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:

Regulation of age-associated B cells by IRF5 in systemic autoimmunity

Abstract

Age-associated B cells (ABCs) are a subset of B cells dependent on the transcription factor T-bet that accumulate prematurely in autoimmune settings. The pathways that regulate ABCs in autoimmunity are largely unknown. SWAP-70 and DEF6 (also known as IBP or SLAT) are the only two members of the SWEF family, a unique family of Rho GTPase–regulatory proteins that control both cytoskeletal dynamics and the activity of the transcription factor IRF4. Notably, DEF6 is a newly identified human risk variant for systemic lupus erythematosus. Here we found that the lupus syndrome that developed in SWEF-deficient mice was accompanied by the accumulation of ABCs that produced autoantibodies after stimulation. ABCs from SWEF-deficient mice exhibited a distinctive transcriptome and a unique chromatin landscape characterized by enrichment for motifs bound by transcription factors of the IRF and AP-1 families and the transcription factor T-bet. Enhanced ABC formation in SWEF-deficient mice was controlled by the cytokine IL-21 and IRF5, whose variants are strongly associated with lupus. The lack of SWEF proteins led to dysregulated activity of IRF5 in response to stimulation with IL-21. These studies thus elucidate a previously unknown signaling pathway that controls ABCs in autoimmunity.

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

Access options

Buy this article

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

Fig. 1: Spontaneous expansion of the ABC population in DKO mice.
Fig. 2: IL-21 regulates the generation of DKO ABCs in vitro and in vivo
Fig. 3: DKO ABCs exhibit a distinctive transcriptome.
Fig. 4: The chromatin landscape of DKO ABCs shows enrichment for IRF and AP-1-BATF motifs.
Fig. 5: Wild-type and DKO ABCs exhibit distinct transcriptional and chromatin profiles.
Fig. 6: IRF5 regulates the IL-21-mediated formation of DKO ABCs.
Fig. 7: Enhanced binding of IRF5 to ABC regulatory regions in the absence of SWEF proteins.
Fig. 8: Monoallelic deletion of Irf5 abolishes the accumulation of ABCs and lupus development in DKO mice.

Similar content being viewed by others

References

  1. Liu, Z. & Davidson, A. Taming lupus-a new understanding of pathogenesis is leading to clinical advances. Nat. Med. 18, 871–882 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Rubtsov, A. V. et al. 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 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hao, Y., O’Neill, P., Naradikian, M. S., Scholz, J. L. & Cancro, M. P. A B-cell subset uniquely responsive to innate stimuli accumulates in aged mice. Blood 118, 1294–1304 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Naradikian, M. S., Hao, Y. & Cancro, M. P. Age-associated B cells: key mediators of both protective and autoreactive humoral responses. Immunol. Rev. 269, 118–129 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Naradikian, M. S. et al. Cutting Edge: IL-4, IL-21, and IFN-γ interact to govern T-bet and CD11c expression in TLR-activated B cells. J. Immunol. 197, 1023–1028 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rubtsova, K., Rubtsov, A. V., van Dyk, L. F., Kappler, J. W. & Marrack, P. 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 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Russell Knode, L. M. et al. Age-associated B cells express a diverse repertoire of VH and Vκ genes with somatic hypermutation. J. Immunol. 198, 1921–1927 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Rubtsov, A. V., Rubtsova, K., Kappler, J. W. & Marrack, P. TLR7 drives accumulation of ABCs and autoantibody production in autoimmune-prone mice. Immunol. Res. 55, 210–216 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wei, C. et al. A new population of cells lacking expression of CD27 represents a notable component of the B cell memory compartment in systemic lupus erythematosus. J. Immunol. 178, 6624–6633 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Sanz, I., Wei, C., Lee, F. E. & Anolik, J. Phenotypic and functional heterogeneity of human memory B cells. Semin. Immunol. 20, 67–82 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rubtsova, K. et al. B cells expressing the transcription factor T-bet drive lupus-like autoimmunity. J. Clin. Invest. 127, 1392–1404 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Rogatsky, I., Chandrasekaran, U., Manni, M., Yi, W. & Pernis, A. B. Epigenetics and the IRFs: a complex interplay in the control of immunity and autoimmunity. Autoimmunity 47, 242–255 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Matta, B., Song, S., Li, D. & Barnes, B. J. Interferon regulatory factor signaling in autoimmune disease. Cytokine 98, 15–26 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Murphy, T. L., Tussiwand, R. & Murphy, K. M. Specificity through cooperation: BATF-IRF interactions control immune-regulatory networks. Nat. Rev. Immunol. 13, 499–509 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Lazzari, E. & Jefferies, C. A. IRF5-mediated signaling and implications for SLE. Clin. Immunol. 153, 343–352 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Cham, C. M., Ko, K. & Niewold, T. B. Interferon regulatory factor 5 in the pathogenesis of systemic lupus erythematosus. Clin. Dev. Immunol. 2012, 780436 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Yasuda, K. et al. Interferon regulatory factor-5 deficiency ameliorates disease severity in the MRL/lpr mouse model of lupus in the absence of a mutation in DOCK2. PLoS One 9, e103478 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Ban, T. et al. Lyn kinase suppresses the transcriptional activity of IRF5 in the TLR-MyD88 pathway to restrain the development of autoimmunity. Immunity 45, 319–332 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Savitsky, D. A., Yanai, H., Tamura, T., Taniguchi, T. & Honda, K. Contribution of IRF5 in B cells to the development of murine SLE-like disease through its transcriptional control of the IgG2a locus. Proc. Natl. Acad. Sci. USA 107, 10154–10159 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Watkins, A. A. et al. IRF5 deficiency ameliorates lupus but promotes atherosclerosis and metabolic dysfunction in a mouse model of lupus-associated atherosclerosis. J. Immunol. 194, 1467–1479 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ikushima, H., Negishi, H. & Taniguchi, T. The IRF family transcription factors at the interface of innate and adaptive immune responses. Cold Spring Harb. Symp. Quant. Biol. 78, 105–116 (2013).

    Article  PubMed  Google Scholar 

  22. Shen, H. et al. Gender-dependent expression of murine Irf5 gene: implications for sex bias in autoimmunity. J. Mol. Cell Biol. 2, 284–290 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fang, C. M. et al. Unique contribution of IRF-5-Ikaros axis to the B-cell IgG2a response. Genes Immun. 13, 421–430 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hotfilder, M., Baxendale, S., Cross, M. A. & Sablitzky, F. Def-2, -3, -6 and -8, novel mouse genes differentially expressed in the haemopoietic system. Br. J. Haematol. 106, 335–344 (1999).

    Article  CAS  PubMed  Google Scholar 

  25. Tanaka, Y. et al. SWAP-70-like adapter of T cells, an adapter protein that regulates early TCR-initiated signaling in Th2 lineage cells. Immunity 18, 403–414 (2003).

    Article  CAS  PubMed  Google Scholar 

  26. Gupta, S. et al. Molecular cloning of IBP, a SWAP-70 homologous GEF, which is highly expressed in the immune system. Hum. Immunol. 64, 389–401 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Borggrefe, T., Wabl, M., Akhmedov, A. T. & Jessberger, R. A B-cell-specific DNA recombination complex. J. Biol. Chem. 273, 17025–17035 (1998).

    Article  CAS  PubMed  Google Scholar 

  28. Bécart, S. SpringerAmpamp; Altman, A. SWAP-70-like adapter of T cells: a novel Lck-regulated guanine nucleotide exchange factor coordinating actin cytoskeleton reorganization and Ca2+ signaling in T cells. Immunol. Rev 232, 319–333 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Biswas, P. S., Bhagat, G. & Pernis, A. B. IRF4 and its regulators: evolving insights into the pathogenesis of inflammatory arthritis? Immunol. Rev. 233, 79–96 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ripich, T. et al. SWEF proteins distinctly control maintenance and differentiation of hematopoietic stem cells. PLoS One 11, e0161060 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Sun, C. et al. High-density genotyping of immune-related loci identifies new SLE risk variants in individuals with Asian ancestry. Nat. Genet. 48, 323–330 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Biswas, P. S. et al. Dual regulation of IRF4 function in T and B cells is required for the coordination of T-B cell interactions and the prevention of autoimmunity. J. Exp. Med. 209, 581–596 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Vinuesa, C. G. & Chang, P. P. Innate B cell helpers reveal novel types of antibody responses. Nat. Immunol. 14, 119–126 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Cannons, J. L., Tangye, S. G. & Schwartzberg, P. L. SLAM family receptors and SAP adaptors in immunity. Annu. Rev. Immunol. 29, 665–705 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21–29 (2015).

  36. Chen, Q. et al. IRF-4-binding protein inhibits interleukin-17 and interleukin-21 production by controlling the activity of IRF-4 transcription factor. Immunity 29, 899–911 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Manni, M. et al. IRF4-dependent and IRF4-independent pathways contribute to DC dysfunction in lupus. PLoS One 10, e0141927 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Kreher, S. et al. Mapping of transcription factor motifs in active chromatin identifies IRF5 as key regulator in classical Hodgkin lymphoma. Proc. Natl. Acad. Sci. USA 111, E4513–E4522 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dominguez, C. X. et al. The transcription factors ZEB2 and T-bet cooperate to program cytotoxic T cell terminal differentiation in response to LCMV viral infection. J. Exp. Med. 212, 2041–2056 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Barwick, B. G., Scharer, C. D., Bally, A. P. R. & Boss, J. M. Plasma cell differentiation is coupled to division-dependent DNA hypomethylation and gene regulation. Nat. Immunol. 17, 1216–1225 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Karwacz, K. et al. Critical role of IRF1 and BATF in forming chromatin landscape during type 1 regulatory cell differentiation. Nat. Immunol. 18, 412–421 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Audzevich, T., Pearce, G., Breucha, M., Günal, G. & Jessberger, R. Control of the STAT6-BCL6 antagonism by SWAP-70 determines IgE production. J. Immunol. 190, 4946–4955 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Ochiai, K. et al. Transcriptional regulation of germinal center B and plasma cell fates by dynamical control of IRF4. Immunity 38, 918–929 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kang, K. et al. Interferon-γ represses M2 gene expression in human macrophages by disassembling enhancers bound by the transcription factor MAF. Immunity 47, 235–250 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Carotta, S. et al. The transcription factors IRF8 and PU.1 negatively regulate plasma cell differentiation. J. Exp. Med. 211, 2169–2181 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Lu, D. et al. The miR-155-PU.1 axis acts on Pax5 to enable efficient terminal B cell differentiation. J. Exp. Med. 211, 2183–2198 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sarra, M., Pallone, F. & Monteleone, G. Interleukin-21 in chronic inflammatory diseases. Biofactors 39, 368–373 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Eames, H. L., Corbin, A. L. & Udalova, I. A. Interferon regulatory factor 5 in human autoimmunity and murine models of autoimmune disease. Transl. Res. 167, 167–182 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Claes, N. et al. Age-associated B cells with proinflammatory characteristics are expanded in a proportion of multiple sclerosis patients. J. Immunol. 197, 4576–4583 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Wang, Z. et al. T-bet-expressing B cells are positively associated with Crohn’s disease activity and support Th1 inflammation. DNA Cell Biol. 35, 628–635 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Biswas, P. S. et al. Phosphorylation of IRF4 by ROCK2 regulates IL-17 and IL-21 production and the development of autoimmunity in mice. J. Clin. Invest. 120, 3280–3295 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Biswas, P. S., Kang, K., Gupta, S., Bhagat, G. & Pernis, A. B. A murine autoimmune model of rheumatoid arthritis and systemic lupus erythematosus associated with deregulated production of IL-17 and IL-21. Methods Mol. Biol 900, 233–251 (2012).

    Article  CAS  PubMed  Google Scholar 

  53. Minnich, M. et al. Multifunctional role of the transcription factor Blimp-1 in coordinating plasma cell differentiation. Nat. Immunol. 17, 331–343 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank I. Rogatsky (Hospital for Special Surgery) for expression constructs encoding full-length wild-type human IRF5 in the plasmid pcDNA3P, and the late P. Pitha-Rowe (Johns Hopkins University) for Irf5fl/fl mice. Supported by the Barbara Volcker Center and a gift made in honor of Anne Kennedy O’Neil (M.M.), the US National Institutes of Health (NIH F31 Ruth L. Kirschstein National Service Award to E.R.; AR064883 and AR070146 to A.B.P; and AI044938 to L.B.I.), the Peter Jay Sharp Foundation, the Tow Foundation (support for the David Z. Rosensweig Genomics Research Center), Giammaria Giuliani, the Ambrose Monell Foundation and the Epigenomics Core and the Flow Cytometry Core Facility of Weill Cornell Medical College (Office of the Director of the National Institutes of Health under Award Number S10OD019986 to Hospital for Special Surgery; technical support).

Author information

Authors and Affiliations

Authors

Contributions

M.M. designed and performed the experiments, interpreted the experiments and wrote the manuscript; S.G., E.R. and M.S. performed the experiments; Y.C. conducted the RNA-seq bioinformatics analysis; S.H.P. conducted the ATAC-seq bioinformatics analysis; T.P. assisted with the histological analysis; R.J. generated the Swap70−/− mice and helped write the manuscript; L.B.I. supervised the bioinformatics analysis and helped write the manuscript; and A.B.P. designed and supervised the study, interpreted the experiments, and wrote the manuscript.

Corresponding author

Correspondence to Alessandra B. Pernis.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Spontaneous population expansion of ABCs in DKO mice

(a) Spleens of WT and DKO female mice (>20 weeks old) were evaluated by flow cytometry for the presence of plasmacytoid dendritic cells. Representative FACS plots for CD11c and PDCA-1 expression after gating on B220+ or B220+CD19+ cells. Graphs show the frequencies and numbers for individual mice and mean value of 3 independent experiments (n = 3 WT and 5 DKO). ns: not significant. (two-tailed Student-t test). (b) Representative FACS plots for CD11b and CD11c expression after gating on B220+CD19+ cells in the spleens of 10 weeks old WT and DKO female mice. Graphs show frequencies and numbers of individual mice and mean value of 2 independent experiments (n = 4 WT and 5 DKO). ** P = 0.0011; *** P = 0.0001. (two-tailed Student-t test). (c) Representative FACS plots for CD11b and CD11c expression after gating on B220+CD19+ cells in skin draining lymph nodes of WT, DKO, Def6–/– and Swap70–/– mice (18–24 weeks old). Graphs show frequencies and numbers of individual mice and mean value of 3 independent experiments (n = 3 WT, 5 DKO, 5 Def6–/–, 4 Swap70–/–). ** P = 0.0018 (WT vs. DKO), P = 0.0011 (DKO vs. Def6–/–), P = 0.0055 (DKO vs. Swap70–/–) (One-way ANOVA followed by Bonferroni’s multiple comparisons test). (d) Representative FACS plots for CD11c and T-bet expression gated on B220+ cells or B220+CD19+ cells. Graphs show frequencies and numbers for individual mice and mean values of 3 independent experiments (n = 3 WT, 5 DKO). * P = 0.0137 (B220+) and P = 0.0131 (B220+CD19+); ** P = 0.068 (B220+) and P = 0.0075 (B220+CD19+). (two-tailed Student-t test). (e) Representative FACS plots for IgG1 and IgG2c expression on B220+ CD19+CD11c+CD11b+ cells from WT and DKO female mice (>20 weeks old). Graphs show frequencies and numbers for individual mice and mean values of 16 independent experiments, (n = 17 WT and 30 DKO). ** P = 0.0062; *** P = 0.0006; **** P < 0.0001. (two-tailed Student-t test). (f) Gating strategy employed to sort ABC and FoB cells.

Supplementary Figure 2 IL-21 regulates the generation of DKO ABCs in vitro and in vivo

(a) Generation of ABCs (B220+CD11c+CD11b+) B cells from cultures of CD23+ B cells purified from WT and DKO female mice (8–10 weeks of age) stimulated with αIgM (5 μg/ml), αCD40 (5 μg/ml), IL-21 (50 ng/ml) or imiquimod (1 μg/ml) for 3 days as assessed by flow cytometry. Representative FACS plot is shown. Graph shows mean and individual value of 5 independent experiments (n = 5 cell culture). *** P = 0.0001. (One-way ANOVA followed by Bonferroni’s multiple comparisons test). (b) Generation of ABCs (B220+CD11c+T-bet+) B cells from cultures of CD23+ B cells purified from WT and DKO female mice (8–10 weeks of age) and stimulated with various combinations of αIgM (5 μg/ml), αCD40 (5 μg/ml), IL-21 (50 ng/ml), IL-4 (10 ng/ml), or IFN-γ (20 ng/ml) as indicated. Representative FACS plot is shown. Graph shows mean and individual values of 3 independent experiments (n = 3 cell cultures). **** P < 0.0001. (One-way ANOVA followed by Bonferroni’s multiple comparisons test). (c-e) Representative FACS plots showing the presence of TFH cells (CD4+CXCR5+PD1+Foxp3-), germinal center (GC) B cells (B220+FAS+GL-7+) and B220intCD138+ plasma cells (PC) in the spleens of WT, DKO, IL21–/– DKO, and SAP–/– DKO female mice (>24 weeks old). (f) Quantification of TFH, GC, and PC of c,d,e. Graphs show percentages and numbers of specific cells types in individual mice and mean of 4 independent experiments (n = 4 WT, 4–8 DKO, 7 IL21–/– DKO, 7 SAP–/– DKO). * P < 0.05; ** P < 0.01; *** P < 0.001; **** P < 0.0001. (One-way ANOVA followed by Bonferroni’s multiple comparisons test).

Supplementary Figure 3 DKO ABCs exhibit a distinctive transcriptome

(a) Selected GSEA Hallmark pathways analysis of WT FoB compared to DKO FoB cells (n = 2 WT and 3 DKO/group). (b) Apoptotic rates as measured by caspase 3 cleavage in B220+CD11cT-betB cells from WT and DKO mice (left panel) or in CD11cT-bet B cells and CD11c+T-bet+ (ABC) B cells from DKO mice (Right panel). Shown is a representative histogram. All data are representative of 3 independent experiments. (n = 4 mice/group). (c) Proliferation of B220+CD11cT-betcells and ABCs (B220+CD11c+T-bet+) was assessed by evaluating dilution of cell trace violet (CTV) by flow cytometry. CD23+ B cells were purified from WT and DKO female mice (6–9 weeks old), labeled with CTV and cultured with αIgM (5 μg/ml), αCD40 (5 μg/ml), and IL-21 (50 ng/ml) for 3 days. Shown is a representative histogram of 5 independent experiments. (n = 5 cell cultures). (d) Cell viability of CD23+ B cells purified from WT and DKO female mice (6–9 weeks old) and stimulated with αIgM (5 μg/ml), αCD40 (5 μg/ml), IL-21 (50 ng/ml) or imiquimod (1 μg/ml) for 3 or 5 days was assessed by staining with CaspGlow or propidium iodide as indicated. Graphs show percentages of live cells +SEM in 3 independent experiments (n = 3 cell cultures). (One-way ANOVA followed by Bonferroni’s multiple comparisons test) (e) Viability was assessed as in d in CD23+B cells stimulated with αIgM (5 μg/ml), αCD40 (5 μg/ml) and IL-21 (50 ng/ml) and gated on CD11c+ or CD11c- cells. Graphs show percentages of live cells +SEM in 3 independent experiments (n = 3 cell cultures). (One-way ANOVA followed by Bonferroni’s multiple comparisons test) (f) Selected GSEA pathways analysis of DKO FoB compared to DKO ABC cells. (n = 3 mice/group).

Supplementary Figure 4 The chromatin landscape of DKO ABCs shows enrichment for IRF and AP-1–BATF motifs

(a) Expression (RNAseq) of total ABC-specific peaks associated genes (n = 2,482) from three independent experiments (n = 3) (b) Functionally enriched Gene Ontology (GO) categories of selected genes associated with ABC-specific peaks of ATAC-seq (n = 487). (c) qPCR analysis of the expression of Il6 mRNA in sorted FoB (B220+CD19+CD11c-CD11b-CD23+) cells from WT and DKO female mice and ABC (B220+CD19+CD11c+CD11b+) cells from DKO female mice as indicated. The data were normalized relative to Ppia mRNA expression. Mean of one representative experiment of 2 independent experiments is shown (n = 2 mice/group). SEM of technical replicates is shown. ** P = 0.0052.

Supplementary Figure 5 WT and DKO ABCs exhibit distinct transcriptional and chromatin profiles

(a) Expression of selected transcription factors in WT and DKO ABC cells as identified by RNA-seq analysis. (n = 2 mice/group). (b) Genomic distribution of WT ABC-specific and DKO ABC-specific peaks of ATAC-seq relative to annotated genomic features. (n = 2 mice/group) (c) Normalized ATAC-seq tag distributions tracks for representative genomic regions at Maf, Mafb and Znhit1 genes. Highlighted are WT ABC or DKO ABC-specific ATAC-seq peaks (n = 2 mice/group).

Supplementary Figure 6 IRF5 regulates the IL-21-mediated formation of DKO ABCs

(a) Flow cytometric analysis of B220+CD11c+CD11b+B cells in the spleens of WT, DKO and Cd11c-Cre Irf4fl/fl DKO female mice (>18 weeks-old). Graphs show frequencies and numbers for individual mice and mean values of 4 independent experiments (n = 4 WT, 5 DKO, 6 Cd11c-Cre Irf4fl/fl DKO). ns: not significant; * P = 0.0485; ** P = 0.0072. (One-way ANOVA followed by Bonferroni’s multiple comparisons test). (b) qPCR analysis of the expression of Irf5 mRNA in cultures of CD23+ B cells purified from WT, DKO and Cd21-Cre Irf5fl/– DKO female mice (8–10 weeks of age) stimulated with αIgM (5 μg/ml) and αCD40 (5 μg/ml). Data were normalized relative to Ppia mRNA expression. Mean of one representative experiment of 4 independent experiments is shown (n = 4 mice/group). SEM of technical replicates is shown. ns: not significant. (One-way ANOVA followed by Bonferroni’s multiple comparisons test).

Supplementary Figure 7 Enhanced binding of IRF5 to ABC regulatory regions in the absence of the SWEF proteins

(a) ChIP assays were performed with either an IRF5 antibody or a T-bet antibody on CD23+ B cells purified from WT, DKO, and Cd21-Cre Irf5fl/– DKO mice and stimulated with αIgM (5 μg/ml) and αCD40 (5 μg/ml) (top panel) or with αIgM (5 μg/ml), αCD40 (5 μg/ml) and IL-21 (50 ng/ml) (bottom panel) for 2 days. qPCR for the ABC specific ATAC-seq peak at the Il6 TSS or the Zeb2 Exon 8 locus was performed. Data are representative of 2 independent experiments. Mean ± SEM is shown (n = 2 mice/grup). ns: not significant; ** P = 0.0079. (One-way ANOVA followed by Bonferroni’s multiple comparisons test). (b) Nuclear extracts were prepared from CD23+ B cells purified from WT, DKO and Cd21-Cre Irf5fl/– DKO female mice (8–10 weeks of age) stimulated with αIgM (5 μg/ml), αCD40 (5 μg/ml) +/− IL-21 (50 ng/ml) or imiquimod (1 μg/ml) for 3 days. Extracts were analyzed by immunoblotting with pSTAT3, STAT3, IRF5, and HDAC1 antibodies. Data are representative of 2 independent experiments. (c) Quantification of immunoblot in (b) by densitometry. P-STAT3, STAT3 and IRF5 were normalized to HDAC1 density. Mean and individual values of 2 independent experiments is shown. (d) Nuclear extracts were prepared from cells stimulated as in a. for 2 days and subjected to ONP assay with a biotinylated oligonucleotide from the Il6 TSS. Precipitated proteins were analyzed by immunoblotting with IRF5 and T-bet antibodies. Data are representative of 2 independent experiments. (e) Binding of IRF5 to the ONP shown in Fig. 7d. 293T cells were transiently transfected with various IRF5 and T-bet constructs as indicated. Nuclear extracts were prepared and subjected to ONP assay with a biotinylated oligonucleotide from the Cxcl10 cluster. Precipitated proteins were analyzed by immunblotting with a FLAG antibody. Data are representative of 2 independent experiments. (f) and (g) 293T cells were transiently transfected with various DEF6, SWAP-70, and IRF5 constructs as indicated. Immunoprecipitations were performed using an anti-HA antibody. Immunoprecipitates were analyzed by immunoblotting using an anti-IRF5 or anti-HA antibody. Data are representative of 2 independent experiments with similar results. (h), (i), and (j) 293T cells were transiently transfected with various constructs as indicated. Immunoprecipitations were performed using an anti-HA antibody. Immunoprecipitates were analyzed by immunoblotting using an anti-FLAG, T-bet, DEF6 or HA antibodies. Data are representative of 2 independent experiments with similar results.

Supplementary Figure 8 Monoallelic deletion of Irf5 abolishes the accumulation of ABCs and lupus development in DKO mice

(a,b) Percentage and numbers of B220+CD19+CD11c+T-bet+ (a) and of B220+CD19+CD21CD23CD11c+CD11b+ (b) B cells in the spleens of WT, Irf5fl/fl DKO, Irf5fl/– DKO, Cd11c-Cre Irf5fl/– DKO and Cd21-Cre Irf5fl/– DKO female mice (>20 weeks-old). Graphs show the frequencies and numbers for individual mice and mean value of 6 independent experiments (n = 7 WT, 5 Irf5fl/fl DKO, 6 Irf5fl/– DKO, 5 Cd11c-Cre Irf5fl/– DKO and 6 Cd21-Cre Irf5fl/– DKO). *** P = 0.0001; **** P < 0.0001. (One-way ANOVA followed by Bonferroni’s multiple comparisons test). (c) Flow cytometric analysis of CD11c+CD11b+ B cells in the spleens of WT, Irf5fl/fl DKO and Cd11c-Cre Irf5fl/– DKO female mice (>20 weeks-old). Representative FACS plots of 10 independent experiments is shown. (d) Spleen size of indicated mice. Graph shows numbers of individual mice and mean value of 10 independent experiments. (n = 9 WT, 10 Irf5fl/fl DKO, 6 Irf5fl/– DKO, 10 Cd11c-Cre Irf5fl/– DKO and 5 Cd21-Cre Irf5fl/– DKO female mice >20 weeks of age). * P = 0.0254; *** P = 0.0004; **** P < 0.0001. (One-way ANOVA followed by Bonferroni’s multiple comparison test). (e) Quantification of flow cytometric analysis of TFH (CD4+CXCR5+PD1+Foxp3), GC (B220+FAS+GL-7+), and PC (B220intCD138+) in spleens of WT, Irf5fl/fl DKO, Irf5fl/– DKO, Cd11c-Cre Irf5fl/– DKO and Cd21-Cre Irf5fl/– DKO female mice (>20 weeks). Graphs show percentages and numbers of specific cells types in individual mice and mean value of 10 independent experiments (n = 10 WT, 10 Irf5fl/fl DKO, 6 Irf5fl/– DKO, 10 Cd11c-Cre Irf5fl/– DKO and 5 Cd21-Cre Irf5fl/– DKO female mice). ** P < 0.01; *** P < 0.001; **** P < 0.0001. (One-way ANOVA followed by Bonferroni’s multiple comparisons test). (f) Quantification of flow cytometric analysis of Treg (CD4+Foxp3+), activated Treg (CD4+Foxp3+CD44+), and activated T cells (CD4+Foxp3CD44+) in spleens of WT, Irf5fl/fl DKO, Irf5fl/– DKO, Cd11c-Cre Irf5fl/– DKO and Cd21-Cre Irf5fl/– DKO female mice (>20 weeks). Graphs show frequencies and numbers of individual mice and mean value of 10 independent experiments (n = 9 WT, 10 Irf5fl/fl DKO, 6 Irf5fl/– DKO, 10 Cd11c-Cre Irf5fl/– DKO and 5 Cd21-Cre Irf5fl/– DKO female mice). ** P < 0.01; *** P < 0.001; **** P < 0.0001. (One-way ANOVA followed by Bonferroni’s multiple comparisons test).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8

Life Sciences Reporting Summary

Supplementary Table 1

Differentially expressed genes in WT and DKO ABC

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Manni, M., Gupta, S., Ricker, E. et al. Regulation of age-associated B cells by IRF5 in systemic autoimmunity. Nat Immunol 19, 407–419 (2018). https://doi.org/10.1038/s41590-018-0056-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0056-8

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