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:

Increased cathepsin S in Prdm1−/− dendritic cells alters the TFH cell repertoire and contributes to lupus

Abstract

Aberrant population expansion of follicular helper T cells (TFH cells) occurs in patients with lupus. An unanswered question is whether an altered repertoire of T cell antigen receptors (TCRs) is associated with such expansion. Here we found that the transcription factor Blimp-1 (encoded by Prdm1) repressed expression of the gene encoding cathepsin S (Ctss), a cysteine protease that cleaves invariant chains and produces antigenic peptides for loading onto major histocompatibility complex (MHC) class II molecules. The increased CTSS expression in dendritic cells (DCs) from female mice with dendritic cell–specific conditional knockout of Prdm1 (CKO mice) altered the presentation of antigen to CD4+ T cells. Analysis of complementarity-determining region 3 (CDR3) regions containing the β-chain variable region (Vβ) demonstrated a more diverse repertoire of TFH cells from female CKO mice than of those from wild-type mice. In vivo treatment of CKO mice with a CTSS inhibitor abolished the lupus-related phenotype and reduced the diversity of the TFH cell TCR repertoire. Thus, Blimp-1 deficiency in DCs led to loss of appropriate regulation of Ctss expression in female mice and thereby modulated antigen presentation and the TFH cell repertoire to contribute to 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

Figure 1: Increased expression of Ctss in DCs from female CKO mice.
Figure 2: Blimp-1 regulates Ctss expression in DCs.
Figure 3: IL-6 has a role in Ctss expression in DCs.
Figure 4: 'Preferential' differentiation of IL-21-producing T cells by co-culture with Blimp-1-deficient DCs in vitro.
Figure 5: The TFH cells of female CKO mice harbor a diverse CDR3 repertoire.
Figure 6: The CTSS inhibitor RO5461111 prevents development of the lupus-like phenotype in female CKO mice.
Figure 7: RO5461111 suppresses the population expansion of TFH cells and germinal-center B cells in CKO mice.

Similar content being viewed by others

References

  1. Pavli, P., Hume, D.A., Van De Pol, E. & Doe, W.F. Dendritic cells, the major antigen-presenting cells of the human colonic lamina propria. Immunology 78, 132–141 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Guilliams, M. et al. Skin-draining lymph nodes contain dermis-derived CD103 dendritic cells that constitutively produce retinoic acid and induce Foxp3+ regulatory T cells. Blood 115, 1958–1968 (2010).

    CAS  PubMed  Google Scholar 

  3. Bousso, P. T-cell activation by dendritic cells in the lymph node: lessons from the movies. Nat. Rev. Immunol. 8, 675–684 (2008).

    CAS  PubMed  Google Scholar 

  4. Zhu, J. & Paul, W.E. CD4 T cells: fates, functions, and faults. Blood 112, 1557–1569 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Rogers, P.R. & Croft, M. Peptide dose, affinity, and time of differentiation can contribute to the Th1/Th2 cytokine balance. J. Immunol. 163, 1205–1213 (1999).

    CAS  PubMed  Google Scholar 

  6. Rogers, P.R., Dubey, C. & Swain, S.L. Qualitative changes accompany memory T cell generation: faster, more effective responses at lower doses of antigen. J. Immunol. 164, 2338–2346 (2000).

    CAS  PubMed  Google Scholar 

  7. Théry, C. & Amigorena, S. The cell biology of antigen presentation in dendritic cells. Curr. Opin. Immunol. 13, 45–51 (2001).

    PubMed  Google Scholar 

  8. Villadangos, J.A. et al. Proteases involved in MHC class II antigen presentation. Immunol. Rev. 172, 109–120 (1999).

    CAS  PubMed  Google Scholar 

  9. Shi, G.P. et al. Human cathepsin S: chromosomal localization, gene structure, and tissue distribution. J. Biol. Chem. 269, 11530–11536 (1994).

    CAS  PubMed  Google Scholar 

  10. Hsieh, C.S., deRoos, P., Honey, K., Beers, C. & Rudensky, A.Y. A role for cathepsin L and cathepsin S in peptide generation for MHC class II presentation. J. Immunol. 168, 2618–2625 (2002).

    CAS  PubMed  Google Scholar 

  11. Beers, C. et al. Cathepsin S controls MHC class II-mediated antigen presentation by epithelial cells in vivo. J. Immunol. 174, 1205–1212 (2005).

    CAS  PubMed  Google Scholar 

  12. Stoeckle, C. et al. Cathepsin S dominates autoantigen processing in human thymic dendritic cells. J. Autoimmun. 38, 332–343 (2012).

    CAS  PubMed  Google Scholar 

  13. Han, J.W. et al. Genome-wide association study in a Chinese Han population identifies nine new susceptibility loci for systemic lupus erythematosus. Nat. Genet. 41, 1234–1237 (2009).

    CAS  PubMed  Google Scholar 

  14. Gateva, V. et al. A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus. Nat. Genet. 41, 1228–1233 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kim, S.J., Gregersen, P.K. & Diamond, B. Regulation of dendritic cell activation by microRNA let-7c and BLIMP1. J. Clin. Invest. 123, 823–833 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Xu, H. et al. Increased frequency of circulating follicular helper T cells in lupus patients is associated with autoantibody production in a CD40L-dependent manner. Cell. Immunol. 295, 46–51 (2015).

    CAS  PubMed  Google Scholar 

  17. Le Coz, C. et al. Circulating TFH subset distribution is strongly affected in lupus patients with an active disease. PLoS One 8, e75319 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Choi, J.Y. et al. Circulating follicular helper-like T cells in systemic lupus erythematosus: association with disease activity. Arthritis Rheumatol. 67, 988–999 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Kim, S.J., Zou, Y.R., Goldstein, J., Reizis, B. & Diamond, B. Tolerogenic function of Blimp-1 in dendritic cells. J. Exp. Med. 208, 2193–2199 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Piskurich, J.F. et al. BLIMP-I mediates extinction of major histocompatibility class II transactivator expression in plasma cells. Nat. Immunol. 1, 526–532 (2000).

    CAS  PubMed  Google Scholar 

  21. Vander Lugt, B. et al. Transcriptional programming of dendritic cells for enhanced MHC class II antigen presentation. Nat. Immunol. 15, 161–167 (2014).

    CAS  PubMed  Google Scholar 

  22. Doody, G.M. et al. An extended set of PRDM1/BLIMP1 target genes links binding motif type to dynamic repression. Nucleic Acids Res. 38, 5336–5350 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Seillet, C. et al. Estradiol promotes functional responses in inflammatory and steady-state dendritic cells through differential requirement for activation function-1 of estrogen receptor α. J. Immunol. 190, 5459–5470 (2013).

    CAS  PubMed  Google Scholar 

  24. Kitamura, H. et al. IL-6-STAT3 controls intracellular MHC class II αβ dimer level through cathepsin S activity in dendritic cells. Immunity 23, 491–502 (2005).

    CAS  PubMed  Google Scholar 

  25. Becker, S., Groner, B. & Müller, C.W. Three-dimensional structure of the Stat3β homodimer bound to DNA. Nature 394, 145–151 (1998).

    CAS  PubMed  Google Scholar 

  26. Hutchins, A.P., Diez, D. & Miranda-Saavedra, D. The IL-10/STAT3-mediated anti-inflammatory response: recent developments and future challenges. Brief. Funct. Genomics 12, 489–498 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Pratama, A. & Vinuesa, C.G. Control of TFH cell numbers: why and how? Immunol. Cell Biol. 92, 40–48 (2014).

    CAS  PubMed  Google Scholar 

  28. Walker, B., Lynas, J.F., Meighan, M.A. & Brömme, D. Evaluation of dipeptide α-keto-β-aldehydes as new inhibitors of cathepsin S. Biochem. Biophys. Res. Commun. 275, 401–405 (2000).

    CAS  PubMed  Google Scholar 

  29. Rupanagudi, K.V. et al. Cathepsin S inhibition suppresses systemic lupus erythematosus and lupus nephritis because cathepsin S is essential for MHC class II-mediated CD4 T cell and B cell priming. Ann. Rheum. Dis. 74, 452–463 (2015).

    CAS  PubMed  Google Scholar 

  30. Klein, L., Hinterberger, M., Wirnsberger, G. & Kyewski, B. Antigen presentation in the thymus for positive selection and central tolerance induction. Nat. Rev. Immunol. 9, 833–844 (2009).

    CAS  PubMed  Google Scholar 

  31. Deng, Y. & Tsao, B.P. Genetic susceptibility to systemic lupus erythematosus in the genomic era. Nat. Rev. Rheumatol. 6, 683–692 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Raj, P. et al. Regulatory polymorphisms modulate the expression of HLA class II molecules and promote autoimmunity. eLife 5, e12089 (2016).

    PubMed  PubMed Central  Google Scholar 

  33. Yang, H., Rittner, H., Weyand, C.M. & Goronzy, J.J. Aberrations in the primary T-cell receptor repertoire as a predisposition for synovial inflammation in rheumatoid arthritis. J. Investig. Med. 47, 236–245 (1999).

    CAS  PubMed  Google Scholar 

  34. Yang, H. et al. Hrd1-mediated BLIMP-1 ubiquitination promotes dendritic cell MHCII expression for CD4 T cell priming during inflammation. J. Exp. Med. 211, 2467–2479 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Honey, K. & Rudensky, A.Y. Lysosomal cysteine proteases regulate antigen presentation. Nat. Rev. Immunol. 3, 472–482 (2003).

    CAS  PubMed  Google Scholar 

  36. Yang, H. et al. Cathepsin S is required for murine autoimmune myasthenia gravis pathogenesis. J. Immunol. 174, 1729–1737 (2005).

    CAS  PubMed  Google Scholar 

  37. Tato, M. et al. Cathepsin S inhibition combines control of systemic and peripheral pathomechanisms of autoimmune tissue injury. Sci. Rep. 7, 2775 (2017).

    PubMed  PubMed Central  Google Scholar 

  38. Yu, D. et al. Roquin represses autoimmunity by limiting inducible T-cell co-stimulator messenger RNA. Nature 450, 299–303 (2007).

    CAS  PubMed  Google Scholar 

  39. Kim, S.J. et al. Increased IL-12 inhibits B cells' differentiation to germinal center cells and promotes differentiation to short-lived plasmablasts. J. Exp. Med. 205, 2437–2448 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Brocker, T. Survival of mature CD4 T lymphocytes is dependent on major histocompatibility complex class II-expressing dendritic cells. J. Exp. Med. 186, 1223–1232 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Goldrath, A.W. & Bevan, M.J. Selecting and maintaining a diverse T-cell repertoire. Nature 402, 255–262 (1999).

    CAS  PubMed  Google Scholar 

  42. Rocha, B. & von Boehmer, H. Peripheral selection of the T cell repertoire. Science 251, 1225–1228 (1991).

    CAS  PubMed  Google Scholar 

  43. Ndifon, W. et al. Chromatin conformation governs T-cell receptor Jβ gene segment usage. Proc. Natl. Acad. Sci. USA 109, 15865–15870 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank H. Borrero and C. Colon for assistance with the flow cytometry; G. Klein and M. DeFranco RN for recruiting PRDM1-genotyped subjects; and C. Chrysostomou for discussions and assistance in bioinformatics analysis. Supported by National Institute of Arthritis and Musculoskeletal and Skin Diseases, the US National Institutes of Health (R01 AR065209 to S.J.K. and B.D.), the Alliance of Lupus Research (B.D.) and the Defense Threat Reduction Agency (HDTRA 1-12-C-0105 for G.G.).

Author information

Authors and Affiliations

Authors

Contributions

S.J.K. designed and performed experiments, analyzed data and interpreted results; S.S. performed the sequencing experiments and analyzed data; S.S.A. and W.H. provided RO5461111, and critically reviewed the manuscript; S.H.J. performed the ChIP experiments and Il6 promoter assay; P.K.G. provided the samples for the human study; G.G. designed the sequencing experiments and interpreted data; B.D. designed the study and analyzed and interpreted results; S.J.K., S.S., G.G. and B.D. participated in the interpretation of the study and wrote the manuscript, and also provided critical review of the paper; and all contributing authors agreed to the submission of this manuscript for publication.

Corresponding author

Correspondence to Betty Diamond.

Ethics declarations

Competing interests

S.S.A. and W.H. are employed by and hold stock in Roche Pharmaceuticals.

Integrated supplementary information

Supplementary Figure 1 Expression of cathepsin-encoding genes in splenic DCs from wild-type or CKO mice.

DCs were purified from the spleens of age matched female wild-type or CKO mice, and RNA was purified for qPCR. Gene specific primers were used for each cathepsin gene and the relative expression of each was calculated by normalization to the level of Hprt. Each dot represents an individual mouse and the bar represents the mean ± SEM (n=5).

Supplementary Figure 2 Increased expression of Il6 by estrogen in BM-DCs.

BM-DCs were differentiated with GM-CSF (200 ng/ml) in hormone-free culture system without or with E2, ICI or E2 and ICI together for 7 days. On day 7, BM-DCs were counted and further cultured with or without LPS (1 μg/ml) for overnight. Total RNA was purified and Il6 transcript was assessed by qPCR. Relative level of Il6 was normalized to housekeeping gene Polr2a. In the box-and-whisker plot, horizontal bars indicate the median, boxes indicate 25th to 75th percentile, and whiskers indicate 10th and 90th percentile (n=7).

Supplementary Figure 3 Diagram of Ctss reporter construct.

Supplementary Figure 4 Gene expression in TFH and non-TFH cells.

Spleens were collected from age-matched female wild-type (open circle) or CKO (closed circle) mice. TFH cells and non-TFH cells were identified as represented in the flow image (left panel), and each population was purified. Total RNA was purified and gene expression was measured by qPCR. Relative level of each gene was normalized to housekeeping gene Polr2a. Each dot represents an individual mouse and the bar represents the mean ± SEM (n=3).

Supplementary Figure 5 Increased IL-21 protein in the supernatant from co-culture of OT-II CD4+ T cells and Blimp-1-deficient DCs.

CD4+ T cells from OT-II mice and splenic DCs (open box is wild-type mice and gray box is CKO mice) were co-cultured in various conditions for 4 days. To measure the secreted cytokines, IL-2 and IL-21, supernatant was collected and cytokines were detected by a multiplex assay. In the box-and-whisker plot, horizontal bars indicate the median, boxes indicate 25th to 75th percentile, and whiskers indicate 10th and 90th percentile (n=6).

Supplementary Figure 6 Cytokine production in the supernatant from day 3 co-cultures of OT-II T cells and DCs.

DCs were purified from the spleens of female wild-type (open box) or CKO (gray box) mice, and CD4+ T cells were purified from female OT-II mice. DCs and T cells were co-cultured for 3 days at a 1:10 ratio (DC:T) with either OVA323-339 peptide (100 ng/ml) or OVA protein (10 μg/ml). Cytokine levels were measured in the supernatant by multiplex assay. In the box-and-whisker plot, horizontal bars indicate the median, boxes indicate 25th to 75th percentile, and whiskers indicate 10th and 90th percentile (n=6).

Supplementary Figure 7 Proliferation and IL-21 production of OT-II T cells by DCs presenting OVA peptide (amino acids 323–339).

OT-II T cells were co-cultured with splenic DCs for 3 days at 1:10 ratio (DC:T cells) in the presence of OVA323-339 peptide. To assess proliferation, T cells were labeled with CFSE (10 mM) before the culture and proliferation was measured by calculation of the division index (left panel). In the box-and-whisker plot, horizontal bars indicate the median, boxes indicate 25th to 75th percentile, and whiskers indicate 10th and 90th percentile (n=3). To assess IL-21 production, culture supernatant was harvested and measured by Multiplex assay (right panel). In the box-and-whisker plot, horizontal bars indicate the median, boxes indicate 25th to 75th percentile, and whiskers indicate 10th and 90th percentile (n=3).

Supplementary Figure 8 Protein level of cytokines, IL-2 and IL-21, measured by intracellular staining of T cells.

OT-II T cells were co-cultured with splenic DCs that had been incubated with HEL (10 μ/ml) or OVA (10 μg/ml). CTSB inhibitor (1 nM) was added during processing of OVA protein antigen. After co-culture for 4 days, T cells were stimulated with PMA (100 ng/ml)/ionomycin (1 μg/ml) for 6 hours. BFA (20 μg/ml) was added during the last 4 hours of stimulation. Cytokine-positive cells were identified by flow cytometry and frequency was calculated and presented in the graph. Each dot represents an individual experiment and the bar represents the mean ± SEM (n=4).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Table 1 (PDF 689 kb)

Life Sciences Reporting (PDF 82 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, S., Schätzle, S., Ahmed, S. et al. Increased cathepsin S in Prdm1−/− dendritic cells alters the TFH cell repertoire and contributes to lupus. Nat Immunol 18, 1016–1024 (2017). https://doi.org/10.1038/ni.3793

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3793

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