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:

Memory-like HCV-specific CD8+ T cells retain a molecular scar after cure of chronic HCV infection

Abstract

In chronic hepatitis C virus (HCV) infection, exhausted HCV-specific CD8+ T cells comprise memory-like and terminally exhausted subsets. However, little is known about the molecular profile and fate of these two subsets after the elimination of chronic antigen stimulation by direct-acting antiviral (DAA) therapy. Here, we report a progenitor–progeny relationship between memory-like and terminally exhausted HCV-specific CD8+ T cells via an intermediate subset. Single-cell transcriptomics implicated that memory-like cells are maintained and terminally exhausted cells are lost after DAA-mediated cure, resulting in a memory polarization of the overall HCV-specific CD8+ T cell response. However, an exhausted core signature of memory-like CD8+ T cells was still detectable, including, to a smaller extent, in HCV-specific CD8+ T cells targeting variant epitopes. These results identify a molecular signature of T cell exhaustion that is maintained as a chronic scar in HCV-specific CD8+ T cells even after the cessation of chronic antigen stimulation.

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: Emergence of TML/HCV subsets is associated with antigen recognition.
Fig. 2: TML/HCV and TTE/HCV cells are transcriptionally distinct.
Fig. 3: Progenitor–progeny relationship of TML/HCV and TTE/HCV cells.
Fig. 4: The molecular scar of chronicity after HCV cure.
Fig. 5: TML/HCV cells retain the chronic scar after HCV cure.
Fig. 6: The chronic scar is determined by antigen recognition.

Similar content being viewed by others

Data availability

The primary read files and expression count files for the scRNA-seq datasets reported in this paper are available to download from the Gene Expression Omnibus (GEO) under accession number GSE150305. The expression count files for the low-input RNA sequencing are available to download from GEO under accession number GSE150345. The raw data files for the low-input RNA sequencing are available from the European Genome-phenome Archive under accession number EGAD00001006259. The gene sets used for GSEA were: (1) exhaustion (chronic TCF-1), memory-like (chronic TCF-1+) and memory signature (https://doi.org/10.1016/j.immuni.2016.07.021)19; (2) PD-1int CD8+ TILs signature (https://doi.org/10.1053/j.gastro.2018.08.030)29 and the TCF-1+ CD8+ TILs signature (https://doi.org/10.1016/j.immuni.2018.12.021)35. The remaining data supporting the findings of this study are available from the corresponding authors upon request.

References

  1. Blank, C. U. et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 19, 665–674 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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 

  3. Moskophidis, D., Lechner, F., Pircher, H. & Zinkernagel, R. M. Virus persistence in acutely infected immunocompetent mice by exhaustion of antiviral cytotoxic effector T cells. Nature 362, 758–761 (1993).

    Article  CAS  PubMed  Google Scholar 

  4. 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 

  5. 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 

  6. McLane, L. M., Abdel-Hakeem, M. S. & Wherry, E. J. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 37, 457–495 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. Bengsch, B. et al. Deep immune profiling by mass cytometry links human T and NK cell differentiation and cytotoxic molecule expression patterns. J. Immunol. Methods 453, 3–10 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Barili, V. et al. Targeting p53 and histone methyltransferases restores exhausted CD8+ T cells in HCV infection. Nat. Commun. 11, 604 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Fisicaro, P. et al. Targeting mitochondrial dysfunction can restore antiviral activity of exhausted HBV-specific CD8+ T cells in chronic hepatitis B. Nat. Med. 23, 327–336 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Kurktschiev, P. D. et al. Dysfunctional CD8+ T cells in hepatitis B and C are characterized by a lack of antigen-specific T-bet induction. J. Exp. Med. 211, 2047–2059 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. 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 

  14. 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 

  15. Yao, C. et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol. 20, 890–901 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. He, R. et al. Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection. Nature 537, 412–428 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Paley, M. A. et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Utzschneider, D. T. et al. T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Utzschneider, D. T. et al. T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat. Immunol. 14, 603–610 (2013).

    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. Aregay, A. et al. Elimination of hepatitis C virus has limited impact on the functional and mitochondrial impairment of HCV-specific CD8+ T cell responses. J. Hepatol. 71, 889–899 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. 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 

  24. Burchill, M. A., Golden-Mason, L., Wind-Rotolo, M. & Rosen, H. R. Memory re-differentiation and reduced lymphocyte activation in chronic HCV-infected patients receiving direct-acting antivirals. J. Viral Hepat. 22, 983–991 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Cox, A. L. et al. Comprehensive analyses of CD8+ T cell responses during longitudinal study of acute human hepatitis C. Hepatology 42, 104–112 (2005).

    Article  PubMed  Google Scholar 

  26. Golden-Mason, L. et al. Upregulation of PD-1 expression on circulating and intrahepatic hepatitis C virus-specific CD8+ T cells associated with reversible immune dysfunction. J. Virol. 81, 9249–9258 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Martin, B. et al. Restoration of HCV-specific CD8+ T cell function by interferon-free therapy. J. Hepatol. 61, 538–543 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. 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 

  29. Kim, H. D. et al. Association between expression level of PD-1 by tumor-infiltrating CD8+ T cells and features of hepatocellular carcinoma. Gastroenterology 155, 1936–1950 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. 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 

  31. Herman, J. S., Sagar & Grun, D. FateID infers cell fate bias in multipotent progenitors from single-cell RNA-seq data. Nat. Methods 15, 379–386 (2018).

    Article  CAS  PubMed  Google Scholar 

  32. Haghverdi, L., Buttner, M., Wolf, F. A., Buettner, F. & Theis, F. J. Diffusion pseudotime robustly reconstructs lineage branching. Nat. Methods 13, 845–848 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Man, K. et al. Transcription factor IRF4 promotes CD8+ T cell exhaustion and limits the development of memory-like T cells during chronic infection. Immunity 47, 1129–1141 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Philip, M. et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 545, 452–456 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Siddiqui, I. et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. 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 

  37. 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 

  38. Abdel-Hakeem, M. S. et al. Comparison of immune restoration in early versus late alpha interferon therapy against hepatitis C virus. J. Virol. 84, 10429–10435 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Angelosanto, J. M., Blackburn, S. D., Crawford, A. & Wherry, E. J. Progressive loss of memory T cell potential and commitment to exhaustion during chronic viral infection. J. Virol. 86, 8161–8170 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Nowicka, M. et al. CyTOF workflow: differential discovery in high-throughput high-dimensional cytometry datasets. F1000Res 6, 748 (2017).

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  42. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Tarasov, A., Vilella, A. J., Cuppen, E., Nijman, I. J. & Prins, P. Sambamba: fast processing of NGS alignment formats. Bioinformatics 31, 2032–2034 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. DeLuca, D. S. et al. RNA-SeQC: RNA-seq metrics for quality control and process optimization. Bioinformatics 28, 1530–1532 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Langfelder, P. & Horvath, S. Fast R functions for robust correlations and hierarchical clustering. J. Stat. Softw. 46, i11 (2012).

  48. Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. Hashimshony, T. et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-seq. Genome Biol. 17, 77 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Baker, S. C. et al. The external RNA controls consortium: a progress report. Nat. Methods 2, 731–734 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Grun, D., Kester, L. & van Oudenaarden, A. Validation of noise models for single-cell transcriptomics. Nat. Methods 11, 637–640 (2014).

    Article  PubMed  CAS  Google Scholar 

  53. Grun, D. et al. De novo prediction of stem cell identity using single-cell transcriptome data. Cell Stem Cell 19, 266–277 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Haghverdi, L., Lun, A. T. L., Morgan, M. D. & Marioni, J. C. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat. Biotechnol. 36, 421–427 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all participants in the current study and H. Pircher, I. Schulien, K. Heim, V. Oberhardt and T. Gross for critical reading of the manuscript. The work presented here was supported by the CRC/TRR 179-Project (no. 01 to R.T., no. 02 to C.N.-H., no. 04 to T.B., no. 09 to R.B., no. 20 to M.H., no. 21 to B.B. and no. Z2 to R.E. of the German Research Foundation (DFG; TRR179 project no. 272983813). M.H. was supported by a Margarete von Wrangell fellowship. D.G. was supported by the Max Planck Society, the Behrens-Weise-Foundation and the DFG (no. GR4980/3-1). D.A.P. was supported by a Wellcome Trust Senior Investigator Award (no. 100326/Z/12/Z).

Author information

Authors and Affiliations

Authors

Contributions

N.H. performed and analyzed experiments with the help of D.W., K.J., J.K. and O.S. S.L.-L., E.G. and D.A.P. provided pMHC-I tetramers. F.E. performed four-digit HLA typing by next-generation sequencing. Z.G. and N.I. analyzed and interpreted low-input RNA-seq data. C.C., R.E., B.B., T.B., C.N.-H. and R.B. contributed to data interpretation. Sagar and D.G. performed scRNA-seq and analyzed the respective data. M.H. and R.T. designed the study, contributed to experimental design and planning, interpreted data and wrote the manuscript.

Corresponding authors

Correspondence to Robert Thimme or Maike Hofmann.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information 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.

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

Extended data

Extended Data Fig. 1 Experimental setup.

Gating strategy of flow cytometric-based analyses and sorting of HCV-specific CD8+ T cells for transcriptome analyses using low-input or single-cell RNAseq. Dead cells, CD14+ (monocytes) and CD19+ (B cells) cells were excluded. Naïve CD8+ T cells (CD45RA+CCR7+) were also excluded. HLA-A*02:01- and HLA-B*27:05-restricted HCV-specific CD8+ T cells were identified by peptide/MHCI tetramers and further analyzed.

Extended Data Fig. 2 TML/HCV and TTE/HCV cells exhibit distinct characteristics.

a, Volcano plot showing DEGs comparing low-input transcriptome data of TML/HCV and TTE/HCV subsets (targeting conserved epitopes) derived from 3 cHCV-infected patients (red: exemplary DEGs are specified). x-axis represents the log2FC; y-axis represents the –log10 adjusted p values (≤ 0.05). Dotted lines indicate filter criteria of log2FC of ± 1 and adj. p value of 0.05. b–d, Flow cytometric analysis including various T-cell memory and exhaustion/effector markers of TML/HCV and TTE/HCV subsets derived from 12 cHCV patients. Representative dot plots including control analyses of non-naïve and naïve bulk CD8+ T cells (b,d), t-SNE representation (c) and bar charts (d, median fluorescence intensity (MFI) normalized to naïve CD8 + T cells) are depicted. e,f, Representative overview of the TCR clonotype distribution (low-input transcriptome data) within TML/HCV and TTE/HCV subsets from one (out of 3) cHCV patient depicted in three-layer donut plots: the inner layer depicts singleton, doubleton and high-order clonotypes; the second layer displays the top percentiles of clonotypes from the higher-order clonotypes and outer layer displays individual abundances of most recurrent clonotypes (e). Clonal overlap was assessed (f; n = 3). Bar charts show the median with IQR. Significance was assessed by Mann-Whitney comparison tests (two-sided) for flow cytometric analysis and by Kruskal-Wallis test (one-sided) including Dunn’s multiple comparisons test for TCR clonotypes analysis. DEG analysis of the volcano plot (two-sided) was done by DESeq2. All p values are corrected by the Benjamini-Hochberg method.

Extended Data Fig. 3 Characterization of HCV-specific CD127lo, CD127int and CD127hi subsets.

a, mRNA expression level of TCF7, BCL2, CCR7 and TOX (single-cell RNA sequencing data; n = 784 cells from 6 cHCV) in cluster 1–3 are depicted by violin plots. b, Flow cytometric analysis including various T cell memory and exhaustion/effector markers of CD127lo, CD127int and CD127hi (representing cluster 1–3) HCV-specific CD8+ T cells derived from 12 cHCV-infected patients. Representative dot plots are depicted including control analyses of non-naïve and naïve bulk CD8+ T cells. Summary graphs display the median fluorescence intensity (MFI) of marker expression of HCV-specific CD8+ T cell subsets normalized to naïve CD8+ T cells. Significance was assessed by Kruskal-Wallis test (one-sided) including Dunn’s multiple comparisons test for flow cytometric analysis and Wilcoxon matched-pairs signed rank test (two-sided) for mRNA expression levels. The violin plots show the frequency with box plots depicting the median with IQR.

Extended Data Fig. 4 Phenotypic characteristics of HCV-specific CD8+ T cells after HCV cure.

Flow cytometric analysis including various T cell memory and exhaustion/effector markers of HCV-specific CD8+ T cells before (n = 12) and after (n = 7) HCV cure and in rHCV (n = 5). a, t-SNE representation with scaled expression (color code: red: high; blue: low) is depicted. b, Representative dot plots (including control analyses of non-naïve and naïve bulk CD8+ T cells) and bar charts summarizing manually gated flow cytometric data (median fluorescence intensity (MFI) normalized to naïve CD8+ T cells) are depicted. c, Serum viral loads (HCV RNA), the frequency and cell counts [/ml] of TML/HCV subsets from patients before (n = 12) and after HCV cure (n = 7). Bar charts show the median with IQR. Significance was assessed by Kruskal-Wallis test (one-sided) including Dunn’s multiple comparisons test (b), Mann-Whitney comparison test (two-sided) (c; right) and Wilcoxon matched-pairs signed rank test (two-sided) (c, left).

Extended Data Fig. 5 Phenotypic characteristics of the TML/HCV subset.

Flow cytometric analysis including various T cell memory and exhaustion/effector markers of TML/HCV cells before (n = 12) and after (n = 7) HCV cure and in rHCV (n = 5). t-SNE representation with scaled expression depicted as color code (red: high; blue: low) a, representative dot plots (including control analyses of non-naïve and naïve bulk CD8+ T cells) and bar charts summarizing manually gated flow cytometric data (b; median fluorescence intensity (MFI) normalized to naïve CD8+ T cells) are depicted. Bar charts show the median with IQR. Significance was assessed by Kruskal-Wallis test (one-sided) including Dunn’s multiple comparisons test.

Extended Data Fig. 6 Characteristics of TML/HCV cells after HCV cure.

a, Flow cytometric analysis of TOX1, PD-1, Bcl-2 and CCR7 in TML/HCV subsets at the end of DAA therapy (EOT) (n = 6) and at long-term (>6 months) follow-up (FU) after HCV cure (n = 7) is depicted as bar chart (median fluorescence intensity (MFI) normalized to naïve CD8+ T cells). b, Principle component analysis (PCA) of the transcriptomes of TML/HCV subsets before and after HCV cure and in rHCV; and of TTE/HCV cells (cHCV) and TM cells (rHCV) (cHCV, cured HCV, rHCV: n = 3 each). (c) WGCNA of low input transcriptome data was performed. Numbers of genes in each defined module are depicted in the bar chart. Bar charts show the median with IQR. Significance was assessed by Mann-Whitney comparison test (two-sided).

Extended Data Fig. 7 Phenotypic characteristics of CD127hi and CD127int subsets before and after HCV cure.

Flow cytometric analysis including various T cell memory and exhaustion/effector markers of CD127int and CD127hi (representing cluster 1 and 2) HCV-specific CD8 + T cells from patients before (n = 12) and after (n = 7) HCV cure. Representative dot plots (including control analyses of non-naïve and naïve bulk CD8+ T cells), bar charts (a; median fluorescence intensity (MFI) normalized to naïve CD8+ T cells) and t-SNE representation b,c, are depicted. Scaled expression is depicted as color code (red: high; blue: low). Bar charts show the median with IQR. Significance was assessed by Kruskal-Wallis test (one-sided) including Dunn’s multiple comparisons test.

Extended Data Fig. 8 Phenotypic characteristics of HCV-specific CD8+ T cells targeting conserved versus variant epitopes.

Flow cytometric analysis including various T cell memory and exhaustion/effector markers of HCV-specific CD8+ T cells in rHCV (n = 5), cHCV and cured HCV targeting either conserved epitopes (AG; cHCV: n = 12, cured HCV: n = 7) or variant epitopes (ESC; cHCV: n = 8, cured HCV: n = 6). t-SNE representation with scaled expression depicted as color code (red: high; blue: low) a, representative dot plots (including control analyses of non-naïve and naïve bulk CD8+ T cells) and bar charts summarizing manually gated flow cytometric data (b; median fluorescence intensity (MFI) normalized to naïve CD8+ T cells) are depicted. Bar charts show the median with IQR. Significance was assessed by Kruskal-Wallis test (one-sided).

Extended Data Fig. 9 Phenotypic characteristics of TML/HCV cells targeting conserved versus variant epitopes.

Flow cytometric analysis including various T cell memory and exhaustion/effector markers of TML/HCV cells in rHCV (n = 5), cHCV and cured HCV targeting either conserved epitopes (AG; cHCV: n = 12, cured HCV: n = 7) or variant epitopes (ESC; cHCV: n = 8, cured HCV: n = 6). t-SNE representation with scaled expression depicted as color code (red: high; blue: low) a, representative dot plots (including control analyses of non-naïve and naïve bulk CD8+ T cells) and bar charts (b; median fluorescence intensity (MFI) normalized to naïve CD8+ T cells) are depicted. Bar charts show the median with IQR. Significance was assessed by Kruskal-Wallis test (one-sided) including Dunn’s multiple comparisons test.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hensel, N., Gu, Z., Sagar 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). https://doi.org/10.1038/s41590-020-00817-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-00817-w

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