Skip to main content
Erschienen in: Seminars in Immunopathology 2/2018

06.11.2017 | Review

Dietary and metabolic modulators of hepatic immunity

verfasst von: Antonella Carambia, Johannes Herkel

Erschienen in: Seminars in Immunopathology | Ausgabe 2/2018

Einloggen, um Zugang zu erhalten

Abstract

The liver is the central metabolic organ of the organism and is thus constantly exposed to gut-derived dietary and microbial antigens. The liver maintains homoeostatic tolerance to these mostly harmless antigens. However, the liver also functions as a barrier organ to harmful pathogens and is thus permissive to liver inflammation. The regulation of the delicate balance between liver tolerance and liver inflammation is of vital importance for the organism. In recent years, a general role for dietary components and metabolites as immune mediators has been emerging. However, although the liver is exposed to a great deal of metabolic mediators, surprisingly, little is known about their actual role in the regulation of hepatic immune responses. Here, we will explore the possible impacts of metabolic mediators for homoeostatic and pathological immunity in the liver, by highlighting selected examples of metabolic immune regulation in the liver.
Literatur
1.
Zurück zum Zitat Wisse E, De Zanger RB, Charels K, Van Der Smissen P, McCuskey RS (1985) The liver sieve: considerations concerning the structure and function of endothelial fenestrae, the sinusoidal wall and the space of Disse. Hepatology 5:683–692PubMedCrossRef Wisse E, De Zanger RB, Charels K, Van Der Smissen P, McCuskey RS (1985) The liver sieve: considerations concerning the structure and function of endothelial fenestrae, the sinusoidal wall and the space of Disse. Hepatology 5:683–692PubMedCrossRef
2.
Zurück zum Zitat Balmer ML, Slack E, de Gottardi A, Lawson MA, Hapfelmeier S, Miele L, Grieco A, Van Vlierberghe H, Fahrner R, Patuto N, Bernsmeier C, Ronchi F, Wyss M, Stroka D, Dickgreber N, Heim MH, McCoy KD, Macpherson AJ (2014) The liver may act as a firewall mediating mutualism between the host and its gut commensal microbiota. Sci Transl Med 6:237ra66PubMedCrossRef Balmer ML, Slack E, de Gottardi A, Lawson MA, Hapfelmeier S, Miele L, Grieco A, Van Vlierberghe H, Fahrner R, Patuto N, Bernsmeier C, Ronchi F, Wyss M, Stroka D, Dickgreber N, Heim MH, McCoy KD, Macpherson AJ (2014) The liver may act as a firewall mediating mutualism between the host and its gut commensal microbiota. Sci Transl Med 6:237ra66PubMedCrossRef
3.
4.
Zurück zum Zitat Chen Y, Jiang G, Yang HR, Gu X, Wang L, Hsieh CC, Chou HS, Fung JJ, Qian S, Lu L (2009) Distinct response of liver myeloid dendritic cells to endotoxin is mediated by IL-27. J Hepatol 51:510–519PubMedPubMedCentralCrossRef Chen Y, Jiang G, Yang HR, Gu X, Wang L, Hsieh CC, Chou HS, Fung JJ, Qian S, Lu L (2009) Distinct response of liver myeloid dendritic cells to endotoxin is mediated by IL-27. J Hepatol 51:510–519PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Uhrig A, Banafsche R, Kremer M, Hegenbarth S, Hamann A, Neurath M, Gerken G, Limmer A, Knolle PA (2005) Development and functional consequences of LPS tolerance in sinusoidal endothelial cells of the liver. J Leukoc Biol 77:626–633PubMedCrossRef Uhrig A, Banafsche R, Kremer M, Hegenbarth S, Hamann A, Neurath M, Gerken G, Limmer A, Knolle PA (2005) Development and functional consequences of LPS tolerance in sinusoidal endothelial cells of the liver. J Leukoc Biol 77:626–633PubMedCrossRef
6.
Zurück zum Zitat Knolle PA, Löser E, Protzer U, Duchmann R, Schmitt E, Zum Büschenfelde KH, Rose-John S, Gerken G (1997) Regulation of endotoxin-induced IL-6 production in liver sinusoidal endothelial cells and Kupffer cells by IL-10. Clin Exp Immunol 107:555–561PubMedPubMedCentralCrossRef Knolle PA, Löser E, Protzer U, Duchmann R, Schmitt E, Zum Büschenfelde KH, Rose-John S, Gerken G (1997) Regulation of endotoxin-induced IL-6 production in liver sinusoidal endothelial cells and Kupffer cells by IL-10. Clin Exp Immunol 107:555–561PubMedPubMedCentralCrossRef
7.
8.
Zurück zum Zitat Thomson AW, Knolle PA (2010) Antigen-presenting cell function in the tolerogenic liver environment. Nat Rev Immunol 10:753–766PubMedCrossRef Thomson AW, Knolle PA (2010) Antigen-presenting cell function in the tolerogenic liver environment. Nat Rev Immunol 10:753–766PubMedCrossRef
10.
11.
12.
Zurück zum Zitat Knolle PA, Uhrig A, Protzer U, Trippler M, Duchmann R, Meyer zum Büschenfelde KH, Gerken G (1998) Interleukin-10 expression is autoregulated at the transcriptional level in human and murine Kupffer cells. Hepatology 27:93–99PubMedCrossRef Knolle PA, Uhrig A, Protzer U, Trippler M, Duchmann R, Meyer zum Büschenfelde KH, Gerken G (1998) Interleukin-10 expression is autoregulated at the transcriptional level in human and murine Kupffer cells. Hepatology 27:93–99PubMedCrossRef
13.
Zurück zum Zitat Bissell DM, Wang SS, Jarnagin WR, Roll FJ (1995) Cell-specific expression of transforming growth factor-beta in rat liver. Evidence for autocrine regulation of hepatocyte proliferation. J Clin Invest 96:447–455PubMedPubMedCentralCrossRef Bissell DM, Wang SS, Jarnagin WR, Roll FJ (1995) Cell-specific expression of transforming growth factor-beta in rat liver. Evidence for autocrine regulation of hepatocyte proliferation. J Clin Invest 96:447–455PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Breous E, Somanathan S, Vandenberghe LH, Wilson JM (2009) Hepatic regulatory T cells and Kupffer cells are crucial mediators of systemic T cell tolerance to antigens targeting murine liver. Hepatology 50:612–621PubMedPubMedCentralCrossRef Breous E, Somanathan S, Vandenberghe LH, Wilson JM (2009) Hepatic regulatory T cells and Kupffer cells are crucial mediators of systemic T cell tolerance to antigens targeting murine liver. Hepatology 50:612–621PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Heymann F, Peusquens J, Ludwig-Portugall I, Kohlhepp M, Ergen C, Niemietz P, Martin C, van Rooijen N, Ochando JC, Randolph GJ, Luedde T, Ginhoux F, Kurts C, Trautwein C, Tacke F (2015) Liver inflammation abrogates immunological tolerance induced by Kupffer cells. Hepatology 62:279–291PubMedCrossRef Heymann F, Peusquens J, Ludwig-Portugall I, Kohlhepp M, Ergen C, Niemietz P, Martin C, van Rooijen N, Ochando JC, Randolph GJ, Luedde T, Ginhoux F, Kurts C, Trautwein C, Tacke F (2015) Liver inflammation abrogates immunological tolerance induced by Kupffer cells. Hepatology 62:279–291PubMedCrossRef
16.
Zurück zum Zitat Lee WY, Moriarty TJ, Wong CH, Zhou H, Strieter RM, van Rooijen N, Chaconas G, Kubes P (2010) An intravascular immune response to Borrelia burgdorferi involves Kupffer cells and iNKT cells. Nat Immunol 11:295–302PubMedPubMedCentralCrossRef Lee WY, Moriarty TJ, Wong CH, Zhou H, Strieter RM, van Rooijen N, Chaconas G, Kubes P (2010) An intravascular immune response to Borrelia burgdorferi involves Kupffer cells and iNKT cells. Nat Immunol 11:295–302PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Sørensen KK, McCourt P, Berg T, Crossley C, Le Couteur D, Wake K, Smedsrød B (2012) The scavenger endothelial cell: a new player in homeostasis and immunity. Am J Physiol Regul Integr Comp Physiol 303:R1217–R1230PubMedCrossRef Sørensen KK, McCourt P, Berg T, Crossley C, Le Couteur D, Wake K, Smedsrød B (2012) The scavenger endothelial cell: a new player in homeostasis and immunity. Am J Physiol Regul Integr Comp Physiol 303:R1217–R1230PubMedCrossRef
18.
Zurück zum Zitat Ganesan LP, Kim J, Wu Y, Mohanty S, Phillips GS, Birmingham DJ, Robinson JM, Anderson CL (2012) FcγRIIb on liver sinusoidal endothelium clears small immune complexes. J Immunol 189:4981–4988PubMedPubMedCentralCrossRef Ganesan LP, Kim J, Wu Y, Mohanty S, Phillips GS, Birmingham DJ, Robinson JM, Anderson CL (2012) FcγRIIb on liver sinusoidal endothelium clears small immune complexes. J Immunol 189:4981–4988PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Schölzel K, Schildberg FA, Welz M, Börner C, Geiger S, Kurts C, Heikenwälder M, Knolle PA, Wohlleber D (2014) Transfer of MHC-class-I molecules among liver sinusoidal cells facilitates hepatic immune surveillance. J Hepatol 61:600–608PubMedCrossRef Schölzel K, Schildberg FA, Welz M, Börner C, Geiger S, Kurts C, Heikenwälder M, Knolle PA, Wohlleber D (2014) Transfer of MHC-class-I molecules among liver sinusoidal cells facilitates hepatic immune surveillance. J Hepatol 61:600–608PubMedCrossRef
20.
Zurück zum Zitat Carambia A, Herkel J (2014) Liver sinusoidal cells collecting MHC-I molecules: you can’t get enough of a good thing. J Hepatol 61:464–465PubMedCrossRef Carambia A, Herkel J (2014) Liver sinusoidal cells collecting MHC-I molecules: you can’t get enough of a good thing. J Hepatol 61:464–465PubMedCrossRef
21.
Zurück zum Zitat Lohse AW, Knolle PA, Bilo K, Uhrig A, Waldmann C, Ibe M, Schmitt E, Gerken G, Meyer zum Büschenfelde KH (1996) Antigen-presenting function and B7 expression of murine sinusoidal endothelial cells and Kupffer cells. Gastroenterology 110:1175–1181PubMedCrossRef Lohse AW, Knolle PA, Bilo K, Uhrig A, Waldmann C, Ibe M, Schmitt E, Gerken G, Meyer zum Büschenfelde KH (1996) Antigen-presenting function and B7 expression of murine sinusoidal endothelial cells and Kupffer cells. Gastroenterology 110:1175–1181PubMedCrossRef
22.
Zurück zum Zitat Schurich A, Böttcher JP, Burgdorf S, Penzler P, Hegenbarth S, Kern M, Dolf A, Endl E, Schultze J, Wiertz E, Stabenow D, Kurts C, Knolle P (2009) Distinct kinetics and dynamics of cross-presentation in liver sinusoidal endothelial cells compared to dendritic cells. Hepatology 50:909–919PubMedCrossRef Schurich A, Böttcher JP, Burgdorf S, Penzler P, Hegenbarth S, Kern M, Dolf A, Endl E, Schultze J, Wiertz E, Stabenow D, Kurts C, Knolle P (2009) Distinct kinetics and dynamics of cross-presentation in liver sinusoidal endothelial cells compared to dendritic cells. Hepatology 50:909–919PubMedCrossRef
23.
Zurück zum Zitat Knolle PA, Schmitt E, Jin S, Germann T, Duchmann R, Hegenbarth S, Gerken G, Lohse AW (1999) Induction of cytokine production in naive CD4(+) T cells by antigen-presenting murine liver sinusoidal endothelial cells but failure to induce differentiation toward Th1 cells. Gastroenterology 116:1428–1440PubMedCrossRef Knolle PA, Schmitt E, Jin S, Germann T, Duchmann R, Hegenbarth S, Gerken G, Lohse AW (1999) Induction of cytokine production in naive CD4(+) T cells by antigen-presenting murine liver sinusoidal endothelial cells but failure to induce differentiation toward Th1 cells. Gastroenterology 116:1428–1440PubMedCrossRef
24.
Zurück zum Zitat Diehl L, Schurich A, Grochtmann R, Hegenbarth S, Chen L, Knolle PA (2008) Tolerogenic maturation of liver sinusoidal endothelial cells promotes B7-homolog 1-dependent CD8+ T cell tolerance. Hepatology 47:296–305PubMedCrossRef Diehl L, Schurich A, Grochtmann R, Hegenbarth S, Chen L, Knolle PA (2008) Tolerogenic maturation of liver sinusoidal endothelial cells promotes B7-homolog 1-dependent CD8+ T cell tolerance. Hepatology 47:296–305PubMedCrossRef
25.
Zurück zum Zitat Carambia A, Frenzel C, Bruns OT, Schwinge D, Reimer R, Hohenberg H, Huber S, Tiegs G, Schramm C, Lohse AW, Herkel J (2013) Inhibition of inflammatory CD4 T cell activity by murine liver sinusoidal endothelial cells. J Hepatol 58:112–118PubMedCrossRef Carambia A, Frenzel C, Bruns OT, Schwinge D, Reimer R, Hohenberg H, Huber S, Tiegs G, Schramm C, Lohse AW, Herkel J (2013) Inhibition of inflammatory CD4 T cell activity by murine liver sinusoidal endothelial cells. J Hepatol 58:112–118PubMedCrossRef
26.
Zurück zum Zitat Limmer A, Ohl J, Kurts C, Ljunggren HG, Reiss Y, Groettrup M, Momburg F, Arnold B, Knolle PA (2000) Efficient presentation of exogenous antigen by liver endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance. Nat Med 6:1348–1354PubMedCrossRef Limmer A, Ohl J, Kurts C, Ljunggren HG, Reiss Y, Groettrup M, Momburg F, Arnold B, Knolle PA (2000) Efficient presentation of exogenous antigen by liver endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance. Nat Med 6:1348–1354PubMedCrossRef
27.
Zurück zum Zitat Carambia A, Freund B, Schwinge D, Heine M, Laschtowitz A, Huber S, Wraith DC, Korn T, Schramm C, Lohse AW, Heeren J, Herkel J (2014) TGF-β-dependent induction of CD4+ CD25+ Foxp3+ Tregs by liver sinusoidal endothelial cells. J Hepatol 61:594–599PubMedCrossRef Carambia A, Freund B, Schwinge D, Heine M, Laschtowitz A, Huber S, Wraith DC, Korn T, Schramm C, Lohse AW, Heeren J, Herkel J (2014) TGF-β-dependent induction of CD4+ CD25+ Foxp3+ Tregs by liver sinusoidal endothelial cells. J Hepatol 61:594–599PubMedCrossRef
28.
Zurück zum Zitat Wing K, Sakaguchi S (2010) Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat Immunol 11:7–13PubMedCrossRef Wing K, Sakaguchi S (2010) Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat Immunol 11:7–13PubMedCrossRef
29.
Zurück zum Zitat Carambia A, Freund B, Schwinge D, Bruns OT, Salmen SC, Ittrich H, Reimer R, Heine M, Huber S, Waurisch C, Eychmüller A, Wraith DC, Korn T, Nielsen P, Weller H, Schramm C, Lüth S, Lohse AW, Heeren J, Herkel J (2015) Nanoparticle-based autoantigen delivery to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity in mice. J Hepatol 62:1349–1356PubMedCrossRef Carambia A, Freund B, Schwinge D, Bruns OT, Salmen SC, Ittrich H, Reimer R, Heine M, Huber S, Waurisch C, Eychmüller A, Wraith DC, Korn T, Nielsen P, Weller H, Schramm C, Lüth S, Lohse AW, Heeren J, Herkel J (2015) Nanoparticle-based autoantigen delivery to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity in mice. J Hepatol 62:1349–1356PubMedCrossRef
30.
Zurück zum Zitat Kern M, Popov A, Scholz K, Schumak B, Djandji D, Limmer A, Eggle D, Sacher T, Zawatzky R, Holtappels R, Reddehase MJ, Hartmann G, Debey-Pascher S, Diehl L, Kalinke U, Koszinowski U, Schultze J, Knolle PA (2010) Virally infected mouse liver endothelial cells trigger CD8+ T-cell immunity. Gastroenterology 138:336–346PubMedCrossRef Kern M, Popov A, Scholz K, Schumak B, Djandji D, Limmer A, Eggle D, Sacher T, Zawatzky R, Holtappels R, Reddehase MJ, Hartmann G, Debey-Pascher S, Diehl L, Kalinke U, Koszinowski U, Schultze J, Knolle PA (2010) Virally infected mouse liver endothelial cells trigger CD8+ T-cell immunity. Gastroenterology 138:336–346PubMedCrossRef
31.
Zurück zum Zitat Liu J, Jiang M, Ma Z, Dietze KK, Zelinskyy G, Yang D, Dittmer U, Schlaak JF, Roggendorf M, Lu M (2013) TLR1/2 ligand-stimulated mouse liver endothelial cells secrete IL-12 and trigger CD8+ T cell immunity in vitro. J Immunol 191:6178–6190PubMedCrossRef Liu J, Jiang M, Ma Z, Dietze KK, Zelinskyy G, Yang D, Dittmer U, Schlaak JF, Roggendorf M, Lu M (2013) TLR1/2 ligand-stimulated mouse liver endothelial cells secrete IL-12 and trigger CD8+ T cell immunity in vitro. J Immunol 191:6178–6190PubMedCrossRef
32.
Zurück zum Zitat Wohlleber D, Kashkar H, Gärtner K, Frings MK, Odenthal M, Hegenbarth S, Börner C, Arnold B, Hämmerling G, Nieswandt B, van Rooijen N, Limmer A, Cederbrant K, Heikenwalder M, Pasparakis M, Protzer U, Dienes HP, Kurts C, Krönke M, Knolle PA (2012) TNF-induced target cell killing by CTL activated through cross-presentation. Cell Rep 2:478–487PubMedCrossRef Wohlleber D, Kashkar H, Gärtner K, Frings MK, Odenthal M, Hegenbarth S, Börner C, Arnold B, Hämmerling G, Nieswandt B, van Rooijen N, Limmer A, Cederbrant K, Heikenwalder M, Pasparakis M, Protzer U, Dienes HP, Kurts C, Krönke M, Knolle PA (2012) TNF-induced target cell killing by CTL activated through cross-presentation. Cell Rep 2:478–487PubMedCrossRef
33.
Zurück zum Zitat Böttcher JP, Schanz O, Wohlleber D, Abdullah Z, Debey-Pascher S, Staratschek-Jox A, Höchst B, Hegenbarth S, Grell J, Limmer A, Atreya I, Neurath MF, Busch DH, Schmitt E, van Endert P, Kolanus W, Kurts C, Schultze JL, Diehl L, Knolle PA (2013) Liver-primed memory T cells generated under noninflammatory conditions provide anti-infectious immunity. Cell Rep 3:779–795PubMedCrossRef Böttcher JP, Schanz O, Wohlleber D, Abdullah Z, Debey-Pascher S, Staratschek-Jox A, Höchst B, Hegenbarth S, Grell J, Limmer A, Atreya I, Neurath MF, Busch DH, Schmitt E, van Endert P, Kolanus W, Kurts C, Schultze JL, Diehl L, Knolle PA (2013) Liver-primed memory T cells generated under noninflammatory conditions provide anti-infectious immunity. Cell Rep 3:779–795PubMedCrossRef
35.
Zurück zum Zitat Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V (2011) Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immunol 29:163–183PubMedPubMedCentralCrossRef Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V (2011) Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immunol 29:163–183PubMedPubMedCentralCrossRef
36.
37.
Zurück zum Zitat Knolle P, Schlaak J, Uhrig A, Kempf P, Meyer zum Büschenfelde KH, Gerken G (1995) Human Kupffer cells secrete IL-10 in response to lipopolysaccharide (LPS) challenge. J Hepatol 22:226–229PubMedCrossRef Knolle P, Schlaak J, Uhrig A, Kempf P, Meyer zum Büschenfelde KH, Gerken G (1995) Human Kupffer cells secrete IL-10 in response to lipopolysaccharide (LPS) challenge. J Hepatol 22:226–229PubMedCrossRef
38.
Zurück zum Zitat Schon HT, Weiskirchen R (2014) Immunomodulatory effects of transforming growth factor-β in the liver. Hepatobiliary Surg Nutr 3:386–406PubMedPubMedCentral Schon HT, Weiskirchen R (2014) Immunomodulatory effects of transforming growth factor-β in the liver. Hepatobiliary Surg Nutr 3:386–406PubMedPubMedCentral
39.
Zurück zum Zitat Bronte V, Serafini P, Mazzoni A, Segal DM, Zanovello P (2003) L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol 24:302–306PubMedCrossRef Bronte V, Serafini P, Mazzoni A, Segal DM, Zanovello P (2003) L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol 24:302–306PubMedCrossRef
41.
Zurück zum Zitat Cantor HM, Dumont AE (1967) Hepatic suppression of sensitization to antigen absorbed into the portal system. Nature 245:744–745CrossRef Cantor HM, Dumont AE (1967) Hepatic suppression of sensitization to antigen absorbed into the portal system. Nature 245:744–745CrossRef
42.
Zurück zum Zitat Calne RY, Sells RA, Pena JR, Davis DR, Millard PR, Herbertson BM, Binns RM, Davies DA (1969) Induction of immunological tolerance by porcine liver allografts. Nature 223:472–476PubMedCrossRef Calne RY, Sells RA, Pena JR, Davis DR, Millard PR, Herbertson BM, Binns RM, Davies DA (1969) Induction of immunological tolerance by porcine liver allografts. Nature 223:472–476PubMedCrossRef
43.
Zurück zum Zitat Sriwatanawongsa V, Davies HS, Calne RY (1995) The essential roles of parenchymal tissues and passenger leukocytes in the tolerance induced by liver grafting in rats. Nat Med 1:428–432PubMedCrossRef Sriwatanawongsa V, Davies HS, Calne RY (1995) The essential roles of parenchymal tissues and passenger leukocytes in the tolerance induced by liver grafting in rats. Nat Med 1:428–432PubMedCrossRef
44.
Zurück zum Zitat Lüth S, Huber S, Schramm C, Buch T, Zander S, Stadelmann C, Brück W, Wraith DC, Herkel J, Lohse AW (2008) Ectopic expression of neural autoantigen in mouse liver suppresses experimental autoimmune neuroinflammation by inducing antigen-specific Tregs. J Clin Invest 118:3403–3410PubMedPubMedCentral Lüth S, Huber S, Schramm C, Buch T, Zander S, Stadelmann C, Brück W, Wraith DC, Herkel J, Lohse AW (2008) Ectopic expression of neural autoantigen in mouse liver suppresses experimental autoimmune neuroinflammation by inducing antigen-specific Tregs. J Clin Invest 118:3403–3410PubMedPubMedCentral
45.
Zurück zum Zitat Luedde T, Kaplowitz N, Schwabe RF (2014) Cell death and cell death responses in liver disease: mechanisms and clinical relevance. Gastroenterology 147:765–783PubMedPubMedCentralCrossRef Luedde T, Kaplowitz N, Schwabe RF (2014) Cell death and cell death responses in liver disease: mechanisms and clinical relevance. Gastroenterology 147:765–783PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Knolle PA, Thimme R (2014) Hepatic immune regulation and its involvement in viral hepatitis infection. Gastroenterology 146:1193–1207PubMedCrossRef Knolle PA, Thimme R (2014) Hepatic immune regulation and its involvement in viral hepatitis infection. Gastroenterology 146:1193–1207PubMedCrossRef
47.
Zurück zum Zitat Huang LR, Wohlleber D, Reisinger F, Jenne CN, Cheng RL, Abdullah Z, Schildberg FA, Odenthal M, Dienes HP, van Rooijen N, Schmitt E, Garbi N, Croft M, Kurts C, Kubes P, Protzer U, Heikenwalder M, Knolle PA (2013) Intrahepatic myeloid-cell aggregates enable local proliferation of CD8(+) T cells and successful immunotherapy against chronic viral liver infection. Nat Immunol 14:574–583PubMedCrossRef Huang LR, Wohlleber D, Reisinger F, Jenne CN, Cheng RL, Abdullah Z, Schildberg FA, Odenthal M, Dienes HP, van Rooijen N, Schmitt E, Garbi N, Croft M, Kurts C, Kubes P, Protzer U, Heikenwalder M, Knolle PA (2013) Intrahepatic myeloid-cell aggregates enable local proliferation of CD8(+) T cells and successful immunotherapy against chronic viral liver infection. Nat Immunol 14:574–583PubMedCrossRef
48.
Zurück zum Zitat Jones GW, Jones SA (2016) Ectopic lymphoid follicles: inducible centres for generating antigen-specific immune responses within tissues. Immunology 147:141–151PubMedCrossRef Jones GW, Jones SA (2016) Ectopic lymphoid follicles: inducible centres for generating antigen-specific immune responses within tissues. Immunology 147:141–151PubMedCrossRef
49.
Zurück zum Zitat Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M (2016) Global epidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 64:73–84PubMedCrossRef Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M (2016) Global epidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 64:73–84PubMedCrossRef
50.
Zurück zum Zitat Petro TM, Bhattacharjee JK (1981) Effect of dietary essential amino acid limitations upon the susceptibility to Salmonella typhimurium and the effect upon humoral and cellular immune responses in mice. Infect Immun 32:251–259PubMedPubMedCentral Petro TM, Bhattacharjee JK (1981) Effect of dietary essential amino acid limitations upon the susceptibility to Salmonella typhimurium and the effect upon humoral and cellular immune responses in mice. Infect Immun 32:251–259PubMedPubMedCentral
51.
Zurück zum Zitat Tsukishiro T, Shimizu Y, Higuchi K, Watanabe A (2000) Effect of branched-chain amino acids on the composition and cytolytic activity of liver-associated lymphocytes in rats. J Gastroenterol Hepatol 15:849–859PubMedCrossRef Tsukishiro T, Shimizu Y, Higuchi K, Watanabe A (2000) Effect of branched-chain amino acids on the composition and cytolytic activity of liver-associated lymphocytes in rats. J Gastroenterol Hepatol 15:849–859PubMedCrossRef
52.
Zurück zum Zitat Nakamura I, Ochiai K, Imai Y, Moriyasu F, Imawari M (2007) Restoration of innate host defense responses by oral supplementation of branched-chain amino acids in decompensated cirrhotic patients. Hepatol Res 37:1062–1067PubMedCrossRef Nakamura I, Ochiai K, Imai Y, Moriyasu F, Imawari M (2007) Restoration of innate host defense responses by oral supplementation of branched-chain amino acids in decompensated cirrhotic patients. Hepatol Res 37:1062–1067PubMedCrossRef
53.
Zurück zum Zitat Barnes PF, Arevalo C, Chan LS, Wong SF, Reynolds TB (1988) A prospective evaluation of bacteremic patients with chronic liver disease. Hepatology 8:1099–1103PubMedCrossRef Barnes PF, Arevalo C, Chan LS, Wong SF, Reynolds TB (1988) A prospective evaluation of bacteremic patients with chronic liver disease. Hepatology 8:1099–1103PubMedCrossRef
54.
Zurück zum Zitat Nakamura I (2014) Impairment of innate immune responses in cirrhotic patients and treatment by branched-chain amino acids. World J Gastroenterol 20:7298–7305PubMedPubMedCentralCrossRef Nakamura I (2014) Impairment of innate immune responses in cirrhotic patients and treatment by branched-chain amino acids. World J Gastroenterol 20:7298–7305PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Navarro LA, Wree A, Povero D, Berk MP, Eguchi A, Ghosh S, Papouchado BG, Erzurum SC, Feldstein AE (2015) Arginase 2 deficiency results in spontaneous steatohepatitis: a novel link between innate immune activation and hepatic de novo lipogenesis. J Hepatol 62:412–420PubMedCrossRef Navarro LA, Wree A, Povero D, Berk MP, Eguchi A, Ghosh S, Papouchado BG, Erzurum SC, Feldstein AE (2015) Arginase 2 deficiency results in spontaneous steatohepatitis: a novel link between innate immune activation and hepatic de novo lipogenesis. J Hepatol 62:412–420PubMedCrossRef
56.
Zurück zum Zitat Liu C, Rajapakse AG, Riedo E, Fellay B, Bernhard MC, Montani JP, Yang Z, Ming XF (2016) Targeting arginase-II protects mice from high-fat-diet-induced hepatic steatosis through suppression of macrophage inflammation. Sci Rep 6:20405PubMedPubMedCentralCrossRef Liu C, Rajapakse AG, Riedo E, Fellay B, Bernhard MC, Montani JP, Yang Z, Ming XF (2016) Targeting arginase-II protects mice from high-fat-diet-induced hepatic steatosis through suppression of macrophage inflammation. Sci Rep 6:20405PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Scheja L, Kluwe J (2015) Arginine and NASH—do macrophages deliver the first hit? J Hepatol 62:260–261PubMedCrossRef Scheja L, Kluwe J (2015) Arginine and NASH—do macrophages deliver the first hit? J Hepatol 62:260–261PubMedCrossRef
58.
Zurück zum Zitat Bhatt S, Qin J, Bennett C, Qian S, Fung JJ, Hamilton TA, Lu L (2014) All-trans retinoic acid induces arginase-1 and inducible nitric oxide synthase-producing dendritic cells with T cell inhibitory function. J Immunol 192:5098–5108PubMedPubMedCentralCrossRef Bhatt S, Qin J, Bennett C, Qian S, Fung JJ, Hamilton TA, Lu L (2014) All-trans retinoic acid induces arginase-1 and inducible nitric oxide synthase-producing dendritic cells with T cell inhibitory function. J Immunol 192:5098–5108PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Stone TW, Stoy N, Darlington LG (2013) An expanding range of targets for kynurenine metabolites of tryptophan. Trends Pharmacol Sci 34:136–143PubMedCrossRef Stone TW, Stoy N, Darlington LG (2013) An expanding range of targets for kynurenine metabolites of tryptophan. Trends Pharmacol Sci 34:136–143PubMedCrossRef
60.
Zurück zum Zitat Ito H, Hoshi M, Ohtaki H, Taguchi A, Ando K, Ishikawa T, Osawa Y, Hara A, Moriwaki H, Saito K, Seishima M (2010) Ability of IDO to attenuate liver injury in alpha-galactosylceramide-induced hepatitis model. J Immunol 185:4554–4560PubMedCrossRef Ito H, Hoshi M, Ohtaki H, Taguchi A, Ando K, Ishikawa T, Osawa Y, Hara A, Moriwaki H, Saito K, Seishima M (2010) Ability of IDO to attenuate liver injury in alpha-galactosylceramide-induced hepatitis model. J Immunol 185:4554–4560PubMedCrossRef
61.
Zurück zum Zitat Ogiso H, Ito H, Ando T, Arioka Y, Kanbe A, Ando K, Ishikawa T, Saito K, Hara A, Moriwaki H, Shimizu M, Seishima M (2016) The deficiency of indoleamine 2,3-dioxygenase aggravates the CCl4-induced liver fibrosis in mice. PLoS One 11:e0162183PubMedPubMedCentralCrossRef Ogiso H, Ito H, Ando T, Arioka Y, Kanbe A, Ando K, Ishikawa T, Saito K, Hara A, Moriwaki H, Shimizu M, Seishima M (2016) The deficiency of indoleamine 2,3-dioxygenase aggravates the CCl4-induced liver fibrosis in mice. PLoS One 11:e0162183PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Cho NE, Bang BR, Gurung P, Li M, Clemens DL, Underhill TM, James LP, Chase JR, Saito T (2016) Retinoid regulation of antiviral innate immunity in hepatocytes. Hepatology 63:1783–1795PubMedPubMedCentralCrossRef Cho NE, Bang BR, Gurung P, Li M, Clemens DL, Underhill TM, James LP, Chase JR, Saito T (2016) Retinoid regulation of antiviral innate immunity in hepatocytes. Hepatology 63:1783–1795PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Kitson MT, Roberts SK (2012) D-livering the message: the importance of vitamin D status in chronic liver disease. J Hepatol 57:897–909PubMedCrossRef Kitson MT, Roberts SK (2012) D-livering the message: the importance of vitamin D status in chronic liver disease. J Hepatol 57:897–909PubMedCrossRef
64.
Zurück zum Zitat Kwok RM, Torres DM, Harrison SA (2013) Vitamin D and nonalcoholic fatty liver disease (NAFLD): is it more than just an association? Hepatology 58:1166–1174PubMedCrossRef Kwok RM, Torres DM, Harrison SA (2013) Vitamin D and nonalcoholic fatty liver disease (NAFLD): is it more than just an association? Hepatology 58:1166–1174PubMedCrossRef
65.
Zurück zum Zitat Thorburn AN, Macia L, Mackay CR (2014) Diet, metabolites, and “western-lifestyle” inflammatory diseases. Immunity 40:833–842PubMedCrossRef Thorburn AN, Macia L, Mackay CR (2014) Diet, metabolites, and “western-lifestyle” inflammatory diseases. Immunity 40:833–842PubMedCrossRef
66.
Zurück zum Zitat Willemsen LE, Koetsier MA, van Deventer SJ, van Tol EA (2003) Short chain fatty acids stimulate epithelial mucin 2 expression through differential effects on prostaglandin E(1) and E(2) production by intestinal myofibroblasts. Gut 52:1442–1447PubMedPubMedCentralCrossRef Willemsen LE, Koetsier MA, van Deventer SJ, van Tol EA (2003) Short chain fatty acids stimulate epithelial mucin 2 expression through differential effects on prostaglandin E(1) and E(2) production by intestinal myofibroblasts. Gut 52:1442–1447PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Shan M, Gentile M, Yeiser JR, Walland AC, Bornstein VU, Chen K, He B, Cassis L, Bigas A, Cols M, Comerma L, Huang B, Blander JM, Xiong H, Mayer L, Berin C, Augenlicht LH, Velcich A, Cerutti A (2013) Mucus enhances gut homeostasis and oral tolerance by delivering immunoregulatory signals. Science 342:447–453PubMedPubMedCentralCrossRef Shan M, Gentile M, Yeiser JR, Walland AC, Bornstein VU, Chen K, He B, Cassis L, Bigas A, Cols M, Comerma L, Huang B, Blander JM, Xiong H, Mayer L, Berin C, Augenlicht LH, Velcich A, Cerutti A (2013) Mucus enhances gut homeostasis and oral tolerance by delivering immunoregulatory signals. Science 342:447–453PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Bellanti F, Villani R, Facciorusso A, Vendemiale G, Serviddio G (2017) Lipid oxidation products in the pathogenesis of non-alcoholic steatohepatitis. Free Radic Biol Med 111:173–185PubMedCrossRef Bellanti F, Villani R, Facciorusso A, Vendemiale G, Serviddio G (2017) Lipid oxidation products in the pathogenesis of non-alcoholic steatohepatitis. Free Radic Biol Med 111:173–185PubMedCrossRef
69.
Zurück zum Zitat Pakula MM, Maier TJ, Vorup-Jensen T (2017) Insight on the impacts of free amino acids and their metabolites on the immune system from a perspective of inborn errors of amino acid metabolism. Expert Opin Ther Targets 21:611–626PubMedCrossRef Pakula MM, Maier TJ, Vorup-Jensen T (2017) Insight on the impacts of free amino acids and their metabolites on the immune system from a perspective of inborn errors of amino acid metabolism. Expert Opin Ther Targets 21:611–626PubMedCrossRef
70.
Zurück zum Zitat Bronte V, Zanovello P (2005) Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol 5:641–654PubMedCrossRef Bronte V, Zanovello P (2005) Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol 5:641–654PubMedCrossRef
71.
Zurück zum Zitat Le Floc’h N, Otten W, Merlot E (2011) Tryptophan metabolism, from nutrition to potential therapeutic applications. Amino Acids 41:1195–1205PubMedCrossRef Le Floc’h N, Otten W, Merlot E (2011) Tryptophan metabolism, from nutrition to potential therapeutic applications. Amino Acids 41:1195–1205PubMedCrossRef
73.
Zurück zum Zitat Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA (2010) An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol 185:3190–3198PubMedPubMedCentralCrossRef Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA (2010) An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol 185:3190–3198PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Di Meglio P, Duarte JH, Ahlfors H, Owens ND, Li Y, Villanova F, Tosi I, Hirota K, Nestle FO, Mrowietz U, Gilchrist MJ, Stockinger B (2014) Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditions. Immunity 40:989–1001PubMedPubMedCentralCrossRef Di Meglio P, Duarte JH, Ahlfors H, Owens ND, Li Y, Villanova F, Tosi I, Hirota K, Nestle FO, Mrowietz U, Gilchrist MJ, Stockinger B (2014) Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditions. Immunity 40:989–1001PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Katt J, Schwinge D, Schoknecht T, Quaas A, Sobottka I, Burandt E, Becker C, Neurath MF, Lohse AW, Herkel J, Schramm C (2013) Increased T helper type 17 response to pathogen stimulation in patients with primary sclerosing cholangitis. Hepatology 58:1084–1093PubMedCrossRef Katt J, Schwinge D, Schoknecht T, Quaas A, Sobottka I, Burandt E, Becker C, Neurath MF, Lohse AW, Herkel J, Schramm C (2013) Increased T helper type 17 response to pathogen stimulation in patients with primary sclerosing cholangitis. Hepatology 58:1084–1093PubMedCrossRef
76.
Zurück zum Zitat Sebode M, Peiseler M, Franke B, Schwinge D, Schoknecht T, Wortmann F, Quaas A, Petersen BS, Ellinghaus E, Baron U, Olek S, Wiegard C, Weiler-Normann C, Lohse AW, Herkel J, Schramm C (2014) Reduced FOXP3(+) regulatory T cells in patients with primary sclerosing cholangitis are associated with IL2RA gene polymorphisms. J Hepatol 60:1010–1016PubMedCrossRef Sebode M, Peiseler M, Franke B, Schwinge D, Schoknecht T, Wortmann F, Quaas A, Petersen BS, Ellinghaus E, Baron U, Olek S, Wiegard C, Weiler-Normann C, Lohse AW, Herkel J, Schramm C (2014) Reduced FOXP3(+) regulatory T cells in patients with primary sclerosing cholangitis are associated with IL2RA gene polymorphisms. J Hepatol 60:1010–1016PubMedCrossRef
77.
Zurück zum Zitat Lahvis GP, Bradfield CA (1998) Ahr null alleles: distinctive or different? Biochem Pharmacol 56:781–787PubMedCrossRef Lahvis GP, Bradfield CA (1998) Ahr null alleles: distinctive or different? Biochem Pharmacol 56:781–787PubMedCrossRef
78.
Zurück zum Zitat Fernandez-Salguero P, Pineau T, Hilbert DM, McPhail T, Lee SS, Kimura S, Nebert DW, Rudikoff S, Ward JM, Gonzalez FJ (1995) Immune system impairment and hepatic fibrosis in mice lacking the dioxin-binding Ah receptor. Science 268:722–726PubMedCrossRef Fernandez-Salguero P, Pineau T, Hilbert DM, McPhail T, Lee SS, Kimura S, Nebert DW, Rudikoff S, Ward JM, Gonzalez FJ (1995) Immune system impairment and hepatic fibrosis in mice lacking the dioxin-binding Ah receptor. Science 268:722–726PubMedCrossRef
79.
Zurück zum Zitat Fernandez-Salguero PM, Ward JM, Sundberg JP, Gonzalez FJ (1997) Lesions of aryl-hydrocarbon receptor-deficient mice. Vet Pathol 34:605–614PubMedCrossRef Fernandez-Salguero PM, Ward JM, Sundberg JP, Gonzalez FJ (1997) Lesions of aryl-hydrocarbon receptor-deficient mice. Vet Pathol 34:605–614PubMedCrossRef
80.
Zurück zum Zitat Bessede A, Gargaro M, Pallotta MT, Matino D, Servillo G, Brunacci C, Bicciato S, Mazza EM, Macchiarulo A, Vacca C, Iannitti R, Tissi L, Volpi C, Belladonna ML, Orabona C, Bianchi R, Lanz TV, Platten M, Della Fazia MA, Piobbico D, Zelante T, Funakoshi H, Nakamura T, Gilot D, Denison MS, Guillemin GJ, DuHadaway JB, Prendergast GC, Metz R, Geffard M, Boon L, Pirro M, Iorio A, Veyret B, Romani L, Grohmann U, Fallarino F, Puccetti P (2014) Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 511:184–190PubMedPubMedCentralCrossRef Bessede A, Gargaro M, Pallotta MT, Matino D, Servillo G, Brunacci C, Bicciato S, Mazza EM, Macchiarulo A, Vacca C, Iannitti R, Tissi L, Volpi C, Belladonna ML, Orabona C, Bianchi R, Lanz TV, Platten M, Della Fazia MA, Piobbico D, Zelante T, Funakoshi H, Nakamura T, Gilot D, Denison MS, Guillemin GJ, DuHadaway JB, Prendergast GC, Metz R, Geffard M, Boon L, Pirro M, Iorio A, Veyret B, Romani L, Grohmann U, Fallarino F, Puccetti P (2014) Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 511:184–190PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Kimura A, Naka T, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T (2009) Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses. J Exp Med 206:2027–2035PubMedPubMedCentralCrossRef Kimura A, Naka T, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T (2009) Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses. J Exp Med 206:2027–2035PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Zhu C, Fuchs CD, Halilbasic E, Trauner M (2016) Bile acids in regulation of inflammation and immunity: friend or foe? Clin Exp Rheumatol 34:25–31PubMed Zhu C, Fuchs CD, Halilbasic E, Trauner M (2016) Bile acids in regulation of inflammation and immunity: friend or foe? Clin Exp Rheumatol 34:25–31PubMed
83.
Zurück zum Zitat Chávez-Talavera O, Tailleux A, Lefebvre P, Staels B (2017) Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease. Gastroenterology 152:1679–1694PubMedCrossRef Chávez-Talavera O, Tailleux A, Lefebvre P, Staels B (2017) Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease. Gastroenterology 152:1679–1694PubMedCrossRef
84.
Zurück zum Zitat Allen K, Jaeschke H, Copple BL (2011) Bile acids induce inflammatory genes in hepatocytes: a novel mechanism of inflammation during obstructive cholestasis. Am J Pathol 178:175–186PubMedPubMedCentralCrossRef Allen K, Jaeschke H, Copple BL (2011) Bile acids induce inflammatory genes in hepatocytes: a novel mechanism of inflammation during obstructive cholestasis. Am J Pathol 178:175–186PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Iwata M, Harada K, Katayanagi K, Saito T, Kaneko S, Kobayashi K, Nakanuma Y (2003) Apoptosis of murine cultured biliary epithelial cells induced by glycochenodeoxycholic acid involves Fas receptor and its ligand. Hepatol Res 25:329–342PubMedCrossRef Iwata M, Harada K, Katayanagi K, Saito T, Kaneko S, Kobayashi K, Nakanuma Y (2003) Apoptosis of murine cultured biliary epithelial cells induced by glycochenodeoxycholic acid involves Fas receptor and its ligand. Hepatol Res 25:329–342PubMedCrossRef
86.
Zurück zum Zitat Li S, Zhao Y, He X, Kim TH, Kuharsky DK, Rabinowich H, Chen J, Du C, Yin XM (2002) Relief of extrinsic pathway inhibition by the Bid-dependent mitochondrial release of Smac in Fas-mediated hepatocyte apoptosis. J Biol Chem 277:26912–26920PubMedCrossRef Li S, Zhao Y, He X, Kim TH, Kuharsky DK, Rabinowich H, Chen J, Du C, Yin XM (2002) Relief of extrinsic pathway inhibition by the Bid-dependent mitochondrial release of Smac in Fas-mediated hepatocyte apoptosis. J Biol Chem 277:26912–26920PubMedCrossRef
87.
Zurück zum Zitat Becker S, Reinehr R, Graf D, vom Dahl S, Häussinger D (2007) Hydrophobic bile salts induce hepatocyte shrinkage via NADPH oxidase activation. Cell Physiol Biochem 19:89–98PubMedCrossRef Becker S, Reinehr R, Graf D, vom Dahl S, Häussinger D (2007) Hydrophobic bile salts induce hepatocyte shrinkage via NADPH oxidase activation. Cell Physiol Biochem 19:89–98PubMedCrossRef
88.
Zurück zum Zitat Adachi T, Kaminaga T, Yasuda H, Kamiya T, Hara H (2014) The involvement of endoplasmic reticulum stress in bile acid-induced hepatocellular injury. J Clin Biochem Nutr 54:129–135PubMedCrossRef Adachi T, Kaminaga T, Yasuda H, Kamiya T, Hara H (2014) The involvement of endoplasmic reticulum stress in bile acid-induced hepatocellular injury. J Clin Biochem Nutr 54:129–135PubMedCrossRef
89.
Zurück zum Zitat Hisamoto S, Shimoda S, Harada K, Iwasaka S, Onohara S, Chong Y, Nakamura M, Bekki Y, Yoshizumi T, Ikegami T, Maehara Y, He XS, Gershwin ME, Akashi K (2016) Hydrophobic bile acids suppress expression of AE2 in biliary epithelial cells and induce bile duct inflammation in primary biliary cholangitis. J Autoimmun 75:150–160PubMedCrossRef Hisamoto S, Shimoda S, Harada K, Iwasaka S, Onohara S, Chong Y, Nakamura M, Bekki Y, Yoshizumi T, Ikegami T, Maehara Y, He XS, Gershwin ME, Akashi K (2016) Hydrophobic bile acids suppress expression of AE2 in biliary epithelial cells and induce bile duct inflammation in primary biliary cholangitis. J Autoimmun 75:150–160PubMedCrossRef
90.
Zurück zum Zitat Gujral JS, Liu J, Farhood A, Hinson JA, Jaeschke H (2004) Functional importance of ICAM-1 in the mechanism of neutrophil-induced liver injury in bile duct-ligated mice. Am J Physiol Gastrointest Liver Physiol 286:G499–G507PubMedCrossRef Gujral JS, Liu J, Farhood A, Hinson JA, Jaeschke H (2004) Functional importance of ICAM-1 in the mechanism of neutrophil-induced liver injury in bile duct-ligated mice. Am J Physiol Gastrointest Liver Physiol 286:G499–G507PubMedCrossRef
91.
Zurück zum Zitat Cai SY, Ouyang X, Chen Y, Soroka CJ, Wang J, Mennone A, Wang Y, Mehal WZ, Jain D, Boyer JL (2017) Bile acids initiate cholestatic liver injury by triggering a hepatocyte-specific inflammatory response. JCI Insight 2:e90780PubMedPubMedCentralCrossRef Cai SY, Ouyang X, Chen Y, Soroka CJ, Wang J, Mennone A, Wang Y, Mehal WZ, Jain D, Boyer JL (2017) Bile acids initiate cholestatic liver injury by triggering a hepatocyte-specific inflammatory response. JCI Insight 2:e90780PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat O'Brien KM, Allen KM, Rockwell CE, Towery K, Luyendyk JP, Copple BL (2013) IL-17A synergistically enhances bile acid-induced inflammation during obstructive cholestasis. Am J Pathol 183:1498–1507PubMedPubMedCentralCrossRef O'Brien KM, Allen KM, Rockwell CE, Towery K, Luyendyk JP, Copple BL (2013) IL-17A synergistically enhances bile acid-induced inflammation during obstructive cholestasis. Am J Pathol 183:1498–1507PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Licata LA, Nguyen CT, Burga RA, Falanga V, Espat NJ, Ayala A, Thorn M, Junghans RP, Katz SC (2013) Biliary obstruction results in PD-1-dependent liver T cell dysfunction and acute inflammation mediated by Th17 cells and neutrophils. J Leukoc Biol 94:813–823PubMedPubMedCentralCrossRef Licata LA, Nguyen CT, Burga RA, Falanga V, Espat NJ, Ayala A, Thorn M, Junghans RP, Katz SC (2013) Biliary obstruction results in PD-1-dependent liver T cell dysfunction and acute inflammation mediated by Th17 cells and neutrophils. J Leukoc Biol 94:813–823PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Yang CY, Ma X, Tsuneyama K, Huang S, Takahashi T, Chalasani NP, Bowlus CL, Yang GX, Leung PS, Ansari AA, Wu L, Coppel RL, Gershwin ME (2014) IL-12/Th1 and IL-23/Th17 biliary microenvironment in primary biliary cirrhosis: implications for therapy. Hepatology 59:1944–1953PubMedPubMedCentralCrossRef Yang CY, Ma X, Tsuneyama K, Huang S, Takahashi T, Chalasani NP, Bowlus CL, Yang GX, Leung PS, Ansari AA, Wu L, Coppel RL, Gershwin ME (2014) IL-12/Th1 and IL-23/Th17 biliary microenvironment in primary biliary cirrhosis: implications for therapy. Hepatology 59:1944–1953PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Sung JJ, Go MY (1999) Reversible Kupffer cell suppression in biliary obstruction is caused by hydrophobic bile acids. J Hepatol 30:413–418PubMedCrossRef Sung JJ, Go MY (1999) Reversible Kupffer cell suppression in biliary obstruction is caused by hydrophobic bile acids. J Hepatol 30:413–418PubMedCrossRef
96.
Zurück zum Zitat Keitel V, Donner M, Winandy S, Kubitz R, Häussinger D (2008) Expression and function of the bile acid receptor TGR5 in Kupffer cells. Biochem Biophys Res Commun 372:78–84PubMedCrossRef Keitel V, Donner M, Winandy S, Kubitz R, Häussinger D (2008) Expression and function of the bile acid receptor TGR5 in Kupffer cells. Biochem Biophys Res Commun 372:78–84PubMedCrossRef
97.
Zurück zum Zitat Keitel V, Reinehr R, Gatsios P, Rupprecht C, Görg B, Selbach O, Häussinger D, Kubitz R (2007) The G-protein coupled bile salt receptor TGR5 is expressed in liver sinusoidal endothelial cells. Hepatology 45:695–704PubMedCrossRef Keitel V, Reinehr R, Gatsios P, Rupprecht C, Görg B, Selbach O, Häussinger D, Kubitz R (2007) The G-protein coupled bile salt receptor TGR5 is expressed in liver sinusoidal endothelial cells. Hepatology 45:695–704PubMedCrossRef
98.
Zurück zum Zitat McMahan RH, Wang XX, Cheng LL, Krisko T, Smith M, El Kasmi K, Pruzanski M, Adorini L, Golden-Mason L, Levi M, Rosen HR (2013) Bile acid receptor activation modulates hepatic monocyte activity and improves nonalcoholic fatty liver disease. J Biol Chem 288:11761–11770PubMedPubMedCentralCrossRef McMahan RH, Wang XX, Cheng LL, Krisko T, Smith M, El Kasmi K, Pruzanski M, Adorini L, Golden-Mason L, Levi M, Rosen HR (2013) Bile acid receptor activation modulates hepatic monocyte activity and improves nonalcoholic fatty liver disease. J Biol Chem 288:11761–11770PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Haselow K, Bode JG, Wammers M, Ehlting C, Keitel V, Kleinebrecht L, Schupp AK, Häussinger D, Graf D (2013) Bile acids PKA-dependently induce a switch of the IL-10/IL-12 ratio and reduce proinflammatory capability of human macrophages. J Leukoc Biol 94:1253–1264PubMedCrossRef Haselow K, Bode JG, Wammers M, Ehlting C, Keitel V, Kleinebrecht L, Schupp AK, Häussinger D, Graf D (2013) Bile acids PKA-dependently induce a switch of the IL-10/IL-12 ratio and reduce proinflammatory capability of human macrophages. J Leukoc Biol 94:1253–1264PubMedCrossRef
100.
Zurück zum Zitat Biagioli M, Carino A, Cipriani S, Francisci D, Marchianò S, Scarpelli P, Sorcini D, Zampella A, Fiorucci S (2017) The bile acid receptor GPBAR1 regulates the M1/M2 phenotype of intestinal macrophages and activation of GPBAR1 rescues mice from murine colitis. J Immunol 199:718–733PubMedCrossRef Biagioli M, Carino A, Cipriani S, Francisci D, Marchianò S, Scarpelli P, Sorcini D, Zampella A, Fiorucci S (2017) The bile acid receptor GPBAR1 regulates the M1/M2 phenotype of intestinal macrophages and activation of GPBAR1 rescues mice from murine colitis. J Immunol 199:718–733PubMedCrossRef
101.
Zurück zum Zitat Wang YD, Chen WD, Yu D, Forman BM, Huang W (2011) The G-protein-coupled bile acid receptor, Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing nuclear factor κ light-chain enhancer of activated B cells(NF-κB) in mice. Hepatology 54:1421–1432PubMedPubMedCentralCrossRef Wang YD, Chen WD, Yu D, Forman BM, Huang W (2011) The G-protein-coupled bile acid receptor, Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing nuclear factor κ light-chain enhancer of activated B cells(NF-κB) in mice. Hepatology 54:1421–1432PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Gong Z, Zhou J, Zhao S, Tian C, Wang P, Xu C, Chen Y, Cai W, Wu J (2016) Chenodeoxycholic acid activates NLRP3 inflammasome and contributes to cholestatic liver fibrosis. Oncotarget 7:83951–83963PubMedPubMedCentral Gong Z, Zhou J, Zhao S, Tian C, Wang P, Xu C, Chen Y, Cai W, Wu J (2016) Chenodeoxycholic acid activates NLRP3 inflammasome and contributes to cholestatic liver fibrosis. Oncotarget 7:83951–83963PubMedPubMedCentral
104.
Zurück zum Zitat Calmus Y, Poupon R (2014) Shaping macrophages function and innate immunity by bile acids: mechanisms and implication in cholestatic liver diseases. Clin Res Hepatol Gastroenterol 38:550–556PubMedCrossRef Calmus Y, Poupon R (2014) Shaping macrophages function and innate immunity by bile acids: mechanisms and implication in cholestatic liver diseases. Clin Res Hepatol Gastroenterol 38:550–556PubMedCrossRef
105.
Zurück zum Zitat Bleier JI, Katz SC, Chaudhry UI, Pillarisetty VG, Kingham TP III, Shah AB, Raab JR, DeMatteo RP (2006) Biliary obstruction selectively expands and activates liver myeloid dendritic cells. J Immunol 176:7189–7195PubMedCrossRef Bleier JI, Katz SC, Chaudhry UI, Pillarisetty VG, Kingham TP III, Shah AB, Raab JR, DeMatteo RP (2006) Biliary obstruction selectively expands and activates liver myeloid dendritic cells. J Immunol 176:7189–7195PubMedCrossRef
106.
Zurück zum Zitat Ichikawa R, Takayama T, Yoneno K, Kamada N, Kitazume MT, Higuchi H, Matsuoka K, Watanabe M, Itoh H, Kanai T, Hisamatsu T, Hibi T (2012) Bile acids induce monocyte differentiation toward interleukin-12 hypo-producing dendritic cells via a TGR5-dependent pathway. Immunology 136:153–162PubMedPubMedCentralCrossRef Ichikawa R, Takayama T, Yoneno K, Kamada N, Kitazume MT, Higuchi H, Matsuoka K, Watanabe M, Itoh H, Kanai T, Hisamatsu T, Hibi T (2012) Bile acids induce monocyte differentiation toward interleukin-12 hypo-producing dendritic cells via a TGR5-dependent pathway. Immunology 136:153–162PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Gadaleta RM, van Erpecum KJ, Oldenburg B, Willemsen EC, Renooij W, Murzilli S, Klomp LW, Siersema PD, Schipper ME, Danese S, Penna G, Laverny G, Adorini L, Moschetta A, van Mil SW (2011) Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 60:463–472PubMedCrossRef Gadaleta RM, van Erpecum KJ, Oldenburg B, Willemsen EC, Renooij W, Murzilli S, Klomp LW, Siersema PD, Schipper ME, Danese S, Penna G, Laverny G, Adorini L, Moschetta A, van Mil SW (2011) Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 60:463–472PubMedCrossRef
108.
Zurück zum Zitat Wang YD, Chen WD, Wang M, Yu D, Forman BM, Huang W (2008) Farnesoid X receptor antagonizes nuclear factor kappaB in hepatic inflammatory response. Hepatology 48:1632–1643PubMedPubMedCentralCrossRef Wang YD, Chen WD, Wang M, Yu D, Forman BM, Huang W (2008) Farnesoid X receptor antagonizes nuclear factor kappaB in hepatic inflammatory response. Hepatology 48:1632–1643PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Mencarelli A, Renga B, Migliorati M, Cipriani S, Distrutti E, Santucci L, Fiorucci S (2009) The bile acid sensor farnesoid X receptor is a modulator of liver immunity in a rodent model of acute hepatitis. J Immunol 183:6657–6666PubMedCrossRef Mencarelli A, Renga B, Migliorati M, Cipriani S, Distrutti E, Santucci L, Fiorucci S (2009) The bile acid sensor farnesoid X receptor is a modulator of liver immunity in a rodent model of acute hepatitis. J Immunol 183:6657–6666PubMedCrossRef
110.
Zurück zum Zitat Wang YD, Chen WD, Li C, Guo C, Li Y, Qi H, Shen H, Kong J, Long X, Yuan F, Wang X, Huang W (2015) Farnesoid X receptor antagonizes JNK signaling pathway in liver carcinogenesis by activating SOD3. Mol Endocrinol 29:322–331PubMedCrossRef Wang YD, Chen WD, Li C, Guo C, Li Y, Qi H, Shen H, Kong J, Long X, Yuan F, Wang X, Huang W (2015) Farnesoid X receptor antagonizes JNK signaling pathway in liver carcinogenesis by activating SOD3. Mol Endocrinol 29:322–331PubMedCrossRef
111.
Zurück zum Zitat Worthmann A, John C, Rühlemann MC, Baguhl M, Heinsen FA, Schaltenberg N, Heine M, Schlein C, Evangelakos I, Mineo C, Fischer M, Dandri M, Kremoser C, Scheja L, Franke A, Shaul PW, Heeren J (2017) Cold-induced conversion of cholesterol to bile acids in mice shapes the gut microbiome and promotes adaptive thermogenesis. Nat Med 23:839–849PubMedCrossRef Worthmann A, John C, Rühlemann MC, Baguhl M, Heinsen FA, Schaltenberg N, Heine M, Schlein C, Evangelakos I, Mineo C, Fischer M, Dandri M, Kremoser C, Scheja L, Franke A, Shaul PW, Heeren J (2017) Cold-induced conversion of cholesterol to bile acids in mice shapes the gut microbiome and promotes adaptive thermogenesis. Nat Med 23:839–849PubMedCrossRef
112.
Zurück zum Zitat Saeed A, Hoekstra M, Hoeke MO, Heegsma J, Faber KN (2017) The interrelationship between bile acid and vitamin A homeostasis. Biochim Biophys Acta 1862:496–512PubMedCrossRef Saeed A, Hoekstra M, Hoeke MO, Heegsma J, Faber KN (2017) The interrelationship between bile acid and vitamin A homeostasis. Biochim Biophys Acta 1862:496–512PubMedCrossRef
113.
Zurück zum Zitat Erkelens MN, Mebius RE (2017) Retinoic acid and immune homeostasis: a balancing act. Trends Immunol 38:168–180PubMedCrossRef Erkelens MN, Mebius RE (2017) Retinoic acid and immune homeostasis: a balancing act. Trends Immunol 38:168–180PubMedCrossRef
114.
Zurück zum Zitat Weiskirchen R, Tacke F (2014) Cellular and molecular functions of hepatic stellate cells in inflammatory responses and liver immunology. Hepatobiliary Surg Nutr 3:344–363PubMedPubMedCentral Weiskirchen R, Tacke F (2014) Cellular and molecular functions of hepatic stellate cells in inflammatory responses and liver immunology. Hepatobiliary Surg Nutr 3:344–363PubMedPubMedCentral
115.
116.
Zurück zum Zitat Derebe MG, Zlatkov CM, Gattu S, Ruhn KA, Vaishnava S, Diehl GE, MacMillan JB, Williams NS, Hooper LV (2014) Serum amyloid A is a retinol binding protein that transports retinol during bacterial infection. elife 3:e03206PubMedPubMedCentralCrossRef Derebe MG, Zlatkov CM, Gattu S, Ruhn KA, Vaishnava S, Diehl GE, MacMillan JB, Williams NS, Hooper LV (2014) Serum amyloid A is a retinol binding protein that transports retinol during bacterial infection. elife 3:e03206PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Raverdeau M, Mills KH (2014) Modulation of T cell and innate immune responses by retinoic acid. J Immunol 192:2953–2958PubMedCrossRef Raverdeau M, Mills KH (2014) Modulation of T cell and innate immune responses by retinoic acid. J Immunol 192:2953–2958PubMedCrossRef
118.
Zurück zum Zitat Ongsakul M, Sirisinha S, Lamb AJ (1985) Impaired blood clearance of bacteria and phagocytic activity in vitamin A-deficient rats. Proc Soc Exp Biol Med 178:204–208PubMedCrossRef Ongsakul M, Sirisinha S, Lamb AJ (1985) Impaired blood clearance of bacteria and phagocytic activity in vitamin A-deficient rats. Proc Soc Exp Biol Med 178:204–208PubMedCrossRef
119.
Zurück zum Zitat Bjelakovic G, Gluud LL, Nikolova D, Bjelakovic M, Nagorni A, Gluud C (2011) Antioxidant supplements for liver diseases. Cochrane Database Syst Rev 3:CD007749 Bjelakovic G, Gluud LL, Nikolova D, Bjelakovic M, Nagorni A, Gluud C (2011) Antioxidant supplements for liver diseases. Cochrane Database Syst Rev 3:CD007749
120.
121.
Zurück zum Zitat Penna G, Adorini L (2000) 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation, maturation, activation, and survival of dendritic cells leading to impaired alloreactive T cell activation. J Immunol 164:2405–2411PubMedCrossRef Penna G, Adorini L (2000) 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation, maturation, activation, and survival of dendritic cells leading to impaired alloreactive T cell activation. J Immunol 164:2405–2411PubMedCrossRef
122.
Zurück zum Zitat Sigmundsdottir H, Pan J, Debes GF, Alt C, Habtezion A, Soler D, Butcher EC (2007) DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27. Nat Immunol 8:285–293PubMedCrossRef Sigmundsdottir H, Pan J, Debes GF, Alt C, Habtezion A, Soler D, Butcher EC (2007) DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27. Nat Immunol 8:285–293PubMedCrossRef
123.
Zurück zum Zitat Adams JS, Sharma OP, Gacad MA, Singer FR (1983) Metabolism of 25-hydroxyvitamin D3 by cultured pulmonary alveolar macrophages in sarcoidosis. J Clin Invest 72:1856–1860PubMedPubMedCentralCrossRef Adams JS, Sharma OP, Gacad MA, Singer FR (1983) Metabolism of 25-hydroxyvitamin D3 by cultured pulmonary alveolar macrophages in sarcoidosis. J Clin Invest 72:1856–1860PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Stoffels K, Overbergh L, Giulietti A, Verlinden L, Bouillon R, Mathieu C (2006) Immune regulation of 25-hydroxyvitamin-D3-1alpha-hydroxylase in human monocytes. J Bone Miner Res 21:37–47PubMedCrossRef Stoffels K, Overbergh L, Giulietti A, Verlinden L, Bouillon R, Mathieu C (2006) Immune regulation of 25-hydroxyvitamin-D3-1alpha-hydroxylase in human monocytes. J Bone Miner Res 21:37–47PubMedCrossRef
125.
Zurück zum Zitat Fritsche J, Mondal K, Ehrnsperger A, Andreesen R, Kreutz M (2003) Regulation of 25-hydroxyvitamin D3-1 alpha-hydroxylase and production of 1 alpha,25-dihydroxyvitamin D3 by human dendritic cells. Blood 102:3314–3316PubMedCrossRef Fritsche J, Mondal K, Ehrnsperger A, Andreesen R, Kreutz M (2003) Regulation of 25-hydroxyvitamin D3-1 alpha-hydroxylase and production of 1 alpha,25-dihydroxyvitamin D3 by human dendritic cells. Blood 102:3314–3316PubMedCrossRef
126.
Zurück zum Zitat Sadeghi K, Wessner B, Laggner U, Ploder M, Tamandl D, Friedl J, Zügel U, Steinmeyer A, Pollak A, Roth E, Boltz-Nitulescu G, Spittler A (2006) Vitamin D3 down-regulates monocyte TLR expression and triggers hyporesponsiveness to pathogen-associated molecular patterns. Eur J Immunol 36:361–370PubMedCrossRef Sadeghi K, Wessner B, Laggner U, Ploder M, Tamandl D, Friedl J, Zügel U, Steinmeyer A, Pollak A, Roth E, Boltz-Nitulescu G, Spittler A (2006) Vitamin D3 down-regulates monocyte TLR expression and triggers hyporesponsiveness to pathogen-associated molecular patterns. Eur J Immunol 36:361–370PubMedCrossRef
127.
Zurück zum Zitat Dickie LJ, Church LD, Coulthard LR, Mathews RJ, Emery P, McDermott MF (2010) Vitamin D3 down-regulates intracellular Toll-like receptor 9 expression and Toll-like receptor 9-induced IL-6 production in human monocytes. Rheumatology (Oxford) 49:1466–1471CrossRef Dickie LJ, Church LD, Coulthard LR, Mathews RJ, Emery P, McDermott MF (2010) Vitamin D3 down-regulates intracellular Toll-like receptor 9 expression and Toll-like receptor 9-induced IL-6 production in human monocytes. Rheumatology (Oxford) 49:1466–1471CrossRef
128.
Zurück zum Zitat Tabrizi R, Moosazadeh M, Lankarani KB, Akbari M, Heydari ST, Kolahdooz F, Samimi M, Asemi Z (2017) The effects of vitamin D supplementation on metabolic profiles and liver function in patients with non-alcoholic fatty liver disease: a systematic review and meta-analysis of randomized controlled trials. Diabetes Metab Syndr. https://doi.org/10.1016/j.dsx.2017.07.025 Tabrizi R, Moosazadeh M, Lankarani KB, Akbari M, Heydari ST, Kolahdooz F, Samimi M, Asemi Z (2017) The effects of vitamin D supplementation on metabolic profiles and liver function in patients with non-alcoholic fatty liver disease: a systematic review and meta-analysis of randomized controlled trials. Diabetes Metab Syndr. https://​doi.​org/​10.​1016/​j.​dsx.​2017.​07.​025
129.
Zurück zum Zitat Corrêa-Oliveira R, Fachi JL, Vieira A, Sato FT, Vinolo MA (2016) Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunology 5:e73PubMedPubMedCentralCrossRef Corrêa-Oliveira R, Fachi JL, Vieira A, Sato FT, Vinolo MA (2016) Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunology 5:e73PubMedPubMedCentralCrossRef
130.
Zurück zum Zitat Alvarez-Curto E, Milligan G (2016) Metabolism meets immunity: the role of free fatty acid receptors in the immune system. Biochem Pharmacol 114:3–13PubMedCrossRef Alvarez-Curto E, Milligan G (2016) Metabolism meets immunity: the role of free fatty acid receptors in the immune system. Biochem Pharmacol 114:3–13PubMedCrossRef
131.
Zurück zum Zitat Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS (2013) The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341:569–573PubMedCrossRef Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS (2013) The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341:569–573PubMedCrossRef
132.
Zurück zum Zitat Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, Takahashi M, Fukuda NN, Murakami S, Miyauchi E, Hino S, Atarashi K, Onawa S, Fujimura Y, Lockett T, Clarke JM, Topping DL, Tomita M, Hori S, Ohara O, Morita T, Koseki H, Kikuchi J, Honda K, Hase K, Ohno H (2013) Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504:446–450PubMedCrossRef Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, Takahashi M, Fukuda NN, Murakami S, Miyauchi E, Hino S, Atarashi K, Onawa S, Fujimura Y, Lockett T, Clarke JM, Topping DL, Tomita M, Hori S, Ohara O, Morita T, Koseki H, Kikuchi J, Honda K, Hase K, Ohno H (2013) Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504:446–450PubMedCrossRef
133.
Zurück zum Zitat Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, Rudensky AY (2013) Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504:451–455PubMedPubMedCentralCrossRef Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, Rudensky AY (2013) Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504:451–455PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Chang PV, Hao L, Offermanns S, Medzhitov R (2014) The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A 111:2247–2252PubMedPubMedCentralCrossRef Chang PV, Hao L, Offermanns S, Medzhitov R (2014) The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A 111:2247–2252PubMedPubMedCentralCrossRef
135.
136.
Zurück zum Zitat Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM, Olefsky JM (2010) GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 142:687–698PubMedPubMedCentralCrossRef Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM, Olefsky JM (2010) GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 142:687–698PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Lee JY, Hwang DH (2006) The modulation of inflammatory gene expression by lipids: mediation through Toll-like receptors. Mol Cells 21:174–185PubMed Lee JY, Hwang DH (2006) The modulation of inflammatory gene expression by lipids: mediation through Toll-like receptors. Mol Cells 21:174–185PubMed
138.
Zurück zum Zitat Ramadori P, Kroy D, Streetz KL (2015) Immunoregulation by lipids during the development of non-alcoholic steatohepatitis. Hepatobiliary Surg Nutr 4:11–23PubMedPubMedCentral Ramadori P, Kroy D, Streetz KL (2015) Immunoregulation by lipids during the development of non-alcoholic steatohepatitis. Hepatobiliary Surg Nutr 4:11–23PubMedPubMedCentral
139.
140.
Zurück zum Zitat Guillemot-Legris O, Mutemberezi V, Muccioli GG (2016) Oxysterols in metabolic syndrome: from bystander molecules to bioactive lipids. Trends Mol Med 22:594–614PubMedCrossRef Guillemot-Legris O, Mutemberezi V, Muccioli GG (2016) Oxysterols in metabolic syndrome: from bystander molecules to bioactive lipids. Trends Mol Med 22:594–614PubMedCrossRef
141.
Zurück zum Zitat Jakobsson T, Treuter E, Gustafsson JÅ, Steffensen KR (2012) Liver X receptor biology and pharmacology: new pathways, challenges and opportunities. Trends Pharmacol Sci 33:394–404PubMedCrossRef Jakobsson T, Treuter E, Gustafsson JÅ, Steffensen KR (2012) Liver X receptor biology and pharmacology: new pathways, challenges and opportunities. Trends Pharmacol Sci 33:394–404PubMedCrossRef
142.
Zurück zum Zitat Traversari C, Sozzani S, Steffensen KR, Russo V (2014) LXR-dependent and -independent effects of oxysterols on immunity and tumor growth. Eur J Immunol 44:1896–1903PubMedCrossRef Traversari C, Sozzani S, Steffensen KR, Russo V (2014) LXR-dependent and -independent effects of oxysterols on immunity and tumor growth. Eur J Immunol 44:1896–1903PubMedCrossRef
144.
Zurück zum Zitat Wang YY, Dahle MK, Agren J, Myhre AE, Reinholt FP, Foster SJ, Collins JL, Thiemermann C, Aasen AO, Wang JE (2006) Activation of the liver X receptor protects against hepatic injury in endotoxemia by suppressing Kupffer cell activation. Shock 25:141–146PubMedCrossRef Wang YY, Dahle MK, Agren J, Myhre AE, Reinholt FP, Foster SJ, Collins JL, Thiemermann C, Aasen AO, Wang JE (2006) Activation of the liver X receptor protects against hepatic injury in endotoxemia by suppressing Kupffer cell activation. Shock 25:141–146PubMedCrossRef
145.
Zurück zum Zitat Geyeregger R, Zeyda M, Bauer W, Kriehuber E, Säemann MD, Zlabinger GJ, Maurer D, Stulnig TM (2007) Liver X receptors regulate dendritic cell phenotype and function through blocked induction of the actin-bundling protein fascin. Blood 109:4288–4295PubMedCrossRef Geyeregger R, Zeyda M, Bauer W, Kriehuber E, Säemann MD, Zlabinger GJ, Maurer D, Stulnig TM (2007) Liver X receptors regulate dendritic cell phenotype and function through blocked induction of the actin-bundling protein fascin. Blood 109:4288–4295PubMedCrossRef
146.
Zurück zum Zitat Xing Y, Zhao T, Gao X, Wu Y (2016) Liver X receptor α is essential for the capillarization of liver sinusoidal endothelial cells in liver injury. Sci Rep 6:21309PubMedPubMedCentralCrossRef Xing Y, Zhao T, Gao X, Wu Y (2016) Liver X receptor α is essential for the capillarization of liver sinusoidal endothelial cells in liver injury. Sci Rep 6:21309PubMedPubMedCentralCrossRef
147.
Zurück zum Zitat Jetten AM (2009) Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism. Nucl Recept Signal 7:e003PubMedPubMedCentral Jetten AM (2009) Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism. Nucl Recept Signal 7:e003PubMedPubMedCentral
148.
Zurück zum Zitat Trivedi PJ, Adams DH (2013) Mucosal immunity in liver autoimmunity: a comprehensive review. J Autoimmun 46:97–111PubMedCrossRef Trivedi PJ, Adams DH (2013) Mucosal immunity in liver autoimmunity: a comprehensive review. J Autoimmun 46:97–111PubMedCrossRef
149.
Zurück zum Zitat Soroosh P, Wu J, Xue X, Song J, Sutton SW, Sablad M, Yu J, Nelen MI, Liu X, Castro G, Luna R, Crawford S, Banie H, Dandridge RA, Deng X, Bittner A, Kuei C, Tootoonchi M, Rozenkrants N, Herman K, Gao J, Yang XV, Sachen K, Ngo K, Fung-Leung WP, Nguyen S, de Leon-Tabaldo A, Blevitt J, Zhang Y, Cummings MD, Rao T, Mani NS, Liu C, McKinnon M, Milla ME, Fourie AM, Sun S (2014) Oxysterols are agonist ligands of RORγt and drive Th17 cell differentiation. Proc Natl Acad Sci U S A 111:12163–12168PubMedPubMedCentralCrossRef Soroosh P, Wu J, Xue X, Song J, Sutton SW, Sablad M, Yu J, Nelen MI, Liu X, Castro G, Luna R, Crawford S, Banie H, Dandridge RA, Deng X, Bittner A, Kuei C, Tootoonchi M, Rozenkrants N, Herman K, Gao J, Yang XV, Sachen K, Ngo K, Fung-Leung WP, Nguyen S, de Leon-Tabaldo A, Blevitt J, Zhang Y, Cummings MD, Rao T, Mani NS, Liu C, McKinnon M, Milla ME, Fourie AM, Sun S (2014) Oxysterols are agonist ligands of RORγt and drive Th17 cell differentiation. Proc Natl Acad Sci U S A 111:12163–12168PubMedPubMedCentralCrossRef
150.
Zurück zum Zitat Sensi C, Daniele S, Parravicini C, Zappelli E, Russo V, Trincavelli ML, Martini C, Abbracchio MP, Eberini I (2014) Oxysterols act as promiscuous ligands of class-A GPCRs: in silico molecular modeling and in vitro validation. Cell Signal 26:2614–2620PubMedCrossRef Sensi C, Daniele S, Parravicini C, Zappelli E, Russo V, Trincavelli ML, Martini C, Abbracchio MP, Eberini I (2014) Oxysterols act as promiscuous ligands of class-A GPCRs: in silico molecular modeling and in vitro validation. Cell Signal 26:2614–2620PubMedCrossRef
151.
Zurück zum Zitat Hannedouche S, Zhang J, Yi T, Shen W, Nguyen D, Pereira JP, Guerini D, Baumgarten BU, Roggo S, Wen B, Knochenmuss R, Noël S, Gessier F, Kelly LM, Vanek M, Laurent S, Preuss I, Miault C, Christen I, Karuna R, Li W, Koo DI, Suply T, Schmedt C, Peters EC, Falchetto R, Katopodis A, Spanka C, Roy MO, Detheux M, Chen YA, Schultz PG, Cho CY, Seuwen K, Cyster JG, Sailer AW (2011) Oxysterols direct immune cell migration via EBI2. Nature 475:524–527PubMedPubMedCentralCrossRef Hannedouche S, Zhang J, Yi T, Shen W, Nguyen D, Pereira JP, Guerini D, Baumgarten BU, Roggo S, Wen B, Knochenmuss R, Noël S, Gessier F, Kelly LM, Vanek M, Laurent S, Preuss I, Miault C, Christen I, Karuna R, Li W, Koo DI, Suply T, Schmedt C, Peters EC, Falchetto R, Katopodis A, Spanka C, Roy MO, Detheux M, Chen YA, Schultz PG, Cho CY, Seuwen K, Cyster JG, Sailer AW (2011) Oxysterols direct immune cell migration via EBI2. Nature 475:524–527PubMedPubMedCentralCrossRef
152.
Zurück zum Zitat Sun S, Liu C (2015) 7α, 25-dihydroxycholesterol-mediated activation of EBI2 in immune regulation and diseases. Front Pharmacol 6:60PubMedPubMedCentral Sun S, Liu C (2015) 7α, 25-dihydroxycholesterol-mediated activation of EBI2 in immune regulation and diseases. Front Pharmacol 6:60PubMedPubMedCentral
153.
Zurück zum Zitat Chiang EY, Johnston RJ, Grogan JL (2013) EBI2 is a negative regulator of type I interferons in plasmacytoid and myeloid dendritic cells. PLoS One 8:e83457PubMedPubMedCentralCrossRef Chiang EY, Johnston RJ, Grogan JL (2013) EBI2 is a negative regulator of type I interferons in plasmacytoid and myeloid dendritic cells. PLoS One 8:e83457PubMedPubMedCentralCrossRef
154.
Zurück zum Zitat Konrad FM, Reutershan J (2012) CXCR2 in acute lung injury. Mediat Inflamm 2012:740987CrossRef Konrad FM, Reutershan J (2012) CXCR2 in acute lung injury. Mediat Inflamm 2012:740987CrossRef
155.
Zurück zum Zitat Van Sweringen HL, Sakai N, Quillin RC, Bailey J, Schuster R, Blanchard J, Goetzman H, Caldwell CC, Edwards MJ, Lentsch AB (2013) Roles of hepatocyte and myeloid CXC chemokine receptor-2 in liver recovery and regeneration after ischemia/reperfusion in mice. Hepatology 57:331–338PubMedPubMedCentralCrossRef Van Sweringen HL, Sakai N, Quillin RC, Bailey J, Schuster R, Blanchard J, Goetzman H, Caldwell CC, Edwards MJ, Lentsch AB (2013) Roles of hepatocyte and myeloid CXC chemokine receptor-2 in liver recovery and regeneration after ischemia/reperfusion in mice. Hepatology 57:331–338PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Raccosta L, Fontana R, Maggioni D, Lanterna C, Villablanca EJ, Paniccia A, Musumeci A, Chiricozzi E, Trincavelli ML, Daniele S, Martini C, Gustafsson JA, Doglioni C, Feo SG, Leiva A, Ciampa MG, Mauri L, Sensi C, Prinetti A, Eberini I, Mora JR, Bordignon C, Steffensen KR, Sonnino S, Sozzani S, Traversari C, Russo V (2013) The oxysterol-CXCR2 axis plays a key role in the recruitment of tumor-promoting neutrophils. J Exp Med 210:1711–1728PubMedPubMedCentralCrossRef Raccosta L, Fontana R, Maggioni D, Lanterna C, Villablanca EJ, Paniccia A, Musumeci A, Chiricozzi E, Trincavelli ML, Daniele S, Martini C, Gustafsson JA, Doglioni C, Feo SG, Leiva A, Ciampa MG, Mauri L, Sensi C, Prinetti A, Eberini I, Mora JR, Bordignon C, Steffensen KR, Sonnino S, Sozzani S, Traversari C, Russo V (2013) The oxysterol-CXCR2 axis plays a key role in the recruitment of tumor-promoting neutrophils. J Exp Med 210:1711–1728PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Strieter RM, Burdick MD, Gomperts BN, Belperio JA, Keane MP (2005) CXC chemokines in angiogenesis. Cytokine Growth Factor Rev 16:593–609PubMedCrossRef Strieter RM, Burdick MD, Gomperts BN, Belperio JA, Keane MP (2005) CXC chemokines in angiogenesis. Cytokine Growth Factor Rev 16:593–609PubMedCrossRef
Metadaten
Titel
Dietary and metabolic modulators of hepatic immunity
verfasst von
Antonella Carambia
Johannes Herkel
Publikationsdatum
06.11.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Seminars in Immunopathology / Ausgabe 2/2018
Print ISSN: 1863-2297
Elektronische ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-017-0659-4

Weitere Artikel der Ausgabe 2/2018

Seminars in Immunopathology 2/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhöhte Mortalität bei postpartalem Brustkrebs

07.05.2024 Mammakarzinom Nachrichten

Auch für Trägerinnen von BRCA-Varianten gilt: Erkranken sie fünf bis zehn Jahre nach der letzten Schwangerschaft an Brustkrebs, ist das Sterberisiko besonders hoch.

Hypertherme Chemotherapie bietet Chance auf Blasenerhalt

07.05.2024 Harnblasenkarzinom Nachrichten

Eine hypertherme intravesikale Chemotherapie mit Mitomycin kann für Patienten mit hochriskantem nicht muskelinvasivem Blasenkrebs eine Alternative zur radikalen Zystektomie darstellen. Kölner Urologen berichten über ihre Erfahrungen.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Medizinstudium Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Vorhofflimmern bei Jüngeren gefährlicher als gedacht

06.05.2024 Vorhofflimmern Nachrichten

Immer mehr jüngere Menschen leiden unter Vorhofflimmern. Betroffene unter 65 Jahren haben viele Risikofaktoren und ein signifikant erhöhtes Sterberisiko verglichen mit Gleichaltrigen ohne die Erkrankung.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.