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:

Dynamics and genomic landscape of CD8+ T cells undergoing hepatic priming

Abstract

The responses of CD8+ T cells to hepatotropic viruses such as hepatitis B range from dysfunction to differentiation into effector cells, but the mechanisms that underlie these distinct outcomes remain poorly understood. Here we show that priming by Kupffer cells, which are not natural targets of hepatitis B, leads to differentiation of CD8+ T cells into effector cells that form dense, extravascular clusters of immotile cells scattered throughout the liver. By contrast, priming by hepatocytes, which are natural targets of hepatitis B, leads to local activation and proliferation of CD8+ T cells but not to differentiation into effector cells; these cells form loose, intravascular clusters of motile cells that coalesce around portal tracts. Transcriptomic and chromatin accessibility analyses reveal unique features of these dysfunctional CD8+ T cells, with limited overlap with those of exhausted or tolerant T cells; accordingly, CD8+ T cells primed by hepatocytes cannot be rescued by treatment with anti-PD-L1, but instead respond to IL-2. These findings suggest immunotherapeutic strategies against chronic hepatitis B infection.

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: Spatiotemporal dynamics of naive CD8+ T cells undergoing intrahepatic priming.
Fig. 2: Transcriptomic and chromatin accessibility analyses of CD8+ T cells undergoing intrahepatic priming.
Fig. 3: Intrahepatically primed, dysfunctional CD8+ T cells can be rescued by IL-2, but not by anti-PD-L1 antibodies.
Fig. 4: Therapeutic potential of IL-2 treatment for restoration of T cells during chronic HBV infection.

Similar content being viewed by others

Data and availability

The RNA-seq and ATAC-seq data on sorted hepatic CD8+ T cells have been deposited in the ArrayExpress database under the accession codes E-MTAB-7462 and E-MTAB-7461, respectively. All other data are available in the main text or the supplementary materials.

References

  1. Sironi, L. et al. In vivo flow mapping in complex vessel networks by single image correlation. Sci. Rep. 4, 7341 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Warren, A. et al. T lymphocytes interact with hepatocytes through fenestrations in murine liver sinusoidal endothelial cells. Hepatology 44, 1182–1190 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Guidotti, L. G. et al. Immunosurveillance of the liver by intravascular effector CD8+ T cells. Cell 161, 486–500 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Wong, Y. C., Tay, S. S., McCaughan, G. W., Bowen, D. G. & Bertolino, P. Immune outcomes in the liver: is CD8 T cell fate determined by the environment? J. Hepatol. 63, 1005–1014 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Holz, L. E. et al. Naive CD8 T cell activation by liver bone marrow-derived cells leads to a “neglected” IL-2low Bimhigh phenotype, poor CTL function and cell death. J. Hepatol. 57, 830–836 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. Guidotti, L. G. & Chisari, F. V. Immunobiology and pathogenesis of viral hepatitis. Annu. Rev. Pathol. 1, 23–61 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Wieland, S. F. & Chisari, F. V. Stealth and cunning: hepatitis B and hepatitis C viruses. J. Virol. 79, 9369–9380 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kennedy, P. T. F., Litwin, S., Dolman, G. E., Bertoletti, A. & Mason, W. S. Immune tolerant chronic hepatitis B: the unrecognized risks. Viruses 9, 96 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  9. European Association for the Study of the Liver. EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. J. Hepatol. 67, 370–398 (2017).

    Article  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. Kennedy, P. T. F. et al. Preserved T-cell function in children and young adults with immune-tolerant chronic hepatitis B. Gastroenterology 143, 637–645 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Isogawa, M., Chung, J., Murata, Y., Kakimi, K. & Chisari, F. V. CD40 activation rescues antiviral CD8+ T cells from PD-1-mediated exhaustion. PLoS Pathog. 9, e1003490 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Guidotti, L. G., Matzke, B., Schaller, H. & Chisari, F. V. High-level hepatitis B virus replication in transgenic mice. J. Virol. 69, 6158–6169 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bertoletti, A. & Ferrari, C. Adaptive immunity in HBV infection. J. Hepatol. 64 (Suppl.), S71–S83 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Guidotti, L. G., Martinez, V., Loh, Y. T., Rogler, C. E. & Chisari, F. V. Hepatitis B virus nucleocapsid particles do not cross the hepatocyte nuclear membrane in transgenic mice. J. Virol. 68, 5469–5475 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Flatz, L. et al. Development of replication-defective lymphocytic choriomeningitis virus vectors for the induction of potent CD8+ T cell immunity. Nat. Med. 16, 339–345 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Guidotti, L. G. et al. Viral clearance without destruction of infected cells during acute HBV infection. Science 284, 825–829 (1999).

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Ishak, K. et al. Histological grading and staging of chronic hepatitis. J. Hepatol. 22, 696–699 (1995).

    Article  CAS  PubMed  Google Scholar 

  19. Klein, I. & Crispe, I. N. Complete differentiation of CD8+ T cells activated locally within the transplanted liver. J. Exp. Med. 203, 437–447 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Böttcher, J. P. et al. IL-6 trans-signaling-dependent rapid development of cytotoxic CD8+ T cell function. Cell Reports 8, 1318–1327 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Sitia, G. et al. Kupffer cells hasten resolution of liver immunopathology in mouse models of viral hepatitis. PLoS Pathog. 7, e1002061 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 

  23. Best, J. A. et al. Transcriptional insights into the CD8+ T cell response to infection and memory T cell formation. Nat. Immunol. 14, 404–412 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Dominguez, C. X. et al. The transcription factors ZEB2 and T-bet cooperate to program cytotoxic T cell terminal differentiation in response to LCMV viral infection. J. Exp. Med. 212, 2041–2056 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Intlekofer, A. M. et al. Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin. Nat. Immunol. 6, 1236–1244 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Cruz-Guilloty, F. et al. Runx3 and T-box proteins cooperate to establish the transcriptional program of effector CTLs. J. Exp. Med. 206, 51–59 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kurachi, M. et al. The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8+ T cells. Nat. Immunol. 15, 373–383 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Chen, J. et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 567, 530–534 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liu, X. et al. Genome-wide analysis identifies NR4A1 as a key mediator of T cell dysfunction. Nature 567, 525–529 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Scott-Browne, J. P. et al. Dynamic changes in chromatin accessibility occur in CD8+ T cells responding to viral infection. Immunity 45, 1327–1340 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Schietinger, A., Delrow, J. J., Basom, R. S., Blattman, J. N. & Greenberg, P. D. Rescued tolerant CD8 T cells are preprogrammed to reestablish the tolerant state. Science 335, 723–727 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sharpe, A. H. & Pauken, K. E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 18, 153–167 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Spolski, R., Li, P. & Leonard, W. J. Biology and regulation of IL-2: from molecular mechanisms to human therapy. Nat. Rev. Immunol. 18, 648–659 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Manske, K. et al. Outcome of anti-viral immunity in the liver is shaped by the level of antigen expressed in infected hepatocytes. Hepatology 68, 2089–2105 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Tolksdorf, F. et al. The PDL1-inducible GTPase Arl4d controls T effector function by limiting IL-2 production. Sci. Rep. 8, 16123 (2018).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  37. Boyman, O., Kovar, M., Rubinstein, M. P., Surh, C. D. & Sprent, J. Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science 311, 1924–1927 (2006).

    Article  ADS  CAS  PubMed  Google Scholar 

  38. Kamimura, D. & Bevan, M. J. Naive CD8+ T cells differentiate into protective memory-like cells after IL-2 anti IL-2 complex treatment in vivo. J. Exp. Med. 204, 1803–1812 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Brown, B. D., Venneri, M. A., Zingale, A., Sergi Sergi, L. & Naldini, L. Endogenous microRNA regulation suppresses transgene expression in hematopoietic lineages and enables stable gene transfer. Nat. Med. 12, 585–591 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Supek, F., Bošnjak, M., Škunca, N. & Šmuc, T. REVIGO summarizes and visualizes long lists of gene ontology terms. PLoS ONE 6, e21800 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Cantore, A. et al. Liver-directed lentiviral gene therapy in a dog model of hemophilia B. Sci. Transl. Med. 7, 277ra28–277ra28 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mátrai, J. et al. Hepatocyte-targeted expression by integrase-defective lentiviral vectors induces antigen-specific tolerance in mice with low genotoxic risk. Hepatology 53, 1696–1707 (2011).

    Article  PubMed  CAS  Google Scholar 

  46. Reeves, J. P., Reeves, P. A. & Chin, L. T. Survival surgery: removal of the spleen or thymus. Curr. Protoc. Immunol. Chapter 1, Unit 1.10 (2001).

  47. Iannacone, M. et al. Platelets mediate cytotoxic T lymphocyte-induced liver damage. Nat. Med. 11, 1167–1169 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Tonti, E. et al. Bisphosphonates target B cells to enhance humoral immune responses. Cell Reports 5, 323–330 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Li, P.-Z., Li, J.-Z., Li, M., Gong, J.-P. & He, K. An efficient method to isolate and culture mouse Kupffer cells. Immunol. Lett. 158, 52–56 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protocols 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  52. Liao, Y., Smyth, G. K. & Shi, W. The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 41, e108–e108 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  54. Robinson, M. D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. 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 

  56. Bullard, J. H., Purdom, E., Hansen, K. D. & Dudoit, S. Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments. BMC Bioinformatics 11, 94 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–9 (2015).

    Article  Google Scholar 

  58. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  60. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Zhu, L. J. et al. ChIPpeakAnno: a Bioconductor package to annotate ChIP-seq and ChIP-chip data. BMC Bioinformatics 11, 237 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Fioravanti, J. et al. Effector CD8+ T cell-derived interleukin-10 enhances acute liver immunopathology. J. Hepatol. 67, 543–548 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Zordan, P. et al. Tuberous sclerosis complex-associated CNS abnormalities depend on hyperactivation of mTORC1 and Akt. J. Clin. Invest. 128, 1688–1706 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Benechet, A. P., Ganzer, L. & Iannacone, M. Intravital microscopy analysis of hepatic T cell dynamics. Methods Mol. Biol. 1514, 49–61 (2017).

    Article  CAS  PubMed  Google Scholar 

  66. Lindh, M., Gonzalez, J. E., Norkrans, G. & Horal, P. Genotyping of hepatitis B virus by restriction pattern analysis of a pre-S amplicon. J. Virol. Methods 72, 163–174 (1998).

    Article  CAS  PubMed  Google Scholar 

  67. Tan, A. T. et al. Host ethnicity and virus genotype shape the hepatitis B virus-specific T-cell repertoire. J. Virol. 82, 10986–10997 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Barbier, L. et al. Two lymph nodes draining the mouse liver are the preferential site of DC migration and T cell activation. J. Hepatol. 57, 352–358 (2012).

    Article  CAS  PubMed  Google Scholar 

  69. Thierry, G. R. et al. The conduit system exports locally secreted IgM from lymph nodes. J. Exp. Med. 245, jem.20180344 (2018).

    Google Scholar 

Download references

Acknowledgements

We thank F. V. Chisari for critical comments and suggestions, and for providing transgenic mouse lineages 1.3.32, MUP-core 50, Cor93 and Env28 TCR that were produced in his laboratory at The Scripps Research Institute in La Jolla; A. Fiocchi, M. Freschi, M. Mainetti, M. Raso and G. Sitia for technical support; M. Silva for secretarial assistance; E. Lugli, A. Mondino, L. Pace, R. Pardi and S. Trifari for critical reading of the manuscript and the members of the Iannacone laboratory for discussions. Confocal immunofluorescence histology was carried out at Alembic, an advanced microscopy laboratory established by the San Raffaele Scientific Institute and the Vita-Salute San Raffaele University. Flow cytometry was carried out at FRACTAL, a flow cytometry resource and advanced cytometry technical applications laboratory established by the San Raffaele Scientific Institute. We would like to acknowledge the PhD program in Basic and Applied Immunology and Oncology at Vita-Salute San Raffaele University, as G.D.S., F.C., V.F. and V.B. conducted this study as partial fulfilment of their PhD in Molecular Medicine within that program. M.I. is supported by European Research Council (ERC) Consolidator Grant 725038, Italian Association for Cancer Research (AIRC) grant 19891 and 22737, Italian Ministry of Health (MoH) grant RF-2018-12365801, Lombardy Foundation for Biomedical Research (FRRB) grant 2015-0010, the European Molecular Biology Organization Young Investigator Program, and a Career Development from the Giovanni Armenise-Harvard Foundation; A.P.B. is the recipient of EMBO Long-Term Fellowship ALTF 694-2016; F.M. is the recipient of Marie Curie Intra-European Fellowship (IEF) for Career Development SEP-210371319; F.A. is the recipient of a Fondazione Umberto Veronesi postdoctoral fellowship; M.K. is supported by the Italian Ministry of Education grant SIR-RBSI14BAO5; L.G.G. is supported by the Italian MoH grant RF-2013-02355209 and the Lombardy Open Innovation grant 229452; R.O. is supported by ERC Starting Grant 759532, Italian Telethon Foundation SR-Tiget Grant Award F04, Italian MoH grant GR-2016-02362156, AIRC MFAG 20247, Cariplo Foundation Grant 2015-0990 and the EU Infect-ERA 126.

Author information

Authors and Affiliations

Authors

Contributions

A.P.B. and G.D.S. designed and performed experiments, analysed data, performed the statistical analyses, prepared the figures and edited the paper; P.D.L., F.M., P.Z., V.F., E.B., L.G., M.K., F.A. performed experiments and analysed data; F.C. generated RNA-seq and ATAC-seq data with help from V.B.; G.B. and E.L. analysed RNA-seq and ATAC-seq data; R.O. supervised F.C., G.B., E.L. and V.B. and prepared figures; N.L.B., K.K., P.T.F.K. and A.B. performed experiments on HBV-infected patients, analysed data, prepared the figures and edited the manuscript; C.B. and F.G. helped with experiments involving Kupffer cells; A.C. and L.N. generated the lentiviral vectors encoding IL-2; G.G.-A. generated recombinant adeno-associated viruses; W.V.B. and D.D.P. generated rLCMV vectors; M.K., R.O. and L.G.G. provided funding, conceptual advice and edited the manuscript; M.I. designed and coordinated the study, provided funding, analysed the data, and wrote the paper.

Corresponding author

Correspondence to Matteo Iannacone.

Ethics declarations

Competing interests

M.I., L.G.G., R.O., A.C. and L.N. are inventors on patents filed, owned and managed by Telethon Foundation and San Raffaele Scientific Institute on LV technology related to the work presented in this manuscript (UK patent application 1907493.9).

Additional information

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

Peer review information Nature thanks Barbara Rehermann and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Fig. 1 Naive CD8+ T cells that recognize hepatocellular antigen are activated locally and expand, but do not develop effector function.

a, Schematic of the experimental setup. Five million Env28 TN cells were transferred into C57BL/6 × BALB/c F1 (WT) or HBV replication-competent transgenic (HBV Tg, C57BL/6 × BALB/c F1) recipients. Livers were collected and analysed 5 days after Env28 TN cell transfer and sera from the same mice were collected daily from day 0 to 5 after transfer. b, c, Absolute numbers of total (b) and IFNγ-producing (c) Env28 T cells in the livers of the indicated mice. d, ALT levels detected in the sera of the indicated mice at the indicated time points. n = 4. e, Schematic of the experimental setup. Five million Cor93 TN cells were transferred into C57BL/6 (WT) or MUP-core recipients. Mice were splenectomized and treated with anti-CD62L antibody 48 h or 4 h before cell transfer, respectively. Untreated wild-type mice that received 5 × 106 Cor93 TN cells were used as controls. Where indicated, mice were injected with 2.5 × 105 infectious units of non-replicating rLCMV-core 4 h before Cor93 TN cell transfer. Liver-draining lymph nodes68 (dLN) and non-draining inguinal lymph nodes (ndLN) were collected at 4 h and 1 day after transfer. f, Representative flow cytometry plots 4 h after Cor93 TN cells transfer. Numbers indicate the percentage of cells within the indicated gate. g, h, Quantification of the absolute numbers of cells recovered from the ndLN (g) and dLN (h) of the indicated mice 4 h and 1 day (d1) after Cor93 TN cell transfer. n = 3. i, Confocal immunofluorescence micrographs of liver sections from wild-type mice, wild-type mice transduced with rLCMV-core, MUP-core mice, and R26-ZsGreen mice injected with 2.5 × 105 infectious units of non-replicating rLCMV-cre. Scale bars, 100 μm. Note that, because HBV core protein did not accumulate at detectable levels in Kupffer cells and hepatic dendritic cells after rLCMV-core injection, we confirmed the tropism of this vector by injecting rLCMV-cre into R26-ZsGreen mice, which express the fluorescent protein ZsGreen after Cre-mediated recombination. j, MFI of CD69 expression on Cor93 T cells in the liver, blood, lung and bone marrow of the indicated mice 4 h after Cor93 TN cell transfer. n = 4. Data are mean ± s.e.m. and representative of at least three independent experiments. ***P < 0.001, two-tailed t-test (b, c) or one-way ANOVA with Bonferroni post-test (g, h, j). Mouse drawings were adapted from ref. 69.

Source data

Extended Data Fig. 2 Spatiotemporal dynamics of naive CD8+ T cells after intrahepatic priming.

Five million fluorescent Cor93 TN cells were transferred into MUP-core mice or wild-type mice transduced with rLCMV-core. Mice were splenectomized and treated with anti-CD62L antibody 48 h or 4 h before Cor93 TN transfer cell, respectively. a, Left, confocal immunofluorescence micrographs of liver sections from the indicated mice at the indicated time points after Cor93 TN cell transfer, showing the distribution of Cor93 T cells (green) relative to portal tracts (highlighted by anti-cytokeratin 7 (CK-7)-antibody-mediated staining of bile ducts in red). Sinusoids are highlighted by anti-LYVE-1+ antibodies (white). Scale bars, 100 μm. Right, immunohistochemical micrographs of liver sections from the indicated mice at the indicated time points after Cor93 TN cell transfer, showing the distribution of leukocyte infiltrates relative to portal tracts (highlighted by anti-CK-7-antibody-mediated staining of bile ducts in brown). Scale bars, 100 μm. b, Distribution of the distances of each Cor93 T cell from the centre of the closest portal triad at the indicated time points. n = 3 mice. Data are representative of at least three independent experiments.

Source data

Extended Data Fig. 3 Kupffer cells, but not dendritic cells, promote CD8+ T cell effector differentiation after rLCMV injection.

a, Schematic of the experimental setup. Five million Cor93 TN cells were transferred into C57BL/6 (WT) recipients. Mice were splenectomized and treated with anti-CD62L antibodies 48 h or 4 h before cell transfer, and injected with 2.5 × 105 infectious units of non-replicating rLCMV-core 4 h before Cor93 TN cell transfer. Where indicated, mice were treated with clodronate liposomes (CLL) 48 h before Cor93 TN cell transfer. b, Confocal microscopy of liver sections from control mice (top) and clodronate liposome-treated mice (bottom) Kupffer cells are depicted in red in all panels, and sinusoids are depicted in grey only in the left panels. Scale bars, 100 μm. c, Absolute numbers of CD11c+MHC-IIhigh dendritic cells (DCs) in the livers of the indicated mice. d, e, Absolute numbers of total (d) and of IFNγ-producing (e) Cor93 T cells in the livers of the indicated mice 5 days after Cor93 TN cell transfer. n = 4 mice (control) and 3 mice (CLL). f, Confocal immunofluorescence micrographs of liver sections from the indicated mice 5 days after Cor93 TN cell transfer. Scale bars, 100 μm. g, Schematic of the experimental setup. Wild-type mice were lethally irradiated and reconstituted with CD11c-DTR bone marrow (BM). One million Cor93 TN cells were transferred into recipients. Mice were injected with 2.5 × 105 infectious units of non-replicating rLCMV-core 4 h before Cor93 TN cell transfer. Indicated mice were treated with 400 ng of diphtheria toxin (DT) 3 days before, 1 day before and 1 day after T cell transfer. Livers were collected and analysed 5 days after Cor93 TN cell transfer. h, Representative flow cytometry plots in the liver of control (left) or diphtheria-toxin-treated (right) mice. i, CD11c+MHC-II+ dendritic cells (expressed as percentage of the total intrahepatic leukocyte population, IHL) in the livers of the indicated mice. n = 3. j, k, Absolute numbers of total (j) and IFNγ-producing (k) Cor93 T cells in the livers of the indicated mice 5 days after Cor93 TN cell transfer. n = 3 (control and WT + rLCMV-core), 4 (control + DT) and 5 (WT + rLCMV-core + DT). Data are mean ± s.e.m. and representative of three independent experiments. **P < 0.01, ***P < 0.001, two-tailed t-test (d, e, i) or one-way ANOVA with Bonferroni post-test (ik). Mouse drawings were adapted from ref. 69.

Source data

Extended Data Fig. 4 A strong reduction in the levels of hepatocellular core antigen expression is per se not sufficient to induce effector differentiation.

a, Schematic of the experimental setup. One million Cor93 TN cells were transferred into C57BL/6 (WT) or MUP-core recipients. Indicated wild-type mice were injected with 3 × 1010 viral genomes of AAV-core 15 days before Cor93 TN cell transfer. Livers were collected and analysed 5 days after Cor93 TN cell transfer. b, Representative confocal immunofluorescence micrographs of a liver section from an AAV-core-injected mouse 15 days after virus injection. Transduced hepatocytes are depicted in green and nuclei in grey. Scale bar, 50 μm. n = 3 mice. ce, Absolute numbers of total (c) and IFNγ-producing (d) Cor93 T cells in the livers of the indicated mice 5 days after Cor93 TN cell transfer. e, ALT levels detected in the sera of the indicated mice. n = 3 (WT and MUP-core) and 5 (AAV-core). f, Schematic of the experimental setup. One million Cor93 TN cells were transferred into 8- or 4-week-old (wo) MUP-core mice. Livers were collected and analysed 5 days after Cor93 TN cell transfer. g, Expression of HBV core antigen (HBcAg) in the livers of the indicated mice was analysed by western blotting. h, Quantification of the western blot shown in g. Core expression, normalized to the housekeeping nuclear protein H3, is expressed as arbitrary units (A.U.). n = 1 (WT) and 3 (MUP-core 8wo and MUP-core 4wo). i, Immunohistochemical micrographs of liver sections from the indicated mice, showing core antigen expression (brown). Scale bars, 50 μm. CV, central vein; PV, portal vein. n = 3 mice. j, k, Absolute numbers of total (j) and of IFNγ-producing (k) Cor93 T cells in the livers of the indicated mice 5 days after Cor93 TN cell transfer. n = 4 mice. l, ALT levels detected in the sera of the indicated mice. n = 4. Data are mean ± s.e.m. and representative of two independent experiments. *P < 0.05, **P < 0.01, one-way ANOVA with Bonferroni post-test (ce) or two-tailed t-test (h, jl). Mouse drawings were adapted from ref. 69.

Source data

Extended Data Fig. 5 Genomic landscape of naive CD8+ T cells undergoing intrahepatic priming.

a, Box plots showing expression levels (log2(RPKM)) in the indicated experimental condition of genes belonging to the categories described in Fig. 2a. Box plots are as in Fig. 3f. Naive (n = 2), WT + rLCMV-core (n = 3), MUP-core (day1 and 3, n = 2; day 7, n = 3). b, Box plots showing ATAC-seq signal intensity (log2(CPM)) in the indicated experimental condition of peaks belonging to the categories described in Fig. 2c. Box plots are as in Fig. 3f. Naive (n = 2), WT + rLCMV-core (day 1 and 7, n = 2; day 3, n = 3), MUP-core (day 1 and 3, n = 2; day 7, n = 3). P values in a and b were determined by two-sided Mann–Whitney U-test. c, Bar plot showing the number of inducible ATAC-seq peaks (logFCCPM > 2.5, FDR < 0.001 versus Cor93 TN) in the indicated conditions. ATAC-seq peaks with higher intensity signal in Cor93 T cells from WT + rLCMV-core (logFCCPM > 1.5, FDR < 0.1) or from MUP-core mice (logFCCPM < −1.5, FDR < 0.1) are shown in blue and red, respectively. Differences in peak signal intensities were evaluated fitting a negative binomial generalized linear model on the dataset and then performing a quasi-likelihood F-test. The Benjamini–Hochberg procedure was applied to correct for multiple tests. Naive (n = 2), WT + rLCMV-core (day 1 and 7, n = 2; day 3, n = 3), MUP-core (day 1 and 3, n = 2; day 7, n = 3).

Extended Data Fig. 6 GO analysis of intrahepatically primed CD8+ T cells.

Heat map showing the NES value associated to the seed GO categories (identified by REVIGO) found enriched in the indicated time points by GSEA. Colour legends indicate NES, with positive values (in blue) reflecting enrichment of GO categories in hepatic CD8+ T cells isolated from wild-type mice injected with rLCMV-core, and negative values (in red) reflecting enrichment of GO categories in hepatic CD8+ T cells isolated from MUP-core mice.

Extended Data Fig. 7 Although priming by hepatocytes initiates a unique dysfunctional program, hepatocellular antigen persistence may gradually trigger an additional exhaustion signature.

ad, Left, number of top 100 genes from Cor93 T cells recovered from the livers of wild-type mice transduced with rLCMV-core (a, c) or of MUP-core (b, d) mice reaching log2(RPKM) >1 in the indicated conditions in RNA-seq data from splenic LCMV-specific effector or exhausted CD8+ T cells30 (a, b) or splenic LCMV-specific exhausted CD8+ T cells31 (c, d). Right, box plots showing the expression levels of top 100 genes from Cor93 T cells recovered from livers of WT + rLCMV-core mice (a, c) or MUP-core mice (b, d) in the indicated conditions in RNA-seq data from splenic LCMV-specific effector or exhausted CD8+ T cells30 (a, b) or splenic LCMV-specific exhausted CD8+ T cells31 (c, d). Naive (n = 2), effector (n = 2), exhausted (n = 2). e, f, Left, number of top 100 genes in Cor93 T cells isolated from livers of wild-type + rLCMV-core mice (e) or from MUP-core mice (f) expressed (log2(normalized data) > 65th percentile of the full distribution) in the indicated conditions in microarray data from tolerant self-antigen-specific CD8+ T cells32. Right, box plots showing the expression levels of genes retrieved in the dataset among the top 100 genes in Cor93 T cells isolated from the livers of wild-type + rLCMV-core (e) or MUP-core (f) mice in the indicated conditions in microarray data from tolerant self-antigen-specific CD8+ T cells32. Only genes for which microarray probes were retrieved were kept for these analyses. Naive (n = 3), tolerant (n = 3). *P < 0.05, **P < 0.01, ***P < 0.001, two-tailed Wilcoxon rank-sum test. All box plots are represented as in Fig. 3f, and dots represent the expression distribution of the set of 100 genes. gi, Enrichment plot showing the results of a GSEAPreanked analysis (Kolmogorow–Smirnov statistics) performed on genes expressed in CD8+ T cells from wild-type + rLCMV-core or MUP-core mice (gene lists ranked by log(FCRPKM)) and using as gene set a curated list of genes induced in exhausted CD8+ T cells (n = 2) but not in effector CD8+ T cells (n = 2) as compared to naive cells (n = 2)30. NES and P values are reported for each time point.

Extended Data Fig. 8 IL-2c substantially rescues the transcriptional program of dysfunctional CD8+ T cells.

Heat map showing expression values (log2(RPKM)) of genes hypo-expressed (top) or hyper-expressed (bottom) in Cor93 CD8+ T cells from livers of MUP-core mice at day 5, which are rescued by treatment with IL-2c.

Extended Data Fig. 9 Therapeutic restoration of intrahepatically primed, dysfunctional CD8+ T cells by IL-2.

a, Schematic of the experimental setup. One million Cor93 TN cells were transferred into HBV Tg mice. Indicated HBV Tg mice received IL-2c treatment 1 day after CD8+ T cell transfer. Livers were collected and analysed 5 days after Cor93 TN cell transfer. Sera were collected before and 5 days after Cor93 TN cell transfer. b, Absolute numbers of IFNγ-producing Cor93 T cells in the livers of the indicated mice. n = 3 (control), 4 (IL-2c). c, ALT levels detected in the sera of the indicated mice. n = 3 (control), 4 (IL-2c). d, HBV DNA quantification (expressed as fold reduction over pre-treatment levels) in sera of the indicated mice before and 5 days after Cor93 TN cell transfer. n = 5. e, HBV DNA quantification by Southern blot analysis in the liver of the indicated mice. Bands corresponding to the expected size of the integrated transgene, relaxed circular (RC), double-stranded linear (DS), and single-stranded (SS) HBV DNAs are indicated. n = 5 mice. f, Representative immunohistochemical micrographs of liver sections from the indicated mice showing HBV core antigen expression (brown). Scale bars, 100 μm. n = 5 mice. Data are mean ± s.e.m. and representative of at least two independent experiments. *P < 0.05, ***P < 0.001, two-tailed t-test. Mouse drawings were adapted from ref. 69.

Source data

Extended Data Fig. 10 Summary of the main findings.

Top, priming by Kupffer cells—which are not natural targets of HBV—leads to differentiation into bona fide effector cells that form dense, extravascular clusters of rather immotile cells scattered throughout the liver. Middle, priming by hepatocytes—which are the natural targets of HBV—leads to local activation and proliferation but lack of differentiation into effector cells; these dysfunctional cells express a unique set of genes including some belonging to GO categories linked to tissue remodelling and they form loose, intravascular clusters of motile cells that coalesce around portal tracts. Bottom, CD8+ T cells primed by hepatocytes can be rescued by IL-2 treatment.

Supplementary information

Supplementary Figure

Gel source data

Reporting Summary

Supplementary Table 1 A list of datasets generated in this study and sequencing information

41586_2019_1620_MOESM4_ESM.xlsx

Supplementary Table 2 Differentially expressed genes from RNA-seq analyses on hepatic Cor93 CD8+ T cells before and after (1, 3 and 7 days) adoptive transfer into rLCMV-core-transduced WT mice or MUP-core mice. Differential gene expression was evaluated fitting a negative binomial generalized linear model on the dataset and then performing a quasi-likelihood (QL) F-test. The Benjamini-Hochberg procedure was applied in order to correct for multiple tests. Sample size: naïve (n = 2), WT+rLCMV-core (n = 3), MUP-core (day1 and day3, n = 2; day7, n = 3).

41586_2019_1620_MOESM5_ESM.xlsx

Supplementary Table 3 Differential ATAC-seq peaks of hepatic Cor93 CD8+ T cells before and after (1, 3 and 7 days) adoptive transfer into rLCMV-core-transduced WT mice or MUP-core mice. Differences in peak signal intensities were evaluated fitting a negative binomial generalized linear model on the dataset and then performing a quasi-likelihood (QL) F-test. The Benjamini-Hochberg procedure was applied in order to correct for multiple tests. Sample size: naïve (n = 2), WT+rLCMV-core (day1 and day7, n = 2; day3, n = 3), MUP-core (day1 and day3, n = 2; day7, n = 3).

41586_2019_1620_MOESM6_ESM.xlsx

Supplementary Table 4 Motif enrichment analyses on differential ATAC-seq peaks in hepatic Cor93 CD8+ T cells before and after (1, 3 and 7 days) adoptive transfer into rLCMV-core-transduced WT mice or MUP-core mice. Motif enrichment was calculated using cumulative binomial distributions. The Benjamini-Hochberg procedure was applied in order to correct for multiple tests. n = 2 (Naïve) or 3 (WT+rLCMV-core and MUP-core) biologically independent samples.

41586_2019_1620_MOESM7_ESM.xlsx

Supplementary Table 5 Differentially expressed genes from RNA-seq analyses on hepatic or splenic Cor93 CD8+ T cells before and after (1, 3 and 7 days) adoptive transfer into rLCMV-core-transduced WT mice. Cor93 CD8+ T cells from the liver of rLCMV-core-injected mice that were not splenectomized nor treated with anti-CD62L upregulated up to 82.8% of the genes induced in Cor93 CD8+ T cells isolated from the spleen of the same mice, including critical effector-related molecules such as Gzma, Gzmb, Gzmc, Gzmk, Il2, Ifng and many others. A negative binomial generalized linear model followed by quasi-likelihood (QL) F-test and Benjamini-Hochberg procedure for multiple tests correction was used for differential gene expression analysis. n = 3 biologically independent samples.

41586_2019_1620_MOESM8_ESM.xlsx

Supplementary Table 6 Enriched Gene Ontologies in RNA-seq data of hepatic Cor93 CD8+ T cells before and after (1, 3 and 7 days) adoptive transfer into rLCMV-core-transduced WT mice or MUP-core mice. n = 2 (Naïve) or 3 (WT+rLCMV-core and MUP-core) biologically independent samples.

41586_2019_1620_MOESM9_ESM.xlsx

Supplementary Table 7 Lists of 100 inducible genes (versus Cor93TN) with highest differential expression in hepatic Cor93 CD8+ T cells 7 days after adoptive transfer into rLCMV-core-transduced WT mice or MUP-core mice.

41586_2019_1620_MOESM10_ESM.xlsx

Supplementary Table 8 Expression levels of genes from Table S7 in datasets from LCMV-specific exhausted CD8+ T cells (refs.30,31) and from tolerant self-Ag-specific CD8+ T cells (ref.32). Numbers of expressed genes from Table S7 in datasets from LCMV-specific exhausted CD8+ T cells (refs.30,31) and from tolerant self-Ag-specific CD8+ T cells (ref.32).

41586_2019_1620_MOESM11_ESM.xlsx

Supplementary Table 9 Differentially expressed genes from RNA-seq analyses on Cor93 CD8+ T cells before and after (5 days) adoptive transfer into WT + rLCMV-core or MUP-core mice upon treatment with vehicle or IL-2c. A negative binomial generalized linear model followed by quasi-likelihood (QL) F-test and Benjamini-Hochberg procedure for multiple tests correction was used for differential gene expression analysis. Sample size: WT + rLCMV-core (n = 3); MUP-core (n = 3); MUP-core+IL-2c (n = 2).

41586_2019_1620_MOESM12_ESM.xlsx

Supplementary Table 10 Clinical and virological details of chronic HBV patients. Patients were categorized into EASL 2017 standard phases using the clinical and virological criteria outlined in the EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection9: i) HBeAg+ chronic infection (eAg+CInf): normal ALT (< 40 IU/L), HBeAg positive and high HBV DNA; ii) HBeAg+ chronic hepatitis (eAg+CHep): elevated ALT, HBeAg positive; iii) HBeAg- chronic hepatitis (eAg-CHep): elevated ALT, anti-HBe positive; and iv) HBeAg- chronic infection (eAg-CInf): normal ALT, anti-HBe positive, low HBV DNA. Patients were followed for at least 1 year with virological and clinical parameters collected every 6 months. The ALT and virological parameters shown in Supplementary Table 10 are the ones present at the time of PBMC isolation. Patients were classified as immunotolerant (IT) if they had HBeAg+ chronic infection (eAg+CInf); alternatively, they were classified as immune active (IA) if they showed signs of immunological activity, that is eAg+CHep or eAg-CHep.

41586_2019_1620_MOESM13_ESM.mov

Video 1 Multiphoton intravital microscopy in the livers of rLCMV-core-transduced WT or MUP-core mice 3 days after Cor93 TN transfer. Cor93 T cells (green) form dense, poorly perfusable clusters that are scattered throughout the liver and are composed of largely immotile cells in WT + rLCMV-core livers (left movies); by contrast Cor93 T cells (green) form looser, intravascular clusters that coalesce around portal tracts and are composed of more motile cells in MUP-core livers (right movies). Blood vasculature is visualized by the intravenous injection of nontargeted Quantum Dots 655 (white). Scale 50 μm (first movie set) or 20µm (second movie set); time in min:s. For four-dimensional analysis of cell migration, 14-15 z-stacks (spacing 4 µm) of 520 µm x 240 µm xy sections were acquired every 27 s for 40 min. Data are representative of at least 5 independent experiments.

41586_2019_1620_MOESM14_ESM.mov

Video 2 Multiphoton intravital microscopy in the livers of rLCMV-core-transduced WT or MUP-core mice 3 days after Cor93 TN transfer. Videos show tracks (yellow) of Cor93 T cells in the livers of rLCMV-core-transduced WT (left movies) or MUP-core mice (right movies). Blood vasculature is visualized by the intravenous injection of nontargeted Quantum Dots 655 (white). Scale 10 μm (first movie set) or 20 µm (second movie set); time in min:s. For four-dimensional analysis of cell migration, 12-17 z-stacks (spacing 4 µm) of 260 µm x 140 µm xy sections were acquired every 23 to 30 s for 38 min to 1 h.

41586_2019_1620_MOESM15_ESM.mov

Video 3 Multiphoton intravital microscopy in the livers of rLCMV-core-transduced WT or MUP-core mice 7 days after Cor93 TN transfer. Cor 93 T cell clusters (green) in WT mice transduced with rLCMV-core (left movies) start to disaggregate as cells move out from the liver into the peripheral blood; by contrast Cor 93 T cell clusters (green) in MUP-core mice (right movies) remain in place, possibly reflecting Ag persistence. Scale 40 μm (first movie set) or 20 µm (second movie set); time in min:s. For four-dimensional analysis of cell migration, 10-18 z-stacks (spacing 4 µm) of 520 µm x 200 µm xy sections were acquired every 18 to 32 s for 25 min.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bénéchet, A.P., De Simone, G., Di Lucia, P. et al. Dynamics and genomic landscape of CD8+ T cells undergoing hepatic priming. Nature 574, 200–205 (2019). https://doi.org/10.1038/s41586-019-1620-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1620-6

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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