Abstract
Tissue-resident memory CD8+ T (CD8+ TRM) cells are localized within peripheral tissues, such as the liver, poised to provide effective immunosurveillance, as well as rapid and enhanced effector functions upon stimulation. Here we review how hepatic CD8+ TRM cells decipher a myriad of environmental signals, ranging from cellular and soluble factors to direct interactions with the underlying stroma and structural tissue niche, which dictate their derivation, retention and function. We discuss insights from both mouse and human studies that have contributed to our understanding of how CD8+ TRM cells can, depending on the context, provide targeted antigen-specific antiviral and antitumour immune responses and elicit antigen-independent tissue-damaging responses that contribute to liver pathology. Specifically, we discuss how the CD8+ TRM cell functional response is shaped by multiple factors and how such environmental cues tip the balance between these dual ‘Jekyll and Hyde’ response modes. Finally, we examine strategies to better identify and characterize hepatic CD8⁺ TRM cells and how the enhanced functionality of CD8+ TRM cells can be harnessed therapeutically in the context of hepatocellular carcinoma.
Key points
-
A population of long-lived hepatic CD8+ T cells interpret, integrates and responds to various environmental signals, ranging from metabolic and soluble mediators to direct cell–cell or cell–extracellular matrix interactions.
-
Localized tissue-resident memory CD8+ T (CD8+ TRM) cells can elicit both immunoprotective and immunopathogenic effector functions within the liver.
-
The functional response mode of hepatic CD8+ TRM cells is shaped and/or regulated by context-specific cues.
-
The enhanced retention within tumours and rapid effector function of CD8+ TRM cells could be harnessed for therapeutic intervention, such as chimeric antigen receptor T cell therapy for hepatocellular carcinoma.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$189.00 per year
only $15.75 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout




Similar content being viewed by others
References
Sender, R. et al. The total mass, number, and distribution of immune cells in the human body. Proc. Natl Acad. Sci. USA 120, e2308511120 (2023).
Kumar, B. V., Connors, T. J. & Farber, D. L. Human T cell development, localization, and function throughout life. Immunity 48, 202–213 (2018).
Masopust, D. & Soerens, A. G. Tissue-resident T cells and other resident leukocytes. Annu. Rev. Immunol. 37, 521–546 (2019).
Mueller, S. N. & Mackay, L. K. Tissue-resident memory T cells: local specialists in immune defence. Nat. Rev. Immunol. 16, 79–89 (2016).
Szabo, P. A., Miron, M. & Farber, D. L. Location, location, location: tissue resident memory T cells in mice and humans. Sci. Immunol. https://doi.org/10.1126/sciimmunol.aas9673 (2019).
Domínguez Conde, C. et al. Cross-tissue immune cell analysis reveals tissue-specific features in humans. Science 376, eabl5197 (2022).
Reina-Campos, M. et al. Tissue-resident memory CD8 T cell diversity is spatiotemporally imprinted. Nature 639, 483–492 (2025).
Hombrink, P. et al. Programs for the persistence, vigilance and control of human CD8+ lung-resident memory T cells. Nat. Immunol. 17, 1467–1478 (2016).
Sasson, S. C., Gordon, C. L., Christo, S. N., Klenerman, P. & Mackay, L. K. Local heroes or villains: tissue-resident memory T cells in human health and disease. Cell. Mol. Immunol. 17, 113–122 (2020).
Maini, M. K. et al. The role of virus-specific CD8+ cells in liver damage and viral control during persistent hepatitis B virus infection. J. Exp. Med. 191, 1269–1280 (2000).
Thimme, R. et al. CD8+ T cells mediate viral clearance and disease pathogenesis during acute hepatitis B virus infection. J. Virol. 77, 68–76 (2003).
Pallett, L. J. et al. IL-2high tissue-resident T cells in the human liver: sentinels for hepatotropic infection. J. Exp. Med. 214, 1567–1580 (2017).
Stelma, F. et al. Human intrahepatic CD69+ CD8+ T cells have a tissue resident memory T cell phenotype with reduced cytolytic capacity. Sci. Rep. 7, 6172 (2017).
Gill, U. S. et al. Fine needle aspirates comprehensively sample intrahepatic immunity. Gut 68, 1493–1503 (2019).
Bénéchet, A. P. et al. Dynamics and genomic landscape of CD8+ T cells undergoing hepatic priming. Nature 574, 200–205 (2019).
Maini, M. K. & Burton, A. R. Restoring, releasing or replacing adaptive immunity in chronic hepatitis B. Nat. Rev. Gastroenterol. Hepatol. 16, 662–675 (2019).
Fernandez-Ruiz, D. et al. Liver-resident memory CD8+ T cells form a front-line defense against malaria liver-stage infection. Immunity 45, 889–902 (2016).
Valencia-Hernandez, A. M. et al. A natural peptide antigen within the plasmodium ribosomal protein RPL6 confers liver TRM cell-mediated immunity against malaria in mice. Cell Host Microbe 27, 950–962.e7 (2020).
Ganley, M. et al. mRNA vaccine against malaria tailored for liver-resident memory T cells. Nat. Immunol. 24, 1487–1498 (2023).
Holz, L. E. et al. CD8+ T cell activation leads to constitutive formation of liver tissue-resident memory T cells that seed a large and flexible niche in the liver. Cell Rep. 25, 68–79.e4 (2018).
Garnelo, M. et al. Interaction between tumour-infiltrating B cells and T cells controls the progression of hepatocellular carcinoma. Gut 66, 342–351 (2017).
Lim, C. J. et al. Multidimensional analyses reveal distinct immune microenvironment in hepatitis B virus-related hepatocellular carcinoma. Gut 68, 916–927 (2019).
Cheng, Y. et al. Non-terminally exhausted tumor-resident memory HBV-specific T cell responses correlate with relapse-free survival in hepatocellular carcinoma. Immunity 54, 1825–1840.e7 (2021).
Barsch, M. et al. T-cell exhaustion and residency dynamics inform clinical outcomes in hepatocellular carcinoma. J. Hepatol. 77, 397–409 (2022).
Koda, Y. et al. CD8+ tissue-resident memory T cells promote liver fibrosis resolution by inducing apoptosis of hepatic stellate cells. Nat. Commun. 12, 4474 (2021).
Pallett, L. J. et al. Longevity and replenishment of human liver-resident memory T cells and mononuclear phagocytes. J. Exp. Med. 217, e20200050 (2020).
Kim, J. et al. Innate-like cytotoxic function of bystander-activated CD8+ T cells is associated with liver injury in acute hepatitis A. Immunity 48, 161–173.e5 (2018).
Kefalakes, H. et al. Liver-resident bystander CD8+ T cells contribute to liver disease pathogenesis in chronic hepatitis D virus infection. Gastroenterology 161, 1567–1583.e9 (2021).
Huang, C.-H. et al. Innate-like bystander-activated CD38+ HLA-DR+ CD8+ T cells play a pathogenic role in patients with chronic hepatitis C. Hepatology 76, 803–818 (2022).
Koh, J.-Y. et al. Identification of a distinct NK-like hepatic T-cell population activated by NKG2C in a TCR-independent manner. J. Hepatol. 77, 1059–1070 (2022).
Nkongolo, S. et al. Longitudinal liver sampling in patients with chronic hepatitis B starting antiviral therapy reveals hepatotoxic CD8+ T cells. J. Clin. Invest. 133, e158903 (2023).
Chu, T. et al. Bystander-activated memory CD8 T cells control early pathogen load in an innate-like, NKG2D-dependent manner. Cell Rep. 3, 701–708 (2013).
Soudja, S. M. et al. Memory-T-cell-derived interferon-γ instructs potent innate cell activation for protective immunity. Immunity 40, 974–988 (2014).
Li, X. et al. Estimating the health impact of vaccination against ten pathogens in 98 low-income and middle-income countries from 2000 to 2030: a modelling study. Lancet 397, 398–408 (2021).
Shin, E.-C., Sung, P. S. & Park, S.-H. Immune responses and immunopathology in acute and chronic viral hepatitis. Nat. Rev. Immunol. 16, 509–523 (2016).
Heim, K. et al. Attenuated effector T cells are linked to control of chronic HBV infection. Nat. Immunol. 25, 1650–1662 (2024).
Dudek, M. et al. Auto-aggressive CXCR6+ CD8 T cells cause liver immune pathology in NASH. Nature 592, 444–449 (2021).
Burtis, A. E. C. et al. Ag-driven CD8+ T cell clonal expansion is a prominent feature of MASH in humans and mice. Hepatology 81, 591–608 (2025).
You, Z. et al. The clinical significance of hepatic CD69+ CD103+ CD8+ resident-memory T cells in autoimmune hepatitis. Hepatology 74, 847–863 (2021).
Li, Y. et al. Itaconate inhibits CD103+ T RM cells and alleviates hepatobiliary injury in mouse models of primary sclerosing cholangitis. Hepatology 79, 25–38 (2024).
Niehaus, C. et al. CXCR6+ CD69+ CD8+ T cells in the ascites are associated with disease severity in patients with liver cirrhosis. JHEP Rep. https://doi.org/10.1016/j.jhepr.2024.101074 (2024).
Yang, H. et al. Intrahepatic infiltration of activated CD8+ T cells and mononuclear phagocyte is associated with idiosyncratic drug-induced liver injury. Front. Immunol. 14, 1138112 (2023).
Pfister, D. et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 592, 450–456 (2021).
Hao, X. et al. HBsAg-specific CD8+ T cells as an indispensable trigger to induce murine hepatocellular carcinoma. Cell. Mol. Immunol. 18, 128–137 (2021).
Lacotte, S. et al. Anti-CD122 antibody restores specific CD8+ T cell response in nonalcoholic steatohepatitis and prevents hepatocellular carcinoma growth. Oncoimmunology 12, 2184991 (2023).
Teutsch, H. F. The modular microarchitecture of human liver. Hepatology 42, 317–325 (2005).
Racanelli, V. & Rehermann, B. The liver as an immunological organ. Hepatology 43, S54–S62 (2006).
Ben-Moshe, S. & Itzkovitz, S. Spatial heterogeneity in the mammalian liver. Nat. Rev. Gastroenterol. Hepatol. 16, 395–410 (2019).
Wei, Y. et al. Liver homeostasis is maintained by midlobular zone 2 hepatocytes. Science 371, eabb1625 (2021).
Wen, Y., Lambrecht, J., Ju, C. & Tacke, F. Hepatic macrophages in liver homeostasis and diseases—diversity, plasticity and therapeutic opportunities. Cell. Mol. Immunol. 18, 45–56 (2021).
Friedman, S. L. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol. Rev. 88, 125–172 (2008).
Knolle, P. A. & Wohlleber, D. Immunological functions of liver sinusoidal endothelial cells. Cell. Mol. Immunol. 13, 347–353 (2016).
Rathmell, J. C., Farkash, E. A., Gao, W. & Thompson, C. B. IL-7 enhances the survival and maintains the size of naive T cells1. J. Immunol. 167, 6869–6876 (2001).
Teague, T. K., Munn, L., Zygourakis, K. & McIntyre, B. W. Analysis of lymphocyte activation and proliferation by video microscopy and digital imaging. Cytometry 14, 772–782 (1993).
Raynor, J. L. & Chi, H. Nutrients: signal 4 in T cell immunity. J. Exp. Med. 221, e20221839 (2024).
Adu-Berchie, K. et al. Generation of functionally distinct T-cell populations by altering the viscoelasticity of their extracellular matrix. Nat. Biomed. Eng. https://doi.org/10.1038/s41551-023-01052-y (2023).
Zhang, J. et al. Osr2 functions as a biomechanical checkpoint to aggravate CD8+ T cell exhaustion in tumor. Cell 187, 3409–3426.e24 (2024).
Pallett, L. J. & Maini, M. K. Liver-resident memory T cells: life in lockdown. Semin. Immunopathol. 44, 813–825 (2022).
Gavil, N. V., Cheng, K. & Masopust, D. Resident memory T cells and cancer. Immunity 57, 1734–1751 (2024).
Scott, M. C. et al. Deep profiling deconstructs features associated with memory CD8+ T cell tissue residence. Immunity 58, 162–181.e10 (2025).
Rosshart, S. P. et al. Laboratory mice born to wild mice have natural microbiota and model human immune responses. Science 365, eaaw4361 (2019).
Beura, L. K. et al. Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature 532, 512–516 (2016).
Zhang, X., Sun, S., Hwang, I., Tough, D. F. & Sprent, J. Potent and selective stimulation of memory-phenotype CD8+ T cells in vivo by IL-15. Immunity 8, 591–599 (1998).
Ku, C. C., Murakami, M., Sakamoto, A., Kappler, J. & Marrack, P. Control of homeostasis of CD8+ memory T cells by opposing cytokines. Science 288, 675–678 (2000).
Sandau, M. M., Kohlmeier, J. E., Woodland, D. L. & Jameson, S. C. IL-15 regulates both quantitative and qualitative features of the memory CD8 T cell pool. J. Immunol.184, 35–44 (2010).
van der Windt, G. J. W. et al. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 36, 68–78 (2012).
O’Sullivan, D. et al. Memory CD8+ T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 41, 75–88 (2014).
Tieu, R. et al. Tissue-resident memory T cell maintenance during antigen persistence requires both cognate antigen and interleukin-15. Sci. Immunol. 8, eadd8454 (2023).
Swadling, L. et al. Human liver memory CD8+ T cells use autophagy for tissue residence. Cell Rep. 30, 687–698.e6 (2020).
Laidlaw, B. J. et al. CD4+ T cell help guides formation of CD103+ lung-resident memory CD8+ T cells during influenza viral infection. Immunity 41, 633–645 (2014).
Mackay, L. K. et al. T-box transcription factors combine with the cytokines TGF-β and IL-15 to control tissue-resident memory T cell fate. Immunity 43, 1101–1111 (2015).
Ihara, A., Koizumi, H., Hashizume, R. & Uchikoshi, T. Expression of epithelial cadherin and α- and β-catenins in nontumoral livers and hepatocellular carcinomas. Hepatology 23, 1441–1447 (1996).
Ks, T. et al. Molecular basis for leukocyte integrin αEβ7 adhesion to epithelial E-cadherin. J. Exp. Med. 191, 1555–1567 (2000).
Bromley, S. K. et al. CD49a regulates cutaneous resident memory CD8+ T cell persistence and response. Cell Rep. 32, 108085 (2020).
Cheuk, S. et al. CD49a expression defines tissue-resident CD8+ T cells poised for cytotoxic function in human skin. Immunity 46, 287–300 (2017).
Skon, C. N. et al. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat. Immunol. 14, 1285–1293 (2013).
Borges da Silva, H. et al. Sensing of ATP via the purinergic receptor P2RX7 promotes CD8+ Trm cell generation by enhancing their sensitivity to the cytokine TGF-β. Immunity 53, 158–171.e6 (2020).
Evrard, M. et al. Single-cell protein expression profiling resolves circulating and resident memory T cell diversity across tissues and infection contexts. Immunity 56, 1664–1680.e9 (2023).
Christo, S. N. et al. Discrete tissue microenvironments instruct diversity in resident memory T cell function and plasticity. Nat. Immunol. 22, 1140–1151 (2021).
McNamara, H. A. et al. Up-regulation of LFA-1 allows liver-resident memory T cells to patrol and remain in the hepatic sinusoids. Sci. Immunol. 2, eaaj1996 (2017).
Taber, A., Konecny, A., Oda, S. K., Scott-Browne, J. & Prlic, M. TGF-β broadly modifies rather than specifically suppresses reactivated memory CD8 T cells in a dose-dependent manner. Proc. Natl Acad. Sci. USA 120, e2313228120 (2023).
Hirai, T. et al. Competition for active TGFβ cytokine allows for selective retention of antigen-specific tissue-resident memory T cells in the epidermal niche. Immunity 54, 84–98.e5 (2021).
Massagué, J. & Sheppard, D. TGF-β signaling in health and disease. Cell 186, 4007–4037 (2023).
Mackay, L. K. et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 352, 459–463 (2016).
Kim, J. H. et al. Functions of human liver CD69+ CD103− CD8+ T cells depend on HIF-2α activity in healthy and pathologic livers. J. Hepatol. 72, 1170–1181 (2020).
Doi, Y. et al. Development of complementary expression patterns of E- and N-cadherin in the mouse liver. Hepatol. Res. 37, 230–237 (2007).
Obers, A. et al. Retinoic acid and TGF-β orchestrate organ-specific programs of tissue residency. Immunity https://doi.org/10.1016/j.immuni.2024.09.015 (2024).
Saeed, A., Dullaart, R. P. F., Schreuder, T. C. M. A., Blokzijl, H. & Faber, K. N. Disturbed vitamin A metabolism in non-alcoholic fatty liver disease (NAFLD). Nutrients 10, 29 (2017).
Czuba, L. C. et al. Altered vitamin A metabolism in human liver slices corresponds to fibrogenesis. Clin. Transl. Sci. 14, 976–989 (2021).
Zhang, X. et al. GARP on hepatic stellate cells is essential for the development of liver fibrosis. J. Hepatol. 79, 1214–1225 (2023).
Yokota, S. et al. IRF-1 promotes liver transplant ischemia/reperfusion injury via hepatocyte IL-15/IL-15Rα production. J. Immunol. 194, 6045–6056 (2015).
Li, M. O. & Flavell, R. A. Contextual regulation of inflammation: a duet by transforming growth factor-β and interleukin-10. Immunity 28, 468–476 (2008).
Jabri, B. & Abadie, V. IL-15 functions as a danger signal to regulate tissue-resident T cells and tissue destruction. Nat. Rev. Immunol. 15, 771–783 (2015).
Ficht, X. & Iannacone, M. Immune surveillance of the liver by T cells. Sci. Immunol. 5, eaba2351 (2020).
Bosch, M. et al. A liver immune rheostat regulates CD8 T cell immunity in chronic HBV infection. Nature https://doi.org/10.1038/s41586-024-07630-7 (2024).
Heesch, K. et al. The function of the chemokine receptor CXCR6 in the T cell response of mice against Listeria monocytogenes. PLoS ONE 9, e97701 (2014).
Remmerie, A. et al. Osteopontin expression identifies a subset of recruited macrophages distinct from Kupffer cells in the fatty liver. Immunity 53, 641–657.e14 (2020).
Zigmond, E. et al. Infiltrating monocyte-derived macrophages and resident Kupffer cells display different ontogeny and functions in acute liver injury. J. Immunol. 193, 344–353 (2014).
Guilliams, M. & Scott, C. L. Liver macrophages in health and disease. Immunity 55, 1515–1529 (2022).
Borst, K. et al. Type I interferon receptor signaling delays Kupffer cell replenishment during acute fulminant viral hepatitis. J. Hepatol. 68, 682–690 (2018).
De Simone, G. et al. Identification of a Kupffer cell subset capable of reverting the T cell dysfunction induced by hepatocellular priming. Immunity 54, 2089–2100.e8 (2021).
Yu, M.-C. et al. Inhibition of T-cell responses by hepatic stellate cells via B7-H1-mediated T-cell apoptosis in mice. Hepatology 40, 1312–1321 (2004).
Mühlbauer, M. et al. PD-L1 is induced in hepatocytes by viral infection and by interferon-α and -γ and mediates T cell apoptosis. J. Hepatol. 45, 520–528 (2006).
Schölzel, K. et al. Transfer of MHC-class-I molecules among liver sinusoidal cells facilitates hepatic immune surveillance. J. Hepatol. 61, 600–608 (2014).
Pallett, L. J. et al. Tissue CD14+ CD8+ T cells reprogrammed by myeloid cells and modulated by LPS. Nature 614, 334–342 (2023).
Legut, M. et al. A genome-scale screen for synthetic drivers of T cell proliferation. Nature 603, 728–735 (2022).
Tilg, H., Adolph, T. E. & Trauner, M. Gut–liver axis: pathophysiological concepts and clinical implications. Cell Metab. 34, 1700–1718 (2022).
Beura, L. K. et al. Intravital mucosal imaging of CD8+ resident memory T cells shows tissue-autonomous recall responses that amplify secondary memory. Nat. Immunol. 19, 173–182 (2018).
Cockburn, I. A. et al. In vivo imaging of CD8+ T cell-mediated elimination of malaria liver stages. Proc. Natl Acad. Sci. USA 110, 9090–9095 (2013).
Guidotti, L. G. et al. Immunosurveillance of the liver by intravascular effector CD8+ T cells. Cell 161, 486–500 (2015).
DeLeve, L. D. Liver sinusoidal endothelial cells in hepatic fibrosis. Hepatology 61, 1740–1746 (2015).
Horn, T., Christoffersen, P. & Henriksen, J. H. Alcoholic liver injury: defenestration in noncirrhotic livers—a scanning electron microscopic study. Hepatology 7, 77–82 (1987).
Alon, R. & Dustin, M. L. Force as a facilitator of integrin conformational changes during leukocyte arrest on blood vessels and antigen-presenting cells. Immunity 26, 17–27 (2007).
Liu, C. S. C. et al. Piezo1 mechanosensing regulates integrin-dependent chemotactic migration in human T cells. eLife 12, RP91903 (2024).
Reilly, E. C. et al. TRM integrins CD103 and CD49a differentially support adherence and motility after resolution of influenza virus infection. Proc. Natl Acad. Sci. USA 117, 12306–12314 (2020).
Richter, M. V. & Topham, D. J. The α1β1 integrin and TNF receptor II protect airway CD8+ effector T cells from apoptosis during influenza infection. J. Immunol. 179, 5054–5063 (2007).
Boyd, D. F. & Thomas, P. G. Towards integrating extracellular matrix and immunological pathways. Cytokine 98, 79–86 (2017).
Higashi, T., Friedman, S. L. & Hoshida, Y. Hepatic stellate cells as key target in liver fibrosis. Adv. Drug Deliv. Rev. 121, 27–42 (2017).
Giles, J. R., Globig, A.-M., Kaech, S. M. & Wherry, E. J. CD8+ T cells in the cancer-immunity cycle. Immunity 56, 2231–2253 (2023).
Schurich, A. et al. Distinct metabolic requirements of exhausted and functional virus-specific CD8 T cells in the same host. Cell Rep. 16, 1243–1252 (2016).
Frizzell, H. et al. Organ-specific isoform selection of fatty acid-binding proteins in tissue-resident lymphocytes. Sci. Immunol. 5, eaay9283 (2020).
Sinclair, L. V. et al. Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat. Immunol. 14, 500–508 (2013).
Pallett, L. J. et al. Metabolic regulation of hepatitis B immunopathology by myeloid-derived suppressor cells. Nat. Med. 21, 591–600 (2015).
Sinclair, L. V., Neyens, D., Ramsay, G., Taylor, P. M. & Cantrell, D. A. Single cell analysis of kynurenine and system L amino acid transport in T cells. Nat. Commun. 9, 1981 (2018).
Cornish, G. H., Sinclair, L. V. & Cantrell, D. A. Differential regulation of T-cell growth by IL-2 and IL-15. Blood 108, 600–608 (2006).
Sinclair, L. V. et al. Autophagy repression by antigen and cytokines shapes mitochondrial, migration and effector machinery in CD8 T cells. Nat. Immunol. 26, 429–443 (2025).
Kabeya, Y. et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 19, 5720–5728 (2000).
Protzer, U., Maini, M. K. & Knolle, P. A. Living in the liver: hepatic infections. Nat. Rev. Immunol. 12, 201–213 (2012).
Baumann, T. et al. Regulatory myeloid cells paralyze T cells through cell–cell transfer of the metabolite methylglyoxal. Nat. Immunol. 21, 555–566 (2020).
Li, X. et al. Prostaglandin E2 facilitates hepatitis B virus replication by impairing CTL function. Mol. Immunol. 103, 243–250 (2018).
Pan, Y. et al. Survival of tissue-resident memory T cells requires exogenous lipid uptake and metabolism. Nature 543, 252–256 (2017).
Manzo, T. et al. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J. Exp. Med. 217, e20191920 (2020).
Xu, S. et al. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+ T cells in tumors. Immunity 54, 1561–1577.e7 (2021).
Varanasi, S. K. et al. Bile acid synthesis impedes tumor-specific T cell responses during liver cancer. Science 387, 192–201 (2025).
Zimmer, C. L. et al. A biliary immune landscape map of primary sclerosing cholangitis reveals a dominant network of neutrophils and tissue-resident T cells. Sci. Transl. Med. 13, eabb3107 (2021).
Zhu, H.-X. et al. Targeting pathogenic CD8+ tissue-resident T cells with chimeric antigen receptor therapy in murine autoimmune cholangitis. Nat. Commun. 15, 2936 (2024).
Antonioli, L., Pacher, P., Vizi, E. S. & Haskó, G. CD39 and CD73 in immunity and inflammation. Trends Mol. Med. 19, 355–367 (2013).
Stark, R. et al. TRM maintenance is regulated by tissue damage via P2RX7. Sci. Immunol. 3, eaau1022 (2018).
Cai, J., Hu, M., Chen, Z. & Ling, Z. The roles and mechanisms of hypoxia in liver fibrosis. J. Transl. Med. 19, 186 (2021).
Doedens, A. L. et al. Hypoxia-inducible factors enhance the effector responses of CD8+ T cells to persistent antigen. Nat. Immunol. 14, 1173–1182 (2013).
Palazon, A., Goldrath, A. W., Nizet, V. & Johnson, R. S. HIF transcription factors, inflammation, and immunity. Immunity 41, 518–528 (2014).
Hasan, F., Chiu, Y., Shaw, R. M., Wang, J. & Yee, C. Hypoxia acts as an environmental cue for the human tissue-resident memory T cell differentiation program. JCI Insight 6, e138970 (2021).
Smiriglia, A. et al. Sex difference in liver diseases: how preclinical models help to dissect the sex-related mechanisms sustaining NAFLD and hepatocellular carcinoma. iScience 26, 108363 (2023).
Schwinge, D. & Schramm, C. Sex-related factors in autoimmune liver diseases. Semin. Immunopathol. 41, 165–175 (2019).
Brown, R., Goulder, P. & Matthews, P. C. Sexual dimorphism in chronic hepatitis B virus (HBV) infection: evidence to inform elimination efforts. Wellcome Open. Res. 7, 32 (2022).
Klein, S. L. & Flanagan, K. L. Sex differences in immune responses. Nat. Rev. Immunol. 16, 626–638 (2016).
Layug, P. J., Vats, H., Kannan, K. & Arsenio, J. Sex differences in CD8+ T cell responses during adaptive immunity. WIREs Mech. Dis. 16, e1645 (2024).
Yuan, B. et al. Estrogen receptor β signaling in CD8+ T cells boosts T cell receptor activation and antitumor immunity through a phosphotyrosine switch. J. Immunother. Cancer 9, e001932 (2021).
Guan, X. et al. Androgen receptor activity in T cells limits checkpoint blockade efficacy. Nature 606, 791–796 (2022).
Yee Mon, K. J. et al. Differential sensitivity to IL-12 drives sex-specific differences in the CD8+ T cell response to infection. ImmunoHorizons 3, 121–132 (2019).
Sayaf, K. et al. Sex drives functional changes in the progression and regression of liver fibrosis. Int. J. Mol. Sci. 24, 16452 (2023).
Rosser, E. C., de Gruijter, N. M. & Matei, D. E. Mini-review: gut-microbiota and the sex-bias in autoimmunity—lessons learnt from animal models. Front. Med. 9, 910561 (2022).
Guan, D. et al. The hepatocyte clock and feeding control chronophysiology of multiple liver cell types. Science 369, 1388–1394 (2020).
Wang, C., Lutes, L. K., Barnoud, C. & Scheiermann, C. The circadian immune system. Sci. Immunol. 7, eabm2465 (2022).
Nobis, C. C. et al. The circadian clock of CD8 T cells modulates their early response to vaccination and the rhythmicity of related signaling pathways. Proc. Natl Acad. Sci. USA 116, 20077–20086 (2019).
Neufeld-Cohen, A. et al. Circadian control of oscillations in mitochondrial rate-limiting enzymes and nutrient utilization by PERIOD proteins. Proc. Natl Acad. Sci. USA 113, E1673–E1682 (2016).
Tahara, Y. & Shibata, S. Circadian rhythms of liver physiology and disease: experimental and clinical evidence. Nat. Rev. Gastroenterol. Hepatol. 13, 217–226 (2016).
Mukherji, A., Bailey, S. M., Staels, B. & Baumert, T. F. The circadian clock and liver function in health and disease. J. Hepatol. 71, 200–211 (2019).
Snyder, M. E. et al. Generation and persistence of human tissue-resident memory T cells in lung transplantation. Sci. Immunol. 4, eaav5581 (2019).
FitzPatrick, M. E. B. et al. Human intestinal tissue-resident memory T cells comprise transcriptionally and functionally distinct subsets. Cell Rep. 34, 108661 (2021).
Jung, I.-Y. et al. Tissue-resident memory CAR T cells with stem-like characteristics display enhanced efficacy against solid and liquid tumors. Cell Rep. Med. 4, 101053 (2023).
Steffin, D. et al. Interleukin-15-armoured GPC3 CAR T cells for patients with solid cancers. Nature 637, 940–946 (2025).
D, A. et al. IL15 enhances CAR-T cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype. Cancer Immunol. Res. 7, 759–772 (2019).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05103631 (2025).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04377932 (2025).
Steinert, E. M. et al. Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell 161, 737–749 (2015).
Guilliams, M. et al. Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches. Cell 185, 379–396.e38 (2022).
Salié, H. et al. Spatial single-cell profiling and neighbourhood analysis reveal the determinants of immune architecture connected to checkpoint inhibitor therapy outcome in hepatocellular carcinoma. Gut https://doi.org/10.1136/gutjnl-2024-332837 (2025).
Olsen, T. M., Stone, B. C., Chuenchob, V. & Murphy, S. C. Prime-and-trap malaria vaccination to generate protective CD8+ liver-resident memory T cells. J. Immunol. 201, 1984–1993 (2018).
Holz, L. E. et al. Glycolipid-peptide vaccination induces liver-resident memory CD8+ T cells that protect against rodent malaria. Sci. Immunol. 5, eaaz8035 (2020).
Noé, A. et al. Deep immune phenotyping and single-cell transcriptomics allow identification of circulating TRM-like cells which correlate with liver-stage immunity and vaccine-induced protection from malaria. Front. Immunol. 13, 795463 (2022).
Acknowledgements
The authors thank M. Maini, P. Knolle, A. Zhuang, A. Das, D. Wohlleber and B. Höchst for their critical review and discussion of the manuscript before submission.
Author information
Authors and Affiliations
Contributions
All authors researched data for the article. L.J.P. and M.D. contributed substantially to discussion of the content. All authors wrote the article. L.J.P. and M.D. reviewed and/or edited the manuscript before submission.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Gastroenterology & Hepatology thanks Patrick Bertolino, Francesco Andreata and Matthew Burchill for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Brown Romero, D., Finney, G.E., Dudek, M. et al. Tissue-resident memory CD8+ T cells: master deciphers of the hepatic environment. Nat Rev Gastroenterol Hepatol (2025). https://doi.org/10.1038/s41575-025-01118-z
Accepted:
Published:
DOI: https://doi.org/10.1038/s41575-025-01118-z