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:

Antigen presentation by cardiac fibroblasts promotes cardiac dysfunction

An Author Correction to this article was published on 09 November 2022

This article has been updated

Abstract

Heart failure (HF) is a leading cause of morbidity and mortality. Studies in animal models and individuals with HF revealed a prominent role for CD4+ T cell immune responses in the pathogenesis of HF and highlighted an active cross-talk between cardiac fibroblasts and interferon (IFN)-γ-producing CD4+ T cells that results in profibrotic myofibroblast transformation. Whether cardiac fibroblasts concomitantly modulate pathogenic cardiac CD4+ T cell immune responses is unknown. Here we report that mouse cardiac fibroblasts express major histocompatibility complex type II (MHCII) in two different experimental models of cardiac inflammation. We demonstrate that cardiac fibroblasts take up and process antigens for presentation to CD4+ T cells via MHCII induced by IFN-γ. Conditional deletion of MhcII in cardiac fibroblasts ameliorates cardiac remodeling and dysfunction induced by cardiac pressure overload. Collectively, we demonstrate that cardiac fibroblasts function as antigen-presenting cells and contribute to cardiac fibrosis and dysfunction through MHCII induced by IFN-γ.

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: Cardiac fibroblasts express MHCII in response to IFN-γ stimulation in vitro.
Fig. 2: Cardiac fibroblasts express MHCII in response to IFN-γ stimulation in vivo.
Fig. 3: Cardiac fibroblasts induce antigen-specific CD4+ T cell proliferation in vitro in the presence of IFN-γ.
Fig. 4: IFN-γ produced by TH1 cells induces MHCII expression by cardiac fibroblasts and promotes antigen-specific CD4+ T cell activation.
Fig. 5: Cardiac fibroblasts take up, process and present antigens to CD4+ T cells in vitro.
Fig. 6: Cardiac fibroblasts express MHCII in response to cardiac pressure overload.
Fig. 7: Cardiac fibroblast MHCII expression is required for cardiac dysfunction to develop in response to pressure overload.

Similar content being viewed by others

Data availability

All data used to generate figures are included in the accompanying files.

Change history

References

  1. Pinto, A. R. et al. Revisiting cardiac cellular composition. Circ. Res. 118, 400–409 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Prabhu, S. D. & Frangogiannis, N. G. The biological basis for cardiac repair after myocardial infarction: from inflammation to fibrosis. Circ. Res. 119, 91–112 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kaur, H. et al. Targeted ablation of periostin-expressing activated fibroblasts prevents adverse cardiac remodeling in mice. Circ. Res. 118, 1906–1917 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Aghajanian, H. et al. Targeting cardiac fibrosis with engineered T cells. Nature 573, 430–433 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Nevers, T. et al. Left ventricular T cell recruitment contributes to the pathogenesis of heart failure. Circ. Heart. Fail. 8, 776–787 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Nevers, T. et al. TH1 effector T cells selectively orchestrate cardiac fibrosis in nonischemic heart failure. J. Exp. Med. 214, 3311–3329 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Laroumanie, F. et al. CD4+ T cells promote the transition from hypertrophy to heart failure during chronic pressure overload. Circulation 129, 2111–2124 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Epelman, S., Liu, P. P. & Mann, D. L. Role of innate and adaptive immune mechanisms in cardiac injury and repair. Nat. Rev. Immunol. 15, 117–129 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kambayashi, T. & Laufer, T. M. Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell. Nat. Rev. Immunol. 14, 719–730 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Umetsu, D. T., Katzen, D., Jabara, H. H. & Geha, R. S. Antigen presentation by human dermal fibroblasts: activation of resting T lymphocytes. J. Immunol. 136, 440–445 (1986).

    CAS  PubMed  Google Scholar 

  11. Kanisicak, O. et al. Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nat. Commun. 7, 12260 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Burgdorf, S., Kautz, A., Bohnert, V., Knolle, P. A. & Kurts, C. Distinct pathways of antigen uptake and intracellular routing in CD4 and CD8 T cell activation. Science 316, 612–616 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Ramachandra, L., Noss, E., Boom, W. H. & Harding, C. V. Phagocytic processing of antigens for presentation by class II major histocompatibility complex molecules. Cell Microbiol. 1, 205–214 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Ngwenyama, N. et al. CXCR3 regulates CD4+ T cell cardiotropism in pressure overload-induced cardiac dysfunction. JCI Insight https://doi.org/10.1172/jci.insight.125527 (2019).

  15. Michailowsky, V. et al. Intercellular adhesion molecule 1 deficiency leads to impaired recruitment of T lymphocytes and enhanced host susceptibility to infection with Trypanosoma cruzi. J. Immunol. 173, 463–470 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Cardillo, F., Voltarelli, J. C., Reed, S. G. & Silva, J. S. Regulation of Trypanosoma cruzi infection in mice by gamma interferon and interleukin 10: role of NK cells. Infect. Immun. 64, 128–134 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Haverson, K., Singha, S., Stokes, C. R. & Bailey, M. Professional and nonprofessional antigen-presenting cells in the porcine small intestine. Immunology 101, 492–500 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Geppert, T. D. & Lipsky, P. E. Antigen presentation by interferon-gamma-treated endothelial cells and fibroblasts: differential ability to function as antigen-presenting cells despite comparable Ia expression. J. Immunol. 135, 3750–3762 (1985).

    CAS  PubMed  Google Scholar 

  19. Spear, T. T., Evavold, B. D., Baker, B. M. & Nishimura, M. I. Understanding TCR affinity, antigen specificity, and cross-reactivity to improve TCR gene-modified T cells for cancer immunotherapy. Cancer Immunol. Immunother. 68, 1881–1889 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Ngwenyama, N. et al. Isolevuglandin-modified cardiac proteins drive CD4+ T cell activation in the heart and promote cardiac dysfunction. Circulation https://doi.org/10.1161/CIRCULATIONAHA.120.051889 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Rieckmann, M. et al. Myocardial infarction triggers cardioprotective antigen-specific T helper cell responses. J. Clin. Invest. 129, 4922–4936 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tallquist, M. D. & Molkentin, J. D. Redefining the identity of cardiac fibroblasts. Nat. Rev. Cardiol. 14, 484–491 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Kong, P., Christia, P. & Frangogiannis, N. G. The pathogenesis of cardiac fibrosis. Cell. Mol. Life Sci. 71, 549–574 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Fields, P. E. et al. B7.1 is a quantitatively stronger costimulus than B7.2 in the activation of naive CD8+ TCR-transgenic T cells. J. Immunol. 161, 5268–5275 (1998).

    CAS  PubMed  Google Scholar 

  25. Pechhold, K. et al. Inflammatory cytokines IFN-gamma plus TNF-alpha induce regulated expression of CD80 (B7-1) but not CD86 (B7-2) on murine fibroblasts. J. Immunol. 158, 4921–4929 (1997).

    CAS  PubMed  Google Scholar 

  26. Smythe, J. A. et al. Human fibroblasts transduced with CD80 or CD86 efficiently trans-costimulate CD4+ and CD8+ T lymphocytes in HLA-restricted reactions: implications for immune augmentation cancer therapy and autoimmunity. J. Immunol. 163, 3239–3249 (1999).

    CAS  PubMed  Google Scholar 

  27. Sansom, D. M. CD28, CTLA-4 and their ligands: who does what and to whom? Immunology 101, 169–177 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hulsmans, M. et al. Cardiac macrophages promote diastolic dysfunction. J. Exp. Med. 215, 423–440 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Seeberg, J. C. et al. Nonprofessional phagocytosis: a general feature of normal tissue cells. Sci Rep. 9, 11875 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Nakaya, M. et al. Cardiac myofibroblast engulfment of dead cells facilitates recovery after myocardial infarction. J. Clin. Invest. 127, 383–401 (2017).

    Article  PubMed  Google Scholar 

  31. Salvador, A. M. et al. Intercellular adhesion molecule 1 regulates left ventricular leukocyte infiltration, cardiac remodeling, and function in pressure overload-induced heart failure. J. Am. Heart Assoc. 5, e003126 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Wang, H. et al. Role of bone marrow-derived CD11c+ dendritic cells in systolic overload-induced left ventricular inflammation, fibrosis and hypertrophy. Basic Res. Cardiol. 112, 25 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Virani, S. S. et al. Heart Disease and Stroke Statistics—2020 update: a report from the American Heart Association. Circulation 141, e139–e596 (2020).

    Article  PubMed  Google Scholar 

  34. Ren, Z. et al. Single-cell reconstruction of progression trajectory reveals intervention principles in pathological cardiac hypertrophy. Circulation 141, 1704–1719 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Wang, L. et al. Single-cell reconstruction of the adult human heart during heart failure and recovery reveals the cellular landscape underlying cardiac function. Nat. Cell Biol. 22, 108–119 (2020).

    Article  PubMed  Google Scholar 

  36. Dubrot, J. et al. Lymph node stromal cells acquire peptide-MHCII complexes from dendritic cells and induce antigen-specific CD4+ T cell tolerance. J. Exp. Med. 211, 1153–1166 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Perez-Shibayama, C. et al. Type I interferon signaling in fibroblastic reticular cells prevents exhaustive activation of antiviral CD8+ T cells. Sci. Immunol. https://doi.org/10.1126/sciimmunol.abb7066 (2020).

  38. Richards, D. A. et al. Distinct phenotypes induced by three degrees of transverse aortic constriction in mice. Sci Rep. 9, 5844 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

These studies were supported by NIH grants R01 HL123658 and R01 HL144477 (to P.A.), NIH T32 HL 69770, NIH T32AI007077-34, NIH F31HL140883 and the American Heart Association grant 18PRE34020084 (to N.N.), NIH R01 HL141187 and HL142624 (to J.D.) and NIH T32AG066574 (to D.B.).

Author information

Authors and Affiliations

Authors

Contributions

N.N. designed and conducted experiments, analyzed data and wrote the manuscript. K.K., R.B., S.P. and B.T. planned and conducted experiments. C.G. and M.A.P. contributed to experiment design and intellectually contributed to data analysis. M.A. performed the TAC surgeries and contributed to experiment design. J.D. and D.B. performed TAC and immunofluorescence in Tcf21iCre/+; R26eGFP mice, sorted GFP+ cardiac fibroblasts and intellectually contributed to the manuscript. P.A. overviewed the design and interpretation of all studies and wrote the paper.

Corresponding author

Correspondence to Pilar Alcaide.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks the anonymous reviewers for their contribution to the peer review of this work.

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 Cardiac fibroblasts express MHCII in vivo in response to acute T. cruzi infection.

(A) Wt mice were inoculated with 20,000 T. cruzi parasites by intraperitoneal injection and the hearts were harvested at 19 days post-infection. (B-E) Ventricular CD31-CD45-MEFSK4+ cardiac fibroblasts were analysed by flow cytometry (≥ 5,000 target cells acquired) to determine surface expression of (B-C) MHCII and (D-E) CD80. n= 4 (mock) and n=3 (Chagas) mice. Error bars represent mean ± SD. (* p≤0.05; *** p<0.001; Mann-Whitney test, two tailed).

Source data

Extended Data Fig. 2 MhcII recombination in cardiac fibroblasts occurred in a cell-specific manner.

Detection of Intact allele (310bp), recombined alleles (475 bp) and GAPDH(156 bp) in sorted CD31-CD45-MEFSK4+ Fibroblasts and leukocytes from Tcf21iCre/+MhcIIfl/fl mice treated with vehicle or Tamoxifen. GAPDH was used as loading control.

Extended Data Fig. 3 Tamoxifen treatment of Tcf21iCre/+/ MhcIIflox/flox mice reduces MHCII surface protein expression specifically in cardiac fibroblasts.

Transverse aortic constriction (TAC) surgery was performed on vehicle and Tamoxifen treated Tcf21iCre/+MhcIIfl/fl mice and the left ventricle was harvested after 4 weeks, digested, stained and analysed by flow cytometry. (A) Representative FACS plot showing gating of CD31+CD45-MESFK4- endothelial cells, CD31-CD45+MESFK4- leukocytes and CD31-CD45-MESFK4+ cardiac fibroblasts. (B-C) Representative FACS plots (B) and quantification of mean fluorescence intensity (MFI) (C) of MHC-II expression in endothelial cells (top), leukocytes (middle) and cardiac fibroblasts (bottom) in the hearts of vehicle and tamoxifen treated mice. n=4 mice/group. Error bars represent mean ± SE. (* p<0.05; Mann-Whitney test, two tailed. C: p value=0.0286).

Source data

Extended Data Fig. 4 Cardiac fibroblast MHCII expression does not affect CD4+ T cell activation in the mLNs in response to TAC.

Transverse aortic constriction (TAC) surgery was performed on vehicle and Tamoxifen treated Tcf21iCre/+MhcIIfl/fl mice and the mediastinal lymph nodes were harvested after 4 weeks, stained and analysed by flow cytometry for the indicated T cell activation markers. (A) Representative FACS plot showing CD62L and CD44 staining. (B-C) quantification of CD62LlowCD44hi effector T CD4+ T cell numbers (B) and frequency (C) in vehicle and tamoxifen treated TAC mice. n=5 mice (vehicle) and n=6 mice (tamoxifen). Error bars represent mean ± SD. (Mann-Whitney test, two tailed. C: p value=0.0286).

Source data

Extended Data Fig. 5 Cardiac fibroblast MHCII expression contributes to cardiomyocyte hypertrophy and to cardiac fibroblast abundance in response to pressure overload.

Transverse aortic constriction (TAC) was performed on vehicle and Tamoxifen treated Tcf21iCre/+MhcIIfl/fl mice and harvested after 4 weeks. Conditional deletion of MhcII on cardiac fibroblasts was induced by administering Tamoxifen via intraperitoneal injections prior to TAC surgery, followed by Tamoxifen citrate chow for 4 weeks. (A) Wheat germ agglutinin (WGA) staining of frozen LV tissue sections was performed and used to calculate (B) mean cardiomyocyte area. Representative images of n=3 hearts (Sham), n=4 (TAC vehicle) and n=7 (TAC TMX). (C) Gross LV mass was acquired and is shown normalized to tibia length. (D-E) LV tissue was digested, stained for CD31 and MESFK4 and analysed by flow cytometry. Representative FACS plot (D) and quantification (E) of vehicle and tamoxifen treated mice hearts (n=4 mice/group). Error bars represent mean ± SE. (* p<0.01; Mann-Whitney test, two tailed: E: p=0286).

Source data

Extended Data Fig. 6 Schematic Diagram showing bidirectionality of cardiac fibroblast and Th1 cell interactions.

Cardiac fibroblasts express the costimulatory molecule CD80 and efficiently capture and process extracellular antigens into small peptides that are uploaded into MHC-II molecules. In response to IFNγ stimulation, cardiac fibroblasts express MHCII and present peptide antigens that induce IFNγ + Th1 cell immune responses and promote cardiac remodelling and dysfunction.

Supplementary information

Source data

Source Data Fig. 1

Statistics and data for graphs in Fig. 1.

Source Data Fig. 2

Statistics and data for graphs in Fig. 2.

Source Data Fig. 3

Statistics and data for graphs in Fig. 3.

Source Data Fig. 4

Statistics and data for graphs in Fig. 4.

Source Data Fig. 5

Statistics and data for graphs in Fig. 5.

Source Data Fig. 6

Statistics and data for graphs in Fig. 6.

Source Data Fig. 7

Statistics and data for graphs in Fig. 7.

Source Data Extended Data Fig. 1

Statistics and data for graphs in Extended data Fig. 1.

Source Data Extended Data Fig. 3

Statistics and data for graphs in Extended data Fig. 3.

Source Data Extended Data Fig. 4

Statistics and data for graphs in Extended data Fig. 4.

Source Data Extended Data Fig. 5

Statistics and data for graphs in Extended data Fig. 5.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Ngwenyama, N., Kaur, K., Bugg, D. et al. Antigen presentation by cardiac fibroblasts promotes cardiac dysfunction. Nat Cardiovasc Res 1, 761–774 (2022). https://doi.org/10.1038/s44161-022-00116-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-022-00116-7

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