Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway

J Park, M Kim, S G Kang, A H Jannasch, B Cooper, J Patterson, C H Kim, J Park, M Kim, S G Kang, A H Jannasch, B Cooper, J Patterson, C H Kim

Abstract

Microbial metabolites, such as short-chain fatty acids (SCFAs), are highly produced in the intestine and potentially regulate the immune system. We studied the function of SCFAs in the regulation of T-cell differentiation into effector and regulatory T cells. We report that SCFAs can directly promote T-cell differentiation into T cells producing interleukin-17 (IL-17), interferon-γ, and/or IL-10 depending on cytokine milieu. This effect of SCFAs on T cells is independent of GPR41 or GPR43, but dependent on direct histone deacetylase (HDAC) inhibitor activity. Inhibition of HDACs in T cells by SCFAs increased the acetylation of p70 S6 kinase and phosphorylation rS6, regulating the mTOR pathway required for generation of Th17 (T helper type 17), Th1, and IL-10(+) T cells. Acetate (C2) administration enhanced the induction of Th1 and Th17 cells during Citrobacter rodentium infection, but decreased anti-CD3-induced inflammation in an IL-10-dependent manner. Our results indicate that SCFAs promote T-cell differentiation into both effector and regulatory T cells to promote either immunity or immune tolerance depending on immunological milieu.

Figures

Fig. 1
Fig. 1
SCFAs promote the generation of Th17 and Th1 effector cells. (a)) Effects of C2 and C3 on naïve CD4+ T cell differentiation into Th17 and Th1 cells were determined. Naive CD4+ T cells, depleted of Treg (CD25+) and memory T (CD44+ CD69+) cells, were activated with anti-CD3/CD28 in a Th17 (IL-1β, IL-6, IL-21, IL-23, TGFβ1, anti-IL-4, and anti-IFN-γ) or Th1 (IL-12, IL-2, and anti-IL-4) condition for 5-6 days. C2 (0, 1, 10, and 20 mM) or C3 (0, 0.1, 0.5, and 1 mM) was added as indicated. (b) Expression patterns of T helper signature genes for Th1 and Th17 cells, generated in the presence of SCFAs, were determined by qRT-PCR. (c) Expression of FoxP3 by T-bet+ or RORγt+ T cells, generated in Th1 or Th17 condition in the presence of SCFAs. Cells were prepared in a Th17 or Th1 condition with C2 (10 mM) or C3 (1 mM). *Significant differences from blank groups (no treatment with C2 or C3) (P≤0.05).
Fig. 2
Fig. 2
The colitogenic activity of C2-treated T cells in Rag1(−/−) mice. (a) Cytokine profiles of cultured T cells in the presence or absence of C2 in a Th17 cell condition. Also shown is the weight change of Rag1(−/−) mice following injection of the cultured T cells. (b) Histological changes in the distal colon of Th17-transferred Rag1(−/−) mice were examined (×100 original magnification; scale bar = 200 μm), and histological scores indicating tissue inflammation are shown. (c) Frequencies of Th1, Th17, and FoxP3+ T cells in the colon of Th17-transferred Rag1(−/−) mice were determined by flow cytometry. Similarly, effector T cells induced in the absence or presence of C2 in a Th1 condition were assessed for their effects on (d) body weight change, (e) histological changes, and (f) frequencies of Th subsets. Representative or pooled data from 2-3 experiments are shown (n=5 for a; 10 for b; 9 for c; 6-7 for d; 4 for e; 6-7 for f). *Significant differences from the control group or between indicated groups (P≤0.05).
Fig. 3
Fig. 3
SCFAs induce IL-10-producing CD4+ or CD8+ T cells admixed with effector T cells. (a) The cytokine phenotype of CD4+ T cells cultured in Tnp, Th17, or Th1 condition with indicated SCFA was determined by flow cytometry. (b) Dose-dependent effects of C2 and C3 on the induction of IL-10+ CD4+ T cells. Conditioned media were examined for secreted IL-10 (c) and IL-10 mRNA (d). (e) The suppressive activity of control and C2-treated T cells on the proliferation of responder T cells was examined. Suppressor T cells were co-cultured with responder cells (CFSE-labeled CD4+CD25− T cells) in the presence of anti-CD3 and irradiated T-cell-depleted splenocytes as antigen presenting cells for 3 days before flow cytometric analysis. C2-treated suppressor T cells were prepared from the culture of naïve CD4+ T cells, isolated from WT or IL-10(−/−) mice, for 5-6 days in the presence of C2 (10 mM) in a Th1 cell-polarization condition. (f) SCFAs also induce expression of IL-10 in CD8+ T cells. Total CD8+ T cells were activated with anti-CD3/28 in a Tc1 (IL-2, IL-12, and anti-IL-4) or Tc17 (TGFβ1, IL-6, IL-1β, IL-23, IL-21, TNFα, anti-IL4, and anti-IFN-γ) polarization condition for 5-6 days. If not indicated, the concentrations of SCFAs were 10 mM (C2) or 1 mM (C3) for all experiments. Representative or pooled (b-f) data obtained from 3-4 experiments are shown. *Significant differences from blank groups or between indicated groups (P≤0.05).
Fig. 4
Fig. 4
Impact of infection on the SCFA effect on effector versus IL-10+ T cells. (a) The concentrations of SCFAs in cecal contents and intestine tissues of C2-fed mice were determined by LC-MS. (b and c) Some of the C2-fed mice were infected with C. rodentium. Changes in Th17, Th1, and IL-10+ T cells in indicated tissues 14 days after oral infection with C. rodentium were examined by flow cytometry. Pooled data (b; n=7-9) and representative dot plots (c) are shown. *Significant differences (P≤0.05).
Fig. 5
Fig. 5
C2 feeding suppressed anti-CD3 induced inflammation in the intestine. (a-c) Mice were fed with C2 for 6-8 weeks, and anti-CD3 antibody (clone 145-2C11) was injected i.p. to stimulate the production of IL-10+ T cells (n=6-9/group). Mice were sacrificed 52 h after the injection of anti-CD3, and the frequencies and numbers of IL-10+ (a), Th17 (b), and Th1 (c) cells were examined by flow cytometry (6-9/group). (d) The inflammatory response in the terminal ileum of the mice fed with C2 and injected with anti-CD3 was examined. Tissue sections were stained with hematoxylin and eosin (×100 original magnification; scale bar= 200 μm). Pooled histological scores of the ileum of control and C2-fed mice (n=6-9/group) obtained from two experiments are shown. *Significant differences from the control group (regular water) or between indicated groups (P≤0.05).
Fig. 6
Fig. 6
T cells poorly express GPR41 and GPR43, and GPR41−/− or GPR43−/− T cells normally respond to C2 or C3. (a and b) Cultured CD4+ T cells, CD326+ colonic epithelial cells, colonic lamina propria CD4+ or CD4− (colonic CD4-neg) cells, bone marrow Gr-1+ cells, and in vitro differentiated BM-DCs were compared for expression of GRP41 and GPR43 mRNA with qRT-PCR (A) and conventional PCR (B). (c and d)In vitro differentiation of GPR41−/− or GPR43−/− CD4+ T cells in response to C2 or C3. Naive CD4+ T cells were cultured in Th1 or Th17 cell condition for 5-6 days with C2 (10 mM) or C3 (1 mM). Frequencies of indicated T cell subsets were determined by flow cytometry. Representative (c) and pooled (d) data obtained from 3 experiments are shown. *Significant differences (P≤0.05) from control groups.
Fig. 7
Fig. 7
Normal production of effector and IL-10+ T cells in mice deficient in GPR41 or GPR43 with or without C2 feeding. (a) Mice were sacrificed at 6-9 weeks of age. Indicated T cell subsets in the spleen, MLN, and various compartments of the intestine were examined by flow cytometry. (b) Mice were fed with regular or C2 water from 3 weeks of age for 6-8 weeks. IFN-γ+, IL-17+, and IL-10+ CD4+ T cells in the colon were examined. Pooled data (n=7-10 for a; 5 for b) are shown. *Significant differences (P≤0.05) from control groups.
Fig. 8
Fig. 8
SCFAs suppress HDACs in a GPR41 or GPR43-independent manner. (a) In-cell HDAC inhibitor activity. T cells were activated for 48 h, pre-incubated with SCFAs or TSA for 2 h, and then assayed for HDAC activity. (b) Comparison of TSA and SCFAs in regulation of T cell differentiation into effector and IL-10+ T cells. Naïve CD4+ T cells were cultured for 5 days in the presence of indicated SCFAs or TSA. Pooled data from three independent experiments are shown. *Significant differences (P≤0.05) from control groups.
Fig. 9
Fig. 9
SCFAs enhance mTOR activity. (a) Activation of the mTOR pathway based on rS6 phosphorylation in CD4+ T cells. T cells were activated with anti-CD3/CD28 and IL-2 in the presence or absence of C2 or C3 and examined by flow cytometry. (b) Acetylation of S6K in activated T cells in the presence of C2, C3 or TSA. The antibodies used for immunoprecipitation (IP) and western blotting (WB) were anti-acetylated-lysine and anti-S6K respectively. (c) The effect of rapamycin on SCFA-dependent generation of Th cell subsets was assessed. Naïve CD4+ T cells were cultured in a Tnp, Th1, or Th17 polarization condition for 5-6 days in the presence of C2 (10 mM), C3 (1 mM), and/or rapamycin (25 nM), and the cytokine phenotype of cultured T cells was examined by flow cytometry and ELISA (for IL-10). (d) AMPK activation suppressed the SCFA effect on T cell differentiation. Naïve CD4+ T cells were cultured with SCFAs and/or metformin in indicated polarization conditions for 5-6 days, and frequencies of indicated T cell subsets were examined. The dot plots were from the data obtained in a Th1 condition. (e) Activation of STAT3 by C2 or C3 was examined. CD4+ T cells were activated for 3 days with anti-CD3/28 and then examined for phosphorylation of STAT3. Representative and pooled data obtained from at least three experiments are shown. *Significant differences from blank or control groups (P≤0.05).
Fig. 10
Fig. 10
Effects of C4 on rS6 phosphorylation (A) and T cell differentiation (B). Naive CD4+ T cells, depleted of Treg (CD25+) and memory T (CD44+ CD69+) cells, were activated with anti-CD3/CD28 for 3 days for rS6 phosphorylation and for 5-6 days in a Tnp, Th17, or Th1 condition for T cell differentiation at indicated concentrations of C4. Representative and pooled data obtained from at least three experiments are shown. *Significant differences from blank or control groups (P≤0.05).

References

    1. Lee YK, Mazmanian SK. Has the microbiota played a critical role in the evolution of the adaptive immune system? Science. 2010;330(6012):1768–1773.
    1. Kamada N, Nunez G. Role of the gut microbiota in the development and function of lymphoid cells. J Immunol. 2013;190(4):1389–1395.
    1. Keeney KM, Finlay BB. Enteric pathogen exploitation of the microbiota-generated nutrient environment of the gut. Curr Opin Microbiol. 2011;14(1):92–98.
    1. Flint HJ, Scott KP, Louis P, Duncan SH. The role of the gut microbiota in nutrition and health. Nat Rev Gastroenterol Hepatol. 2012;9(10):577–589.
    1. Macfarlane S, Macfarlane GT. Regulation of short-chain fatty acid production. The Proceedings of the Nutrition Society. 2003;62(1):67–72.
    1. Kamp F, Hamilton JA. How fatty acids of different chain length enter and leave cells by free diffusion. Prostaglandins, leukotrienes, and essential fatty acids. 2006;75(3):149–159.
    1. Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. 1987;28(10):1221–1227.
    1. Ziegler TR, Evans ME, Fernandez-Estivariz C, Jones DP. Trophic and cytoprotective nutrition for intestinal adaptation, mucosal repair, and barrier function. Annu Rev Nutr. 2003;23:229–261.
    1. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461(7268):1282–1286.
    1. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell Homeostasis. Science. 2013
    1. Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology. 2013;145(2):396–406. e391–310.
    1. Zuniga LA, Jain R, Haines C, Cua DJ. Th17 cell development: from the cradle to the grave. Immunological reviews. 2013;252(1):78–88.
    1. McAleer JP, Kolls JK. Mechanisms controlling Th17 cytokine expression and host defense. Journal of leukocyte biology. 2011;90(2):263–270.
    1. Symons A, Budelsky AL, Towne JE. Are Th17 cells in the gut pathogenic or protective? Mucosal immunology. 2012;5(1):4–6.
    1. Khader SA, Gaffen SL, Kolls JK. Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa. Mucosal immunology. 2009;2(5):403–411.
    1. Atarashi K, Honda K. Microbiota in autoimmunity and tolerance. Curr Opin Immunol. 2011;23(6):761–768.
    1. Ivanov II, Littman DR. Segmented filamentous bacteria take the stage. Mucosal immunology. 2010;3(3)
    1. Sartor RB. Key questions to guide a better understanding of host-commensal microbiota interactions in intestinal inflammation. Mucosal immunology. 2011;4(2):127–132.
    1. Vinolo MA, Ferguson GJ, Kulkarni S, Damoulakis G, Anderson K, Bohlooly YM, et al. SCFAs induce mouse neutrophil chemotaxis through the GPR43 receptor. PLoS One. 2011;6(6):e21205.
    1. Kamanaka M, Kim ST, Wan YY, Sutterwala FS, Lara-Tejero M, Galan JE, et al. Expression of interleukin-10 in intestinal lymphocytes detected by an interleukin-10 reporter knockin tiger mouse. Immunity. 2006;25(6):941–952.
    1. Merger M, Viney JL, Borojevic R, Steele-Norwood D, Zhou P, Clark DA, et al. Defining the roles of perforin, Fas/FasL, and tumour necrosis factor alpha in T cell induced mucosal damage in the mouse intestine. Gut. 2002;51(2):155–163.
    1. Tazoe H, Otomo Y, Kaji I, Tanaka R, Karaki SI, Kuwahara A. Roles of short-chain fatty acids receptors, GPR41 and GPR43 on colonic functions. J Physiol Pharmacol. 2008;59(Suppl 2):251–262.
    1. Kiefer J, Beyer-Sehlmeyer G, Pool-Zobel BL. Mixtures of SCFA, composed according to physiologically available concentrations in the gut lumen, modulate histone acetylation in human HT29 colon cancer cells. Br J Nutr. 2006;96(5):803–810.
    1. Hinnebusch BF, Meng S, Wu JT, Archer SY, Hodin RA. The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation. J Nutr. 2002;132(5):1012–1017.
    1. Aoyama M, Kotani J, Usami M. Butyrate and propionate induced activated or non-activated neutrophil apoptosis via HDAC inhibitor activity but without activating GPR-41/GPR-43 pathways. Nutrition. 2010;26(6):653–661.
    1. Delgoffe GM, Kole TP, Zheng Y, Zarek PE, Matthews KL, Xiao B, et al. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity. 2009;30(6):832–844.
    1. Lee K, Gudapati P, Dragovic S, Spencer C, Joyce S, Killeen N, et al. Mammalian target of rapamycin protein complex 2 regulates differentiation of Th1 and Th2 cell subsets via distinct signaling pathways. Immunity. 2010;32(6):743–753.
    1. Ochanuna Z, Geiger-Maor A, Dembinsky-Vaknin A, Karussis D, Tykocinski ML, Rachmilewitz J. Inhibition of effector function but not T cell activation and increase in FoxP3 expression in T cells differentiated in the presence of PP14. PLoS One. 2010;5(9):e12–68.
    1. Fenton TR, Gwalter J, Cramer R, Gout IT. S6K1 is acetylated at lysine 516 in response to growth factor stimulation. Biochemical and biophysical research communications. 2010;398(3):400–405.
    1. Fenton TR, Gwalter J, Ericsson J, Gout IT. Histone acetyltransferases interact with and acetylate p70 ribosomal S6 kinases in vitro and in vivo. The international journal of biochemistry & cell biology. 2010;42(2):359–366.
    1. Bolster DR, Crozier SJ, Kimball SR, Jefferson LS. AMP-activated protein kinase suppresses protein synthesis in rat skeletal muscle through down-regulated mammalian target of rapamycin (mTOR) signaling. The Journal of biological chemistry. 2002;277(27):23977–23980.
    1. Wei L, Laurence A, O’Shea JJ. New insights into the roles of Stat5a/b and Stat3 in T cell development and differentiation. Seminars in cell & developmental biology. 2008;19(4):394–400.
    1. Saraiva M, Christensen JR, Veldhoen M, Murphy TL, Murphy KM, O’Garra A. Interleukin-10 production by Th1 cells requires interleukin-12-induced STAT4 transcription factor and ERK MAP kinase activation by high antigen dose. Immunity. 2009;31(2):209–219.
    1. Hadjiagapiou C, Schmidt L, Dudeja PK, Layden TJ, Ramaswamy K. Mechanism(s) of butyrate transport in Caco-2 cells: role of monocarboxylate transporter 1. American journal of physiology Gastrointestinal and liver physiology. 2000;279(4):G775–780.
    1. Binder HJ. Role of colonic short-chain fatty acid transport in diarrhea. Annual review of physiology. 2010;72:297–313.
    1. Beier UH, Akimova T, Liu Y, Wang L, Hancock WW. Histone/protein deacetylases control Foxp3 expression and the heat shock response of T-regulatory cells. Curr Opin Immunol. 2011;23(5):670–678.
    1. Haberland M, Montgomery RL, Olson EN. The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nature reviews Genetics. 2009;10(1):32–42.
    1. Bhavsar P, Ahmad T, Adcock IM. The role of histone deacetylases in asthma and allergic diseases. The Journal of allergy and clinical immunology. 2008;121(3):580–584.
    1. Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, et al. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nature immunology. 2011;12(4):295–303.
    1. Haxhinasto S, Mathis D, Benoist C. The AKT-mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells. J Exp Med. 2008;205(3):565–574.
    1. Zeiser R, Leveson-Gower DB, Zambricki EA, Kambham N, Beilhack A, Loh J, et al. Differential impact of mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells. Blood. 2008;111(1):453–462.
    1. Huss DJ, Winger RC, Peng H, Yang Y, Racke MK, Lovett-Racke AE. TGF-beta enhances effector Th1 cell activation but promotes self-regulation via IL-10. J Immunol. 2010;184(10):5628–5636.
    1. Jankovic D, Kugler DG, Sher A. IL-10 production by CD4+ effector T cells: a mechanism for self-regulation. Mucosal immunology. 2010;3(3):239–246.
    1. Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014;40(1):128–139.
    1. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500(7461):232–236.
    1. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–450.
    1. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–455.
    1. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nature medicine. 2014;20(2):159–166.
    1. Symonds EL, Riedel CU, O’Mahony D, Lapthorne S, O’Mahony L, Shanahan F. Involvement of T helper type 17 and regulatory T cell activity in Citrobacter rodentium invasion and inflammatory damage. Clin Exp Immunol. 2009;157(1):148–154.

Source: PubMed

3
Suscribir