Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Diversity, cellular origin and autoreactivity of antibody-secreting cell population expansions in acute systemic lupus erythematosus

Abstract

Acute systemic lupus erythematosus (SLE) courses with surges of antibody-secreting cells (ASCs) whose origin, diversity and contribution to serum autoantibodies remain unknown. Here, deep sequencing, proteomic profiling of autoantibodies and single-cell analysis demonstrated highly diversified ASCs punctuated by clones expressing the variable heavy-chain region VH4-34 that produced dominant serum autoantibodies. A fraction of ASC clones contained autoantibodies without mutation, a finding consistent with differentiation outside the germinal centers. A substantial ASC segment was derived from a distinct subset of newly activated naive cells of considerable clonality that persisted in the circulation for several months. Thus, selection of SLE autoreactivities occurred during polyclonal activation, with prolonged recruitment of recently activated naive B cells. Our findings shed light on the pathogenesis of SLE, help explain the benefit of agents that target B cells and should facilitate the design of future therapies.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: SLE flares are characterized by large polyclonal expansions of ASC populations.
Figure 2: Isotype distribution and SHM in patients with SLE and healthy vaccinated control subjects.
Figure 3: Over-representation of IGH VH4-34 among ASC clonal expansions in patients with SLE.
Figure 4: Contribution of naive cells to ASC populations in SLE flares.
Figure 5: Characterization of acN cells in SLE.
Figure 6: Clonality and connectivity of acN cells in SLE.
Figure 7: Serological consequence and cellular derivation of expanded ASC clones in SLE.
Figure 8: Autoreactivity of 9G4+ ASCs in SLE.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

References

  1. Lee, F.E. et al. 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 (2011).

    CAS  PubMed  Google Scholar 

  2. Wrammert, J. et al. Rapid cloning of high-affinity human monoclonal antibodies against influenza virus. Nature 453, 667–671 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Odendahl, M. et al. Disturbed peripheral B lymphocyte homeostasis in systemic lupus erythematosus. J. Immunol. 165, 5970–5979 (2000).

    CAS  PubMed  Google Scholar 

  4. Cappione, A. III et al. Germinal center exclusion of autoreactive B cells is defective in human systemic lupus erythematosus. J. Clin. Invest. 115, 3205–3216 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Isenberg, D.A. et al. Correlation of 9G4 idiotope with disease activity in patients with systemic lupus erythematosus. Ann. Rheum. Dis. 57, 566–570 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Jackson, K.J. et al. Human responses to influenza vaccination show seroconversion signatures and convergent antibody rearrangements. Cell Host Microbe 16, 105–114 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Palanichamy, A. et al. Immunoglobulin class-switched B cells form an active immune axis between CNS and periphery in multiple sclerosis. Sci. Transl. Med. 6, 248ra106 (2014).

    PubMed  PubMed Central  Google Scholar 

  8. Stern, J.N. et al. B cells populating the multiple sclerosis brain mature in the draining cervical lymph nodes. Sci. Transl. Med. 6, 248ra107 (2014).

    PubMed  PubMed Central  Google Scholar 

  9. Boyd, S.D. et al. Measurement and clinical monitoring of human lymphocyte clonality by massively parallel VDJ pyrosequencing. Sci. Transl. Med. 1, 12ra23 (2009).

    PubMed  PubMed Central  Google Scholar 

  10. Moody, M.A. et al. HIV-1 gp120 vaccine induces affinity maturation in both new and persistent antibody clonal lineages. J. Virol. 86, 7496–7507 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Dogan, I. et al. Multiple layers of B cell memory with different effector functions. Nat. Immunol. 10, 1292–1299 (2009).

    CAS  PubMed  Google Scholar 

  12. Breden, F. et al. Comparison of antibody repertoires produced by HIV-1 infection, other chronic and acute infections, and systemic autoimmune disease. PLoS ONE 6, e16857 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Richardson, C. et al. Molecular basis of 9G4 B cell autoreactivity in human systemic lupus erythematosus. J. Immunol. 191, 4926–4939 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Jenks, S.A. et al. 9G4+ autoantibodies are an important source of apoptotic cell reactivity associated with high levels of disease activity in systemic lupus erythematosus. Arthritis Rheum. 65, 3165–3175 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Klinman, D.M. & Steinberg, A.D. Systemic autoimmune disease arises from polyclonal B cell activation. J. Exp. Med. 165, 1755–1760 (1987).

    CAS  PubMed  Google Scholar 

  16. Pugh-Bernard, A.E. et al. Regulation of inherently autoreactive VH4–34 B cells in the maintenance of human B cell tolerance. J. Clin. Invest. 108, 1061–1070 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. William, J., Euler, C., Christensen, S. & Shlomchik, M.J. Evolution of autoantibody responses via somatic hypermutation outside of germinal centers. Science 297, 2066–2070 (2002).

    CAS  PubMed  Google Scholar 

  18. Shlomchik, M.J. Sites and stages of autoreactive B cell activation and regulation. Immunity 28, 18–28 doi:10.1016/j.immuni.2007.12.004 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Sweet, R.A., Lee, S.K. & Vinuesa, C.G. Developing connections amongst key cytokines and dysregulated germinal centers in autoimmunity. Curr. Opin. Immunol. 24, 658–664 (2012).

    CAS  PubMed  Google Scholar 

  20. Wirths, S. & Lanzavecchia, A. ABCB1 transporter discriminates human resting naive B cells from cycling transitional and memory B cells. Eur. J. Immunol. 35, 3433–3441 (2005).

    CAS  PubMed  Google Scholar 

  21. Sims, G.P. et al. Identification and characterization of circulating human transitional B cells. Blood 105, 4390–4398 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Masilamani, M., Kassahn, D., Mikkat, S., Glocker, M.O. & Illges, H. B cell activation leads to shedding of complement receptor type II (CR2/CD21). Eur. J. Immunol. 33, 2391–2397 (2003).

    CAS  PubMed  Google Scholar 

  23. Wehr, C. et al. A new CD21low B cell population in the peripheral blood of patients with SLE. Clin. Immunol. 113, 161–171 (2004).

    CAS  PubMed  Google Scholar 

  24. William, J., Euler, C. & Shlomchik, M.J. Short-lived plasmablasts dominate the early spontaneous rheumatoid factor response: differentiation pathways, hypermutating cell types, and affinity maturation outside the germinal center. J. Immunol. 174, 6879–6887 (2005).

    CAS  PubMed  Google Scholar 

  25. Barak, M., Zuckerman, N.S., Edelman, H., Unger, R. & Mehr, R. IgTree: creating Immunoglobulin variable region gene lineage trees. J. Immunol. Methods 338, 67–74 (2008).

    CAS  PubMed  Google Scholar 

  26. Shlomchik, M.J., Marshak-Rothstein, A., Wolfowicz, C.B., Rothstein, T.L. & Weigert, M.G. The role of clonal selection and somatic mutation in autoimmunity. Nature 328, 805–811 (1987).

    CAS  PubMed  Google Scholar 

  27. Cheung, W.C. et al. A proteomics approach for the identification and cloning of monoclonal antibodies from serum. Nat. Biotechnol. 30, 447–452 (2012).

    CAS  PubMed  Google Scholar 

  28. Tan, E.M. Antinuclear antibodies: diagnostic markers for autoimmune diseases and probes for cell biology. Adv. Immunol. 44, 93–151 (1989).

    CAS  PubMed  Google Scholar 

  29. Sanz, I. & Lee, F.E. B cells as therapeutic targets in SLE. Nat. Rev. Rheumatol 6, 326–337 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Bernasconi, N.L., Traggiai, E. & Lanzavecchia, A. Maintenance of serological memory by polyclonal activation of human memory B cells. Science 298, 2199–2202 (2002).

    CAS  PubMed  Google Scholar 

  31. Gharavi, A.E., Chu, J.L. & Elkon, K.B. Autoantibodies to intracellular proteins in human systemic lupus erythematosus are not due to random polyclonal B cell activation. Arthritis Rheum. 31, 1337–1345 (1988).

    CAS  PubMed  Google Scholar 

  32. Choi, J., Kim, S.T. & Craft, J. The pathogenesis of systemic lupus erythematosus-an update. Curr. Opin. Immunol. 24, 651–657 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Bernasconi, N.L., Onai, N. & Lanzavecchia, A. A role for Toll-like receptors in acquired immunity: up-regulation of TLR9 by BCR triggering in naive B cells and constitutive expression in memory B cells. Blood 101, 4500–4504 (2003).

    CAS  PubMed  Google Scholar 

  34. Zubler, R.H. & Kanagawa, O. Requirement for three signals in B cell responses. II. Analysis of antigen- and Ia-restricted T helper cell-B cell interaction. J. Exp. Med. 156, 415–429 (1982).

    CAS  PubMed  Google Scholar 

  35. Palanichamy, A. et al. Novel human transitional B cell populations revealed by B cell depletion therapy. J. Immunol. 182, 5982–5993 (2009).

    CAS  PubMed  Google Scholar 

  36. Hanten, J.A. et al. Comparison of human B cell activation by TLR7 and TLR9 agonists. BMC Immunol. 9, 39 (2008).

    PubMed  PubMed Central  Google Scholar 

  37. Yokota, A. et al. Two species of human Fc ɛ receptor II (FcɛRII/CD23): tissue-specific and IL-4-specific regulation of gene expression. Cell 55, 611–618 (1988).

    CAS  PubMed  Google Scholar 

  38. Delespesse, G., Sarfati, M. & Peleman, R. Influence of recombinant IL-4, IFN-α, and IFN-γ on the production of human IgE-binding factor (soluble CD23). J. Immunol. 142, 134–138 (1989).

    CAS  PubMed  Google Scholar 

  39. Stern, J.N.H. et al. B cells populating the multiple sclerosis brain mature in the draining cervical lymph nodes. Sci. Transl. Med. 6, 248ra107 (2014).

    PubMed  PubMed Central  Google Scholar 

  40. Palanichamy, A. et al. Immunoglobulin class-switched B cells form an active immune axis between CNS and periphery in multiple sclerosis. Sci. Transl. Med. 6, 248ra106 (2014).

    PubMed  PubMed Central  Google Scholar 

  41. Joo, H. et al. Serum from patients with SLE instructs monocytes to promote IgG and IgA plasmablast differentiation. J. Exp. Med. 209, 1335–1348 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Wardemann, H. et al. Predominant autoantibody production by early human B cell precursors. Science 301, 1374–1377 (2003).

    CAS  PubMed  Google Scholar 

  43. Sabouri, Z. et al. Redemption of autoantibodies on anergic B cells by variable-region glycosylation and mutation away from self-reactivity. Proc. Natl. Acad. Sci. USA 111, E2567–E2575 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Mietzner, B. et al. Autoreactive IgG memory antibodies in patients with systemic lupus erythematosus arise from nonreactive and polyreactive precursors. Proc. Natl. Acad. Sci. USA 105, 9727–9732 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Chevrier, S., Kratina, T., Emslie, D., Karnowski, A. & Corcoran, L.M. Germinal center-independent, IgM-mediated autoimmunity in sanroque mice lacking Obf1. Immunol. Cell Biol. 92, 12–19 (2014).

    CAS  PubMed  Google Scholar 

  46. Fulcher, D.A. & Basten, A. B cell life span: a review. Immunol. Cell Biol. 75, 446–455 (1997).

    CAS  PubMed  Google Scholar 

  47. Cyster, J.G. & Goodnow, C.C. Antigen-induced exclusion from follicles and anergy are separate and complementary processes that influence peripheral B cell fate. Immunity 3, 691–701 (1995).

    CAS  PubMed  Google Scholar 

  48. Macallan, D.C. et al. B-cell kinetics in humans: rapid turnover of peripheral blood memory cells. Blood 105, 3633–3640 (2005).

    CAS  PubMed  Google Scholar 

  49. Stohl, W. et al. Belimumab reduces autoantibodies, normalizes low complement levels, and reduces select B cell populations in patients with systemic lupus erythematosus. Arthritis Rheum. 64, 2328–2337 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Infantino, S. et al. The tyrosine kinase Lyn limits the cytokine responsiveness of plasma cells to restrict their accumulation in mice. Sci. Signal. 7, 77 (2014).

    Google Scholar 

  51. Thaunat, O. et al. Asymmetric segregation of polarized antigen on B cell division shapes presentation capacity. Science 335, 475–479 (2012).

    CAS  PubMed  Google Scholar 

  52. Jacobi, A.M. et al. Differential effects of epratuzumab on peripheral blood B cells of patients with systemic lupus erythematosus versus normal controls. Ann. Rheum. Dis. 67, 450–457 (2008).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank F. Stevenson (University of Southampton) for the 9G4 hybridoma; E. Meffre (Yale University) for expression vectors; and the blood donors and research coordinators involved in this study. Supported by the Autoimmunity Center of Excellence (U19 AI110483, 5P01AI078907 and 5R37AI049660) and the United States-Israel Binational Science Foundation (2013432).

Author information

Authors and Affiliations

Authors

Contributions

C.M.T. obtained most samples, conducted sample preparation and cell sorting, designed and conducted the NGS studies, analyzed and interpreted the data, helped to design figures and helped to write the manuscript; C.F.F. and A.F.R. wrote the programs used for NGS analysis, helped in data interpretation, produced visualization of the data, and helped to design and produce the figures; J.D., I.G., S.S. and W.C.C. conducted and analyzed the proteomics studies; A.C. conducted the experiments with single-cell monoclonal antibodies; T.I. conducted the ELISPOT experiments; J.H. obtained samples from patients with pemphigus, conducted some sequencing studies and helped with the analysis; S.J. helped with sequencing analysis; R.J.F. provided samples from patients with pemphigus; R.M. provided the IgTree program and aided in analysis; C.W. provided flow cytometry data and helped with its analysis; F.E.-H.L. provided the samples from vaccinated subjects and aided in data analysis; and I.S. designed and supervised the project, helped in experimental design and analysis, and wrote the manuscript.

Corresponding author

Correspondence to Iñaki Sanz.

Ethics declarations

Competing interests

I.S. is a member of the Pfizer Visiting Professor Board and has consulted for Genentech on B cell--depleting agents, and F.E.-H.L. has grants from Genentech.

Integrated supplementary information

Supplementary Figure 1 Data-processing pipeline.

Steps for processing data from multiple sorted B cell populations derived from a single individual at a single time point are shown. Parameters used are indicated to the right of the corresponding box. These steps are implemented by a combination of publicly available software (fastq-joiner), public resources (IMGT/HighV-quest), and custom software written in Perl and Matlab by the authors. After raw reads are joined, the sequences are each confirmed to meet a length threshold of 200bp and are eliminated if they contain more than 0, 10, or 15 bp that are less than Q10, Q20, and Q30 respectively. A subset of 150,000 sequences from the total is randomly chosen to relieve computational stress and these sequences are aligned with IMGT.org's HighV-Quest tool. Alignment results are filtered by removing “unproductive” and “unknown” sequences. Custom software is then used to identify clusters of sequences based on the clonal identification metric (see methods). 50,000 sequences from the final set are then randomly chosen, again to relieve computational stress of the downstream analyses, to retain similar number of sequences in each data set, and for display purposes. All of IMGT/HighV-quest's data is retained through the process and used for mutation calculations and alignment analyses.

Supplementary Figure 2 Validation of identity thresholds for clonality identification metric.

Grouped lineages of ASCs between sample replicates, cell populations and individual subjects are shown under different analytical requirements: (a) Circos plots are used to display connectivity between: a single CD138- ASC sample split into 3 separate fractions; a CD138+ ASC sample from the same individual (FLU 4, gray); and one CD138- ASC sample from a different individual (FLU 1, blue). Percentages in the upper left corner of each Circos plot indicate the level of CDR3 sequence similarity used to assign membership within the same clone. The two right-hand plots also require identical junction regions, as identified by the 3bp before and after the VH-D split and the 3bp before and after the D-JH split. The clone labeled 1 in the 85% threshold plot is split into multiple clones (1a, 1b, and 1c) when the identical junction requirement is used. (b) Plot of lineage sizes of two replicates of CD138- ASCs from FLU 4 based on 85% shows a high correlation between the sizes of the largest clones in each data set. Deviation from similar sized clones within the pair of data sets primarily occurs in small clones, which would be more prone to reflect sampling and/or sequencing error. The high correlation of large clones within replicates further validates the ability to identify expanded clones in the circulating B cell and ASC populations. Blue numbers indicate number of sequences per clone (non-log) for very small clones.

Source data

Supplementary Figure 3 Effect of the requirement for identical V-D and D-J junctions on the clonal identification metric.

(a) Alignment of a sample of VH sequences from the top FLU 4 CD138- ASC clone defined in Supplementary Fig. 2a using a requirement of 85% HCDR3 identity. Consistent with a shared clonal origin, this alignment demonstrates both shared as well as unique mutations in a step-wise fashion within VH rearrangements that share a highly conserved HCDR3 including conserved VH-D junction. However, the added requirement of complete conservation of both the VH-D and D-JH junction splits these sequences into 3 separate clones. Red indicates base differences from germline VH4-38 and differences within the clonally related HCDR3 sequences. (b) Alignments of example sequences from each of the lineages in a, focusing on CDR3 and surrounding region to illustrate the degree of HCDR3 and junctional conservation.

Source data

Supplementary Figure 4 Distribution of lineage sizes for 13 samples.

Lineages are lined up in size order from bottom to top along the extent of the y-axis representing 100% of all the sequences. Horizontal lines delineate the individual lineages. The x-axis is the normalized lineage size (percentage of the total number of sequences). Identical sequences are included in lineage composition.

Supplementary Figure 5 Distribution of lineage sizes for 13 samples.

Lineages are lined up in size order from bottom to top along the extent of the y-axis representing 100% of all the sequences. Horizontal lines delineate the individual lineages. The x-axis is the normalized lineage size (percentage of the total number of sequences). Identical sequences are included in lineage composition.

Supplementary Figure 6 VH4-34+ ASC clonal lineages at multiple time points following a flare.

Time points are from patient SLE-3 at early flare and 5, 6, 7 and 8 weeks thereafter. Each vertical bar represents the repertoire of ASC (CD138− and CD138+) at a single time point, with lineages ordered from smallest to largest, and the y-axis representing the cumulative % of sequences. The largest 10 VH4-34 clones found at active flare are indicated by colored curves connecting corresponding lineages at multiple time points. All but one of these were found at all five time points and in all cases, the abundance of these VH4-34 clones within the most expanded clones diminishes beyond the early flare.

Source data

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6, Supplementary Table, and Supplementary Note (PDF 7191 kb)

Supplementary Data Set 1

Bioinformatics pipeline for the analysis of NGS data. (ZIP 834 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tipton, C., Fucile, C., Darce, J. et al. Diversity, cellular origin and autoreactivity of antibody-secreting cell population expansions in acute systemic lupus erythematosus. Nat Immunol 16, 755–765 (2015). https://doi.org/10.1038/ni.3175

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3175

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing