Abstract
T cells, a cornerstone of the adaptive immune system, have pivotal roles at the host–microorganism interface. The gut microbiome profoundly influences T cell biology by producing a diverse repertoire of small molecules that are sensed by host cells. These microbial metabolites regulate all aspects of the T cell lifecycle, from cell development to differentiation and activation to exhaustion. Recent studies have uncovered microbially derived molecules, including short-chain fatty acids, secondary bile acids and tryptophan metabolites, as potent regulators of T cell function. However, the full scope of microbial metabolite–T cell interactions remains largely unexplored. This Review presents a mechanistic framework linking gut microbial metabolites to discrete stages of T cell fate and function. Expanding our understanding of these intricate host–microbiome interactions will reveal new aspects of immune regulation and inspire microbiome-guided therapeutic strategies for infections, autoimmune diseases and cancer immunotherapy.
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
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout



Similar content being viewed by others
References
Gilbert, J. A. et al. Current understanding of the human microbiome. Nat. Med. 24, 392–400 (2018).
Ivanov, I. I. et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4, 337–349 (2008).
Sefik, E. et al. Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells. Science 349, 993–997 (2015).
Ohnmacht, C. et al. The microbiota regulates type 2 immunity through RORγt+ T cells. Science 349, 989–993 (2015).
Bachem, A. et al. Microbiota-derived short-chain fatty acids promote the memory potential of antigen-activated CD8+ T cells. Immunity 51, 285–297 (2019).
Vétizou, M. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350, 1079–1084 (2015).
Ashby, K. M. & Hogquist, K. A. A guide to thymic selection of T cells. Nat. Rev. Immunol. 24, 103–117 (2024).
Owen, D. L., Sjaastad, L. E. & Farrar, M. A. Regulatory T cell development in the thymus. J. Immunol. 203, 2031–2041 (2019).
Legoux, F. et al. Microbial metabolites control the thymic development of mucosal-associated invariant T cells. Science 366, 494–499 (2019).
Zegarra-Ruiz, D. F. et al. Thymic development of gut-microbiota-specific T cells. Nature 594, 413–417 (2021).
Tsuchiya, M. Immunological abnormalities involving the thymus in ulcerative colitis and therapeutic effects of thymectomy. Gastroenterol. Jpn. 19, 232–246 (1984).
Mizuno, Y. et al. Inflammatory bowel diseases and thymus disorder: reactivity of thymocytes with monoclonal antibodies. Bull. Tokyo Dent. Coll. 31, 137–141 (1990).
Fredin, M. F. et al. Dextran sulfate sodium‐induced colitis generates a transient thymic involution — impact on thymocyte subsets. Scand. J. Immunol. 65, 421–429 (2007).
Sasaki, S., Ishida, Y., Nishio, N., Ito, S. & Isobe, K. Thymic involution correlates with severe ulcerative colitis induced by oral administration of dextran sulphate sodium in C57BL/6 mice but not in BALB/c mice. Inflammation 31, 319–328 (2008).
Bealmear, M. & Wilson, R. Histological comparison of the thymus of germfree (axenic) and conventional CFW mice. Anat. Rec. 154, 261–273 (1966).
Wilson, R., Bealmear, M. & Sobonya, R. Growth and regression of the germfree (axenic) thymus. Proc. Soc. Exp. Biol. Med. 118, 97–99 (1965).
van der Waaij, D. The influence of the intestinal microflora on the relative thymus weight. Med. Microbiol. Immunol. 175, 335–340 (1986).
Ennamorati, M. et al. Intestinal microbes influence development of thymic lymphocytes in early life. Proc. Natl Acad. Sci. USA 117, 2570–2578 (2020).
Nakajima, A. et al. Commensal bacteria regulate thymic Aire expression. PLoS ONE 9, e105904 (2014).
Nakajima, A. et al. Maternal high fiber diet during pregnancy and lactation influences regulatory T cell differentiation in offspring in mice. J. Immunol. 199, 3516–3524 (2017).
Treiner, E. et al. Selection of evolutionarily conserved mucosal-associated invariant T cells by MR1. Nature 422, 164–169 (2003).
Eckle, S. B. G. et al. Recognition of vitamin B precursors and byproducts by mucosal associated invariant T cells. J. Biol. Chem. 290, 30204–30211 (2015).
Besten, G. et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 54, 2325–2340 (2013).
Fredriksson, R., Lagerström, M. C., Lundin, L.-G. & Schiöth, H. B. The G-protein-coupled receptors in the human genome form five main families. phylogenetic analysis, paralogon groups, and fingerprints. Mol. Pharmacol. 63, 1256–1272 (2003).
Ehrlich, A. T. et al. Expression map of 78 brain-expressed mouse orphan GPCRs provides a translational resource for neuropsychiatric research. Commun. Biol. 1, 102 (2018).
Regard, J. B., Sato, I. T. & Coughlin, S. R. Anatomical profiling of G protein-coupled receptor expression. Cell 135, 561–571 (2008).
Colosimo, D. A. et al. Mapping interactions of microbial metabolites with human G-protein-coupled receptors. Cell Host Microbe 26, 273–282 (2019).
Millard, A. L. et al. Butyrate affects differentiation, maturation and function of human monocyte-derived dendritic cells and macrophages. Clin. Exp. Immunol. 130, 245–255 (2002).
Trompette, A. et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 20, 159–166 (2013).
Mariño, E. et al. Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes. Nat. Immunol. 18, 552–562 (2016).
Singh, N. et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 40, 128–139 (2014).
Ridlon, J. M. & Hylemon, P. B. Identification and characterization of two bile acid coenzyme A transferases from Clostridium scindens, a bile acid 7α-dehydroxylating intestinal bacterium. J. Lipid Res. 53, 66–76 (2012).
Hu, J. et al. Gut microbiota-mediated secondary bile acids regulate dendritic cells to attenuate autoimmune uveitis through TGR5 signaling. Cell Rep. 36, 109726 (2021).
Campbell, C. et al. Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature 581, 475–479 (2020).
Li, W. et al. A bacterial bile acid metabolite modulates Treg activity through the nuclear hormone receptor NR4A1. Cell Host Microbe 29, 1366–1377 (2021).
Paik, D. et al. Human gut bacteria produce ΤΗ17-modulating bile acid metabolites. Nature 603, 907–912 (2022).
Sinha, S. R. et al. Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe 27, 659–670 (2020).
Zhou, C., Wang, Y., Li, C., Xie, Z. & Dai, L. Amelioration of colitis by a gut bacterial consortium producing anti-inflammatory secondary bile acids. Microbiol. Spectr. 11, e03330-22 (2023).
Ghimire, S. et al. Indoxyl 3-sulfate inhibits maturation and activation of human monocyte-derived dendritic cells. Immunobiology 223, 239–245 (2018).
Hezaveh, K. et al. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 55, 324–340 (2022).
Shen, P.-X. et al. Urolithin A ameliorates experimental autoimmune encephalomyelitis by targeting aryl hydrocarbon receptor. eBioMedicine 64, 103227 (2021).
Lee, J. & Lee, J. Indole as an intercellular signal in microbial communities. FEMS Microbiol. Rev. 34, 426–444 (2010).
Selma, M. V., Tomás-Barberán, F. A., Beltrán, D., García-Villalba, R. & Espín, J. C. Gordonibacter urolithinfaciens sp. nov., a urolithin-producing bacterium isolated from the human gut. Int. J. Syst. Evol. Microbiol. 64, 2346–2352 (2014).
Selma, M. V. et al. Isolation of human intestinal bacteria capable of producing the bioactive metabolite isourolithin A from ellagic acid. Front. Microbiol. 08, 1521 (2017).
Pidgeon, R. et al. Diet-derived urolithin A is produced by a dehydroxylase encoded by human gut Enterocloster species. Nat. Commun. 16, 999 (2025).
Wu, K. et al. The gut microbial metabolite trimethylamine N-oxide aggravates GVHD by inducing M1 macrophage polarization in mice. Blood 136, 501–515 (2020).
Rath, S., Rud, T., Pieper, D. H. & Vital, M. Potential TMA-producing bacteria are ubiquitously found in Mammalia. Front. Microbiol. 10, 2966 (2020).
Craciun, S. & Balskus, E. P. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc. Natl Acad. Sci. USA 109, 21307–21312 (2012).
Mirji, G. et al. The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer. Sci. Immunol. 7, eabn0704 (2022).
Postow, M. A., Callahan, M. K. & Wolchok, J. D. Immune checkpoint blockade in cancer therapy. J. Clin. Oncol. 33, 1974–1982 (2015).
Wykes, M. N. & Lewin, S. R. Immune checkpoint blockade in infectious diseases. Nat. Rev. Immunol. 18, 91–104 (2017).
Dyck, L. & Mills, K. H. G. Immune checkpoints and their inhibition in cancer and infectious diseases. Eur. J. Immunol. 47, 765–779 (2017).
Buchbinder, E. I. & Desai, A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am. J. Clin. Oncol. 39, 98–106 (2016).
Steed, A. L. et al. The microbial metabolite desaminotyrosine protects from influenza through type I interferon. Science 357, 498–502 (2017).
Wang, Q. et al. Lactiplantibacillus pentoses CCFM1227 produces desaminotyrosine to protect against influenza virus H1N1 infection through the type I interferon in mice. Nutrients 15, 3659 (2023).
Zhou, J., Han, J., Wei, Y. & Wang, Y. Desaminotyrosine is a redox‐active microbial metabolite that bolsters macrophage antimicrobial functions while attenuating IL‐6 production. FASEB J. 38, e23844 (2024).
Joachim, L. et al. The microbial metabolite desaminotyrosine enhances T-cell priming and cancer immunotherapy with immune checkpoint inhibitors. eBioMedicine 97, 104834 (2023).
Atarashi, K. et al. ATP drives lamina propria TH17 cell differentiation. Nature 455, 808–812 (2008).
Zhang, Q. et al. Lactobacillus plantarum-derived indole-3-lactic acid ameliorates colorectal tumorigenesis via epigenetic regulation of CD8+ T cell immunity. Cell Metab. 35, 943–960 (2023).
Lee, R. et al. Short chain fatty acids facilitate protective immunity by macrophages and T cells during acute fowl adenovirus-4 infection. Sci. Rep. 13, 17999 (2023).
Inamoto, T. et al. Short‐chain fatty acids stimulate dendrite elongation in dendritic cells by inhibiting histone deacetylase. FEBS J. 290, 5794–5810 (2023).
Föh, B. et al. Microbial metabolite butyrate promotes induction of IL-10+IgM+ plasma cells. PLoS ONE 17, e0266071 (2022).
Hsu, P. et al. IL-10 potentiates differentiation of human induced regulatory T cells via STAT3 and Foxo1. J. Immunol. 195, 3665–3674 (2015).
Brooks, D. G., Walsh, K. B., Elsaesser, H. & Oldstone, M. B. A. IL-10 directly suppresses CD4 but not CD8 T cell effector and memory responses following acute viral infection. Proc. Natl Acad. Sci. USA 107, 3018–3023 (2010).
Martin-Gallausiaux, C. et al. Butyrate produced by gut commensal bacteria activates TGF-β1 expression through the transcription factor SP1 in human intestinal epithelial cells. Sci. Rep. 8, 9742 (2018).
Kim, M. H., Kang, S. G., Park, J. H., Yanagisawa, M. & Kim, C. H. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 145, 396–406 (2013).
Smith, P. M. et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573 (2013).
Mager, L. F. et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 369, 1481–1489 (2020).
Ohta, A. & Sitkovsky, M. Extracellular adenosine-mediated modulation of regulatory T cells. Front. Immunol. 5, 304 (2014).
Barrett, E., Ross, R. P., O’Toole, P. W., Fitzgerald, G. F. & Stanton, C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 113, 411–417 (2012).
Otaru, N. et al. GABA production by human intestinal Bacteroides spp.: prevalence, regulation, and role in acid stress tolerance. Front. Microbiol. 12, 656895 (2021).
Cohen, L. J. et al. Commensal bacteria make GPCR ligands that mimic human signalling molecules. Nature 549, 48–53 (2017).
Chen, H. et al. A forward chemical genetic screen reveals gut microbiota metabolites that modulate host physiology. Cell 177, 1217–1231 (2019).
Pols, T. W. H. et al. Lithocholic acid controls adaptive immune responses by inhibition of TH1 activation through the vitamin D receptor. PLoS ONE 12, e0176715 (2017).
Song, X. et al. Microbial bile acid metabolites modulate gut RORγ+ regulatory T cell homeostasis. Nature 577, 410–415 (2019).
Hang, S. et al. Bile acid metabolites control TH17 and Treg cell differentiation. Nature 576, 143–148 (2019).
Salsinha, A. S., Pimentel, L. L., Fontes, A. L., Gomes, A. M. & Rodríguez-Alcalá, L. M. Microbial production of conjugated linoleic acid and conjugated linolenic acid relies on a multienzymatic system. Microbiol. Mol. Biol. Rev. 82, https://doi.org/10.1128/mmbr.00019-18 (2018).
Song, X. et al. Gut microbial fatty acid isomerization modulates intraepithelial T cells. Nature 619, 837–843 (2023).
Cervantes-Barragan, L. et al. Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 357, 806–810 (2017).
Gutiérrez-Vázquez, C. & Quintana, F. J. Regulation of the immune response by the aryl hydrocarbon receptor. Immunity 48, 19–33 (2018).
Dean, J. W. et al. The aryl hydrocarbon receptor cell intrinsically promotes resident memory CD8+ T cell differentiation and function. Cell Rep. 42, 111963 (2023).
Zheng, Y. et al. Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature 463, 808–812 (2010).
Waldecker, M., Kautenburger, T., Daumann, H., Busch, C. & Schrenk, D. Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. J. Nutr. Biochem. 19, 587–593 (2008).
Park, J. et al. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR–S6K pathway. Mucosal Immunol. 8, 80–93 (2015).
Arpaia, N. et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504, 451–455 (2013).
Furusawa, Y. et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446–450 (2013).
He, Y. et al. Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8+ T cell immunity. Cell Metab. 33, 988–1000 (2021).
Luu, M. et al. Regulation of the effector function of CD8+ T cells by gut microbiota-derived metabolite butyrate. Sci. Rep. 8, 14430 (2018).
Sun, L., Fu, J. & Zhou, Y. Metabolism controls the balance of TH17/T-regulatory cells. Front. Immunol. 8, 1632 (2017).
Chang, C.-H. et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251 (2013).
Peng, M. et al. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science 354, 481–484 (2016).
Delgoffe, G. M. et al. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat. Immunol. 12, 295–303 (2011).
Sanidad, K. Z. et al. Gut bacteria-derived serotonin promotes immune tolerance in early life. Sci. Immunol. 9, eadj4775 (2024).
Matsumoto, M. et al. Impact of intestinal microbiota on intestinal luminal metabolome. Sci. Rep. 2, 233 (2012).
Nakamura, A. et al. Symbiotic polyamine metabolism regulates epithelial proliferation and macrophage differentiation in the colon. Nat. Commun. 12, 2105 (2021).
Carriche, G. M. et al. Regulating T-cell differentiation through the polyamine spermidine. J. Allergy Clin. Immunol. 147, 335–348 (2021).
Luu, M. et al. The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic–epigenetic crosstalk in lymphocytes. Nat. Commun. 10, 760 (2019).
Trompette, A. et al. Dietary fiber confers protection against flu by shaping Ly6c− patrolling monocyte hematopoiesis and CD8+ T cell metabolism. Immunity 48, 992–1005 (2018).
Qiu, J. et al. Acetate promotes T cell effector function during glucose restriction. Cell Rep. 27, 2063–2074 (2019).
Sun, M. et al. Microbiota-derived short-chain fatty acids promote Th1 cell IL-10 production to maintain intestinal homeostasis. Nat. Commun. 9, 3555 (2018).
Belikov, A. V., Schraven, B. & Simeoni, L. T cells and reactive oxygen species. J. Biomed. Sci. 22, 85 (2015).
Kamimura, D. & Bevan, M. J. Endoplasmic reticulum stress regulator XBP-1 contributes to effector CD8+ T cell differentiation during acute infection. J. Immunol. 181, 5433–5441 (2008).
Varanasi, S. K. et al. Bile acid synthesis impedes tumor-specific T cell responses during liver cancer. Science 387, 192–201 (2025).
Martin, M. D. & Badovinac, V. P. Defining memory CD8 T cell. Front. Immunol. 9, 2692 (2018).
Künzli, M. & Masopust, D. CD4+ T cell memory. Nat. Immunol. 24, 903–914 (2023).
Jiang, Y., Li, Y. & Zhu, B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 6, e1792 (2015).
Östman, S., Rask, C., Wold, A. E., Hultkrantz, S. & Telemo, E. Impaired regulatory T cell function in germ-free mice. Eur. J. Immunol. 36, 2336–2346 (2006).
van der Windt, G. J. W. & Pearce, E. L. Metabolic switching and fuel choice during T-cell differentiation and memory development. Immunol. Rev. 249, 27–42 (2012).
Balmer, M. L. et al. Memory CD8+ T cells require increased concentrations of acetate induced by stress for optimal function. Immunity 44, 1312–1324 (2016).
Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).
Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018).
Park, J. S. et al. Targeting PD-L2-RGMb overcomes microbiome-related immunotherapy resistance. Nature 617, 377–385 (2023).
Bender, M. J. et al. Dietary tryptophan metabolite released by intratumoral Lactobacillus reuteri facilitates immune checkpoint inhibitor treatment. Cell 186, 1846–1862 (2023).
Al-Habsi, M. et al. Spermidine activates mitochondrial trifunctional protein and improves antitumor immunity in mice. Science 378, eabj3510 (2022).
Nomura, M. et al. Association of short-chain fatty acids in the gut microbiome with clinical response to treatment with nivolumab or pembrolizumab in patients with solid cancer tumors. JAMA Netw. Open 3, e202895 (2020).
Jia, D. et al. Microbial metabolite enhances immunotherapy efficacy by modulating T cell stemness in pan-cancer. Cell 187, 1651–1665 (2024).
Coutzac, C. et al. Systemic short chain fatty acids limit antitumor effect of CTLA-4 blockade in hosts with cancer. Nat. Commun. 11, 2168 (2020).
Acknowledgements
This work was supported by National Institutes of Health grants R01 DK110559 (J.R.H. and A.S.D.), R35 GM128618 (A.S.D.) and a Harvard Medical School Van Maanen Fellowship (M.T.).
Author information
Authors and Affiliations
Contributions
M.T., J.R.H. and A.S.D. conceptualized the Review and wrote the manuscript.
Corresponding authors
Ethics declarations
Competing interests
A.S.D. is a consultant for Axial Therapeutics. J.R.H. is a consultant for CJ CheilJedang, hy and Interon Laboratories and an advisor on the Samsung Bio Advisory Board. M.T. declares no competing interests.
Peer review
Peer review information
Nature Immunology thanks Jochen Mattner and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Laurie A. Dempsey, in collaboration with the Nature Immunology team.
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
Tran, M., Huh, J.R. & Devlin, A.S. The role of gut microbial metabolites in the T cell lifecycle. Nat Immunol 26, 1246–1257 (2025). https://doi.org/10.1038/s41590-025-02227-2
Received:
Accepted:
Published:
Version of record:
Issue date:
DOI: https://doi.org/10.1038/s41590-025-02227-2


