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:

Bat3 promotes T cell responses and autoimmunity by repressing Tim-3–mediated cell death and exhaustion

This article has been updated

Abstract

T cell immunoglobulin and mucin domain–containing 3 (Tim-3) is an inhibitory receptor that is expressed on exhausted T cells during infection with HIV-1 and hepatitis C virus. By contrast, Tim-3 expression and function are defective in multiple human autoimmune diseases. However, the molecular mechanisms modulating Tim-3 function are not well understood. Here we show that human leukocyte antigen B (HLA-B)-associated transcript 3 (Bat3) binds to, and represses the function of, Tim-3. Bat3 protects T helper type 1 (TH1) cells from galectin-9–mediated cell death and promotes both proliferation and proinflammatory cytokine production. Bat3-deficient T cells have elevated expression of exhaustion-associated molecules such as Tim-3, Lag3, Prdm1 and Pbx3, and Bat3 knockdown in myelin-antigen–specific CD4+ T cells markedly inhibits the development of experimental autoimmune encephalomyelitis while promoting the expansion of a dysfunctional Tim-3hi, interferon-γ (IFN-γ)loCD4+ cell population. Furthermore, expression of Bat3 is reduced in exhausted Tim-3+ T cells from mouse tumors and HIV-1–infected individuals. These data indicate that Bat3 acts as an inhibitor of Tim-3–dependent exhaustion and cell death. Bat3 may thus represent a viable therapeutic target in autoimmune disorders, chronic infections and cancers.

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: Bat3 binds to the Tim-3 tail in a galectin-9–dependent manner.
Figure 2: Bat3 promotes TH1 cell function and protects from galectin-9–mediated cell death.
Figure 3: Bat3 expression in the hematopoietic compartment promotes CNS autoimmunity.
Figure 4: Loss of Bat3 function in T cells induces an exhausted-like phenotype.
Figure 5: Bat3 expression in CD4+ T cells is required for TH1 cell–driven EAE.
Figure 6: Bat3 expression is reduced in Tim-3+ exhausted T cells.

Similar content being viewed by others

Change history

  • 21 August 2012

     In the version of this article initially published online, the x axis in Figure 6c was incorrectly labeled. The error has been corrected for the print, PDF and HTML versions of this article.

References

  1. Dunn, G.P. et al. Interferon-γ and cancer immunoediting. Immunol. Res. 32, 231–245 (2005).

    Article  CAS  Google Scholar 

  2. Myrmel, H., Ulvestad, E. & Asjo, B. The hepatitis C virus enigma. APMIS 117, 427–439 (2009).

    Article  CAS  Google Scholar 

  3. Romagnani, S. Th1/Th2 cells. Inflamm. Bowel Dis. 5, 285–294 (1999).

    Article  CAS  Google Scholar 

  4. Moss, R.B. et al. Th1/Th2 cells in inflammatory disease states: therapeutic implications. Expert Opin. Biol. Ther. 4, 1887–1896 (2004).

    Article  CAS  Google Scholar 

  5. Monney, L. et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature 415, 536–541 (2002).

    Article  CAS  Google Scholar 

  6. Zhu, C. et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat. Immunol. 6, 1245–1252 (2005).

    Article  CAS  Google Scholar 

  7. Sabatos, C.A. et al. Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat. Immunol. 4, 1102–1110 (2003).

    Article  CAS  Google Scholar 

  8. Sánchez-Fueyo, A. et al. Tim-3 inhibits T helper type 1-mediated auto- and alloimmune responses and promotes immunological tolerance. Nat. Immunol. 4, 1093–1101 (2003).

    Article  Google Scholar 

  9. Koguchi, K. et al. Dysregulated T cell expression of TIM3 in multiple sclerosis. J. Exp. Med. 203, 1413–1418 (2006).

    Article  CAS  Google Scholar 

  10. Yang, L., Anderson, D.E., Kuchroo, J. & Hafler, D.A. Lack of TIM-3 immunoregulation in multiple sclerosis. J. Immunol. 180, 4409–4414 (2008).

    Article  CAS  Google Scholar 

  11. Morimoto, K. et al. Dysregulated upregulation of T-cell immunoglobulin and mucin domain-3 on mucosal T helper 1 cells in patients with Crohn's disease. Scand. J. Gastroenterol. 46, 701–709 (2011).

    Article  CAS  Google Scholar 

  12. Jones, R.B. et al. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J. Exp. Med. 205, 2763–2779 (2008).

    Article  CAS  Google Scholar 

  13. Golden-Mason, L. et al. Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J. Virol. 83, 9122–9130 (2009).

    Article  CAS  Google Scholar 

  14. Jin, H.T. et al. Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proc. Natl. Acad. Sci. USA 107, 14733–14738 (2010).

    Article  CAS  Google Scholar 

  15. Takamura, S. et al. Premature terminal exhaustion of Friend virus-specific effector CD8+ T cells by rapid induction of multiple inhibitory receptors. J. Immunol. 184, 4696–4707 (2010).

    Article  CAS  Google Scholar 

  16. Sakuishi, K. et al. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 207, 2187–2194 (2010).

    Article  CAS  Google Scholar 

  17. Fourcade, J. et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J. Exp. Med. 207, 2175–2186 (2010).

    Article  CAS  Google Scholar 

  18. Zhou, Q. et al. Co-expression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood 117, 4501–4510 (2011).

    Article  CAS  Google Scholar 

  19. Elahi, S. et al. Protective HIV-specific CD8+ T cells evade Treg cell suppression. Nat. Med. 17, 989–995 (2011).

    Article  CAS  Google Scholar 

  20. Desmots, F., Russell, H.R., Lee, Y., Boyd, K. & McKinnon, P.J. The reaper-binding protein scythe modulates apoptosis and proliferation during mammalian development. Mol. Cell. Biol. 25, 10329–10337 (2005).

    Article  CAS  Google Scholar 

  21. Sasaki, T. et al. HLA-B–associated transcript 3 (Bat3)/Scythe is essential for p300-mediated acetylation of p53. Genes Dev. 21, 848–861 (2007).

    Article  CAS  Google Scholar 

  22. Kuchroo, V.K. et al. Experimental allergic encephalomyelitis mediated by cloned T cells specific for a synthetic peptide of myelin proteolipid protein. Fine specificity and T cell receptor V β usage. J. Immunol. 148, 3776–3782 (1992).

    CAS  Google Scholar 

  23. van de Weyer, P.S. et al. A highly conserved tyrosine of Tim-3 is phosphorylated upon stimulation by its ligand galectin-9. Biochem. Biophys. Res. Commun. 351, 571–576 (2006).

    Article  CAS  Google Scholar 

  24. Bettelli, E. et al. Myelin oligodendrocyte glycoprotein-specific T cell receptor transgenic mice develop spontaneous autoimmune optic neuritis. J. Exp. Med. 197, 1073–1081 (2003).

    Article  CAS  Google Scholar 

  25. Tsunoda, I. & Fujinami, R.S. Two models for multiple sclerosis: experimental allergic encephalomyelitis and Theiler's murine encephalomyelitis virus. J. Neuropathol. Exp. Neurol. 55, 673–686 (1996).

    Article  CAS  Google Scholar 

  26. Jäger, A., Dardalhon, V., Sobel, R.A., Bettelli, E. & Kuchroo, V.K. Th1, Th17, and Th9 effector cells induce experimental autoimmune encephalomyelitis with different pathological phenotypes. J. Immunol. 183, 7169–7177 (2009).

    Article  Google Scholar 

  27. Wherry, E.J. T cell exhaustion. Nat. Immunol. 12, 492–499 (2011).

    Article  CAS  Google Scholar 

  28. Blackburn, S.D. & Wherry, E.J. IL-10, T cell exhaustion and viral persistence. Trends Microbiol. 15, 143–146 (2007).

    Article  CAS  Google Scholar 

  29. Priatel, J.J. et al. Chronic immunodeficiency in mice lacking RasGRP1 results in CD4 T cell immune activation and exhaustion. J. Immunol. 179, 2143–2152 (2007).

    Article  CAS  Google Scholar 

  30. Shin, H. et al. A role for the transcriptional repressor Blimp-1 in CD8+ T cell exhaustion during chronic viral infection. Immunity 31, 309–320 (2009).

    Article  CAS  Google Scholar 

  31. Wherry, E.J. et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007).

    Article  CAS  Google Scholar 

  32. Blackburn, S.D. et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 10, 29–37 (2009).

    Article  CAS  Google Scholar 

  33. Lee, J. et al. Phosphotyrosine-dependent coupling of Tim-3 to T-cell receptor signaling pathways. Mol. Cell. Biol. 31, 3963–3974 (2011).

    Article  CAS  Google Scholar 

  34. Mustelin, T. & Tasken, K. Positive and negative regulation of T-cell activation through kinases and phosphatases. Biochem. J. 371, 15–27 (2003).

    Article  CAS  Google Scholar 

  35. Nika, K. et al. Constitutively active Lck kinase in T cells drives antigen receptor signal transduction. Immunity 32, 766–777 (2010).

    Article  CAS  Google Scholar 

  36. Oosterwegel, M.A., Greenwald, R.J., Mandelbrot, D.A., Lorsbach, R.B. & Sharpe, A.H. CTLA-4 and T cell activation. Curr. Opin. Immunol. 11, 294–300 (1999).

    Article  CAS  Google Scholar 

  37. Okazaki, T., Maeda, A., Nishimura, H., Kurosaki, T. & Honjo, T. PD-1 immunoreceptor inhibits B cell receptor–mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc. Natl. Acad. Sci. USA 98, 13866–13871 (2001).

    Article  CAS  Google Scholar 

  38. Chemnitz, J.M., Parry, R.V., Nichols, K.E., June, C.H. & Riley, J.L. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J. Immunol. 173, 945–954 (2004).

    Article  CAS  Google Scholar 

  39. Yokosuka, T. et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 209, 1201–1217 (2012).

    Article  CAS  Google Scholar 

  40. Thress, K., Song, J., Morimoto, R.I. & Kornbluth, S. Reversible inhibition of Hsp70 chaperone function by Scythe and Reaper. EMBO J. 20, 1033–1041 (2001).

    Article  CAS  Google Scholar 

  41. Winnefeld, M. et al. Human SGT interacts with Bag-6/Bat-3/Scythe and cells with reduced levels of either protein display persistence of few misaligned chromosomes and mitotic arrest. Exp. Cell Res. 312, 2500–2514 (2006).

    Article  CAS  Google Scholar 

  42. Desmots, F., Russell, H.R., Michel, D. & McKinnon, P.J. Scythe regulates apoptosis-inducing factor stability during endoplasmic reticulum stress-induced apoptosis. J. Biol. Chem. 283, 3264–3271 (2008).

    Article  CAS  Google Scholar 

  43. Tsukahara, T. et al. Scythe/BAT3 regulates apoptotic cell death induced by papillomavirus binding factor in human osteosarcoma. Cancer Sci. 100, 47–53 (2009).

    Article  CAS  Google Scholar 

  44. Kwak, J.H., Kim, S.I., Kim, J.K. & Choi, M.E. BAT3 interacts with transforming growth factor-β (TGF-β) receptors and enhances TGF-β1–induced type I collagen expression in mesangial cells. J. Biol. Chem. 283, 19816–19825 (2008).

    Article  CAS  Google Scholar 

  45. Degli-Esposti, M.A. et al. Ancestral haplotypes reveal the role of the central MHC in the immunogenetics of IDDM. Immunogenetics 36, 345–356 (1992).

    Article  CAS  Google Scholar 

  46. Hara, H. et al. The apoptotic protease-activating factor 1–mediated pathway of apoptosis is dispensable for negative selection of thymocytes. J. Immunol. 168, 2288–2295 (2002).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Kozoriz, D. Lee, J. Sullivan and M. King for technical assistance; J. Zhang (Brigham and Women's Hospital, Boston) for reagents; B.H. Waksman for valuable guidance and discussion; and B. Walker for critical reading of the manuscript. M.R. was previously supported by Fellowships from the Multiple Sclerosis Society of Canada (MSSOC) and the Canadian Institutes of Health Research (CIHR) and is currently supported by the EMD Serono, Canada and an MS Research and Training Network Transitional Career Development Award from the MSSOC and the Multiple Sclerosis Scientific Research Foundation. K.S. holds a Fellowship from the Sankyo Foundation of Life Science. These studies were funded by grants from the US National Institutes of Health to V.K.K. (NS045937, AI073748 and NS038037) and S.X. (K01DK090105), the Ragon Institute of MGH, MIT and Harvard (V.K.K., C.Z. and M.M.A.), the American Cancer Society (A.C.A.) and the Harvard University Center for AIDS Research (M.M.A.; P30 AI060354).

Author information

Authors and Affiliations

Authors

Contributions

M.R. and C.Z. designed and performed experiments, and collected data. M.R. wrote the manuscript. K.S., S.X., J.K., A.C. and M.A. provided help in performing experiments. A.W. helped generate fetal liver chimeric mice. E.A.G. helped generate Bat3 antiserum. R.A.S. analyzed histopathological data. H.O. generated the Bag6−/− strain. P.J.M. and T.W.M. provided Bag6+/− mice and fetal liver chimeras, respectively. M.M.A. and A.C.A. designed and supervised experiments involving HIV samples and tumor mice, respectively. V.K.K. supervised the project and edited the manuscript.

Corresponding author

Correspondence to Vijay K Kuchroo.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–14, Supplementary Table 1 and Supplementary Methods (PDF 1138 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Rangachari, M., Zhu, C., Sakuishi, K. et al. Bat3 promotes T cell responses and autoimmunity by repressing Tim-3–mediated cell death and exhaustion. Nat Med 18, 1394–1400 (2012). https://doi.org/10.1038/nm.2871

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2871

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