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:

Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans

Abstract

T cell exhaustion is an induced state of dysfunction that arises in response to chronic infection and cancer. Exhausted CD8+ T cells acquire a distinct epigenetic state, but it is not known whether that chromatin landscape is fixed or plastic following the resolution of a chronic infection. Here we show that the epigenetic state of exhaustion is largely irreversible, even after curative therapy. Analysis of chromatin accessibility in HCV- and HIV-specific responses identifies a core epigenetic program of exhaustion in CD8+ T cells, which undergoes only limited remodeling before and after resolution of infection. Moreover, canonical features of exhaustion, including super-enhancers near the genes TOX and HIF1A, remain ‘epigenetically scarred.’ T cell exhaustion is therefore a conserved epigenetic state that becomes fixed and persists independent of chronic antigen stimulation and inflammation. Therapeutic efforts to reverse T cell exhaustion may require new approaches that increase the epigenetic plasticity of exhausted T cells.

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: Distinct epigenetic changes underlie CD8+ T cell responses to Flu and chronic HCV infection.
Fig. 2: Epigenetic signature of T cell exhaustion is conserved across chronic viral infections.
Fig. 3: HCV-specific CD8+ T cells retain epigenetic scars of exhaustion despite cure of the chronic infection.
Fig. 4: Chronic TCR signaling, and not the inflammatory milieu, drives epigenetic scarring in exhausted CD8+ T cells.
Fig. 5: Scarred regions identify critical regulators of exhaustion.
Fig. 6: Epigenetic scars of exhaustion are retained long-term following infection cure.

Similar content being viewed by others

Data availability

All sequencing data from this study will be made publicly available through the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/) and/or NCBI database of Genotypes and Phenotypes (dbGaP; https://www.ncbi.nlm.nih.gov/gap/). All other relevant data are available from the corresponding authors on request.

References

  1. Hashimoto, M. et al. CD8 T cell exhaustion in chronic infection and cancer: opportunities for interventions. Annu. Rev. Med. 69, 301–318 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  3. Doering, T. A. et al. Network analysis reveals centrally connected genes and pathways involved in CD8+ T cell exhaustion versus memory. Immunity 37, 1130–1144 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165–1169 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gallimore, A. et al. Induction and exhaustion of lymphocytic choriomeningitis virus–specific cytotoxic T lymphocytes visualized using soluble tetrameric major histocompatibility complex class I–peptide complexes. J. Exp. Med. 187, 1383–1393 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zajac, A. J. et al. Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188, 2205–2213 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wherry, E. J., Blattman, J. N., Murali-Krishna, K., van der Most, R. & Ahmed, R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J. Virol. 77, 4911–4927 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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  PubMed  Google Scholar 

  9. Day, C. L. et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443, 350–354 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Gruener, N. H. et al. Sustained dysfunction of antiviral CD8+ T lymphocytes after infection with hepatitis C virus. J. Virol. 75, 5550–5558 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wedemeyer, H. et al. Impaired effector function of hepatitis C virus-specific CD8+ T cells in chronic hepatitis C virus infection. J. Immunol. 169, 3447–3458 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Radziewicz, H. et al. Liver-infiltrating lymphocytes in chronic human hepatitis C virus infection display an exhausted phenotype with high levels of PD-1 and low levels of CD127 expression. J. Virol. 81, 2545–2553 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Wolski, D. et al. Early transcriptional divergence marks virus-specific primary human CD8+ T cells in chronic versus acute infection. Immunity 47, 648–663 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Micallef, J. M., Kaldor, J. M. & Dore, G. J. Spontaneous viral clearance following acute hepatitis C infection: a systematic review of longitudinal studies. J. Viral Hepat. 13, 34–41 (2006).

    Article  CAS  PubMed  Google Scholar 

  15. Cox, A. L. et al. Cellular immune selection with hepatitis C virus persistence in humans. J. Exp. Med. 201, 1741–1752 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kuntzen, T. et al. Viral sequence evolution in acute hepatitis C virus infection. J. Virol. 81, 11658–11668 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rutebemberwa, A. et al. High-programmed death-1 levels on hepatitis C virus-specific T cells during acute infection are associated with viral persistence and require preservation of cognate antigen during chronic infection. J. Immunol. 181, 8215–8225 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Feld, J. J. et al. Treatment of HCV with ABT-450/r-ombitasvir and dasabuvir with ribavirin. N. Engl. J. Med. 370, 1594–1603 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Ferenci, P. et al. ABT-450/r-ombitasvir and dasabuvir with or without ribavirin for HCV. N. Engl. J. Med. 370, 1983–1992 (2014).

    Article  PubMed  CAS  Google Scholar 

  20. Baumert, T. F., Berg, T., Lim, J. K. & Nelson, D. R. Status of direct-acting antiviral therapy for hepatitis C virus infection and remaining challenges. Gastroenterology 156, 431–445 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Wieland, D. et al. TCF1+ hepatitis C virus-specific CD8+ T cells are maintained after cessation of chronic antigen stimulation. Nat. Commun. 8, 15050 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Hensel, N. et al. Memory-like HCV-specific CD8+ T cells retain a molecular scar after cure of chronic HCV infection. Nat. Immunol. 22, 229–239 (2021).

    Article  CAS  PubMed  Google Scholar 

  23. Joshi, N. S. et al. Inflammation directs memory precursor and short-lived effector CD8+ T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Martinez, G. J. et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 42, 265–278 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gate, R. E. et al. Genetic determinants of co-accessible chromatin regions in activated T cells across humans. Nat. Genet. 50, 1140–1150 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Rasool, S. T. et al. Increased level of IL-32 during human immunodeficiency virus infection suppresses HIV replication. Immunol. Lett. 117, 161–167 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. El-Far, M. et al. Proinflammatory isoforms of IL-32 as novel and robust biomarkers for control failure in HIV-infected slow progressors. Sci. Rep. 6, 22902 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nguyen, S. et al. Elite control of HIV is associated with distinct functional and transcriptional signatures in lymphoid tissue CD8+ T cells. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aax4077 (2019).

  29. Zaidan, S. M. et al. Upregulation of IL-32 isoforms in virologically suppressed HIV-infected individuals: potential role in persistent inflammation and transcription from stable HIV-1 reservoirs. J. Acquir. Immune Defic. Syndr. 82, 503–513 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chinnaswamy, S. Genetic variants at the IFNL3 locus and their association with hepatitis C virus infections reveal novel insights into host-virus interactions. J. Interferon Cytokine Res. 34, 479–497 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Monteleone, K. et al. Interleukin-32 isoforms: expression, interaction with interferon-regulated genes and clinical significance in chronically HIV-1-infected patients. Med. Microbiol. Immunol. 203, 207–216 (2014).

    CAS  PubMed  Google Scholar 

  32. Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486–499 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chipumuro, E. et al. CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 159, 1126–1139 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Adam, R. C. et al. Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice. Nature 521, 366–370 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Vahedi, G. et al. Super-enhancers delineate disease-associated regulatory nodes in T cells. Nature 520, 558–562 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ma, S. et al. Chromatin potential identified by shared single-cell profiling of RNA and chromatin. Cell 183, 1103–1116 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Scott, A. C. et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Alfei, F. et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Urbani, S. et al. PD-1 expression in acute hepatitis C virus (HCV) infection is associated with HCV-specific CD8 exhaustion. J. Virol. 80, 11398–11403 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bengsch, B. et al. Coexpression of PD-1, 2B4, CD160 and KLRG1 on exhausted HCV-specific CD8+ T cells is linked to antigen recognition and T cell differentiation. PLoS Pathog. 6, e1000947 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Pauken, K. E. et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 354, 1160–1165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Alatrakchi, N. et al. Hepatitis C virus (HCV)-specific CD8+ cells produce transforming growth factor β that can suppress HCV-specific T-cell responses. J. Virol. 81, 5882–5892 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Brooks, D. G. et al. Interleukin-10 determines viral clearance or persistence in vivo. Nat. Med. 12, 1301–1309 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ejrnaes, M. et al. Resolution of a chronic viral infection after interleukin-10 receptor blockade. J. Exp. Med. 203, 2461–2472 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Tinoco, R., Alcalde, V., Yang, Y., Sauer, K. & Zuniga, E. I. Cell-intrinsic transforming growth factor-β signaling mediates virus-specific CD8+ T cell deletion and viral persistence in vivo. Immunity 31, 145–157 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kroy, D. C. et al. Liver environment and HCV replication affect human T-cell phenotype and expression of inhibitory receptors. Gastroenterology 146, 550–561 (2014).

    Article  PubMed  Google Scholar 

  50. Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293–300 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Rutishauser, R. L. et al. Early and delayed antiretroviral therapy results in comparable reductions in CD8+ T cell exhaustion marker expression. AIDS Res. Hum. Retroviruses 33, 658–667 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Youngblood, B. et al. Cutting edge: prolonged exposure to HIV reinforces a poised epigenetic program for PD-1 expression in virus-specific CD8 T cells. J. Immunol. 191, 540–544 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. The SPARTAC Trial Investigators. Short-course antiretroviral therapy in primary HIV infection. N. Engl. J. Med. 368, 207–217 (2013).

  54. Hoffmann, M. et al. Exhaustion of activated CD8 T cells predicts disease progression in primary HIV-1 infection. PLoS Pathog. 12, e1005661 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Amemiya, H. M., Kundaje, A. & Boyle, A. P. The ENCODE blacklist: identification of problematic regions of the genome. Sci. Rep. 9, 9354 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank the members of the Haining laboratory for their input and the research participants for their participation. This work was supported by NIH grant no. U19 AI086230 and funding from the Parker Institute for Cancer Immunotherapy. S.A.W. was supported by NIH grant nos. T32 GM007753 and T32 CA207021; R.T.C. was supported by AI136715 and the MGH Research Scholars Program. We also thank the Center for Virology and Vaccine Research Flow Cytometry Core at Beth Israel Deaconess Medical Center for their assistance.

Author information

Authors and Affiliations

Authors

Contributions

D.R.S. and W.N.H. conceived the study and designed the experiments. D.R.S., K.B.Y., G.E.M., U.G., R.A.A., D.E.C. and S.A.W. performed experiments and/or data analysis. P.T., D.W., D.C.T., R.T.C., T.M.A., A.Y.K. and G.M.L. contributed to the HCV clinical trial design, patient recruitment, sample processing, viral sequencing studies and/or transcriptional analysis. G.E.M., S.F., J. Frater. and J. Fox contributed to the HIV clinical trial design, patient recruitment and/or sample processing. D.R.S. and W.N.H. wrote the manuscript; all authors reviewed and edited the manuscript.

Corresponding authors

Correspondence to W. Nicholas Haining or Debattama R. Sen.

Ethics declarations

Competing interests

AbbVie sponsored the clinical trial (NCT02476617) and gave input to the trial design as well as the clinical and biological sample collection schedule. W.N.H. is an employee of Merck and Company and holds equity in Tango Therapeutics and Arsenal Biosciences. The other authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Hazem Ghoneim, Barbara Rehermann and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Isolation and chromatin accessibility profiling of Flu and HCV multimer+ CD8+ T cells in HCV infection.

a, Representative flow cytometry sorting strategy for Flu and HCV multimer+ CD8+ T cells. b, Recovered numbers of Flu (top) and HCV (bottom) multimer+ cells for each donor during chronic HCV infection. c, Recovered numbers of HCV multimer+ cells for each donor during resolved HCV infection. d, Combined ATAC signal across all TSSs for each biological condition from each donor. Green and range bands indicate ranges for ideal and acceptable values, respectively, for TSS enrichment per ENCODE standards. e, Boxplots of pairwise Pearson correlations for data from individual donors within each biological condition. Center, median; box limits, first and third percentiles; whiskers, min and max. N = 6 donors. f, Combined ATAC signal across all H3K27ac peaks (red), H3K27me3 (green) and H3K4me3 (purple). Histone mark peaks were determined from the following samples on ENCODE: ENCFF653OGM - H3K27ac, ENCFF285FID - H3K4me3, ENCFF367HSC - H3K27me3. g, Clustered similarity matrix between the indicated biological conditions in the chronically-infected and spontaneously resolved cohort. h, Density of overlapping GWAS SNPs per 1000 bp in Flu-specific, HCV-specific or non-differential ChARs.

Extended Data Fig. 2 Isolation and chromatin accessibility profiling of HIV multimer+ CD8+ T cells in HIV infection.

a, Representative flow cytometry sorting strategy for HIV multimer+ CD8+ T cells. b, Boxplots of pairwise Pearson correlations for data from individual donors within each biological condition. Center, median; box limits, first and third percentiles; whiskers, min and max. N = 11 donors. c, Combined principal component analysis of naïve, HIV-, HCV- and Flu- specific CD8+ T cells from the HIV and HCV cohorts.

Extended Data Fig. 3 Features of HCV-specific CD8+ T cells before and after DAA therapy.

a, PD-1 and CD39 staining on tetramer populations before (top) and after (bottom) DAA therapy. b, Partitioning of scarred and reversed regions into those overlapping promoters, UTRs, exons, introns and intergenic areas as indicated. c, Classification of SNPs falling within scarred, reversed or gained ChARs. SNPs that were subcategorized into those associated with chronic viral infection are summarized in Supplementary Table 3.

Extended Data Fig. 4 Minimal impact of the inflammatory milieu on chromatin accessibility within bystander populations in chronic HCV.

a, CCR7 and CD45RA staining on all CD8+ T cells (left) and Flu-specific T cells (right). b, Venn diagram of ChAR overlap in Flu tet+ cells (bottom) from HCV-infected donors and healthy donors. c, PD-1 and CD39 staining on tetramer populations before (top) and after (bottom) DAA therapy. d, Combined principal component analysis of naïve, effector memory, HCV-, and Flu- specific CD8+ T cells from the healthy and HCV cohorts. e, Heatmap showing pathway enrichment (rows) within clustered ChARs from Fig. 4g (columns).

Extended Data Fig. 5 Validation of ATAC-seq-based super-enhancer inference across multiple datasets.

a, H3K27ac ChIP-seq signal (top) and ATAC-seq signal (bottom) at the ETS1 gene locus. b, ROC plots of varying cutoff for ATAC-based ranking of super-enhancer to predict bona fide super-enhancers defined using matched tissue-specific H3K27ac ChIP-seq. c, Boxplots of tissue-specific mRNA expression, partitioned by genes with or without an associated super-enhancer. Center, median; box limits, first and third percentiles. d, Tox mRNA expression in HCV tetramer populations before and after DAA therapy. Mean ± s.d., two-sided Student’s t-test with Welch’s correction; *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001. N = 6 donors.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yates, K.B., Tonnerre, P., Martin, G.E. et al. Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans. Nat Immunol 22, 1020–1029 (2021). https://doi.org/10.1038/s41590-021-00979-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00979-1

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