vaccines. Nat. Immunol 15, 195–204.
    1. Lingwood D, McTamney PM, Yassine HM, Whittle JRR, Guo X, Boyington JC, Wei C-J, and Nabel GJ (2012). Structural and genetic basis for development of broadly neutralizing influenza antibodies. Nature 489, 566–570.
    1. Liu Y, Pan J, Jenni S, Raymond DD, Caradonna T, Do KT, Schmidt AG, Harrison SC, and Grigorieff N (2017). CryoEM structure of an influenza virus receptor-binding site antibody-antigen interface. J. Mol. Biol 429, 1829–1839.
    1. Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O’Shea MA, Roby G, Kottilil S, Arthos J, Proschan MA, et al. (2008). Evidence for HIV-associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV-infected viremic individuals. J. Exp. Med 205, 1797–1805.
    1. Moticka EJ, and Streilein JW (1978). Hypothesis: nonspecific polyclonal activation of memory B cells by antigen as a mechanism for the preservation of long term immunologic anamnesis. Cell. Immunol 41, 406–413.
    1. Muramatsu M, Kinoshita K, Fagarasan S, Yamada S, Shinkai Y, and Honjo T (2000). Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell 102, 553–563.
    1. Okada T, Ngo VN, Ekland EH, Förster R, Lipp M, Littman DR, and Cyster JG (2002). Chemokine requirements for B cell entry to lymph nodes and Peyer’s patches. J. Exp. Med 196, 65–75.
    1. Pappas L, Foglierini M, Piccoli L, Kallewaard NL, Turrini F, Silacci C, Fernandez-Rodriguez B, Agatic G, Giacchetto-Sasselli I, Pellicciotta G, et al. (2014). Rapid development of broadly influenza neutralizing antibodies through redundant mutations. Nature 516, 418–422.
    1. Pène J, Gauchat J-F, Lécart S, Drouet E, Guglielmi P, Boulay V, Del-wail A, Foster D, Lecron J-C, and Yssel H (2004). Cutting edge: IL-21 is a switch factor for the production of IgG1 and IgG3 by human B cells. J. Immunol 172, 5154–5157.
    1. Price MN, Dehal PS, and Arkin AP (2010). FastTree 2—approximately maximum-likelihood trees for large alignments. PLoS ONE 5, e9490.
    1. Raymond DD, Bajic G, Ferdman J, Suphaphiphat P, Settembre EC, Moody MA, Schmidt AG, and Harrison SC (2018). Conserved epitope on influenza-virus hemagglutinin head defined by a vaccine-induced antibody. Proc. Natl. Acad. Sci. USA 115, 168–173.
    1. Rubtsov AV, Rubtsova K, Fischer A, Meehan RT, Gillis JZ, Kappler JW, and Marrack P (2011). Toll-like receptor 7 (TLR7)-driven accumulation of a novel CD11c+ B-cell population is important for the development of autoimmunity. Blood 118, 1305–1315.
    1. Rubtsova K, Rubtsov AV, van Dyk LF, Kappler JW, and Marrack P (2013). T-box transcription factor T-bet, a key player in a unique type of B-cell activation essential for effective viral clearance. Proc. Natl. Acad. Sci. USA 110, E3216–E3224.
    1. Rubtsova K, Rubtsov AV, Thurman JM, Mennona JM, Kappler JW, and Marrack P (2017). B cells expressing the transcription factor T-bet drive lupus-like autoimmunity. J. Clin. Invest 127, 1392–1404.
    1. Schmidt AG, Do KT, McCarthy KR, Kepler TB, Liao H-X, Moody MA, Haynes BF, and Harrison SC (2015). Immunogenic stimulus for germline precursors of antibodies that engage the influenza hemagglutinin receptor-binding site. Cell Rep. 13, 2842–2850.
    1. Stavnezer J (1996). Immunoglobulin class switching. Curr. Opin. Immunol 8, 199–205.
    1. The Tabula Muris Consortium (2018). Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372.
    1. Treanor J, Keitel W, Belshe R, Campbell J, Schiff G, Zangwill K, Wolff M, Klimov A, Levandowski R, and Lambert L (2002). Evaluation of a single dose of half strength inactivated influenza vaccine in healthy adults. Vaccine 20, 1099–1105.
    1. van der Maaten L, and Hinton G (2008). Visualizing data using t-SNE. J. Mach. Learn. Res 9, 2579–2605.
    1. Vollmers C, Sit RV, Weinstein JA, Dekker CL, and Quake SR (2013). Genetic measurement of memory B-cell recall using antibody repertoire sequencing. Proc. Natl. Acad. Sci. USA 110, 13463–13468.
    1. Wang NS, McHeyzer-Williams LJ, Okitsu SL, Burris TP, Reiner SL, and McHeyzer-Williams MG (2012). Divergent transcriptional programming of class-specific B cell memory by T-bet and RORα. Nat. Immunol 13, 604–611.
    1. Whittle JRR, Zhang R, Khurana S, King LR, Manischewitz J, Golding H, Dormitzer PR, Haynes BF, Walter EB, Moody MA, et al. (2011). Broadly neutralizing human antibody that recognizes the receptor-binding pocket of influenza virus hemagglutinin. Proc. Natl. Acad. Sci. USA 108, 14216–14221.
    1. Wolf FA, Angerer P, and Theis FJ (2018). SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15.
    1. Wrammert J, Smith K, Miller J, Langley WA, Kokko K, Larsen C, Zheng N-Y, Mays I, Garman L, Helms C, et al. (2008). Rapid cloning of high-affinity human monoclonal antibodies against influenza virus. Nature 453, 667–671.
    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.

Source: PubMed

3
Abonner