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:

Genetic dissection of TLR9 reveals complex regulatory and cryptic proinflammatory roles in mouse lupus

Abstract

In lupus, Toll-like receptor 7 (TLR7) and TLR9 mediate loss of tolerance to RNA and DNA, respectively. Yet, TLR7 promotes disease, while TLR9 protects from disease, implying differences in signaling. To dissect this ‘TLR paradox’, we generated two TLR9 point mutants (lacking either ligand (TLR9K51E) or MyD88 (TLR9P915H) binding) in lupus-prone MRL/lpr mice. Ameliorated disease of Tlr9K51E mice compared to Tlr9−/− controls revealed a TLR9 ‘scaffold’ protective function that is ligand and MyD88 independent. Unexpectedly, Tlr9P915H mice were more protected than both Tlr9K51E and Tlr9WT mice, suggesting that TLR9 also possesses ligand-dependent, but MyD88-independent, regulatory signaling and MyD88-mediated proinflammatory signaling. Triple-mixed bone marrow chimeras showed that TLR9–MyD88-independent regulatory roles were B cell intrinsic and restrained differentiation into pathogenic age-associated B cells and plasmablasts. These studies reveal MyD88-independent regulatory roles of TLR9, shedding light on the biology of endosomal TLRs.

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: TLR9 has a scaffold protective role in lupus.
Fig. 2: TLR9–MyD88 signaling promotes disease.
Fig. 3: TLR9–MyD88-independent protection can be ligand dependent.
Fig. 4: Effects of TLR9 and sex on TLR7 expression and distribution.
Fig. 5: TLR9 expression does not impact TLR7 signaling.
Fig. 6: TLR9–MyD88-independent functions are B cell intrinsic.
Fig. 7: TLR9 controls ABC gene programs independently of MyD88.
Fig. 8: Tlr9P915H and Tlr9−/− induce distinct ABC and PB states.

Similar content being viewed by others

Data availability

RNA-seq data of the TLR7-induced gene signature and of the in vitro TLR7-stimulated BALB/c B cells are deposited in the NCBI’s Gene Expression Omnibus (GEO) database and are publicly available under accession numbers GSE202108 and GSE202105.

All ATAC-seq data have been deposited in the NCBI GEO database and are publicly available under accession number GSE202103.

RNA-seq data of the sorted FO cells, MZ cells and ABCs across Tlr9 genotypes from the three-way bone marrow chimeric mice are deposited in the NCBI’s GEO and are publicly available under accession number GSE181283.

The mm10 genome database (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20/) was used to align sequences for the RNA-seq analysis and to align sequencing reads for the ATAC-seq analysis. Source data are provided with this paper.

References

  1. Tsokos, G. C., Lo, M. S., Costa Reis, P. & Sullivan, K. E. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat. Rev. Rheumatol. 12, 716–730 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Marshak-Rothstein, A. Toll-like receptors in systemic autoimmune disease. Nat. Rev. Immunol. 6, 823–835 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Teichmann, L. L., Schenten, D., Medzhitov, R., Kashgarian, M. & Shlomchik, M. J. Signals via the adaptor MyD88 in B cells and DCs make distinct and synergistic contributions to immune activation and tissue damage in lupus. Immunity 38, 528–540 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Christensen, S. R. et al. Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing inflammatory and regulatory roles in a murine model of lupus. Immunity 25, 417–428 (2006).

    Article  CAS  PubMed  Google Scholar 

  5. Bossaller, L. et al. TLR9 deficiency leads to accelerated renal disease and myeloid lineage abnormalities in pristane-induced murine lupus. J. Immunol. 197, 1044–1053 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Fairhurst, A. M. et al. Yaa autoimmune phenotypes are conferred by overexpression of TLR7. Eur. J. Immunol. 38, 1971–1978 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Nickerson, K. M., Wang, Y., Bastacky, S. & Shlomchik, M. J. Toll-like receptor 9 suppresses lupus disease in Fas-sufficient MRL mice. PLoS ONE 12, e0173471 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Jackson, S. W. et al. Opposing impact of B cell-intrinsic TLR7 and TLR9 signals on autoantibody repertoire and systemic inflammation. J. Immunol. 192, 4525–4532 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Scofield, R. H. et al. Klinefelter’s syndrome (47,XXY) in male systemic lupus erythematosus patients: support for the notion of a gene-dose effect from the X chromosome. Arthritis Rheum. 58, 2511–2517 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Lee, Y. H., Choi, S. J., Ji, J. D. & Song, G. G. Association between Toll-like receptor polymorphisms and systemic lupus erythematosus: a meta-analysis update. Lupus 25, 593–601 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Garcia-Ortiz, H. et al. Association of TLR7 copy number variation with susceptibility to childhood-onset systemic lupus erythematosus in Mexican population. Ann. Rheum. Dis. 69, 1861–1865 (2010).

    Article  CAS  PubMed  Google Scholar 

  12. Brown, G. J. et al. TLR7 gain-of-function genetic variation causes human lupus. Nature 605, 349–356 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nickerson, K. M. et al. TLR9 regulates TLR7- and MyD88-dependent autoantibody production and disease in a murine model of lupus. J. Immunol. 184, 1840–1848 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. Kim, Y. M., Brinkmann, M. M., Paquet, M. E. & Ploegh, H. L. UNC93B1 delivers nucleotide-sensing Toll-like receptors to endolysosomes. Nature 452, 234–238 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Fukui, R. et al. Unc93B1 biases Toll-like receptor responses to nucleic acid in dendritic cells toward DNA- but against RNA-sensing. J. Exp. Med. 206, 1339–1350 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Fukui, R. et al. Unc93B1 restricts systemic lethal inflammation by orchestrating Toll-like receptor 7 and 9 trafficking. Immunity 35, 69–81 (2011).

    Article  CAS  PubMed  Google Scholar 

  17. Stoehr, A. D. et al. TLR9 in peritoneal B-1b cells is essential for production of protective self-reactive IgM to control TH17 cells and severe autoimmunity. J. Immunol. 187, 2953–2965 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Tilstra, J. S. et al. B cell-intrinsic TLR9 expression is protective in murine lupus. J. Clin. Invest. 130, 3172–3187 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Peter, M. E., Kubarenko, A. V., Weber, A. N. & Dalpke, A. H. Identification of an N-terminal recognition site in TLR9 that contributes to CpG-DNA-mediated receptor activation. J. Immunol. 182, 7690–7697 (2009).

    Article  CAS  PubMed  Google Scholar 

  20. Christensen, S. R. et al. Toll-like receptor 9 controls anti-DNA autoantibody production in murine lupus. J. Exp. Med. 202, 321–331 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nickerson, K. M., Cullen, J. L., Kashgarian, M. & Shlomchik, M. J. Exacerbated autoimmunity in the absence of TLR9 in MRL.Faslpr mice depends on Ifnar1. J. Immunol. 190, 3889–3894 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Poltorak, A. et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 282, 2085–2088 (1998).

    Article  CAS  PubMed  Google Scholar 

  23. Syrett, C. M., Sierra, I., Beethem, Z. T., Dubin, A. H. & Anguera, M. C. Loss of epigenetic modifications on the inactive X chromosome and sex-biased gene expression profiles in B cells from NZB/W F1 mice with lupus-like disease. J. Autoimmun. 107, 102357 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Souyris, M. et al. TLR7 escapes X chromosome inactivation in immune cells. Sci. Immunol. 3, eaap8855 (2018).

    Article  PubMed  Google Scholar 

  25. Azulay-Debby, H., Edry, E. & Melamed, D. CpG DNA stimulates autoreactive immature B cells in the bone marrow. Eur. J. Immunol. 37, 1463–1475 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Nickerson, K. M. et al. TLR9 promotes tolerance by restricting survival of anergic anti-DNA B cells, yet is also required for their activation. J. Immunol. 190, 1447–1456 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Scharer, C. D. et al. Epigenetic programming underpins B cell dysfunction in human SLE. Nat. Immunol. 20, 1071–1082 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang, S. et al. IL-21 drives expansion and plasma cell differentiation of autoreactive CD11chiT-bet+ B cells in SLE. Nat. Commun. 9, 1758 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Saelee, P., Kearly, A., Nutt, S. L. & Garrett-Sinha, L. A. Genome-wide identification of target genes for the key B cell transcription factor Ets1. Front Immunol. 8, 383 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Kikuchi, H., Nakayama, M., Takami, Y., Kuribayashi, F. & Nakayama, T. EBF1 acts as a powerful repressor of Blimp-1 gene expression in immature B cells. Biochem. Biophys. Res. Commun. 422, 780–785 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Schmidlin, H. et al. Spi-B inhibits human plasma cell differentiation by repressing BLIMP1 and XBP-1 expression. Blood 112, 1804–1812 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Willis, S. N. et al. Environmental sensing by mature B cells is controlled by the transcription factors PU.1 and SpiB. Nat. Commun. 8, 1426 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. 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 

  34. Garrett-Sinha, L. A., Kearly, A. & Satterthwaite, A. B. The role of the transcription factor Ets1 in lupus and other autoimmune diseases. Crit. Rev. Immunol. 36, 485–510 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Thomsen, I. et al. RUNX1 regulates a transcription program that affects the dynamics of cell cycle entry of naive resting B cells. J. Immunol. 207, 2976–2991 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Emslie, D. et al. Oct2 enhances antibody-secreting cell differentiation through regulation of IL-5 receptor α chain expression on activated B cells. J. Exp. Med. 205, 409–421 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Scharer, C. D., Barwick, B. G., Guo, M., Bally, A. P. R. & Boss, J. M. Plasma cell differentiation is controlled by multiple cell division-coupled epigenetic programs. Nat. Commun. 9, 1698 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Goel, R. R. et al. Interferon λ promotes immune dysregulation and tissue inflammation in TLR7-induced lupus. Proc. Natl Acad. Sci. USA 117, 5409–5419 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Syedbasha, M. et al. Interferon-λ enhances the differentiation of naive B cells into plasmablasts via the mTORC1 pathway. Cell Rep. 33, 108211 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Huang, W., Ghisletti, S., Perissi, V., Rosenfeld, M. G. & Glass, C. K. Transcriptional integration of TLR2 and TLR4 signaling at the NCoR derepression checkpoint. Mol. Cell 35, 48–57 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Pascual, G. et al. A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-γ. Nature 437, 759–763 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Setoguchi, K. et al. Peroxisome proliferator-activated receptor-γ haploinsufficiency enhances B cell proliferative responses and exacerbates experimentally induced arthritis. J. Clin. Invest. 108, 1667–1675 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Barish, G. D. et al. Bcl-6 and NF-κB cistromes mediate opposing regulation of the innate immune response. Genes Dev. 24, 2760–2765 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Xu, F. et al. Bcl6 sets a threshold for antiviral signaling by restraining IRF7 transcriptional program. Sci. Rep. 6, 18778 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Tan, C. et al. NR4A nuclear receptors restrain B cell responses to antigen when second signals are absent or limiting. Nat. Immunol. 21, 1267–1279 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Carroll, M. C. A protective role for innate immunity in systemic lupus erythematosus. Nat. Rev. Immunol. 4, 825–831 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Nakano-Yokomizo, T. et al. The immunoreceptor adapter protein DAP12 suppresses B lymphocyte-driven adaptive immune responses. J. Exp. Med. 208, 1661–1671 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Matsumura, T., Kawamura-Tsuzuku, J., Yamamoto, T., Semba, K. & Inoue, J. TRAF-interacting protein with a forkhead-associated domain B (TIFAB) is a negative regulator of the TRAF6-induced cellular functions. J. Biochem. 146, 375–381 (2009).

    Article  CAS  PubMed  Google Scholar 

  49. Mashima, R., Hishida, Y., Tezuka, T. & Yamanashi, Y. The roles of Dok family adapters in immunoreceptor signaling. Immunol. Rev. 232, 273–285 (2009).

    Article  CAS  PubMed  Google Scholar 

  50. Nakamura, A. et al. Increased susceptibility to LPS-induced endotoxin shock in secretory leukoprotease inhibitor (SLPI)-deficient mice. J. Exp. Med. 197, 669–674 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sancho, D. & Reis e Sousa, C. Signaling by myeloid C-type lectin receptors in immunity and homeostasis. Annu. Rev. Immunol. 30, 491–529 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Komai, T. et al. Transforming growth factor-β and interleukin-10 synergistically regulate humoral immunity via modulating metabolic signals. Front. Immunol. 9, 1364 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  53. van der Vuurst de Vries, A. R., Clevers, H., Logtenberg, T. & Meyaard, L. Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is differentially expressed during human B cell differentiation and inhibits B cell receptor-mediated signaling. Eur. J. Immunol. 29, 3160–3167 (1999).

    Article  PubMed  Google Scholar 

  54. Colombo, B. M. et al. Defective expression and function of the leukocyte associated Ig-like receptor 1 in B lymphocytes from systemic lupus erythematosus patients. PLoS ONE 7, e31903 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Silva, R. et al. CD300a is expressed on human B cells, modulates BCR-mediated signaling, and its expression is down-regulated in HIV infection. Blood 117, 5870–5880 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Pelka, K. et al. The chaperone UNC93B1 regulates Toll-like receptor stability independently of endosomal TLR transport. Immunity 48, 911–922 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Nundel, K. et al. Cell-intrinsic expression of TLR9 in autoreactive B cells constrains BCR/TLR7-dependent responses. J. Immunol. 194, 2504–2512 (2015).

    Article  PubMed  Google Scholar 

  58. Celhar, T. et al. Toll-like receptor 9 deficiency breaks tolerance to RNA-associated antigens and up-regulates Toll-like receptor 7 protein in Sle1 mice. Arthritis Rheumatol. 70, 1597–1609 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. ten Broeke, T., Wubbolts, R. & Stoorvogel, W. MHC class II antigen presentation by dendritic cells regulated through endosomal sorting. Cold Spring Harb. Perspect. Biol. 5, a016873 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Akkaya, M. et al. Toll-like receptor 9 antagonizes antibody affinity maturation. Nat. Immunol. 19, 255–266 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Sindhava, V. J. et al. A TLR9-dependent checkpoint governs B cell responses to DNA-containing antigens. J. Clin. Invest. 127, 1651–1663 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Sanjuan, M. A. et al. Toll-like receptor signalling in macrophages links the autophagy pathway to phagocytosis. Nature 450, 1253–1257 (2007).

    Article  CAS  PubMed  Google Scholar 

  63. Dinarello, C. A. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol. Rev. 281, 8–27 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Pelletier, S., Gingras, S. & Green, D. R. Mouse genome engineering via CRISPR–Cas9 for study of immune function. Immunity 42, 18–27 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Teichmann, L. L. et al. B cell-derived IL-10 does not regulate spontaneous systemic autoimmunity in MRL.Faslpr mice. J. Immunol. 188, 678–685 (2012).

    Article  CAS  PubMed  Google Scholar 

  66. Blanco, F. K., Isenberg, J. & Analysis, D. A. of antibodies to RNA in patients with systemic lupus erythematosus and other autoimmune rheumatic diseases. Clin. Exp. Immunol. 86, 66–70 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Murakami, Y. et al. The protective effect of the anti-Toll-like receptor 9 antibody against acute cytokine storm caused by immunostimulatory DNA. Sci. Rep. 7, 44042 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  72. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  73. Elsner, R. A. & Shlomchik, M. J. IL-12 blocks TFH cell differentiation during salmonella infection, thereby contributing to germinal center suppression. Cell Rep. 29, 2796–2809 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Marshak-Rothstein (University of Massachusetts School of Medicine) and J. Tilstra for helpful discussions. We thank L. Conter, K. Minjung, M. Price and T. Marinov for technical support. We thank S. Watkins for advice on confocal analysis and Center for Biologic Imaging (CBI) for contributing as provider for instrumentation (funding 1S10OD019973-01). cDNA generation, library preparation and sequencing were performed by the University of Pittsburgh Health Science Sequencing Core at the University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh. This work benefitted from ImageStreamX Mark II funded by NIH 1S10OD019942-01 (principal investigator L. Borghesi). This work was supported by NIH grant R37-AI118841.

Author information

Authors and Affiliations

Authors

Contributions

C.L., S.J. and M.J.S. conceived the project and designed experiments. S.J. designed and validated the Tlr9K51E and Tlr9P915H constructs. S.G. created the mice. C.L. and K.B.T. performed experiments, and C.L. analyzed data. S.S. assisted with ATAC-seq and RNA-seq data analysis. R.A.G., K.M.N. and R.A.E. assisted with data interpretation and experimental design. R.C.L. performed the in vitro RNA-seq experiments. R.A.E. and S.J. designed and performed the multiparametric flow cytometry panels. D.J.C. performed the ATAC-seq analysis. S.B. conducted pathologic analysis of the kidney tissue. R.F. and K.M. provided TLR9 antibody (Nar9 clone). C.L. and M.J.S. wrote the manuscript. M.J.S. supervised the study.

Corresponding author

Correspondence to Mark J. Shlomchik.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Marcus Clark and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. L.A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Additional information

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

Extended data

Extended Data Fig. 1 TLR9 has a scaffold protective role in lupus.

a-b, Proportion of activated CD21int (a) or CD21hi (b) B cells with nuclear NF-κB (black gate, Fig. 1c) before (black bar) and after (red bar) stimulation. Five week old MRL/lpr mice, n = 3 per group, (* p < 0.05; *** p < 0.001, two-tailed paired-t test between the unstimulated and stimulated conditions for each Tlr9 genotype; (a) Tlr9 WT unstimulated versus stimulated p = 0.0007, Tlr9K51E/K51E unstimulated versus stimulated p = 0.0452, (b) Tlr9 WT unstimulated versus stimulated p = 0.0013, Tlr9K51E/K51E unstimulated versus stimulated p = 0.0480; # p < 0.05, ### p < 0.001, one-way ANOVA between the stimulated conditions). c-g, Evaluation of the clinical parameters, separated by gender (Tlr9WT male, n = 54; Tlr9−/− male, n = 21; Tlr9K51E/K51E male, n = 15; Tlr9WT female, n = 46; Tlr9−/− female, n = 22; Tlr9K51E/K51E female, n = 16; (c female) Tlr9WT versus Tlr9−/− p < 0.0001, Tlr9−/− versus Tlr9K51E/K51E p = 0.0001; (d) Tlr9WT versus Tlr9−/− p = 0.0056, Tlr9−/− versus Tlr9K51E/K51E p < 0.0001,Tlr9WT versus Tlr9K51E/K51E p = 0.038; (e) Tlr9WT versus Tlr9−/− p < 0.0001, Tlr9−/− versus Tlr9K51E/K51E p < 0.0001; (f) Tlr9WT versus Tlr9−/− p = 0.0173, Tlr9−/− versus Tlr9K51E/K51E p = 0.0003; (g) Tlr9WT versus Tlr9−/− p = 0.0632, Tlr9−/− versus Tlr9K51E/K51E p = 0.0115). h, Representative gating strategy of B cell populations (upper panel) and T cell (lower panel) subsets i, Percent TCRCD19 + B cells of live splenocytes in male and female mice of the indicated genotypes (Tlr9WT, n = 86; Tlr9−/−, n = 36; Tlr9K51E/K51E, n = 29; (i) Tlr9WT versus Tlr9−/− p = 0.0420, Tlr9−/− versus Tlr9K51E/K51E p = 0.0078). j-k, Percent CD21hi CD23lo MZ, and CD11b+ CD11c+ ABC of live B cells in mice of the indicated genotypes. ((j) Tlr9WT, n = 86; Tlr9−/−, n = 36; Tlr9K51E/K51E, n = 29; Tlr9WT versus Tlr9−/− p = 0.0081, Tlr9−/− versus Tlr9K51E/K51E p = 0.0196; (k) Tlr9WT, n = 86; Tlr9−/−, n = 36; Tlr9K51E/K51E, n = 24; (k) Tlr9WT versus Tlr9−/− p = 0.0871, Tlr9−/− versus Tlr9K51E/K51E p = 0.0215) l, Percent TCRCD44hiCD138hi plasmablasts of live splenocytes in mice of the indicated genotypes (Tlr9WT, n = 86; Tlr9−/−, n = 36; Tlr9K51E/K51E, n = 29; Tlr9WT versus Tlr9−/− p = 0.0372, Tlr9−/− versus Tlr9K51E/K51E p = 0.0015). m, Percent TCR+CD4+ and TCR+CD8+ cells of live splenocytes in male and female mice of the indicated genotypes (Tlr9WT, n = 79; Tlr9−/−, n = 36; Tlr9K51E/K51E, n = 28; for CD4 + , Tlr9−/− versus Tlr9K51E/K51E p = 0.0002, Tlr9WT versus Tlr9K51E/K51E p = 0.0023; for CD8 + , Tlr9WT versus Tlr9−/− p = 0.001, Tlr9−/− versus Tlr9K51E/K51E p = 0.0304). n-o, Percent naïve (CD62Lhi CD44lo), activated (CD62hi CD44hi) and memory (CD62Llo CD44hi) of CD4 and CD8 splenocytes (Tlr9WT, n = 79; Tlr9−/−, n = 36; Tlr9K51E/K51E, n = 28; for naïve CD4 + , Tlr9WT versus Tlr9−/− p = 0.0017, Tlr9−/− versus Tlr9K51E/K51E p = 0.0071; for activated CD4 + , Tlr9WT versus Tlr9−/− p < 0.0001, Tlr9−/− versus Tlr9K51E/K51E p = 0.0089; for naïve CD8 + , Tlr9WT versus Tlr9−/− p = 0.0012; for memory CD8 + , Tlr9WT versus Tlr9−/− p = 0.0095). For all panels, data points indicate individual mice and bars the mean ± SEM. For statistics in (c-o) * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001, one-way ANOVA with Tukey’s multiple comparisons test).

Source data

Extended Data Fig. 2 TLR9-MyD88 signaling promotes disease.

a-e, Evaluation of the clinical parameters separated by gender (Tlr9−/− male, n = 21; Tlr9P915H/P915H male, n = 37; Tlr9+/− male, n = 24; Tlr9−/− female, n = 22; Tlr9P915H/P915H female, n = 25; Tlr9+/− female, n = 28; (a, female) Tlr9−/− versus Tlr9P915H/P915H p = 0.0022, Tlr9−/− versus Tlr9+/− p = 0.0431; (b) Tlr9−/− versus Tlr9P915H/P915H p < 0.0001, Tlr9P915H/P915H versus Tlr9+/− p = 0.0005; (c) Tlr9−/− versus Tlr9P915H/P915H p = 0.0007, Tlr9P915H/P915H versus Tlr9+/− p = 0.0445; (d) Tlr9−/− versus Tlr9P915H/P915H p < 0.0001, Tlr9P915H/P915H versus Tlr9+/− p = 0.0005; (e) Tlr9P915H/P915H versus Tlr9+/− p = 0.0521) f, Percent TCRCD19+ B cells of live splenocytes in male and female mice of the indicated genotypes (Tlr9−/−, n = 36; Tlr9P915H/P915H, n = 55; Tlr9+/−, n = 44; Tlr9−/− versus Tlr9+/− p = 0.0260). g-h, Percent CD21hi CD23lo MZ (Tlr9−/−, n = 36; Tlr9P915H/P915H, n = 55; Tlr9+/−, n = 44), and CD11b+ CD11c+ ABC of live B cells in mice of the indicated genotypes (Tlr9−/−, n = 36; Tlr9P915H/P915H, n = 53; Tlr9+/−, n = 41; (g) Tlr9−/− versus Tlr9P915H/P915H p = 0.0109, Tlr9P915H/P915H versus Tlr9+/− p = 0.0032; (h) Tlr9−/− versus Tlr9P915H/P915H p = 0.0228). i, Percent TCRCD44hiCD138hi plasmablasts of live splenocytes in mice of the indicated genotypes (Tlr9−/−, n = 36; Tlr9P915H/P915H, n = 55; Tlr9+/−, n = 44; Tlr9−/− versus Tlr9P915H/P915H p = 0.0264). j, Percent TCR+CD4+ and TCR+CD8+ cells of live splenocytes in male and female mice of the indicated genotypes (Tlr9−/−, n = 36; Tlr9P915H/P915H, n = 52; Tlr9+/−, n = 44; CD4+, Tlr9−/− versus Tlr9P915H/P915H p = 0.0047, Tlr9P915H/P915H versus Tlr9+/− p = 0.0131; CD8+, Tlr9−/− versus Tlr9P915H/P915H p = 0.0001, Tlr9P915H/P915H versus Tlr9+/− p = 0.0005). k-l, Percent naïve (CD62Lhi CD44lo), activated (CD62Lhi CD44hi) and memory (CD62Llo CD44hi) of CD4 and CD8 splenocytes (Tlr9−/−, n = 36; Tlr9P915H/P915H, n = 52; Tlr9+/−, n = 44, (k) naïve Tlr9−/− versus Tlr9P915H/P915H p < 0.0001, Tlr9P915H/P915H versus Tlr9+/− p = 0.0018; activated Tlr9−/− versus Tlr9P915H/P915H p = 0.0345; (l) activated Tlr9−/− versus Tlr9P915H/P915H p = 0.0324, Tlr9P915H/P915H versus Tlr9+/− p = 0.0034; memory Tlr9−/− versus Tlr9P915H/P915H p = 0.0013, Tlr9P915H/P915H versus Tlr9+/− p = 0.0006). (a-l, For all panels, data points indicate individual mice and bars the mean ± SEM. For statistics in (a-l) * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001, one-way ANOVA with Tukey’s multiple comparisons test).

Source data

Extended Data Fig. 3 Effects of TLR9 and gender on TLR7 expression and distribution.

a-d, Representative flow cytometry plots of live, singlet, TCRβCD19+ B cells of 13 week old BALB/c (a) or 5 week old MRL/lpr (d) mice. Numbers indicate the frequencies in the gate. b,c, Comparison of TLR7 and TLR9 MFI between Tlr9 WT male and Tlr9 WT female BALB/c mice is shown for each of the defined B cell subsets (male, n = 8; female, n = 8 from two experiments; (b) male versus female MZ B cells p = 0.0333; male FO versus male MZ B cells p < 0.0001; (c) male FO versus male MZ B cells p = 0.0009). e,f, Comparison of TLR7 and TLR9 MFI between Tlr9 WT male and Tlr9 WT female 5 week old MRL/lpr mice is shown for each of the defined B cell subsets (male, n = 9 mice; female, n = 9 mice from two experiments; (e) male versus female FO B cells p < 0.0001; male versus female MZ B cells p < 0.0001; male versus female ABC B cells p = 0.0282; male FO versus male MZ B cells p < 0.0001; male FO versus male ABC B cells p < 0.0001; male MZ versus male ABC B cells p = 0.0330 (f) male FO versus male MZ B cells p < 0.0001; male FO versus male ABC B cells p < 0.0001; male MZ versus male ABC B cells p < 0.0001). (For b,c,e,f, data points indicate individual mice and bars the mean ± SEM of two experiments pooled. * p < 0.05, **** p < 0.0001, two tailed unpaired t-test between male and female groups; b,c, # p < 0.05; ## p < 0.01; ### p < 0.001; #### p < 0.0001, two tailed paired-t-test between subsets in the male group; e,f # p < 0.05; ## p < 0.01; ### p < 0.001; #### p < 0.0001 RM one way ANOVA with Tukey’s multiple comparisons test between subsets in the male group.) g-i, Quantification of TLR7 MFI (ratio of TLR7 MFI over average TLR7 MFI of the corresponding male or female WT group), in FO, MZ and ABC B cells of 5 week old female and male MRL/lpr mice (Tlr9K51E/K51E male, n = 4; female, n = 4; Tlr9 WT male, n = 4, female, n = 8; Tlr9P915H/P915H male, n = 4, female, n = 7; Tlr9+/− male, n = 3, female, n = 4; Tlr9−/− male, n = 3, female, n = 7; (g, male) Tlr9K51E/K51E versus Tlr9P915H/P915H p = 0.0097; Tlr9K51E/K51E versus Tlr9−/− p = 0.0061, Tlr9+/− versus Tlr9−/− p = 0.0430; (g, female) Tlr9 WT versus Tlr9+/− p = 0.0232; Tlr9 WT versus Tlr9−/− p < 0.0001; Tlr9K51E/K51E versus Tlr9+/− p = 0.0386, Tlr9K51E/K51E versus Tlr9−/- p < 0.0001, Tlr9P915H/P915H versus Tlr9 -/- p = 0.0002; (h, male) Tlr9 WT versus Tlr9P915H/P915H p = 0.0018; Tlr9 WT versus Tlr9 -/- p = 0.0261; Tlr9K51E/K51E versus Tlr9P915H/P915H p < 0.0001; Tlr9K51E/K51E versus Tlr9−/− p = 0.0005; Tlr9P915H/P915H versus Tlr9+/− p = 0.0001; Tlr9−/− versus Tlr9+/− p = 0.0012; (h, female) Tlr9 WT versus Tlr9−/− p = 0.0033; Tlr9K51E/K51E versus Tlr9−/− p < 0.0021; Tlr9−/− versus Tlr9+/− p = 0.0074; (i, male) Tlr9 WT versus Tlr9K51E/K51E p = 0.0398, Tlr9K51E/K51E versus Tlr9P915H/P915H p = 0.0143, Tlr9K51E/K51E versus Tlr9−/− p = 0.0284; (i, female) Tlr9 WT versus Tlr9+/− p = 0.0011, Tlr9 WT versus Tlr9−/− P < 0.0001, Tlr9K51E/K51E versus Tlr9+/− p = 0.0004, Tlr9K51E/K51E versus Tlr9−/− p < 0.0001, Tlr9P915H/P915H versus Tlr9+/− p = 0.004, Tlr9P915H/P915H versus Tlr9−/− p = 0.0002). j-l, Quantification of TLR9 MFI (ratio of TLR9 MFI over mean TLR9 MFI of the corresponding male or female WT group), in FO, MZ and ABC B cells of 5 week old female and male MRL/lpr mice (Tlr9K51E/K51E male, n = 4; female, n = 4; Tlr9 WT male, n = 4, female, n = 8; Tlr9P915H/P915H male, n = 4, female, n = 7; Tlr9+/− male, n = 3, female, n = 4; Tlr9−/− male, n = 3, female, n = 7; (j, male) Tlr9 WT versus Tlr9P915H/P915H p = 0.0001, Tlr9 WT versus Tlr9+/− p < 0.0001, Tlr9 WT versus Tlr9−/− p < 0.0001, Tlr9K51E/K51E versus Tlr9P915H/P915H p = 0.0004, Tlr9K51E/K51E versus Tlr9+/− p < 0.0001, Tlr9K51E/K51E versus Tlr9−/− p < 0.0001, Tlr9P915H/P915H versus Tlr9−/− p = 0.0004, Tlr9+/− versus Tlr9−/− p = 0.0104; (j, female) Tlr9 WT versus Tlr9P915H/P915H p < 0.0001, Tlr9 WT versus Tlr9+/− p < 0.0001, Tlr9 WT versus Tlr9−/− p < 0.0001, Tlr9K51E/K51E versus Tlr9P915H/P915H p < 0.0001, Tlr9K51E/K51E versus Tlr9+/− p < 0.0001, Tlr9K51E/K51E versus Tlr9−/− p < 0.0001, Tlr9P915H/P915H versus Tlr9−/− p < 0.0001, Tlr9+/− versus Tlr9−/− p < 0.0001; (k, male) Tlr9 WT versus Tlr9P915H/P915H p < 0.0001, Tlr9 WT versus Tlr9+/− p < 0.0001, Tlr9 WT versus Tlr9−/− p < 0.0001, Tlr9K51E/K51E versus Tlr9P915H/P915H p < 0.0001, Tlr9K51E/K51E versus Tlr9+/− p < 0.0001, Tlr9K51E/K51E versus Tlr9−/− p < 0.0001, Tlr9P915H/P915H versus Tlr9−/− p = 0.0008, Tlr9+/− versus Tlr9−/− p = 0.0015; (k, female) Tlr9 WT versus Tlr9P915H/P915H p < 0.0001, Tlr9 WT versus Tlr9+/− p < 0.0001, Tlr9 WT versus Tlr9−/− p < 0.0001, Tlr9K51E/K51E versus Tlr9P915H/P915H p < 0.0001, Tlr9K51E/K51E versus Tlr9+/− p < 0.0001, Tlr9K51E/K51E versus Tlr9−/− p < 0.0001, Tlr9P915H/P915H versus Tlr9+/− p = 0.0467, Tlr9P915H/P915H versus Tlr9−/− p < 0.0001, Tlr9+/− versus Tlr9−/− p < 0.0001; (l, male) Tlr9 WT versus Tlr9P915H/P915H p < 0.0001, Tlr9 WT versus Tlr9+/− p < 0.0001, Tlr9 WT versus Tlr9−/− p < 0.0001, Tlr9K51E/K51E versus Tlr9P915H/P915H p < 0.0001, Tlr9K51E/K51E versus Tlr9+/− p < 0.0001, Tlr9K51E/K51E versus Tlr9−/− p < 0.0001, Tlr9P915H/P915H versus Tlr9−/− p < 0.0001, Tlr9+/− versus Tlr9−/− p < 0.0001; (l, female) Tlr9 WT versus Tlr9P915H/P915H p < 0.0001, Tlr9 WT versus Tlr9+/− p < 0.0001, Tlr9 WT versus Tlr9−/− p < 0.0001, Tlr9K51E/K51E versus Tlr9P915H/P915H p = 0.0018, Tlr9K51E/K51E versus Tlr9+/− p = 0.0108, Tlr9K51E/K51E versus Tlr9−/− p < 0.0001, Tlr9P915H/P915H versus Tlr9−/− p = 0.0008, Tlr9+/− versus Tlr9−/− p = 0.0024). (For g-l, data points indicate individual mice and bars the mean ± SEM of one or two experiments pooled; * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001, one-way ANOVA with Tukey’s multiple comparisons test). m, The percent of TLR7 positivity in EEA1+ or LAMP-1+ endosomes of TLR9WT B cells is unchanged after a 2 hour stimulation with TLR7 agonist (CL097). (TLR7+EEA1+ unstimulated B cells, n = 17 from one mouse; TLR7+EEA1+ stimulated B cells, n = 25 from one mouse; TLR7+LAMP1+ unstimulated B cells, n = 27 from two mice; TLR7+LAMP1+ stimulated B cells, n = 40 from two mice). Data points indicate individual fields (1-3 cells per field) analyzed, and bars the mean ± SEM of two mice.

Source data

Extended Data Fig. 4 TLR9–MyD88-independent functions are B cell-intrinsic.

a-b, The competitive advantage of Tlr9WT vs Tlr9−/− or Tlr9P915H/P915H in different subsets of immature (n = 11 mice from three cohorts), transitional and naïve B cells (n = 15 mice from three cohorts), at the late time point (19-20 weeks post chimerism). Values greater than one on the log2 scale indicate TLR9WT advantage and below one indicate Tlr9−/− or Tlr9P915H/P915H advantage ((a) T2, p = 0.066; FO, p < 0.0001; (b) Fr. A-C, p < 0.0434; Fr. D, p = 0.0063; Fr. E, p = 0.0092). c,d, The competitive advantage of Tlr9WT vs Tlr9−/− or Tlr9P915H/P915H in different subsets of mature and differentiated B cells at the late time point (FO, MZ, GC, ABC, PB, CD80 + PDL2 + n = 15 mice from three cohorts; PC, n = 11 mice from three cohorts). Values greater than one on the log2 scale indicate TLR9WT advantage and below one indicate Tlr9−/− or Tlr9P915H advantage ((c) FO, p < 0.0001; CD80+PDL2+, p = 0.0162; PB, p = 0.0003; PC, p = 0.0356; FO versus MZ, p < 0.0001 (d) ABC, p = 0.0025; CD80+PDL2+, p = 0.0052; PB, p = 0.0014; PC, p = 0.0288). e, The competitive advantage of Tlr9−/− vs Tlr9P915H/P915H in different subsets of mature and differentiated B cells at the late time point (FO, MZ, GC, ABC, PB, CD80 + PDL2 + n = 15 mice from three cohorts; PC, n = 11 mice from three cohorts). Values greater than one indicate Tlr9−/− advantage and below one indicate Tlr9P915H advantage (FO, p < 0.0001; GC, p = 0.0002; ABC, p = 0.0005; CD80+PDL2+, p = 0.0002; PB, p < 0.0001; PC, p = 0.0056). (a-e) Data were pooled from three cohorts (two females and one male). Symbols represent individual mice and bars the mean ± SEM. Each group was compared to the hypothetical mean of no competitive advantage (one), by two-tailed one-sample t test (# p < 0.05; ## p < 0.01; ### p < 0.001; #### p < 0.0001), shown above each individual bar. Additionally, for (c), statistical significance between FO and the other B cell subsets was calculated by a mixed-effect analysis with Dunnett’s multiple comparisons test (* p < 0.05). f,g, Quantification of TLR7 (f) and TLR9 (g) in Tlr9WT transitional (T1,T2) (TLR7 n = 5; TLR9, n = 5 mice), FO (TLR7, n = 18; TLR9, n = 12 mice), MZ (TLR7, n = 18; TLR9, n = 12 mice), ABC (TLR7, n = 18; TLR9, n = 12 mice) and memory-like CD80+PDL2+ (TLR7, n = 11; TLR9, n = 5 mice) B cells of the chimera mice, at the early time point. TLR7 expression is minimal in immature B cells and maximal in ABCs ((f) T1 versus FO, p = 0.0180; T1 versus MZ, p = 0.0122; T1 versus ABC, p = 0.0017; T1 versus CD80+PDL2+, p = 0.0059; T2 versus FO, p = 0.0127; T2 versus MZ, p = 0.0119; T2 versus ABC, p = 0.0015; T2 versus CD80+PDL2+, p = 0.0064; FO versus MZ, p < 0.0001; FO versus ABC, p < 0.0001; FO versus CD80+PDL2+, p < 0.0001; MZ versus ABC, p < 0.0001; ABC versus CD80+PDL2+, p < 0.0001; (g) T1 versus ABC, p = 0.0193; T2 versus ABC, p = 0.0240; FO versus ABC, p < 0.0001; MZ versus ABC, p < 0.0001; ABC versus CD80+PDL2+, p = 0.0144). ((f-g), Symbols represent individual mice and bars the mean ± SEM. * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001 Mixed-effects analysis between subsets, with Tukey’s multiple comparisons test). h,i, Quantification of TLR7 MFI (ratio of TLR7 MFI in Tlr9−/− or Tlr9P915H/P915H cells over Tlr9WT subset per mouse), in FO (early, n = 18; late, n = 14), MZ (early, n = 18; late, n = 14), ABC (early, n = 18; late, n = 14) and CD80+PDL2+ (early, n = 11; late, n = 14) B cells, at the early (h) and late (i) time point. Each group was compared to the hypothetical mean of one (indicating similar TLR7 expression in mutated Tlr9 and Tlr9WT mice), by two-tailed one-sample t test (# p < 0.05; ## p < 0.01; ### p < 0.001; #### p < 0.0001), shown above each individual bar; (Symbols represent individual mice and bars the mean ± SEM; (h) Tlr9−/− FO, p < 0.0001; Tlr9P915H/P915H FO, p = 0.0002; Tlr9−/− MZ, p < 0.0001; Tlr9P915H/P915H MZ, p < 0.0001; Tlr9−/− ABC, p < 0.0001; Tlr9P915H/P915H ABC, p = 0.0024; Tlr9P915H/P915H CD80+PDL2+, p = 0.0174; (i) Tlr9−/− FO, p = 0.0019; Tlr9P915H/P915H FO, p < 0.0001; Tlr9−/− MZ, p = 0.0008; Tlr9P915H/P915H MZ, p < 0.0001; Tlr9−/− ABC, p < 0.0001; Tlr9P915H/P915H ABC, p = 0.0065). j,k, Quantification of TLR9 MFI in T1 (n = 5), T2 (n = 5), FO (n = 12), MZ (n = 12), ABC (n = 12) and CD80+PDL2+ (n = 5) B cells of the 3-way BM chimera mice at the early time point. Symbols represent individual mice and bars the mean ± SEM; For statistics, * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001, RM one-way ANOVA with Tukey’s multiple comparisons test; (j) T1, Tlr9WT versus Tlr9−/−, p = 0.0004; Tlr9WT versus Tlr9P915H/P915H, p = 0.0022; Tlr9−/− versus Tlr9P915H/P915H, p = 0.0004; T2, Tlr9WT versus Tlr9−/−, p = 0.0006; Tlr9WT versus Tlr9P915H/P915H, p = 0.0003; Tlr9−/− versus Tlr9P915H/P915H, p = 0.0009; FO, Tlr9WT versus Tlr9−/−, p < 0.0001; Tlr9WT versus Tlr9P915H/P915H, p < 0.0001; Tlr9−/− versus Tlr9P915H/P915H, p < 0.0001; (k) MZ, Tlr9WT versus Tlr9−/−, p < 0.0001; Tlr9WT versus Tlr9P915H/P915H, p < 0.0001; Tlr9−/− versus Tlr9P915H/P915H, p < 0.0001; ABC, Tlr9WT versus Tlr9−/−, p < 0.0001; Tlr9WT versus Tlr9P915H/P915H, p < 0.0001; Tlr9−/− versus Tlr9P915H/P915H, p < 0.0001; CD80+PDL2+, Tlr9WT versus Tlr9−/−, p = 0.0001; Tlr9WT versus Tlr9P915H/P915H, p = 0.0026; Tlr9−/− versus Tlr9P915H/P915H, p = 0.0006; l-p, Frequencies of T1 (n = 5), T2 (n = 3), FO (n = 4), ABC (n = 5) B cells and PB (n = 5) of the 3-way BM chimera mice in S or G2/M phase, determined by Ki67 and DAPI staining, and frequencies of activated caspase 3+ cells for each B cell subset (n = 6 mice). ((l-p), data points indicate individual mice and bars the mean ± SEM. For statistics * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001, RM one-way ANOVA with Turkey’s multiple comparisons test. (l), G2/M T1, Tlr9WT versus Tlr9P915H/P915H, p = 0.0240; Caspase 3+ Tlr9−/− versus Tlr9P915H/P915H, p = 0.0020; Caspase 3+ Tlr9WT versus Tlr9P915H/P915H, p = 0.0076; (m), S T2, Tlr9−/− versus Tlr9P915H/P915H, p = 0.0485; G2/M T2, Tlr9WT versus Tlr9−/−, p = 0.0172; Caspase 3+ Tlr9−/− versus Tlr9P915H/P915H, p = 0.0333; Caspase 3+ Tlr9WT versus Tlr9P915H/P915H, p = 0.0361; (n) G2/M FO, Tlr9WT versus Tlr9−/−, p = 0.0109; Tlr9WT versus Tlr9P915H/P915H, p = 0.0113; (o) Caspase 3+ Tlr9−/− versus Tlr9P915H/P915H, p = 0.0473).

Source data

Extended Data Fig. 5 TLR9 controls ABC gene programs independently of MyD88.

a, PCA plot showing differences in accessible regions between the ABCs of the three genotypes Tlr9WT, Tlr9−/− and Tlr9P915H, sorted from the 3-way BM chimera mice. The different genotypes are represented as different shapes. b, PCA plot showing transcriptional differences between the ABCs of the three genotypes Tlr9WT, Tlr9−/− and Tlr9P915H, sorted from the 3-way BM chimera mice. c, Venn diagram representing the number of up-regulated genes (from Fig. 7d), shared or unique between Tlr9WT and Tlr9P915H ABCs in comparison to Tlr9−/− ABCs. d, Gene Set Enrichment Analysis (GSEA) was performed to look for plasma cell gene signature. GSEA shows significant enrichment of a plasma cell gene signature in Tlr9WT compared to Tlr9P915H ABCs (left panel), but no significant enrichment between Tlr9WT and Tlr9−/− ABCs (right panel). The p value was calculated using rankSumTestWithCorrelation function from the R limma package. e, XY plot showing the transcripts of the inhibitory pathway identified in Fig. 7d that were uniquely upregulated in Tlr9P915H ABCs vs. the transcripts upregulated by both Tlr9P915H and Tlr9WT ABCs, compared to Tlr9−/− ABCs. f, Quantification by qPCR (lower panels) of the transcripts of key inhibitory markers, found uniquely upregulated in Tlr9P915H ABCs compared to Tlr9WT and Tlr9−/− ABCs by RNA seq (upper panels), in sorted Tlr9−/−, Tlr9P915H and Tlr9WT ABCs from another cohort of 3-way BM chimera MRL/lpr mice (n = 4). g, Gene Set Enrichment Analysis (GSEA) was performed to look for TLR7-induced gene-set signature. GSEA shows no significant enrichment of TLR7-induced gene-set signature in Tlr9−/− ABCs compared to Tlr9P915H ABCs of the 3-way BM chimera MRL/lpr mice. The p value was calculated using rankSumTestWithCorrelation function from the R limma package. h, Venn diagram representing that only 20 genes were upregulated in Tlr9−/− ABCs compared to both Tlr9P915H and Tlr9WT ABCs.

Source data

Extended Data Fig. 6 Effects of TLR9 genotypes on FO and MZ B cell gene programs.

RNA-seq analysis of sorted FO B cells (TCRβ CD19+ CD23hi CD21lo) and MZ B cells (TCRβ CD19+ CD23lo CD21hi) of the different Tlr9 genotypes from the 3-way BM chimera MRL/lpr mice (described in Fig. 6a), all over 19 weeks post-chimerization. a, PCA plot shows transcriptional differences between the FO B cells of the three genotypes Tlr9WT, Tlr9−/− and Tlr9P915H, sorted from the 3-way BM chimera mice. b, Number of DEGs identified using Limma R package (Log2 fold-change >1 and FDR-corrected p value < 0.05) between the different Tlr9 genotypes from sorted FO B cells of the 3-way BM chimera MRL/lpr mice (n = 4 replicates). c, Venn diagram representing the number of up-regulated genes (from Fig. E6b), in Tlr9WT FO B cells compared to Tlr9P915H and Tlr9−/− FO B cells. d, XY plot showing the DEG from Fig. E6b: (i) uniquely upregulated in Tlr9WT FO B cells compared to Tlr9−/− FO B cells; (ii) upregulated in Tlr9WT FO B cells compared to Tlr9P915H FO B cells; or (iii) upregulated in comparison to both Tlr9P915H and Tlr9−/− FO B cells. e, PCA plot showing transcriptional differences between the MZ B cells of the three genotypes Tlr9WT, Tlr9−/− and Tlr9P915H, sorted from the 3-way BM chimera mice. f, Number of DEGs identified using Limma R package (Log2 fold-change > 1 and FDR-corrected p value < 0.05) between the different Tlr9 genotypes from sorted MZ B cells of the 3-way BM chimera MRL/lpr mice (n = 4 replicates). g, Venn diagram representing the number of up-regulated genes from Fig. E6f, in Tlr9WT MZ B cells compared to Tlr9P915H and Tlr9−/− MZ B cells. h, XY plot showing the DEG from Fig. E6f): (i) uniquely upregulated in Tlr9WT MZ B cells compared to Tlr9−/− MZ B cells, (ii) upregulated in Tlr9WT MZ B cells compared to Tlr9P915H MZ B cells, or (iii) upregulated in comparison to both Tlr9P915H and Tlr9−/− MZ B cells.

Extended Data Fig. 7 Tlr9P915H and Tlr9−/− induce distinct ABC and PB states.

a,b, Costimulatory molecules and PB-associated transcripts from the RNA-seq analysis that were more highly expressed in Tlr9−/− and Tlr9WT ABCs compared to Tlr9P915H ABCs (upper row, n = 4) were investigated at the protein level by flow cytometry (lower row) in a separate cohort of 3-way BM chimera MRL/lpr mice (n = 12) ((a) CD80, Tlr9−/− versus Tlr9P915H, p < 0.0001; Tlr9P915H versus Tlr9WT,p = 0.0023; CD86, Tlr9−/− versus Tlr9P915H, p < 0.0001; Tlr9P915H versus Tlr9WT,p = 0.0017; CD200, Tlr9−/− versus Tlr9P915H, p = 0.0040; Tlr9−/− versus Tlr9WT,p = 0.0073; Itgb7, Tlr9−/− versus Tlr9P915H, p = 0.0003; Tlr9P915H versus Tlr9WT,p = 0.0057; (b) CD44, Tlr9−/− versus Tlr9P915H, p = 0.0003; Tlr9P915H versus Tlr9WT,p = 0.0076; CXCR4, Tlr9−/− versus Tlr9P915H, p < 0.0001; Tlr9P915H versus Tlr9WT,p = 0.0008; CD138, Tlr9−/− versus Tlr9P915H, p < 0.0001; Tlr9P915H versus Tlr9WT,p = 0.0094). c,d, Flow cytometry analysis shows differential expression (MFI) of selected transcription factors (c) and the inhibitory molecule CD300a (d) among Tlr9 genotypes in ABCs of the 3-way BM chimera MRL/lpr mice ((c) IRF4, Tlr9−/− versus Tlr9P915H, p = 0.0002; BCL6, Tlr9−/− versus Tlr9P915H, p = 0.0015; Tlr9P915H versus Tlr9WT, p = 0.0011; Tlr9−/− versus Tlr9WT,p = 0.0025; TBET, Tlr9−/− versus Tlr9P915H, p = 0.0002; Tlr9−/− versus Tlr9WT,p = 0.0107; (d) CD300a, Tlr9−/− versus Tlr9P915H, p < 0.0001; Tlr9P915H versus Tlr9WT,p = 0.0188). (a-d) Splenocytes from twelve 3-way BM chimera mice (n = 6 females, and n = 6 males from 2 experiments) were stained for the indicated markers and the CD45.1/CD45.2 congenic markers. (Each line represents a 3-way BM chimera MRL/lpr mouse, * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001, RM one-way ANOVA with Turkey’s multiple comparisons test).

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Leibler, C., John, S., Elsner, R.A. et al. Genetic dissection of TLR9 reveals complex regulatory and cryptic proinflammatory roles in mouse lupus. Nat Immunol 23, 1457–1469 (2022). https://doi.org/10.1038/s41590-022-01310-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01310-2

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