메뉴 건너뛰기




Volumn 154, Issue 2, 2018, Pages 220-229

Immune regulation by microbiome metabolites

Author keywords

barrier function; bile acids; immunity; indole; inflammation; metabolites; microbiome; short chain fatty acids

Indexed keywords

AMINO ACID DERIVATIVE; AROMATIC HYDROCARBON RECEPTOR; BILE ACID; CELL RECEPTOR; CHOLESTEROL; FARNESOID X RECEPTOR; G PROTEIN COUPLED RECEPTOR; G PROTEIN COUPLED RECEPTOR 109A; G PROTEIN COUPLED RECEPTOR 41; G PROTEIN COUPLED RECEPTOR 43; INDOLE DERIVATIVE; LIPID; PREGNANE X RECEPTOR; PURINERGIC P2X7 RECEPTOR; SHORT CHAIN FATTY ACID; TGR5 RECEPTOR; UNCLASSIFIED DRUG; IMMUNOGLOBULIN RECEPTOR; INDOLE; VOLATILE FATTY ACID;

EID: 85047872974     PISSN: 00192805     EISSN: 13652567     Source Type: Journal    
DOI: 10.1111/imm.12930     Document Type: Review
Times cited : (242)

References (133)
  • 1
    • 85017165971 scopus 로고    scopus 로고
    • Gut microbiota functions: metabolism of nutrients and other food components
    • Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I et al. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr 2017; 57:1–24.
    • (2017) Eur J Nutr , vol.57 , pp. 1-24
    • Rowland, I.1    Gibson, G.2    Heinken, A.3    Scott, K.4    Swann, J.5    Thiele, I.6
  • 2
    • 84971519476 scopus 로고    scopus 로고
    • From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites
    • Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 2016; 165:1332–45.
    • (2016) Cell , vol.165 , pp. 1332-1345
    • Koh, A.1    De Vadder, F.2    Kovatcheva-Datchary, P.3    Backhed, F.4
  • 3
    • 84991053856 scopus 로고    scopus 로고
    • Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism
    • Wahlstrom A, Sayin SI, Marschall HU, Backhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab 2016; 24:41–50.
    • (2016) Cell Metab , vol.24 , pp. 41-50
    • Wahlstrom, A.1    Sayin, S.I.2    Marschall, H.U.3    Backhed, F.4
  • 4
    • 84883139076 scopus 로고    scopus 로고
    • The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism
    • den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud DJ, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 2013; 54:2325–40.
    • (2013) J Lipid Res , vol.54 , pp. 2325-2340
    • den Besten, G.1    van Eunen, K.2    Groen, A.K.3    Venema, K.4    Reijngoud, D.J.5    Bakker, B.M.6
  • 5
    • 84971201113 scopus 로고    scopus 로고
    • Gut microbiota, metabolites and host immunity
    • Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016; 16:341–52.
    • (2016) Nat Rev Immunol , vol.16 , pp. 341-352
    • Rooks, M.G.1    Garrett, W.S.2
  • 6
    • 85014707675 scopus 로고    scopus 로고
    • Microbiome-modulated metabolites at the interface of host immunity
    • Blacher E, Levy M, Tatirovsky E, Elinav E. Microbiome-modulated metabolites at the interface of host immunity. J Immunol 2017; 198:572–80.
    • (2017) J Immunol , vol.198 , pp. 572-580
    • Blacher, E.1    Levy, M.2    Tatirovsky, E.3    Elinav, E.4
  • 7
    • 85020788732 scopus 로고    scopus 로고
    • Understanding the holobiont: how microbial metabolites affect human health and shape the immune system
    • Postler TS, Ghosh S. Understanding the holobiont: how microbial metabolites affect human health and shape the immune system. Cell Metab 2017; 26:110–30.
    • (2017) Cell Metab , vol.26 , pp. 110-130
    • Postler, T.S.1    Ghosh, S.2
  • 8
    • 84924758166 scopus 로고    scopus 로고
    • Gut microbiota-derived short-chain fatty acids, T cells, and inflammation
    • Kim CH, Park J, Kim M. Gut microbiota-derived short-chain fatty acids, T cells, and inflammation. Immune Netw 2014; 14:277–88.
    • (2014) Immune Netw , vol.14 , pp. 277-288
    • Kim, C.H.1    Park, J.2    Kim, M.3
  • 9
    • 85006379794 scopus 로고    scopus 로고
    • Formation of propionate and butyrate by the human colonic microbiota
    • Louis P, Flint HJ. Formation of propionate and butyrate by the human colonic microbiota. Environ Microbiol 2017; 19:29–41.
    • (2017) Environ Microbiol , vol.19 , pp. 29-41
    • Louis, P.1    Flint, H.J.2
  • 10
    • 0029665112 scopus 로고    scopus 로고
    • The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7)
    • Surprenant A, Rassendren F, Kawashima E, North RA, Buell G. The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7). Science 1996; 272:735–8.
    • (1996) Science , vol.272 , pp. 735-738
    • Surprenant, A.1    Rassendren, F.2    Kawashima, E.3    North, R.A.4    Buell, G.5
  • 11
    • 0033026760 scopus 로고    scopus 로고
    • Endogenous bile acids are ligands for the nuclear receptor FXR/BAR
    • Wang H, Chen J, Hollister K, Sowers LC, Forman BM. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol Cell 1999; 3:543–53.
    • (1999) Mol Cell , vol.3 , pp. 543-553
    • Wang, H.1    Chen, J.2    Hollister, K.3    Sowers, L.C.4    Forman, B.M.5
  • 13
    • 0022647681 scopus 로고
    • Ah receptor mediating induction of aryl hydrocarbon hydroxylase: detection in human lung by binding of 2,3,7,8-[3H]tetrachlorodibenzo-p-dioxin
    • Roberts EA, Golas CL, Okey AB. Ah receptor mediating induction of aryl hydrocarbon hydroxylase: detection in human lung by binding of 2,3,7,8-[3H]tetrachlorodibenzo-p-dioxin. Cancer Res 1986; 46:3739–43.
    • (1986) Cancer Res , vol.46 , pp. 3739-3743
    • Roberts, E.A.1    Golas, C.L.2    Okey, A.B.3
  • 14
    • 0032498303 scopus 로고    scopus 로고
    • An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway
    • Kliewer SA, Moore JT, Wade L, Staudinger JL, Watson MA, Jones SA et al. An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell 1998; 92:73–82.
    • (1998) Cell , vol.92 , pp. 73-82
    • Kliewer, S.A.1    Moore, J.T.2    Wade, L.3    Staudinger, J.L.4    Watson, M.A.5    Jones, S.A.6
  • 15
    • 84907597269 scopus 로고    scopus 로고
    • Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4
    • Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity 2014; 41:296–310.
    • (2014) Immunity , vol.41 , pp. 296-310
    • Venkatesh, M.1    Mukherjee, S.2    Wang, H.3    Li, H.4    Sun, K.5    Benechet, A.P.6
  • 16
    • 0026055308 scopus 로고
    • A classification of glycosyl hydrolases based on amino acid sequence similarities
    • Henrissat B. A classification of glycosyl hydrolases based on amino acid sequence similarities. Biochem J 1991; 280:309–16.
    • (1991) Biochem J , vol.280 , pp. 309-316
    • Henrissat, B.1
  • 17
    • 0029166485 scopus 로고
    • Conserved catalytic machinery and the prediction of a common fold for several families of glycosyl hydrolases
    • Henrissat B, Callebaut I, Fabrega S, Lehn P, Mornon JP, Davies G. Conserved catalytic machinery and the prediction of a common fold for several families of glycosyl hydrolases. Proc Natl Acad Sci USA 1995; 92:7090–4.
    • (1995) Proc Natl Acad Sci USA , vol.92 , pp. 7090-7094
    • Henrissat, B.1    Callebaut, I.2    Fabrega, S.3    Lehn, P.4    Mornon, J.P.5    Davies, G.6
  • 18
    • 0033167973 scopus 로고    scopus 로고
    • The cellulosome concept as an efficient microbial strategy for the degradation of insoluble polysaccharides
    • Shoham Y, Lamed R, Bayer EA. The cellulosome concept as an efficient microbial strategy for the degradation of insoluble polysaccharides. Trends Microbiol 1999; 7:275–81.
    • (1999) Trends Microbiol , vol.7 , pp. 275-281
    • Shoham, Y.1    Lamed, R.2    Bayer, E.A.3
  • 19
    • 0025338715 scopus 로고
    • The bacterial phosphoenolpyruvate: glycose phosphotransferase system
    • Meadow ND, Fox DK, Roseman S. The bacterial phosphoenolpyruvate: glycose phosphotransferase system. Annu Rev Biochem 1990; 59:497–542.
    • (1990) Annu Rev Biochem , vol.59 , pp. 497-542
    • Meadow, N.D.1    Fox, D.K.2    Roseman, S.3
  • 20
    • 84862200127 scopus 로고    scopus 로고
    • Complex carbohydrate utilization by the healthy human microbiome
    • Cantarel BL, Lombard V, Henrissat B. Complex carbohydrate utilization by the healthy human microbiome. PLoS One 2012; 7:e28742.
    • (2012) PLoS One , vol.7
    • Cantarel, B.L.1    Lombard, V.2    Henrissat, B.3
  • 21
    • 0027715254 scopus 로고
    • In vitro production of short-chain fatty acids by bacterial fermentation of dietary fiber compared with effects of those fibers on hepatic sterol synthesis in rats
    • Stark AH, Madar Z. In vitro production of short-chain fatty acids by bacterial fermentation of dietary fiber compared with effects of those fibers on hepatic sterol synthesis in rats. J Nutr 1993; 123:2166–73.
    • (1993) J Nutr , vol.123 , pp. 2166-2173
    • Stark, A.H.1    Madar, Z.2
  • 22
    • 0037509909 scopus 로고    scopus 로고
    • Regulation of short-chain fatty acid production
    • Macfarlane S, Macfarlane GT. Regulation of short-chain fatty acid production. Proc Nutr Soc 2003; 62:67–72.
    • (2003) Proc Nutr Soc , vol.62 , pp. 67-72
    • Macfarlane, S.1    Macfarlane, G.T.2
  • 23
    • 5444248493 scopus 로고    scopus 로고
    • Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium
    • Derrien M, Vaughan EE, Plugge CM, de Vos WM. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol 2004; 54:1469–76.
    • (2004) Int J Syst Evol Microbiol , vol.54 , pp. 1469-1476
    • Derrien, M.1    Vaughan, E.E.2    Plugge, C.M.3    de Vos, W.M.4
  • 24
    • 84908608590 scopus 로고    scopus 로고
    • The gut microbiota, bacterial metabolites and colorectal cancer
    • Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol 2014; 12:661–72.
    • (2014) Nat Rev Microbiol , vol.12 , pp. 661-672
    • Louis, P.1    Hold, G.L.2    Flint, H.J.3
  • 25
    • 0038153876 scopus 로고    scopus 로고
    • SLC5A8, a sodium transporter, is a tumor suppressor gene silenced by methylation in human colon aberrant crypt foci and cancers
    • Li H, Myeroff L, Smiraglia D, Romero MF, Pretlow TP, Kasturi L et al. SLC5A8, a sodium transporter, is a tumor suppressor gene silenced by methylation in human colon aberrant crypt foci and cancers. Proc Natl Acad Sci USA 2003; 100:8412–7.
    • (2003) Proc Natl Acad Sci USA , vol.100 , pp. 8412-8417
    • Li, H.1    Myeroff, L.2    Smiraglia, D.3    Romero, M.F.4    Pretlow, T.P.5    Kasturi, L.6
  • 26
    • 54049151915 scopus 로고    scopus 로고
    • Cellular expression of a sodium-dependent monocarboxylate transporter (Slc5a8) and the MCT family in the mouse kidney
    • Yanase H, Takebe K, Nio-Kobayashi J, Takahashi-Iwanaga H, Iwanaga T. Cellular expression of a sodium-dependent monocarboxylate transporter (Slc5a8) and the MCT family in the mouse kidney. Histochem Cell Biol 2008; 130:957–66.
    • (2008) Histochem Cell Biol , vol.130 , pp. 957-966
    • Yanase, H.1    Takebe, K.2    Nio-Kobayashi, J.3    Takahashi-Iwanaga, H.4    Iwanaga, T.5
  • 27
    • 77649119543 scopus 로고    scopus 로고
    • Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene
    • Louis P, Young P, Holtrop G, Flint HJ. Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene. Environ Microbiol 2010; 12:304–14.
    • (2010) Environ Microbiol , vol.12 , pp. 304-314
    • Louis, P.1    Young, P.2    Holtrop, G.3    Flint, H.J.4
  • 28
    • 84961200851 scopus 로고    scopus 로고
    • Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism
    • Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016; 7:189–200.
    • (2016) Gut Microbes , vol.7 , pp. 189-200
    • Morrison, D.J.1    Preston, T.2
  • 29
    • 84863990011 scopus 로고    scopus 로고
    • Ruminococcus bromii is a keystone species for the degradation of resistant starch in the human colon
    • Ze X, Duncan SH, Louis P, Flint HJ. Ruminococcus bromii is a keystone species for the degradation of resistant starch in the human colon. ISME J 2012; 6:1535–43.
    • (2012) ISME J , vol.6 , pp. 1535-1543
    • Ze, X.1    Duncan, S.H.2    Louis, P.3    Flint, H.J.4
  • 30
    • 85015839014 scopus 로고    scopus 로고
    • Regulation of humoral immunity by gut microbial products
    • Kim M, Kim CH. Regulation of humoral immunity by gut microbial products. Gut Microbes 2017; 8:392–9.
    • (2017) Gut Microbes , vol.8 , pp. 392-399
    • Kim, M.1    Kim, C.H.2
  • 31
    • 84927934357 scopus 로고    scopus 로고
    • Dietary gut microbial metabolites, short-chain fatty acids, and host metabolic regulation
    • Kasubuchi M, Hasegawa S, Hiramatsu T, Ichimura A, Kimura I. Dietary gut microbial metabolites, short-chain fatty acids, and host metabolic regulation. Nutrients 2015; 7:2839–49.
    • (2015) Nutrients , vol.7 , pp. 2839-2849
    • Kasubuchi, M.1    Hasegawa, S.2    Hiramatsu, T.3    Ichimura, A.4    Kimura, I.5
  • 32
    • 0029882856 scopus 로고    scopus 로고
    • Pathways of acetate, propionate, and butyrate formation by the human fecal microbial flora
    • Miller TL, Wolin MJ. Pathways of acetate, propionate, and butyrate formation by the human fecal microbial flora. Appl Environ Microbiol 1996; 62:1589–92.
    • (1996) Appl Environ Microbiol , vol.62 , pp. 1589-1592
    • Miller, T.L.1    Wolin, M.J.2
  • 33
    • 0038363378 scopus 로고    scopus 로고
    • The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids
    • Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem 2003; 278:11 312–9.
    • (2003) J Biol Chem , vol.278 , pp. 11 312-9
    • Brown, A.J.1    Goldsworthy, S.M.2    Barnes, A.A.3    Eilert, M.M.4    Tcheang, L.5    Daniels, D.6
  • 34
    • 0037453280 scopus 로고    scopus 로고
    • Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids
    • Nilsson NE, Kotarsky K, Owman C, Olde B. Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids. Biochem Biophys Res Commun 2003; 303:1047–52.
    • (2003) Biochem Biophys Res Commun , vol.303 , pp. 1047-1052
    • Nilsson, N.E.1    Kotarsky, K.2    Owman, C.3    Olde, B.4
  • 35
    • 66149084058 scopus 로고    scopus 로고
    • GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon
    • Thangaraju M, Cresci GA, Liu K, Ananth S, Gnanaprakasam JP, Browning DD et al. GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res 2009; 69:2826–32.
    • (2009) Cancer Res , vol.69 , pp. 2826-2832
    • Thangaraju, M.1    Cresci, G.A.2    Liu, K.3    Ananth, S.4    Gnanaprakasam, J.P.5    Browning, D.D.6
  • 36
    • 84875047650 scopus 로고    scopus 로고
    • Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation
    • Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci USA 2013; 110:4410–5.
    • (2013) Proc Natl Acad Sci USA , vol.110 , pp. 4410-4415
    • Pluznick, J.L.1    Protzko, R.J.2    Gevorgyan, H.3    Peterlin, Z.4    Sipos, A.5    Han, J.6
  • 37
    • 84880620577 scopus 로고    scopus 로고
    • Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice
    • 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:e1–10.
    • (2013) Gastroenterology , vol.145 , pp. e1-10
    • Kim, M.H.1    Kang, S.G.2    Park, J.H.3    Yanagisawa, M.4    Kim, C.H.5
  • 38
    • 0038491435 scopus 로고    scopus 로고
    • Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation
    • Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem 2003; 278:25 481–9.
    • (2003) J Biol Chem , vol.278 , pp. 25 481-9
    • Le Poul, E.1    Loison, C.2    Struyf, S.3    Springael, J.Y.4    Lannoy, V.5    Decobecq, M.E.6
  • 39
    • 84922163095 scopus 로고    scopus 로고
    • Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway
    • Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson 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 2015; 8:80–93.
    • (2015) Mucosal Immunol , vol.8 , pp. 80-93
    • Park, J.1    Kim, M.2    Kang, S.G.3    Jannasch, A.H.4    Cooper, B.5    Patterson, J.6
  • 40
    • 85022328114 scopus 로고    scopus 로고
    • The short chain fatty acid receptor GPR43 regulates inflammatory signals in adipose tissue M2-type macrophages
    • Nakajima A, Nakatani A, Hasegawa S, Irie J, Ozawa K, Tsujimoto G et al. The short chain fatty acid receptor GPR43 regulates inflammatory signals in adipose tissue M2-type macrophages. PLoS One 2017; 12:e0179696.
    • (2017) PLoS One , vol.12
    • Nakajima, A.1    Nakatani, A.2    Hasegawa, S.3    Irie, J.4    Ozawa, K.5    Tsujimoto, G.6
  • 41
    • 79955018521 scopus 로고    scopus 로고
    • Amino acid metabolism in intestinal bacteria: links between gut ecology and host health
    • Dai ZL, Wu G, Zhu WY. Amino acid metabolism in intestinal bacteria: links between gut ecology and host health. Front Biosci (Landmark Ed) 2011; 16:1768–86.
    • (2011) Front Biosci (Landmark Ed) , vol.16 , pp. 1768-1786
    • Dai, Z.L.1    Wu, G.2    Zhu, W.Y.3
  • 42
  • 43
    • 84944278561 scopus 로고    scopus 로고
    • Remaining mysteries of molecular biology: the role of polyamines in the cell
    • Miller-Fleming L, Olin-Sandoval V, Campbell K, Ralser M. Remaining mysteries of molecular biology: the role of polyamines in the cell. J Mol Biol 2015; 427:3389–406.
    • (2015) J Mol Biol , vol.427 , pp. 3389-3406
    • Miller-Fleming, L.1    Olin-Sandoval, V.2    Campbell, K.3    Ralser, M.4
  • 44
    • 84881646440 scopus 로고    scopus 로고
    • Pharmacological potential of biogenic amine-polyamine interactions beyond neurotransmission
    • Sanchez-Jimenez F, Ruiz-Perez MV, Urdiales JL, Medina MA. Pharmacological potential of biogenic amine-polyamine interactions beyond neurotransmission. Br J Pharmacol 2013; 170:4–16.
    • (2013) Br J Pharmacol , vol.170 , pp. 4-16
    • Sanchez-Jimenez, F.1    Ruiz-Perez, M.V.2    Urdiales, J.L.3    Medina, M.A.4
  • 45
    • 0035134087 scopus 로고    scopus 로고
    • Biogenic amine production by Lactobacillus
    • Arena ME, Manca de Nadra MC. Biogenic amine production by Lactobacillus. J Appl Microbiol 2001; 90:158–62.
    • (2001) J Appl Microbiol , vol.90 , pp. 158-162
    • Arena, M.E.1    Manca de Nadra, M.C.2
  • 46
    • 36448955373 scopus 로고    scopus 로고
    • Effects of amino acid-derived luminal metabolites on the colonic epithelium and physiopathological consequences
    • Blachier F, Mariotti F, Huneau JF, Tome D. Effects of amino acid-derived luminal metabolites on the colonic epithelium and physiopathological consequences. Amino Acids 2007; 33:547–62.
    • (2007) Amino Acids , vol.33 , pp. 547-562
    • Blachier, F.1    Mariotti, F.2    Huneau, J.F.3    Tome, D.4
  • 47
    • 0031438749 scopus 로고    scopus 로고
    • Expression of the human aryl hydrocarbon receptor complex in yeast. Activation of transcription by indole compounds
    • Miller CA 3rd. Expression of the human aryl hydrocarbon receptor complex in yeast. Activation of transcription by indole compounds. J Biol Chem 1997; 272:32 824–9.
    • (1997) J Biol Chem , vol.272 , pp. 32 824-9
    • Miller, C.A.1
  • 49
    • 0026046412 scopus 로고
    • Aromatic hydrocarbon responsiveness-receptor agonists generated from indole-3-carbinol in vitro and in vivo: comparisons with 2,3,7,8-tetrachlorodibenzo-p-dioxin
    • Bjeldanes LF, Kim JY, Grose KR, Bartholomew JC, Bradfield CA. Aromatic hydrocarbon responsiveness-receptor agonists generated from indole-3-carbinol in vitro and in vivo: comparisons with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Proc Natl Acad Sci USA 1991; 88:9543–7.
    • (1991) Proc Natl Acad Sci USA , vol.88 , pp. 9543-9547
    • Bjeldanes, L.F.1    Kim, J.Y.2    Grose, K.R.3    Bartholomew, J.C.4    Bradfield, C.A.5
  • 50
    • 34447117473 scopus 로고    scopus 로고
    • Dietary phytochemicals regulate whole-body CYP1A1 expression through an arylhydrocarbon receptor nuclear translocator-dependent system in gut
    • Ito S, Chen C, Satoh J, Yim S, Gonzalez FJ. Dietary phytochemicals regulate whole-body CYP1A1 expression through an arylhydrocarbon receptor nuclear translocator-dependent system in gut. J Clin Invest 2007; 117:1940–50.
    • (2007) J Clin Invest , vol.117 , pp. 1940-1950
    • Ito, S.1    Chen, C.2    Satoh, J.3    Yim, S.4    Gonzalez, F.J.5
  • 51
    • 0001679413 scopus 로고
    • Biohydrogenation of unsaturated fatty acids by rumen bacteria
    • Polan CE, McNeill JJ, Tove SB. Biohydrogenation of unsaturated fatty acids by rumen bacteria. J Bacteriol 1964; 88:1056–64.
    • (1964) J Bacteriol , vol.88 , pp. 1056-1064
    • Polan, C.E.1    McNeill, J.J.2    Tove, S.B.3
  • 52
    • 84887122496 scopus 로고    scopus 로고
    • Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition
    • Kishino S, Takeuchi M, Park SB, Hirata A, Kitamura N, Kunisawa J et al. Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition. Proc Natl Acad Sci USA 2013; 110:17 808–13.
    • (2013) Proc Natl Acad Sci USA , vol.110 , pp. 17 808-13
    • Kishino, S.1    Takeuchi, M.2    Park, S.B.3    Hirata, A.4    Kitamura, N.5    Kunisawa, J.6
  • 53
    • 33947124087 scopus 로고    scopus 로고
    • Metabolism of linoleic acid by human gut bacteria: different routes for biosynthesis of conjugated linoleic acid
    • Devillard E, McIntosh FM, Duncan SH, Wallace RJ. Metabolism of linoleic acid by human gut bacteria: different routes for biosynthesis of conjugated linoleic acid. J Bacteriol 2007; 189:2566–70.
    • (2007) J Bacteriol , vol.189 , pp. 2566-2570
    • Devillard, E.1    McIntosh, F.M.2    Duncan, S.H.3    Wallace, R.J.4
  • 54
    • 84903524994 scopus 로고    scopus 로고
    • Metabolism of cholesterol and bile acids by the gut microbiota
    • Gerard P. Metabolism of cholesterol and bile acids by the gut microbiota. Pathogens 2013; 3:14–24.
    • (2013) Pathogens , vol.3 , pp. 14-24
    • Gerard, P.1
  • 55
    • 85015364857 scopus 로고    scopus 로고
    • Trimethylamine and trimethylamine N-Oxide, a Flavin-Containing Monooxygenase 3 (FMO3)-mediated host-microbiome metabolic axis implicated in health and disease
    • Fennema D, Phillips IR, Shephard EA. Trimethylamine and trimethylamine N-Oxide, a Flavin-Containing Monooxygenase 3 (FMO3)-mediated host-microbiome metabolic axis implicated in health and disease. Drug Metab Dispos 2016; 44:1839–50.
    • (2016) Drug Metab Dispos , vol.44 , pp. 1839-1850
    • Fennema, D.1    Phillips, I.R.2    Shephard, E.A.3
  • 57
    • 0029046931 scopus 로고
    • Identification of a nuclear receptor that is activated by farnesol metabolites
    • Forman BM, Goode E, Chen J, Oro AE, Bradley DJ, Perlmann T et al. Identification of a nuclear receptor that is activated by farnesol metabolites. Cell 1995; 81:687–93.
    • (1995) Cell , vol.81 , pp. 687-693
    • Forman, B.M.1    Goode, E.2    Chen, J.3    Oro, A.E.4    Bradley, D.J.5    Perlmann, T.6
  • 58
    • 1542513682 scopus 로고    scopus 로고
    • Expression and activation of the farnesoid X receptor in the vasculature
    • Bishop-Bailey D, Walsh DT, Warner TD. Expression and activation of the farnesoid X receptor in the vasculature. Proc Natl Acad Sci USA 2004; 101:3668–73.
    • (2004) Proc Natl Acad Sci USA , vol.101 , pp. 3668-3673
    • Bishop-Bailey, D.1    Walsh, D.T.2    Warner, T.D.3
  • 60
    • 84895520482 scopus 로고    scopus 로고
    • The bile acid TGR5 membrane receptor: from basic research to clinical application
    • Duboc H, Tache Y, Hofmann AF. The bile acid TGR5 membrane receptor: from basic research to clinical application. Dig Liver Dis 2014; 46:302–12.
    • (2014) Dig Liver Dis , vol.46 , pp. 302-312
    • Duboc, H.1    Tache, Y.2    Hofmann, A.F.3
  • 61
    • 85037168132 scopus 로고    scopus 로고
    • Distinct fecal and oral microbiota composition in human type 1 diabetes, an observational study
    • de Groot PF, Belzer C, Aydin O, Levin E, Levels JH, Aalvink S et al. Distinct fecal and oral microbiota composition in human type 1 diabetes, an observational study. PLoS One 2017; 12:e0188475.
    • (2017) PLoS One , vol.12
    • de Groot, P.F.1    Belzer, C.2    Aydin, O.3    Levin, E.4    Levels, J.H.5    Aalvink, S.6
  • 62
    • 84874005935 scopus 로고    scopus 로고
    • Gut microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study
    • Murri M, Leiva I, Gomez-Zumaquero JM, Tinahones FJ, Cardona F, Soriguer F et al. Gut microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study. BMC Med 2013; 11:46.
    • (2013) BMC Med , vol.11 , pp. 46
    • Murri, M.1    Leiva, I.2    Gomez-Zumaquero, J.M.3    Tinahones, F.J.4    Cardona, F.5    Soriguer, F.6
  • 63
    • 0029693922 scopus 로고    scopus 로고
    • Short-chain fatty acids in the human colon: relation to gastrointestinal health and disease
    • Mortensen PB, Clausen MR. Short-chain fatty acids in the human colon: relation to gastrointestinal health and disease. Scand J Gastroenterol Suppl 1996; 216:132–48.
    • (1996) Scand J Gastroenterol Suppl , vol.216 , pp. 132-148
    • Mortensen, P.B.1    Clausen, M.R.2
  • 64
    • 84876920542 scopus 로고    scopus 로고
    • Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma
    • Chen HM, Yu YN, Wang JL, Lin YW, Kong X, Yang CQ et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr 2013; 97:1044–52.
    • (2013) Am J Clin Nutr , vol.97 , pp. 1044-1052
    • Chen, H.M.1    Yu, Y.N.2    Wang, J.L.3    Lin, Y.W.4    Kong, X.5    Yang, C.Q.6
  • 65
    • 64149112596 scopus 로고    scopus 로고
    • Faecal short chain fatty acid pattern and allergy in early childhood
    • Sandin A, Braback L, Norin E, Bjorksten B. Faecal short chain fatty acid pattern and allergy in early childhood. Acta Paediatr 2009; 98:823–7.
    • (2009) Acta Paediatr , vol.98 , pp. 823-827
    • Sandin, A.1    Braback, L.2    Norin, E.3    Bjorksten, B.4
  • 66
    • 0033692480 scopus 로고    scopus 로고
    • Microflora-associated characteristics in faeces from allergic and nonallergic infants
    • Bottcher MF, Nordin EK, Sandin A, Midtvedt T, Bjorksten B. Microflora-associated characteristics in faeces from allergic and nonallergic infants. Clin Exp Allergy 2000; 30:1590–6.
    • (2000) Clin Exp Allergy , vol.30 , pp. 1590-1596
    • Bottcher, M.F.1    Nordin, E.K.2    Sandin, A.3    Midtvedt, T.4    Bjorksten, B.5
  • 67
    • 84922390026 scopus 로고    scopus 로고
    • Faecal metabolite profiling identifies medium-chain fatty acids as discriminating compounds in IBD
    • De Preter V, Machiels K, Joossens M, Arijs I, Matthys C, Vermeire S et al. Faecal metabolite profiling identifies medium-chain fatty acids as discriminating compounds in IBD. Gut 2015; 64:447–58.
    • (2015) Gut , vol.64 , pp. 447-458
    • De Preter, V.1    Machiels, K.2    Joossens, M.3    Arijs, I.4    Matthys, C.5    Vermeire, S.6
  • 69
    • 84966526506 scopus 로고    scopus 로고
    • CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands
    • Lamas B, Richard ML, Leducq V, Pham HP, Michel ML, Da Costa G et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med 2016; 22:598–605.
    • (2016) Nat Med , vol.22 , pp. 598-605
    • Lamas, B.1    Richard, M.L.2    Leducq, V.3    Pham, H.P.4    Michel, M.L.5    Da Costa, G.6
  • 71
    • 0020696321 scopus 로고
    • Stimulatory effect of short chain fatty acids on the epithelial cell proliferation in rat large intestine
    • Sakata T, von Engelhardt W. Stimulatory effect of short chain fatty acids on the epithelial cell proliferation in rat large intestine. Comp Biochem Physiol A Comp Physiol 1983; 74:459–62.
    • (1983) Comp Biochem Physiol A Comp Physiol , vol.74 , pp. 459-462
    • Sakata, T.1    von Engelhardt, W.2
  • 72
    • 0028143270 scopus 로고
    • Dietary fibers stimulate colonic cell proliferation by different mechanisms at different sites
    • Zhang J, Lupton JR. Dietary fibers stimulate colonic cell proliferation by different mechanisms at different sites. Nutr Cancer 1994; 22:267–76.
    • (1994) Nutr Cancer , vol.22 , pp. 267-276
    • Zhang, J.1    Lupton, J.R.2
  • 73
    • 0242577147 scopus 로고    scopus 로고
    • Acetate and butyrate are the major substrates for de novo lipogenesis in rat colonic epithelial cells
    • Zambell KL, Fitch MD, Fleming SE. Acetate and butyrate are the major substrates for de novo lipogenesis in rat colonic epithelial cells. J Nutr 2003; 133:3509–15.
    • (2003) J Nutr , vol.133 , pp. 3509-3515
    • Zambell, K.L.1    Fitch, M.D.2    Fleming, S.E.3
  • 75
    • 0028144716 scopus 로고
    • Propionate-initiated changes in intracellular pH in rabbit colonocytes
    • DeSoignie R, Sellin JH. Propionate-initiated changes in intracellular pH in rabbit colonocytes. Gastroenterology 1994; 107:347–56.
    • (1994) Gastroenterology , vol.107 , pp. 347-356
    • DeSoignie, R.1    Sellin, J.H.2
  • 76
    • 0032917770 scopus 로고    scopus 로고
    • Regulation of intracellular pH gradients by identified Na/H exchanger isoforms and a short-chain fatty acid
    • Gonda T, Maouyo D, Rees SE, Montrose MH. Regulation of intracellular pH gradients by identified Na/H exchanger isoforms and a short-chain fatty acid. Am J Physiol 1999; 276:G259–70.
    • (1999) Am J Physiol , vol.276 , pp. G259-G270
    • Gonda, T.1    Maouyo, D.2    Rees, S.E.3    Montrose, M.H.4
  • 77
    • 0028286187 scopus 로고
    • Potentiation by specific short-chain fatty acids of differentiation and apoptosis in human colonic carcinoma cell lines
    • Heerdt BG, Houston MA, Augenlicht LH. Potentiation by specific short-chain fatty acids of differentiation and apoptosis in human colonic carcinoma cell lines. Cancer Res 1994; 54:3288–93.
    • (1994) Cancer Res , vol.54 , pp. 3288-3293
    • Heerdt, B.G.1    Houston, M.A.2    Augenlicht, L.H.3
  • 78
    • 0036252352 scopus 로고    scopus 로고
    • The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation
    • 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; 132:1012–7.
    • (2002) J Nutr , vol.132 , pp. 1012-1017
    • Hinnebusch, B.F.1    Meng, S.2    Wu, J.T.3    Archer, S.Y.4    Hodin, R.A.5
  • 79
    • 84904543667 scopus 로고    scopus 로고
    • Gut microbial metabolism drives transformation of MSH2-deficient colon epithelial cells
    • Belcheva A, Irrazabal T, Robertson SJ, Streutker C, Maughan H, Rubino S et al. Gut microbial metabolism drives transformation of MSH2-deficient colon epithelial cells. Cell 2014; 158:288–99.
    • (2014) Cell , vol.158 , pp. 288-299
    • Belcheva, A.1    Irrazabal, T.2    Robertson, S.J.3    Streutker, C.4    Maughan, H.5    Rubino, S.6
  • 80
    • 0036737806 scopus 로고    scopus 로고
    • Enhanced production of IL-18 in butyrate-treated intestinal epithelium by stimulation of the proximal promoter region
    • Kalina U, Koyama N, Hosoda T, Nuernberger H, Sato K, Hoelzer D et al. Enhanced production of IL-18 in butyrate-treated intestinal epithelium by stimulation of the proximal promoter region. Eur J Immunol 2002; 32:2635–43.
    • (2002) Eur J Immunol , vol.32 , pp. 2635-2643
    • Kalina, U.1    Koyama, N.2    Hosoda, T.3    Nuernberger, H.4    Sato, K.5    Hoelzer, D.6
  • 81
    • 0038523850 scopus 로고    scopus 로고
    • Expression of the cathelicidin LL-37 is modulated by short chain fatty acids in colonocytes: relevance of signalling pathways
    • Schauber J, Svanholm C, Termen S, Iffland K, Menzel T, Scheppach W et al. Expression of the cathelicidin LL-37 is modulated by short chain fatty acids in colonocytes: relevance of signalling pathways. Gut 2003; 52:735–41.
    • (2003) Gut , vol.52 , pp. 735-741
    • Schauber, J.1    Svanholm, C.2    Termen, S.3    Iffland, K.4    Menzel, T.5    Scheppach, W.6
  • 82
    • 69549114504 scopus 로고    scopus 로고
    • Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers
    • Peng L, Li ZR, Green RS, Holzman IR, Lin J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J Nutr 2009; 139:1619–25.
    • (2009) J Nutr , vol.139 , pp. 1619-1625
    • Peng, L.1    Li, Z.R.2    Green, R.S.3    Holzman, I.R.4    Lin, J.5
  • 83
    • 74449085010 scopus 로고    scopus 로고
    • HDAC inhibitors regulate claudin-1 expression in colon cancer cells through modulation of mRNA stability
    • Krishnan M, Singh AB, Smith JJ, Sharma A, Chen X, Eschrich S et al. HDAC inhibitors regulate claudin-1 expression in colon cancer cells through modulation of mRNA stability. Oncogene 2010; 29:305–12.
    • (2010) Oncogene , vol.29 , pp. 305-312
    • Krishnan, M.1    Singh, A.B.2    Smith, J.J.3    Sharma, A.4    Chen, X.5    Eschrich, S.6
  • 84
    • 66549120804 scopus 로고    scopus 로고
    • The regulation of intestinal mucin MUC2 expression by short-chain fatty acids: implications for epithelial protection
    • Burger-van Paassen N, Vincent A, Puiman PJ, van der Sluis M, Bouma J, Boehm G et al. The regulation of intestinal mucin MUC2 expression by short-chain fatty acids: implications for epithelial protection. Biochem J 2009; 420:211–9.
    • (2009) Biochem J , vol.420 , pp. 211-219
    • Burger-van Paassen, N.1    Vincent, A.2    Puiman, P.J.3    van der Sluis, M.4    Bouma, J.5    Boehm, G.6
  • 85
    • 85031497192 scopus 로고    scopus 로고
    • Microbial-derived butyrate promotes epithelial barrier function through IL-10 receptor-dependent repression of Claudin-2
    • Zheng L, Kelly CJ, Battista KD, Schaefer R, Lanis JM, Alexeev EE et al. Microbial-derived butyrate promotes epithelial barrier function through IL-10 receptor-dependent repression of Claudin-2. J Immunol 2017; 199: 2976–84
    • (2017) J Immunol , vol.199 , pp. 2976-2984
    • Zheng, L.1    Kelly, C.J.2    Battista, K.D.3    Schaefer, R.4    Lanis, J.M.5    Alexeev, E.E.6
  • 87
    • 84892449521 scopus 로고    scopus 로고
    • Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis
    • 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; 40:128–39.
    • (2014) Immunity , vol.40 , pp. 128-139
    • Singh, N.1    Gurav, A.2    Sivaprakasam, S.3    Brady, E.4    Padia, R.5    Shi, H.6
  • 88
    • 84893859801 scopus 로고    scopus 로고
    • The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition
    • Chang PV, Hao L, Offermanns S, Medzhitov R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci USA 2014; 111:2247–52.
    • (2014) Proc Natl Acad Sci USA , vol.111 , pp. 2247-2252
    • Chang, P.V.1    Hao, L.2    Offermanns, S.3    Medzhitov, R.4
  • 89
    • 84878414731 scopus 로고    scopus 로고
    • Butyrate attenuates inflammation and lipolysis generated by the interaction of adipocytes and macrophages
    • Ohira H, Fujioka Y, Katagiri C, Mamoto R, Aoyama-Ishikawa M, Amako K et al. Butyrate attenuates inflammation and lipolysis generated by the interaction of adipocytes and macrophages. J Atheroscler Thromb 2013; 20:425–42.
    • (2013) J Atheroscler Thromb , vol.20 , pp. 425-442
    • Ohira, H.1    Fujioka, Y.2    Katagiri, C.3    Mamoto, R.4    Aoyama-Ishikawa, M.5    Amako, K.6
  • 91
    • 85014936442 scopus 로고    scopus 로고
    • Diet-derived short chain fatty acids stimulate intestinal epithelial cells to induce mucosal tolerogenic dendritic cells
    • Goverse G, Molenaar R, Macia L, Tan J, Erkelens MN, Konijn T et al. Diet-derived short chain fatty acids stimulate intestinal epithelial cells to induce mucosal tolerogenic dendritic cells. J Immunol 2017; 198:2172–81.
    • (2017) J Immunol , vol.198 , pp. 2172-2181
    • Goverse, G.1    Molenaar, R.2    Macia, L.3    Tan, J.4    Erkelens, M.N.5    Konijn, T.6
  • 92
    • 84867433001 scopus 로고    scopus 로고
    • n-Butyrate anergized effector CD4 + T cells independent of regulatory T cell generation or activity
    • Fontenelle B, Gilbert KM. n-Butyrate anergized effector CD4 + T cells independent of regulatory T cell generation or activity. Scand J Immunol 2012; 76:457–63.
    • (2012) Scand J Immunol , vol.76 , pp. 457-463
    • Fontenelle, B.1    Gilbert, K.M.2
  • 93
    • 84881068658 scopus 로고    scopus 로고
    • The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis
    • 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; 341:569–73.
    • (2013) Science , vol.341 , pp. 569-573
    • Smith, P.M.1    Howitt, M.R.2    Panikov, N.3    Michaud, M.4    Gallini, C.A.5    Bohlooly, Y.M.6
  • 94
    • 84890550163 scopus 로고    scopus 로고
    • Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation
    • 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; 504:451–5.
    • (2013) Nature , vol.504 , pp. 451-455
    • Arpaia, N.1    Campbell, C.2    Fan, X.3    Dikiy, S.4    van der Veeken, J.5    deRoos, P.6
  • 95
    • 84890564250 scopus 로고    scopus 로고
    • Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells
    • 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; 504:446–50.
    • (2013) Nature , vol.504 , pp. 446-450
    • Furusawa, Y.1    Obata, Y.2    Fukuda, S.3    Endo, T.A.4    Nakato, G.5    Takahashi, D.6
  • 96
    • 3042697412 scopus 로고    scopus 로고
    • Dietary fructooligosaccharides up-regulate immunoglobulin A response and polymeric immunoglobulin receptor expression in intestines of infant mice
    • Nakamura Y, Nosaka S, Suzuki M, Nagafuchi S, Takahashi T, Yajima T et al. Dietary fructooligosaccharides up-regulate immunoglobulin A response and polymeric immunoglobulin receptor expression in intestines of infant mice. Clin Exp Immunol 2004; 137:52–8.
    • (2004) Clin Exp Immunol , vol.137 , pp. 52-58
    • Nakamura, Y.1    Nosaka, S.2    Suzuki, M.3    Nagafuchi, S.4    Takahashi, T.5    Yajima, T.6
  • 97
    • 0033386877 scopus 로고    scopus 로고
    • An increase in histone acetylation and IL-2 antagonizing the immunoinhibitory effect are necessary for augmentation by butyrate of in vitro anti-TNP antibody production
    • Okamura T, Gohda E, Kohge T, Yamamoto I. An increase in histone acetylation and IL-2 antagonizing the immunoinhibitory effect are necessary for augmentation by butyrate of in vitro anti-TNP antibody production. Biol Pharm Bull 1999; 22:1288–92.
    • (1999) Biol Pharm Bull , vol.22 , pp. 1288-1292
    • Okamura, T.1    Gohda, E.2    Kohge, T.3    Yamamoto, I.4
  • 98
    • 84979735744 scopus 로고    scopus 로고
    • Gut microbial metabolites fuel host antibody responses
    • Kim M, Qie Y, Park J, Kim CH. Gut microbial metabolites fuel host antibody responses. Cell Host Microbe 2016; 20:202–14.
    • (2016) Cell Host Microbe , vol.20 , pp. 202-214
    • Kim, M.1    Qie, Y.2    Park, J.3    Kim, C.H.4
  • 99
    • 70350666634 scopus 로고    scopus 로고
    • Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43
    • 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; 461:1282–6.
    • (2009) Nature , vol.461 , pp. 1282-1286
    • Maslowski, K.M.1    Vieira, A.T.2    Ng, A.3    Kranich, J.4    Sierro, F.5    Yu, D.6
  • 100
    • 73349123182 scopus 로고    scopus 로고
    • G protein-coupled receptor 43 is essential for neutrophil recruitment during intestinal inflammation
    • Sina C, Gavrilova O, Forster M, Till A, Derer S, Hildebrand F et al. G protein-coupled receptor 43 is essential for neutrophil recruitment during intestinal inflammation. J Immunol 2009; 183:7514–22.
    • (2009) J Immunol , vol.183 , pp. 7514-7522
    • Sina, C.1    Gavrilova, O.2    Forster, M.3    Till, A.4    Derer, S.5    Hildebrand, F.6
  • 102
    • 77955472405 scopus 로고    scopus 로고
    • Effect of butyrate enemas on inflammation and antioxidant status in the colonic mucosa of patients with ulcerative colitis in remission
    • Hamer HM, Jonkers DM, Vanhoutvin SA, Troost FJ, Rijkers G, de Bruine A et al. Effect of butyrate enemas on inflammation and antioxidant status in the colonic mucosa of patients with ulcerative colitis in remission. Clin Nutr 2010; 29:738–44.
    • (2010) Clin Nutr , vol.29 , pp. 738-744
    • Hamer, H.M.1    Jonkers, D.M.2    Vanhoutvin, S.A.3    Troost, F.J.4    Rijkers, G.5    de Bruine, A.6
  • 103
    • 0036843980 scopus 로고    scopus 로고
    • Short-chain fatty acid enemas fail to decrease colonic hypersensitivity and inflammation in TNBS-induced colonic inflammation in rats
    • Tarrerias AL, Millecamps M, Alloui A, Beaughard C, Kemeny JL, Bourdu S et al. Short-chain fatty acid enemas fail to decrease colonic hypersensitivity and inflammation in TNBS-induced colonic inflammation in rats. Pain 2002; 100:91–7.
    • (2002) Pain , vol.100 , pp. 91-97
    • Tarrerias, A.L.1    Millecamps, M.2    Alloui, A.3    Beaughard, C.4    Kemeny, J.L.5    Bourdu, S.6
  • 104
    • 84975168249 scopus 로고    scopus 로고
    • Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways
    • Tan J, McKenzie C, Vuillermin PJ, Goverse G, Vinuesa CG, Mebius RE et al. Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways. Cell Rep 2016; 15:2809–24.
    • (2016) Cell Rep , vol.15 , pp. 2809-2824
    • Tan, J.1    McKenzie, C.2    Vuillermin, P.J.3    Goverse, G.4    Vinuesa, C.G.5    Mebius, R.E.6
  • 105
    • 85016122461 scopus 로고    scopus 로고
    • Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes
    • Marino E, Richards JL, McLeod KH, Stanley D, Yap YA, Knight J et al. Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes. Nat Immunol 2017; 18:552–62.
    • (2017) Nat Immunol , vol.18 , pp. 552-562
    • Marino, E.1    Richards, J.L.2    McLeod, K.H.3    Stanley, D.4    Yap, Y.A.5    Knight, J.6
  • 107
    • 84962022192 scopus 로고    scopus 로고
    • Chronically elevated levels of short-chain fatty acids induce T cell-mediated ureteritis and hydronephrosis
    • Park J, Goergen CJ, HogenEsch H, Kim CH. Chronically elevated levels of short-chain fatty acids induce T cell-mediated ureteritis and hydronephrosis. J Immunol 2016; 196:2388–400.
    • (2016) J Immunol , vol.196 , pp. 2388-2400
    • Park, J.1    Goergen, C.J.2    HogenEsch, H.3    Kim, C.H.4
  • 109
    • 42649095378 scopus 로고    scopus 로고
    • Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor
    • Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, Bettelli E et al. Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature 2008; 453:65–71.
    • (2008) Nature , vol.453 , pp. 65-71
    • Quintana, F.J.1    Basso, A.S.2    Iglesias, A.H.3    Korn, T.4    Farez, M.F.5    Bettelli, E.6
  • 110
    • 84864146433 scopus 로고    scopus 로고
    • Aiolos promotes TH17 differentiation by directly silencing Il2 expression
    • Quintana FJ, Jin H, Burns EJ, Nadeau M, Yeste A, Kumar D et al. Aiolos promotes TH17 differentiation by directly silencing Il2 expression. Nat Immunol 2012; 13:770–7.
    • (2012) Nat Immunol , vol.13 , pp. 770-777
    • Quintana, F.J.1    Jin, H.2    Burns, E.J.3    Nadeau, M.4    Yeste, A.5    Kumar, D.6
  • 111
    • 47749107262 scopus 로고    scopus 로고
    • Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells
    • Kimura A, Naka T, Nohara K, Fujii-Kuriyama Y, Kishimoto T. Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells. Proc Natl Acad Sci USA 2008; 105:9721–6.
    • (2008) Proc Natl Acad Sci USA , vol.105 , pp. 9721-9726
    • Kimura, A.1    Naka, T.2    Nohara, K.3    Fujii-Kuriyama, Y.4    Kishimoto, T.5
  • 112
    • 77955903212 scopus 로고    scopus 로고
    • The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27
    • Apetoh L, Quintana FJ, Pot C, Joller N, Xiao S, Kumar D et al. The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat Immunol 2010; 11:854–61.
    • (2010) Nat Immunol , vol.11 , pp. 854-861
    • Apetoh, L.1    Quintana, F.J.2    Pot, C.3    Joller, N.4    Xiao, S.5    Kumar, D.6
  • 113
    • 85031751449 scopus 로고    scopus 로고
    • Aryl hydrocarbon receptor controls monocyte differentiation into dendritic cells versus macrophages
    • Goudot C, Coillard A, Villani AC, Gueguen P, Cros A, Sarkizova S et al. Aryl hydrocarbon receptor controls monocyte differentiation into dendritic cells versus macrophages. Immunity 2017; 47:e6.
    • (2017) Immunity , vol.47
    • Goudot, C.1    Coillard, A.2    Villani, A.C.3    Gueguen, P.4    Cros, A.5    Sarkizova, S.6
  • 114
    • 84957625583 scopus 로고    scopus 로고
    • Dietary indoles suppress delayed-type hypersensitivity by inducing a switch from proinflammatory Th17 cells to anti-inflammatory regulatory T cells through regulation of MicroRNA
    • Singh NP, Singh UP, Rouse M, Zhang J, Chatterjee S, Nagarkatti PS et al. Dietary indoles suppress delayed-type hypersensitivity by inducing a switch from proinflammatory Th17 cells to anti-inflammatory regulatory T cells through regulation of MicroRNA. J Immunol 2016; 196:1108–22.
    • (2016) J Immunol , vol.196 , pp. 1108-1122
    • Singh, N.P.1    Singh, U.P.2    Rouse, M.3    Zhang, J.4    Chatterjee, S.5    Nagarkatti, P.S.6
  • 115
    • 78650301414 scopus 로고    scopus 로고
    • Elevated epidermal ornithine decarboxylase activity suppresses contact hypersensitivity
    • Keough MP, Hayes CS, DeFeo K, Gilmour SK. Elevated epidermal ornithine decarboxylase activity suppresses contact hypersensitivity. J Invest Dermatol 2011; 131:158–66.
    • (2011) J Invest Dermatol , vol.131 , pp. 158-166
    • Keough, M.P.1    Hayes, C.S.2    DeFeo, K.3    Gilmour, S.K.4
  • 117
    • 0034242623 scopus 로고    scopus 로고
    • Spermine differentially regulates the production of interleukin-12 p40 and interleukin-10 and suppresses the release of the T helper 1 cytokine interferon-gamma
    • Hasko G, Kuhel DG, Marton A, Nemeth ZH, Deitch EA, Szabo C. Spermine differentially regulates the production of interleukin-12 p40 and interleukin-10 and suppresses the release of the T helper 1 cytokine interferon-gamma. Shock 2000; 14:144–9.
    • (2000) Shock , vol.14 , pp. 144-149
    • Hasko, G.1    Kuhel, D.G.2    Marton, A.3    Nemeth, Z.H.4    Deitch, E.A.5    Szabo, C.6
  • 119
    • 84925500413 scopus 로고    scopus 로고
    • Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile
    • Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 2015; 517:205–8.
    • (2015) Nature , vol.517 , pp. 205-208
    • Buffie, C.G.1    Bucci, V.2    Stein, R.R.3    McKenney, P.T.4    Ling, L.5    Gobourne, A.6
  • 120
    • 84879888338 scopus 로고    scopus 로고
    • Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome
    • Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013; 499:97–101.
    • (2013) Nature , vol.499 , pp. 97-101
    • Yoshimoto, S.1    Loo, T.M.2    Atarashi, K.3    Kanda, H.4    Sato, S.5    Oyadomari, S.6
  • 121
    • 85023208268 scopus 로고    scopus 로고
    • The bile acid receptor GPBAR1 regulates the M1/M2 phenotype of intestinal macrophages and activation of GPBAR1 rescues mice from murine colitis
    • Biagioli M, Carino A, Cipriani S, Francisci D, Marchiano S, Scarpelli P et al. The bile acid receptor GPBAR1 regulates the M1/M2 phenotype of intestinal macrophages and activation of GPBAR1 rescues mice from murine colitis. J Immunol 2017; 199:718–33.
    • (2017) J Immunol , vol.199 , pp. 718-733
    • Biagioli, M.1    Carino, A.2    Cipriani, S.3    Francisci, D.4    Marchiano, S.5    Scarpelli, P.6
  • 122
    • 84872852300 scopus 로고    scopus 로고
    • The bile acid sensor FXR is required for immune-regulatory activities of TLR-9 in intestinal inflammation
    • Renga B, Mencarelli A, Cipriani S, D'Amore C, Carino A, Bruno A et al. The bile acid sensor FXR is required for immune-regulatory activities of TLR-9 in intestinal inflammation. PLoS One 2013; 8:e54472.
    • (2013) PLoS One , vol.8
    • Renga, B.1    Mencarelli, A.2    Cipriani, S.3    D'Amore, C.4    Carino, A.5    Bruno, A.6
  • 123
    • 34147170945 scopus 로고    scopus 로고
    • Pregnane X receptor activation ameliorates DSS-induced inflammatory bowel disease via inhibition of NF-kappaB target gene expression
    • Shah YM, Ma X, Morimura K, Kim I, Gonzalez FJ. Pregnane X receptor activation ameliorates DSS-induced inflammatory bowel disease via inhibition of NF-kappaB target gene expression. Am J Physiol Gastrointest Liver Physiol 2007; 292:G1114–22.
    • (2007) Am J Physiol Gastrointest Liver Physiol , vol.292 , pp. G1114-G1122
    • Shah, Y.M.1    Ma, X.2    Morimura, K.3    Kim, I.4    Gonzalez, F.J.5
  • 124
    • 84876041065 scopus 로고    scopus 로고
    • TGR5 signalling inhibits the production of pro-inflammatory cytokines by in vitro differentiated inflammatory and intestinal macrophages in Crohn's disease
    • Yoneno K, Hisamatsu T, Shimamura K, Kamada N, Ichikawa R, Kitazume MT et al. TGR5 signalling inhibits the production of pro-inflammatory cytokines by in vitro differentiated inflammatory and intestinal macrophages in Crohn's disease. Immunology 2013; 139:19–29.
    • (2013) Immunology , vol.139 , pp. 19-29
    • Yoneno, K.1    Hisamatsu, T.2    Shimamura, K.3    Kamada, N.4    Ichikawa, R.5    Kitazume, M.T.6
  • 125
    • 84860253913 scopus 로고    scopus 로고
    • Bile acids induce monocyte differentiation toward interleukin-12 hypo-producing dendritic cells via a TGR5-dependent pathway
    • Ichikawa R, Takayama T, Yoneno K, Kamada N, Kitazume MT, Higuchi H et al. Bile acids induce monocyte differentiation toward interleukin-12 hypo-producing dendritic cells via a TGR5-dependent pathway. Immunology 2012; 136:153–62.
    • (2012) Immunology , vol.136 , pp. 153-162
    • Ichikawa, R.1    Takayama, T.2    Yoneno, K.3    Kamada, N.4    Kitazume, M.T.5    Higuchi, H.6
  • 126
    • 57249104897 scopus 로고    scopus 로고
    • A rat model of chronic postinflammatory visceral pain induced by deoxycholic acid
    • Traub RJ, Tang B, Ji Y, Pandya S, Yfantis H, Sun Y. A rat model of chronic postinflammatory visceral pain induced by deoxycholic acid. Gastroenterology 2008; 135:2075–83.
    • (2008) Gastroenterology , vol.135 , pp. 2075-2083
    • Traub, R.J.1    Tang, B.2    Ji, Y.3    Pandya, S.4    Yfantis, H.5    Sun, Y.6
  • 128
    • 85006100780 scopus 로고    scopus 로고
    • Deoxycholic acid triggers NLRP3 inflammasome activation and aggravates DSS-induced colitis in mice
    • Zhao S, Gong Z, Zhou J, Tian C, Gao Y, Xu C et al. Deoxycholic acid triggers NLRP3 inflammasome activation and aggravates DSS-induced colitis in mice. Front Immunol 2016; 7:536.
    • (2016) Front Immunol , vol.7 , pp. 536
    • Zhao, S.1    Gong, Z.2    Zhou, J.3    Tian, C.4    Gao, Y.5    Xu, C.6
  • 129
    • 84994887772 scopus 로고    scopus 로고
    • Bile acids control inflammation and metabolic disorder through inhibition of NLRP3 inflammasome
    • Guo C, Xie S, Chi Z, Zhang J, Liu Y, Zhang L et al. Bile acids control inflammation and metabolic disorder through inhibition of NLRP3 inflammasome. Immunity 2016; 45:944.
    • (2016) Immunity , vol.45 , pp. 944
    • Guo, C.1    Xie, S.2    Chi, Z.3    Zhang, J.4    Liu, Y.5    Zhang, L.6
  • 130
    • 33746701380 scopus 로고    scopus 로고
    • P2X receptors as cell-surface ATP sensors in health and disease
    • Khakh BS, North RA. P2X receptors as cell-surface ATP sensors in health and disease. Nature 2006; 442:527–32.
    • (2006) Nature , vol.442 , pp. 527-532
    • Khakh, B.S.1    North, R.A.2
  • 132
    • 67649365966 scopus 로고    scopus 로고
    • Autocrine regulation of T-cell activation by ATP release and P2X7 receptors
    • Yip L, Woehrle T, Corriden R, Hirsh M, Chen Y, Inoue Y et al. Autocrine regulation of T-cell activation by ATP release and P2X7 receptors. FASEB J 2009; 23:1685–93.
    • (2009) FASEB J , vol.23 , pp. 1685-1693
    • Yip, L.1    Woehrle, T.2    Corriden, R.3    Hirsh, M.4    Chen, Y.5    Inoue, Y.6
  • 133
    • 85020908467 scopus 로고    scopus 로고
    • Contraction of intestinal effector T cells by retinoic acid-induced purinergic receptor P2X7
    • Hashimoto-Hill S, Friesen L, Kim M, Kim CH. Contraction of intestinal effector T cells by retinoic acid-induced purinergic receptor P2X7. Mucosal Immunol 2017; 10:912–23.
    • (2017) Mucosal Immunol , vol.10 , pp. 912-923
    • Hashimoto-Hill, S.1    Friesen, L.2    Kim, M.3    Kim, C.H.4


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.