메뉴 건너뛰기




Volumn 5, Issue 2, 2015, Pages 158-167

Role of farnesoid X receptor and bile acids in alcoholic liver disease

Author keywords

Alcoholic liver disease; Autophagy; Bile acids; Farnesoid X receptor; FoxO3

Indexed keywords

BILE ACID; CHOLESTEROL 7ALPHA MONOOXYGENASE; FARNESOID X RECEPTOR; FIBROBLAST GROWTH FACTOR RECEPTOR 4; GAMMA GLUTAMYLTRANSFERASE; LIVER RECEPTOR HOMOLOG 1; PEROXISOME PROLIFERATOR ACTIVATED RECEPTOR ALPHA; STEROL REGULATORY ELEMENT BINDING PROTEIN 1; TAUROCHOLIC ACID; TRANSCRIPTION FACTOR FKHRL1; URSODEOXYCHOLIC ACID;

EID: 84947202485     PISSN: 22113835     EISSN: 22113843     Source Type: Journal    
DOI: 10.1016/j.apsb.2014.12.011     Document Type: Review
Times cited : (80)

References (145)
  • 1
    • 79958847922 scopus 로고    scopus 로고
    • The emerging role of autophagy in alcoholic liver disease
    • Ding WX, Manley S, Ni HM The emerging role of autophagy in alcoholic liver disease. Exp Biol Med 2011, 236:546-556.
    • (2011) Exp Biol Med , vol.236 , pp. 546-556
    • Ding, W.X.1    Manley, S.2    Ni, H.M.3
  • 2
    • 80054872790 scopus 로고    scopus 로고
    • Alcoholic liver disease: pathogenesis and new therapeutic targets
    • Gao B, Bataller R. Alcoholic liver disease: pathogenesis and new therapeutic targets. Gastroenterology 2011, 141:1572-1585.
    • (2011) Gastroenterology , vol.141 , pp. 1572-1585
    • Gao, B.1    Bataller, R.2
  • 3
    • 84919816311 scopus 로고    scopus 로고
    • New advances in molecular mechanisms and emerging therapeutic targets in alcoholic liver diseases
    • Williams JA, Manley S, Ding WX. New advances in molecular mechanisms and emerging therapeutic targets in alcoholic liver diseases. World J Gastroenterol 2014, 20:12908-12933.
    • (2014) World J Gastroenterol , vol.20 , pp. 12908-12933
    • Williams, J.A.1    Manley, S.2    Ding, W.X.3
  • 4
    • 84935840850 scopus 로고    scopus 로고
    • Autophagy in alcohol-induced multiorgan injury: mechanisms and potential therapeutic targets
    • Li Y, Wang S, Ni HM, Huang HQ, Ding WX. Autophagy in alcohol-induced multiorgan injury: mechanisms and potential therapeutic targets. BioMed Res Int 2014, 2014:498491.
    • (2014) BioMed Res Int , vol.2014 , pp. 498491
    • Li, Y.1    Wang, S.2    Ni, H.M.3    Huang, H.Q.4    Ding, W.X.5
  • 5
    • 33847008303 scopus 로고    scopus 로고
    • Genetics of alcoholic liver disease and nonalcoholic fatty liver disease
    • Wilfred de Alwis NM, Day CP. Genetics of alcoholic liver disease and nonalcoholic fatty liver disease. Semin Liver Dis 2007, 27:44-54.
    • (2007) Semin Liver Dis , vol.27 , pp. 44-54
    • Wilfred de Alwis, N.M.1    Day, C.P.2
  • 8
    • 0033171439 scopus 로고    scopus 로고
    • Cholestasis and alcoholic liver disease
    • Tung BY, Carithers R.L.Jr Cholestasis and alcoholic liver disease. Clin Liver Dis 1999, 3:585-601.
    • (1999) Clin Liver Dis , vol.3 , pp. 585-601
    • Tung, B.Y.1    Carithers, R.L.J.2
  • 9
  • 10
    • 84901365815 scopus 로고    scopus 로고
    • Bile acids are nutrient signaling hormones
    • Zhou H, Hylemon PB. Bile acids are nutrient signaling hormones. Steroids 2014, 86:62-68.
    • (2014) Steroids , vol.86 , pp. 62-68
    • Zhou, H.1    Hylemon, P.B.2
  • 11
    • 84857775690 scopus 로고    scopus 로고
    • Bile acid signaling in liver metabolism and diseases
    • Li TG, Chiang JYL. Bile acid signaling in liver metabolism and diseases. J Lipids 2012, 2012:754067.
    • (2012) J Lipids , vol.2012 , pp. 754067
    • Li, T.G.1    Chiang, J.Y.L.2
  • 12
    • 84905257719 scopus 로고    scopus 로고
    • Bile acid signaling in metabolic disease and drug therapy
    • Li TG, Chiang JYL Bile acid signaling in metabolic disease and drug therapy. Pharmacol Rev 2014, 66:948-983.
    • (2014) Pharmacol Rev , vol.66 , pp. 948-983
    • Li, T.G.1    Chiang, J.Y.L.2
  • 13
    • 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, 81:687-693.
    • (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
  • 14
    • 0028836714 scopus 로고
    • Isolation of proteins that interact specifically with the retinoid X receptor: two novel orphan receptors
    • Seol W, Choi HS, Moore DD. Isolation of proteins that interact specifically with the retinoid X receptor: two novel orphan receptors. Mol Endocrinol 1995, 9:72-85.
    • (1995) Mol Endocrinol , vol.9 , pp. 72-85
    • Seol, W.1    Choi, H.S.2    Moore, D.D.3
  • 17
    • 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, 3:543-553.
    • (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
  • 18
    • 0035976638 scopus 로고    scopus 로고
    • Nuclear receptors and lipid physiology: opening the X-files
    • Chawla A, Repa JJ, Evans RM, Mangelsdorf DJ. Nuclear receptors and lipid physiology: opening the X-files. Science 2001, 294:1866-1870.
    • (2001) Science , vol.294 , pp. 1866-1870
    • Chawla, A.1    Repa, J.J.2    Evans, R.M.3    Mangelsdorf, D.J.4
  • 19
    • 58249110568 scopus 로고    scopus 로고
    • Role of bile acids and bile acid receptors in metabolic regulation
    • Lefebvre P, Cariou B, Lien F, Kuipers F, Staels B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev 2009, 89:147-191.
    • (2009) Physiol Rev , vol.89 , pp. 147-191
    • Lefebvre, P.1    Cariou, B.2    Lien, F.3    Kuipers, F.4    Staels, B.5
  • 20
    • 0034616147 scopus 로고    scopus 로고
    • Identification of the DNA binding specificity and potential target genes for the farnesoid X-activated receptor
    • Laffitte BA, Kast HR, Nguyen CM, Zavacki AM, Moore DD, Edwards PA. Identification of the DNA binding specificity and potential target genes for the farnesoid X-activated receptor. J Biol Chem 2000, 275:10638-10647.
    • (2000) J Biol Chem , vol.275 , pp. 10638-10647
    • Laffitte, B.A.1    Kast, H.R.2    Nguyen, C.M.3    Zavacki, A.M.4    Moore, D.D.5    Edwards, P.A.6
  • 21
    • 37249015421 scopus 로고    scopus 로고
    • Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine
    • Kim I, Ahn SH, Inagaki T, Choi M, Ito S, Guo GL, et al. Differential regulation of bile acid homeostasis by the farnesoid X receptor in liver and intestine. J Lipid Res 2007, 48:2664-2672.
    • (2007) J Lipid Res , vol.48 , pp. 2664-2672
    • Kim, I.1    Ahn, S.H.2    Inagaki, T.3    Choi, M.4    Ito, S.5    Guo, G.L.6
  • 22
    • 27844546989 scopus 로고    scopus 로고
    • Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis
    • Inagaki T, Choi M, Moschetta A, Peng L, Cummins CL, McDonald JG, et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab 2005, 2:217-225.
    • (2005) Cell Metab , vol.2 , pp. 217-225
    • Inagaki, T.1    Choi, M.2    Moschetta, A.3    Peng, L.4    Cummins, C.L.5    McDonald, J.G.6
  • 23
    • 58949097425 scopus 로고    scopus 로고
    • Bile acids activate fibroblast growth factor 19 signaling in human hepatocytes to inhibit cholesterol 7α-hydroxylase gene expression
    • Song KH, Li T, Owsley E, Strom S, Chiang JY. Bile acids activate fibroblast growth factor 19 signaling in human hepatocytes to inhibit cholesterol 7α-hydroxylase gene expression. Hepatology 2009, 49:297-305.
    • (2009) Hepatology , vol.49 , pp. 297-305
    • Song, K.H.1    Li, T.2    Owsley, E.3    Strom, S.4    Chiang, J.Y.5
  • 25
    • 64349115048 scopus 로고    scopus 로고
    • Discovery of XL335 (WAY-362450), a highly potent, selective, and orally active agonist of the farnesoid X receptor (FXR)
    • Flatt B, Martin R, Wang TL, Mahaney P, Murphy B, Gu XH, et al. Discovery of XL335 (WAY-362450), a highly potent, selective, and orally active agonist of the farnesoid X receptor (FXR). J Med Chem 2009, 52:904-907.
    • (2009) J Med Chem , vol.52 , pp. 904-907
    • Flatt, B.1    Martin, R.2    Wang, T.L.3    Mahaney, P.4    Murphy, B.5    Gu, X.H.6
  • 26
    • 0033637121 scopus 로고    scopus 로고
    • A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis
    • Goodwin B, Jones SA, Price RR, Watson MA, McKee DD, Moore LB, et al. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell 2000, 6:517-526.
    • (2000) Mol Cell , vol.6 , pp. 517-526
    • Goodwin, B.1    Jones, S.A.2    Price, R.R.3    Watson, M.A.4    McKee, D.D.5    Moore, L.B.6
  • 28
    • 35648935499 scopus 로고    scopus 로고
    • Involvement of corepressor complex subunit GPS2 in transcriptional pathways governing human bile acid biosynthesis
    • Sanyal S, Båvner A, Haroniti A, Nilsson LM, Lundåsen T, Rehnmark S, et al. Involvement of corepressor complex subunit GPS2 in transcriptional pathways governing human bile acid biosynthesis. Proc Natl Acad Sci USA 2007, 104:15665-15670.
    • (2007) Proc Natl Acad Sci USA , vol.104 , pp. 15665-15670
    • Sanyal, S.1    Båvner, A.2    Haroniti, A.3    Nilsson, L.M.4    Lundåsen, T.5    Rehnmark, S.6
  • 29
    • 0034664729 scopus 로고    scopus 로고
    • Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis
    • Sinal CJ, Tohkin M, Miyata M, Ward JM, Lambert G, Gonzalez FJ. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 2000, 102:731-744.
    • (2000) Cell , vol.102 , pp. 731-744
    • Sinal, C.J.1    Tohkin, M.2    Miyata, M.3    Ward, J.M.4    Lambert, G.5    Gonzalez, F.J.6
  • 30
    • 0017566067 scopus 로고
    • Cholestasis in acute alcoholic liver disease
    • Glover SC, McPhie JL, Brunt PW. Cholestasis in acute alcoholic liver disease. Lancet 1977, 2:1305-1307.
    • (1977) Lancet , vol.2 , pp. 1305-1307
    • Glover, S.C.1    McPhie, J.L.2    Brunt, P.W.3
  • 31
    • 0015487213 scopus 로고
    • Effect of ethanol on cholesterol and bile acid metabolism
    • Lefevre AF, DeCarli LM, Lieber CS. Effect of ethanol on cholesterol and bile acid metabolism. J Lipid Res 1972, 13:48-55.
    • (1972) J Lipid Res , vol.13 , pp. 48-55
    • Lefevre, A.F.1    DeCarli, L.M.2    Lieber, C.S.3
  • 32
    • 0021369331 scopus 로고
    • Depression of biliary glutathione excretion by chronic ethanol feeding in the rat
    • Vendemiale G, Jayatilleke E, Shaw S, Lieber CS. Depression of biliary glutathione excretion by chronic ethanol feeding in the rat. Life Sci 1984, 34:1065-1073.
    • (1984) Life Sci , vol.34 , pp. 1065-1073
    • Vendemiale, G.1    Jayatilleke, E.2    Shaw, S.3    Lieber, C.S.4
  • 33
    • 84890857617 scopus 로고    scopus 로고
    • Activation of farnesoid X receptor attenuates hepatic injury in a murine model of alcoholic liver disease
    • Wu W, Zhu B, Peng X, Zhou M, Jia D, Gu J. Activation of farnesoid X receptor attenuates hepatic injury in a murine model of alcoholic liver disease. Biochem Biophys Res Commun 2014, 443:68-73.
    • (2014) Biochem Biophys Res Commun , vol.443 , pp. 68-73
    • Wu, W.1    Zhu, B.2    Peng, X.3    Zhou, M.4    Jia, D.5    Gu, J.6
  • 34
    • 84883395327 scopus 로고    scopus 로고
    • Alteration of bile acid metabolism in the rat induced by chronic ethanol consumption
    • Xie G, Zhong W, Li H, Li Q, Qiu Y, Zheng X, et al. Alteration of bile acid metabolism in the rat induced by chronic ethanol consumption. FASEB J 2013, 27:3583-3593.
    • (2013) FASEB J , vol.27 , pp. 3583-3593
    • Xie, G.1    Zhong, W.2    Li, H.3    Li, Q.4    Qiu, Y.5    Zheng, X.6
  • 35
    • 0021739393 scopus 로고
    • Physicochemical properties of bile acids and their relationship to biological properties: an overview of the problem
    • Hofmann AF, Roda A. Physicochemical properties of bile acids and their relationship to biological properties: an overview of the problem. J Lipid Res 1984, 25:1477-1489.
    • (1984) J Lipid Res , vol.25 , pp. 1477-1489
    • Hofmann, A.F.1    Roda, A.2
  • 36
  • 37
    • 0031709570 scopus 로고    scopus 로고
    • Taurine: protective properties against ethanol-induced hepatic steatosis and lipid peroxidation during chronic ethanol consumption in rats
    • Kerai MD, Waterfield CJ, Kenyon SH, Asker DS, Timbrell JA. Taurine: protective properties against ethanol-induced hepatic steatosis and lipid peroxidation during chronic ethanol consumption in rats. Amino Acids 1998, 15:53-76.
    • (1998) Amino Acids , vol.15 , pp. 53-76
    • Kerai, M.D.1    Waterfield, C.J.2    Kenyon, S.H.3    Asker, D.S.4    Timbrell, J.A.5
  • 38
    • 0025979461 scopus 로고
    • Ethanol has an acute effect on bile acid biosynthesis in man
    • Axelson M, Mörk B, Sjövall J. Ethanol has an acute effect on bile acid biosynthesis in man. FEBS Lett 1991, 281:155-159.
    • (1991) FEBS Lett , vol.281 , pp. 155-159
    • Axelson, M.1    Mörk, B.2    Sjövall, J.3
  • 40
    • 84880671539 scopus 로고    scopus 로고
    • Hepatic cannabinoid receptor type 1 mediates alcohol-induced regulation of bile acid enzyme genes expression via CREBH
    • Chanda D, Kim YH, Li T, Misra J, Kim DK, Kim JR, et al. Hepatic cannabinoid receptor type 1 mediates alcohol-induced regulation of bile acid enzyme genes expression via CREBH. PLoS One 2013, 8:e68845.
    • (2013) PLoS One , vol.8 , pp. e68845
    • Chanda, D.1    Kim, Y.H.2    Li, T.3    Misra, J.4    Kim, D.K.5    Kim, J.R.6
  • 41
    • 0033636789 scopus 로고    scopus 로고
    • Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors
    • Lu TT, Makishima M, Repa JJ, Schoonjans K, Kerr TA, Auwerx J, et al. Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell 2000, 6:507-515.
    • (2000) Mol Cell , vol.6 , pp. 507-515
    • Lu, T.T.1    Makishima, M.2    Repa, J.J.3    Schoonjans, K.4    Kerr, T.A.5    Auwerx, J.6
  • 42
    • 84888262636 scopus 로고    scopus 로고
    • Critical role of FoxO3a in alcohol-induced autophagy and hepatotoxicity
    • Ni HM, Du K, You M, Ding WX. Critical role of FoxO3a in alcohol-induced autophagy and hepatotoxicity. Am J Pathol 2013, 183:1815-1825.
    • (2013) Am J Pathol , vol.183 , pp. 1815-1825
    • Ni, H.M.1    Du, K.2    You, M.3    Ding, W.X.4
  • 44
    • 0031659023 scopus 로고    scopus 로고
    • Dietary fat and alcoholic liver disease
    • Mezey E. Dietary fat and alcoholic liver disease. Hepatology 1998, 28:901-905.
    • (1998) Hepatology , vol.28 , pp. 901-905
    • Mezey, E.1
  • 45
    • 33751113602 scopus 로고    scopus 로고
    • Mammalian sirtuins-emerging roles in physiology, aging, and calorie restriction
    • Haigis MC, Guarente LP. Mammalian sirtuins-emerging roles in physiology, aging, and calorie restriction. Genes Dev 2006, 20:2913-2921.
    • (2006) Genes Dev , vol.20 , pp. 2913-2921
    • Haigis, M.C.1    Guarente, L.P.2
  • 46
    • 0037101810 scopus 로고    scopus 로고
    • 6α-Ethyl-chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity
    • Pellicciari R, Fiorucci S, Camaioni E, Clerici C, Costantino G, Maloney PR, et al. 6α-Ethyl-chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity. J Med Chem 2002, 45:3569-3572.
    • (2002) J Med Chem , vol.45 , pp. 3569-3572
    • Pellicciari, R.1    Fiorucci, S.2    Camaioni, E.3    Clerici, C.4    Costantino, G.5    Maloney, P.R.6
  • 50
    • 84908663793 scopus 로고    scopus 로고
    • Farnesoid X receptor regulates forkhead Box O3a activation in ethanol-induced autophagy and hepatotoxicity
    • Manley S, Ni HM, Williams JA, Kong B, DiTacchio L, Guo G, et al. Farnesoid X receptor regulates forkhead Box O3a activation in ethanol-induced autophagy and hepatotoxicity. Redox Biol 2014, 2:991-1002.
    • (2014) Redox Biol , vol.2 , pp. 991-1002
    • Manley, S.1    Ni, H.M.2    Williams, J.A.3    Kong, B.4    DiTacchio, L.5    Guo, G.6
  • 51
    • 84947201588 scopus 로고    scopus 로고
    • Hepatoprotective effects of farnesoid X receptor in a mouse chronic alcohol feeding model
    • Kong B., Zhan L., Shen J., Forostyan T., Wang L., Ding W.X., et al. Hepatoprotective effects of farnesoid X receptor in a mouse chronic alcohol feeding model. Hepatology 2014, 60:766A-771A.
    • (2014) Hepatology , vol.60 , pp. 766A-771A
    • Kong, B.1    Zhan, L.2    Shen, J.3    Forostyan, T.4    Wang, L.5    Ding, W.X.6
  • 52
  • 53
    • 0037204850 scopus 로고    scopus 로고
    • Effects of high taurocholate load on activities of hepatic alcohol metabolizing enzymes
    • Kim YH, Shin MJ. Effects of high taurocholate load on activities of hepatic alcohol metabolizing enzymes. Exp Mol Med 2002, 34:123-130.
    • (2002) Exp Mol Med , vol.34 , pp. 123-130
    • Kim, Y.H.1    Shin, M.J.2
  • 54
    • 0036725044 scopus 로고    scopus 로고
    • Ursodeoxycholic acid in cholestatic liver disease: mechanisms of action and therapeutic use revisited
    • Paumgartner G, Beuers U. Ursodeoxycholic acid in cholestatic liver disease: mechanisms of action and therapeutic use revisited. Hepatology 2002, 36:525-531.
    • (2002) Hepatology , vol.36 , pp. 525-531
    • Paumgartner, G.1    Beuers, U.2
  • 55
    • 0029120317 scopus 로고
    • Effect of tauroursodeoxycholic and ursodeoxycholic acid on ethanol-induced cell injuries in the human Hep G2 cell line
    • Neuman MG, Cameron RG, Shear NH, Bellentani S, Tiribelli C. Effect of tauroursodeoxycholic and ursodeoxycholic acid on ethanol-induced cell injuries in the human Hep G2 cell line. Gastroenterology 1995, 109:555-563.
    • (1995) Gastroenterology , vol.109 , pp. 555-563
    • Neuman, M.G.1    Cameron, R.G.2    Shear, N.H.3    Bellentani, S.4    Tiribelli, C.5
  • 56
  • 57
    • 0027185531 scopus 로고
    • Stable expression of human cytochrome P4502E1 in HepG2 cells: characterization of catalytic activities and production of reactive oxygen intermediates
    • Dai Y, Rashba-Step J, Cederbaum AI. Stable expression of human cytochrome P4502E1 in HepG2 cells: characterization of catalytic activities and production of reactive oxygen intermediates. Biochemistry 1993, 32:6928-6937.
    • (1993) Biochemistry , vol.32 , pp. 6928-6937
    • Dai, Y.1    Rashba-Step, J.2    Cederbaum, A.I.3
  • 58
    • 1642524322 scopus 로고    scopus 로고
    • Toxicity of ethanol and acetaldehyde in hepatocytes treated with ursodeoxycholic or tauroursodeoxycholic acid
    • Henzel K, Thorborg C, Hofmann M, Zimmer G, Leuschner U. Toxicity of ethanol and acetaldehyde in hepatocytes treated with ursodeoxycholic or tauroursodeoxycholic acid. Biochim Biophys Acta 2004, 1644:37-45.
    • (2004) Biochim Biophys Acta , vol.1644 , pp. 37-45
    • Henzel, K.1    Thorborg, C.2    Hofmann, M.3    Zimmer, G.4    Leuschner, U.5
  • 60
  • 61
    • 0033529806 scopus 로고    scopus 로고
    • Cyp2e1 and Cyp3a1/2 gene expression is not associated with the ursodeoxycholate effect on ethanol-induced lipoperoxidation
    • Nguyen TD, Oliva L, Villard PH, Puyoou F, Sauze C, Montet AM, et al. Cyp2e1 and Cyp3a1/2 gene expression is not associated with the ursodeoxycholate effect on ethanol-induced lipoperoxidation. Life Sci 1999, 65:1103-1113.
    • (1999) Life Sci , vol.65 , pp. 1103-1113
    • Nguyen, T.D.1    Oliva, L.2    Villard, P.H.3    Puyoou, F.4    Sauze, C.5    Montet, A.M.6
  • 62
    • 0031912463 scopus 로고    scopus 로고
    • Ethanol-induced changes of intracellular thiol compartmentation and protein redox status in the rat liver: effect of tauroursodeoxycholate
    • Vendemiale G, Grattagliano I, Signorile A, Altomare E. Ethanol-induced changes of intracellular thiol compartmentation and protein redox status in the rat liver: effect of tauroursodeoxycholate. J Hepatol 1998, 28:46-53.
    • (1998) J Hepatol , vol.28 , pp. 46-53
    • Vendemiale, G.1    Grattagliano, I.2    Signorile, A.3    Altomare, E.4
  • 63
    • 0035679490 scopus 로고    scopus 로고
    • Effect of ursodeoxycholic acid on prostaglandin metabolism and microsomal membranes in alcoholic fatty liver
    • Lukivskaya OY, Maskevich AA, Buko VU. Effect of ursodeoxycholic acid on prostaglandin metabolism and microsomal membranes in alcoholic fatty liver. Alcohol 2001, 25:99-105.
    • (2001) Alcohol , vol.25 , pp. 99-105
    • Lukivskaya, O.Y.1    Maskevich, A.A.2    Buko, V.U.3
  • 65
    • 0037382139 scopus 로고    scopus 로고
    • A randomized controlled trial of ursodeoxycholic acid in patients with alcohol-induced cirrhosis and jaundice
    • Pelletier G, Roulot D, Davion T, Masliah C, Causse X, Oberti F, et al. A randomized controlled trial of ursodeoxycholic acid in patients with alcohol-induced cirrhosis and jaundice. Hepatology 2003, 37:887-892.
    • (2003) Hepatology , vol.37 , pp. 887-892
    • Pelletier, G.1    Roulot, D.2    Davion, T.3    Masliah, C.4    Causse, X.5    Oberti, F.6
  • 66
    • 84938514318 scopus 로고    scopus 로고
    • Alterations of the gut microbiome and metabolome in alcoholic liver disease
    • Zhong W, Zhou ZX. Alterations of the gut microbiome and metabolome in alcoholic liver disease. World J Gastrointest Pathophysiol 2014, 5:514-522.
    • (2014) World J Gastrointest Pathophysiol , vol.5 , pp. 514-522
    • Zhong, W.1    Zhou, Z.X.2
  • 67
    • 84898824175 scopus 로고    scopus 로고
    • Interactions between the intestinal microbiome and liver diseases
    • Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 2014, 146:1513-1524.
    • (2014) Gastroenterology , vol.146 , pp. 1513-1524
    • Schnabl, B.1    Brenner, D.A.2
  • 68
    • 84901242455 scopus 로고    scopus 로고
    • Host-microbiome interactions in alcoholic liver disease
    • Chen P, Schnabl B. Host-microbiome interactions in alcoholic liver disease. Gut Liver 2014, 8:237-241.
    • (2014) Gut Liver , vol.8 , pp. 237-241
    • Chen, P.1    Schnabl, B.2
  • 69
    • 84896731920 scopus 로고    scopus 로고
    • The impact of alcohol consumption and cholecystectomy on small intestinal bacterial overgrowth
    • Gabbard SL, Lacy BE, Levine GM, Crowell MD. The impact of alcohol consumption and cholecystectomy on small intestinal bacterial overgrowth. Dig Dis Sci 2014, 59:638-644.
    • (2014) Dig Dis Sci , vol.59 , pp. 638-644
    • Gabbard, S.L.1    Lacy, B.E.2    Levine, G.M.3    Crowell, M.D.4
  • 72
  • 73
    • 84872246853 scopus 로고    scopus 로고
    • Metagenomic analyses of alcohol induced pathogenic alterations in the intestinal microbiome and the effect of Lactobacillus rhamnosus GG treatment
    • Bull-Otterson L, Feng W, Kirpich I, Wang Y, Qin X, Liu Y, et al. Metagenomic analyses of alcohol induced pathogenic alterations in the intestinal microbiome and the effect of Lactobacillus rhamnosus GG treatment. PloS One 2013, 8:e53028.
    • (2013) PloS One , vol.8 , pp. e53028
    • Bull-Otterson, L.1    Feng, W.2    Kirpich, I.3    Wang, Y.4    Qin, X.5    Liu, Y.6
  • 75
    • 0036312492 scopus 로고    scopus 로고
    • Microbes and mucosa in the regulation of intracolonic acetaldehyde concentration during ethanol challenge
    • Visapää JP, Tillonen J, Salaspuro M. Microbes and mucosa in the regulation of intracolonic acetaldehyde concentration during ethanol challenge. Alcohol Alcohol 2002, 37:322-326.
    • (2002) Alcohol Alcohol , vol.37 , pp. 322-326
    • Visapää, J.P.1    Tillonen, J.2    Salaspuro, M.3
  • 76
    • 0022657702 scopus 로고
    • Role of intestinal bacterial overgrowth in ethanol production and metabolism in rats
    • Baraona E, Julkunen R, Tannenbaum L, Lieber CS. Role of intestinal bacterial overgrowth in ethanol production and metabolism in rats. Gastroenterology 1986, 90:103-110.
    • (1986) Gastroenterology , vol.90 , pp. 103-110
    • Baraona, E.1    Julkunen, R.2    Tannenbaum, L.3    Lieber, C.S.4
  • 77
    • 68949178506 scopus 로고    scopus 로고
    • Endotoxemia and gut barrier dysfunction in alcoholic liver disease
    • Rao R. Endotoxemia and gut barrier dysfunction in alcoholic liver disease. Hepatology 2009, 50:638-644.
    • (2009) Hepatology , vol.50 , pp. 638-644
    • Rao, R.1
  • 79
    • 79960714764 scopus 로고    scopus 로고
    • Characterization of fecal microbial communities in patients with liver cirrhosis
    • Chen Y, Yang F, Lu H, Wang B, Chen Y, Lei D, et al. Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology 2011, 54:562-572.
    • (2011) Hepatology , vol.54 , pp. 562-572
    • Chen, Y.1    Yang, F.2    Lu, H.3    Wang, B.4    Chen, Y.5    Lei, D.6
  • 81
    • 70349335851 scopus 로고    scopus 로고
    • Intestinal dysbiosis: a possible mechanism of alcohol-induced endotoxemia and alcoholic steatohepatitis in rats
    • Mutlu E, Keshavarzian A, Engen P, Forsyth CB, Sikaroodi M, Gillevet P. Intestinal dysbiosis: a possible mechanism of alcohol-induced endotoxemia and alcoholic steatohepatitis in rats. Alcohol Clin Exp Res 2009, 33:1836-1846.
    • (2009) Alcohol Clin Exp Res , vol.33 , pp. 1836-1846
    • Mutlu, E.1    Keshavarzian, A.2    Engen, P.3    Forsyth, C.B.4    Sikaroodi, M.5    Gillevet, P.6
  • 82
    • 61349161828 scopus 로고    scopus 로고
    • Lactobacillus GG treatment ameliorates alcohol-induced intestinal oxidative stress, gut leakiness, and liver injury in a rat model of alcoholic steatohepatitis
    • Forsyth CB, Farhadi A, Jakate SM, Tang Y, Shaikh M, Keshavarzian A. Lactobacillus GG treatment ameliorates alcohol-induced intestinal oxidative stress, gut leakiness, and liver injury in a rat model of alcoholic steatohepatitis. Alcohol 2009, 43:163-172.
    • (2009) Alcohol , vol.43 , pp. 163-172
    • Forsyth, C.B.1    Farhadi, A.2    Jakate, S.M.3    Tang, Y.4    Shaikh, M.5    Keshavarzian, A.6
  • 83
    • 0028220726 scopus 로고
    • Lactobacillus feeding reduces endotoxemia and severity of experimental alcoholic liver (disease)
    • Nanji AA, Khettry U, Sadrzadeh SM. Lactobacillus feeding reduces endotoxemia and severity of experimental alcoholic liver (disease). Proc Soc Exp Biol Med 1994, 205:243-247.
    • (1994) Proc Soc Exp Biol Med , vol.205 , pp. 243-247
    • Nanji, A.A.1    Khettry, U.2    Sadrzadeh, S.M.3
  • 84
    • 84857840914 scopus 로고    scopus 로고
    • Effects of live Lactobacillus paracasei on plasma lipid concentration in rats fed an ethanol-containing diet
    • Komatsuzaki N, Shima J. Effects of live Lactobacillus paracasei on plasma lipid concentration in rats fed an ethanol-containing diet. Biosci Biotechnol Biochem 2012, 76:232-237.
    • (2012) Biosci Biotechnol Biochem , vol.76 , pp. 232-237
    • Komatsuzaki, N.1    Shima, J.2
  • 85
    • 84885556267 scopus 로고    scopus 로고
    • The protective effect of VSL#3 on intestinal permeability in a rat model of alcoholic intestinal injury
    • Chang B, Sang LX, Wang Y, Tong J, Zhang D, Wang BY. The protective effect of VSL#3 on intestinal permeability in a rat model of alcoholic intestinal injury. BMC Gastroenterol 2013, 13:151.
    • (2013) BMC Gastroenterol , vol.13 , pp. 151
    • Chang, B.1    Sang, L.X.2    Wang, Y.3    Tong, J.4    Zhang, D.5    Wang, B.Y.6
  • 86
    • 84904393604 scopus 로고    scopus 로고
    • Pyrroloquinoline quinone-secreting probiotic Escherichia coli Nissle 1917 ameliorates ethanol-induced oxidative damage and hyperlipidemia in rats
    • Singh AK, Pandey SK, Naresh Kumar G. Pyrroloquinoline quinone-secreting probiotic Escherichia coli Nissle 1917 ameliorates ethanol-induced oxidative damage and hyperlipidemia in rats. Alcohol Clin Exp Res 2014, 38:2127-2137.
    • (2014) Alcohol Clin Exp Res , vol.38 , pp. 2127-2137
    • Singh, A.K.1    Pandey, S.K.2    Naresh Kumar, G.3
  • 87
    • 56349169101 scopus 로고    scopus 로고
    • Probiotics restore bowel flora and improve liver enzymes in human alcohol-induced liver injury: a pilot study
    • Kirpich IA, Solovieva NV, Leikhter SN, Shidakova NA, Lebedeva OV, Sidorov PI, et al. Probiotics restore bowel flora and improve liver enzymes in human alcohol-induced liver injury: a pilot study. Alcohol 2008, 42:675-682.
    • (2008) Alcohol , vol.42 , pp. 675-682
    • Kirpich, I.A.1    Solovieva, N.V.2    Leikhter, S.N.3    Shidakova, N.A.4    Lebedeva, O.V.5    Sidorov, P.I.6
  • 88
    • 43049181923 scopus 로고    scopus 로고
    • Effect of probiotic treatment on deranged neutrophil function and cytokine responses in patients with compensated alcoholic cirrhosis
    • Stadlbauer V, Mookerjee RP, Hodges S, Wright GA, Davies NA, Jalan R. Effect of probiotic treatment on deranged neutrophil function and cytokine responses in patients with compensated alcoholic cirrhosis. J Hepatol 2008, 48:945-951.
    • (2008) J Hepatol , vol.48 , pp. 945-951
    • Stadlbauer, V.1    Mookerjee, R.P.2    Hodges, S.3    Wright, G.A.4    Davies, N.A.5    Jalan, R.6
  • 89
    • 33244467651 scopus 로고    scopus 로고
    • Bile salt biotransformations by human intestinal bacteria
    • Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 2006, 47:241-259.
    • (2006) J Lipid Res , vol.47 , pp. 241-259
    • Ridlon, J.M.1    Kang, D.J.2    Hylemon, P.B.3
  • 90
    • 0015579966 scopus 로고
    • Metabolism of steroid and amino acid moieties of conjugated bile acids in man. 3. Cholyltaurine (taurocholic acid)
    • Hepner GW, Sturman JA, Hofmann AF, Thomas PJ. Metabolism of steroid and amino acid moieties of conjugated bile acids in man. 3. Cholyltaurine (taurocholic acid). J Clin Invest 1973, 52:433-440.
    • (1973) J Clin Invest , vol.52 , pp. 433-440
    • Hepner, G.W.1    Sturman, J.A.2    Hofmann, A.F.3    Thomas, P.J.4
  • 91
    • 84863170205 scopus 로고    scopus 로고
    • Identification of N-acetyltaurine as a novel metabolite of ethanol through metabolomics-guided biochemical analysis
    • Shi X, Yao D, Chen C. Identification of N-acetyltaurine as a novel metabolite of ethanol through metabolomics-guided biochemical analysis. J Biol Chem 2012, 287:6336-6349.
    • (2012) J Biol Chem , vol.287 , pp. 6336-6349
    • Shi, X.1    Yao, D.2    Chen, C.3
  • 92
    • 33644752102 scopus 로고    scopus 로고
    • Mechanism of growth inhibition by free bile acids in lactobacilli and bifidobacteria
    • Kurdi P, Kawanishi K, Mizutani K, Yokota A. Mechanism of growth inhibition by free bile acids in lactobacilli and bifidobacteria. J Bacteriol 2006, 188:1979-1986.
    • (2006) J Bacteriol , vol.188 , pp. 1979-1986
    • Kurdi, P.1    Kawanishi, K.2    Mizutani, K.3    Yokota, A.4
  • 93
    • 80054862011 scopus 로고    scopus 로고
    • Bile acid is a host factor that regulates the composition of the cecal microbiota in rats
    • Islam KB, Fukiya S, Hagio M, Fujii N, Ishizuka S, Ooka T, et al. Bile acid is a host factor that regulates the composition of the cecal microbiota in rats. Gastroenterology 2011, 141:1773-1781.
    • (2011) Gastroenterology , vol.141 , pp. 1773-1781
    • Islam, K.B.1    Fukiya, S.2    Hagio, M.3    Fujii, N.4    Ishizuka, S.5    Ooka, T.6
  • 95
    • 33644867569 scopus 로고    scopus 로고
    • Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor
    • Inagaki T, Moschetta A, Lee YK, Peng L, Zhao G, Downes M, et al. Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci U S A 2006, 103:3920-3925.
    • (2006) Proc Natl Acad Sci U S A , vol.103 , pp. 3920-3925
    • Inagaki, T.1    Moschetta, A.2    Lee, Y.K.3    Peng, L.4    Zhao, G.5    Downes, M.6
  • 96
    • 75749122303 scopus 로고    scopus 로고
    • Methods in mammalian autophagy research
    • Mizushima N, Yoshimori T, Levine B. Methods in mammalian autophagy research. Cell 2010, 140:313-326.
    • (2010) Cell , vol.140 , pp. 313-326
    • Mizushima, N.1    Yoshimori, T.2    Levine, B.3
  • 97
    • 0034537290 scopus 로고    scopus 로고
    • Autophagy as a regulated pathway of cellular degradation
    • Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science 2000, 290:1717-1721.
    • (2000) Science , vol.290 , pp. 1717-1721
    • Klionsky, D.J.1    Emr, S.D.2
  • 98
    • 0034329418 scopus 로고    scopus 로고
    • LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing
    • Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J 2000, 19:5720-5728.
    • (2000) EMBO J , vol.19 , pp. 5720-5728
    • Kabeya, Y.1    Mizushima, N.2    Ueno, T.3    Yamamoto, A.4    Kirisako, T.5    Noda, T.6
  • 99
    • 0035286734 scopus 로고    scopus 로고
    • Molecular dissection of autophagy: two ubiquitin-like systems
    • Ohsumi Y. Molecular dissection of autophagy: two ubiquitin-like systems. Nat Rev Mol Cell Biol 2001, 2:211-216.
    • (2001) Nat Rev Mol Cell Biol , vol.2 , pp. 211-216
    • Ohsumi, Y.1
  • 101
    • 77951214016 scopus 로고    scopus 로고
    • Mammalian autophagy: core molecular machinery and signaling regulation
    • Yang Z, Klionsky DJ. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol 2010, 22:124-131.
    • (2010) Curr Opin Cell Biol , vol.22 , pp. 124-131
    • Yang, Z.1    Klionsky, D.J.2
  • 102
    • 84881566784 scopus 로고    scopus 로고
    • Role of p62/SQSTM1 in liver physiology and pathogenesis
    • Manley S, Williams JA, Ding WX. Role of p62/SQSTM1 in liver physiology and pathogenesis. Exp Biol Med 2013, 238:525-538.
    • (2013) Exp Biol Med , vol.238 , pp. 525-538
    • Manley, S.1    Williams, J.A.2    Ding, W.X.3
  • 103
    • 36849089101 scopus 로고    scopus 로고
    • Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice
    • Komatsu M, Waguri S, Koike M, Sou YS, Ueno T, Hara T, et al. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 2007, 131:1149-1163.
    • (2007) Cell , vol.131 , pp. 1149-1163
    • Komatsu, M.1    Waguri, S.2    Koike, M.3    Sou, Y.S.4    Ueno, T.5    Hara, T.6
  • 104
    • 84861466718 scopus 로고    scopus 로고
    • Liver-specific loss of Atg5 causes persistent activation of NRF2 and protects against acetaminophen-induced liver injury
    • Ni HM, Boggess N, McGill MR, Lebofsky M, Borude P, Apte U, et al. Liver-specific loss of Atg5 causes persistent activation of NRF2 and protects against acetaminophen-induced liver injury. Toxicol Sci 2012, 127:438-450.
    • (2012) Toxicol Sci , vol.127 , pp. 438-450
    • Ni, H.M.1    Boggess, N.2    McGill, M.R.3    Lebofsky, M.4    Borude, P.5    Apte, U.6
  • 105
    • 60849099049 scopus 로고    scopus 로고
    • A role for NBR1 in autophagosomal degradation of ubiquitinated substrates
    • Kirkin V, Lamark T, Sou YS, Bjørkøy G, Nunn JL, Bruun JA, et al. A role for NBR1 in autophagosomal degradation of ubiquitinated substrates. Mol Cell 2009, 33:505-516.
    • (2009) Mol Cell , vol.33 , pp. 505-516
    • Kirkin, V.1    Lamark, T.2    Sou, Y.S.3    Bjørkøy, G.4    Nunn, J.L.5    Bruun, J.A.6
  • 107
    • 84867773087 scopus 로고    scopus 로고
    • Mitophagy: mechanisms, pathophysiological roles, and analysis
    • Ding WX, Yin XM. Mitophagy: mechanisms, pathophysiological roles, and analysis. Biol Chem 2012, 393:547-564.
    • (2012) Biol Chem , vol.393 , pp. 547-564
    • Ding, W.X.1    Yin, X.M.2
  • 109
    • 43949123866 scopus 로고    scopus 로고
    • Autophagy in the liver
    • Yin XM, Ding WX, Gao W. Autophagy in the liver. Hepatology 2008, 47:1773-1785.
    • (2008) Hepatology , vol.47 , pp. 1773-1785
    • Yin, X.M.1    Ding, W.X.2    Gao, W.3
  • 110
    • 79958847617 scopus 로고    scopus 로고
    • Role of autophagy in liver physiology and pathophysiology
    • Ding WX. Role of autophagy in liver physiology and pathophysiology. World J Biol Chem 2010, 1:3-12.
    • (2010) World J Biol Chem , vol.1 , pp. 3-12
    • Ding, W.X.1
  • 111
    • 79551554585 scopus 로고    scopus 로고
    • Selective taste of ethanol-induced autophagy for mitochondria and lipid droplets
    • Ding WX, Li M, Yin XM. Selective taste of ethanol-induced autophagy for mitochondria and lipid droplets. Autophagy 2011, 7:248-249.
    • (2011) Autophagy , vol.7 , pp. 248-249
    • Ding, W.X.1    Li, M.2    Yin, X.M.3
  • 112
    • 78049467743 scopus 로고    scopus 로고
    • Autophagy reduces acute ethanol-induced hepatotoxicity and steatosis in mice
    • Ding WX, Li M, Chen X, Ni HM, Lin CW, Gao W, et al. Autophagy reduces acute ethanol-induced hepatotoxicity and steatosis in mice. Gastroenterology 2010, 139:1740-1752.
    • (2010) Gastroenterology , vol.139 , pp. 1740-1752
    • Ding, W.X.1    Li, M.2    Chen, X.3    Ni, H.M.4    Lin, C.W.5    Gao, W.6
  • 113
    • 84876287362 scopus 로고    scopus 로고
    • Pharmacological promotion of autophagy alleviates steatosis and injury in alcoholic and non-alcoholic fatty liver conditions in mice
    • Lin CW, Zhang H, Li M, Xiong X, Chen X, Chen X, et al. Pharmacological promotion of autophagy alleviates steatosis and injury in alcoholic and non-alcoholic fatty liver conditions in mice. J Hepatol 2013, 58:993-999.
    • (2013) J Hepatol , vol.58 , pp. 993-999
    • Lin, C.W.1    Zhang, H.2    Li, M.3    Xiong, X.4    Chen, X.5    Chen, X.6
  • 114
    • 34548289502 scopus 로고    scopus 로고
    • Dynamic FoxO transcription factors
    • Huang H, Tindall DJ. Dynamic FoxO transcription factors. J Cell Sci 2007, 120:2479-2487.
    • (2007) J Cell Sci , vol.120 , pp. 2479-2487
    • Huang, H.1    Tindall, D.J.2
  • 115
    • 41849087003 scopus 로고    scopus 로고
    • FOXO animal models reveal a variety of diverse roles for FOXO transcription factors
    • Arden KC. FOXO animal models reveal a variety of diverse roles for FOXO transcription factors. Oncogene 2008, 27:2345-2350.
    • (2008) Oncogene , vol.27 , pp. 2345-2350
    • Arden, K.C.1
  • 116
    • 84880415788 scopus 로고    scopus 로고
    • Forkhead box class O transcription factors in liver function and disease
    • Tikhanovich I, Cox J, Weinman SA. Forkhead box class O transcription factors in liver function and disease. J Gastroenterol Hepatol 2013, 28 Suppl 1:S125-S131.
    • (2013) J Gastroenterol Hepatol , pp. S125-S131
    • Tikhanovich, I.1    Cox, J.2    Weinman, S.A.3
  • 117
    • 80052970809 scopus 로고    scopus 로고
    • FoxO transcription factors; regulation by AKT and 14-3-3 proteins
    • Tzivion G, Dobson M, Ramakrishnan G. FoxO transcription factors; regulation by AKT and 14-3-3 proteins. Biochim Biophys Acta 2011, 1813:1938-1945.
    • (2011) Biochim Biophys Acta , vol.1813 , pp. 1938-1945
    • Tzivion, G.1    Dobson, M.2    Ramakrishnan, G.3
  • 118
    • 79953186624 scopus 로고    scopus 로고
    • FoxO transcription factors promote cardiomyocyte survival upon induction of oxidative stress
    • Sengupta A, Molkentin JD, Paik JH, DePinho RA, Yutzey KE. FoxO transcription factors promote cardiomyocyte survival upon induction of oxidative stress. J Biol Chem 2011, 286:7468-7478.
    • (2011) J Biol Chem , vol.286 , pp. 7468-7478
    • Sengupta, A.1    Molkentin, J.D.2    Paik, J.H.3    DePinho, R.A.4    Yutzey, K.E.5
  • 119
    • 70350500068 scopus 로고    scopus 로고
    • FoxO transcription factors promote autophagy in cardiomyocytes
    • Sengupta A, Molkentin JD, Yutzey KE. FoxO transcription factors promote autophagy in cardiomyocytes. J Biol Chem 2009, 284:28319-28331.
    • (2009) J Biol Chem , vol.284 , pp. 28319-28331
    • Sengupta, A.1    Molkentin, J.D.2    Yutzey, K.E.3
  • 121
    • 36448968532 scopus 로고    scopus 로고
    • FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells
    • Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, et al. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab 2007, 6:472-483.
    • (2007) Cell Metab , vol.6 , pp. 472-483
    • Zhao, J.1    Brault, J.J.2    Schild, A.3    Cao, P.4    Sandri, M.5    Schiaffino, S.6
  • 122
  • 123
    • 84894798787 scopus 로고    scopus 로고
    • Regulation of FOXO3 by phosphorylation and methylation in hepatitis C virus infection and alcohol exposure
    • Tikhanovich I, Kuravi S, Campbell RV, Kharbanda KK, Artigues A, Villar MT, et al. Regulation of FOXO3 by phosphorylation and methylation in hepatitis C virus infection and alcohol exposure. Hepatology 2014, 59:58-70.
    • (2014) Hepatology , vol.59 , pp. 58-70
    • Tikhanovich, I.1    Kuravi, S.2    Campbell, R.V.3    Kharbanda, K.K.4    Artigues, A.5    Villar, M.T.6
  • 124
    • 53549104898 scopus 로고    scopus 로고
    • Multiple cyclin kinase inhibitors promote bile acid-induced apoptosis and autophagy in primary hepatocytes via p53-CD95-dependent signaling
    • Zhang G, Park MA, Mitchell C, Walker T, Hamed H, Studer E, et al. Multiple cyclin kinase inhibitors promote bile acid-induced apoptosis and autophagy in primary hepatocytes via p53-CD95-dependent signaling. J Biol Chem 2008, 283:24343-24358.
    • (2008) J Biol Chem , vol.283 , pp. 24343-24358
    • Zhang, G.1    Park, M.A.2    Mitchell, C.3    Walker, T.4    Hamed, H.5    Studer, E.6
  • 125
    • 84859741607 scopus 로고    scopus 로고
    • The decreased expression of Beclin-1 correlates with progression to esophageal adenocarcinoma: the role of deoxycholic acid
    • Roesly HB, Khan MR, Chen HDR, Hill KA, Narendran N, Watts GS, et al. The decreased expression of Beclin-1 correlates with progression to esophageal adenocarcinoma: the role of deoxycholic acid. Am J Physiol Gastrointest Liver Physiol 2012, 302:G864-G872.
    • (2012) Am J Physiol Gastrointest Liver Physiol , vol.302 , pp. G864-G872
    • Roesly, H.B.1    Khan, M.R.2    Chen, H.D.R.3    Hill, K.A.4    Narendran, N.5    Watts, G.S.6
  • 126
    • 70349572089 scopus 로고    scopus 로고
    • Deoxycholate, an endogenous cytotoxin/genotoxin, induces the autophagic stress-survival pathway: implications for colon carcinogenesis
    • Payne CM, Crowley-Skillicorn C, Holubec H, Dvorak K, Moyer MP, Garewal H, et al. Deoxycholate, an endogenous cytotoxin/genotoxin, induces the autophagic stress-survival pathway: implications for colon carcinogenesis. J Toxicol 2009, 2009:785907.
    • (2009) J Toxicol , vol.2009 , pp. 785907
    • Payne, C.M.1    Crowley-Skillicorn, C.2    Holubec, H.3    Dvorak, K.4    Moyer, M.P.5    Garewal, H.6
  • 127
    • 84893396046 scopus 로고    scopus 로고
    • Suppression of autophagic flux by bile acids in hepatocytes
    • Manley S, Ni HM, Kong B, Apte U, Guo G, Ding WX. Suppression of autophagic flux by bile acids in hepatocytes. Toxicol Sci 2014, 137:478-490.
    • (2014) Toxicol Sci , vol.137 , pp. 478-490
    • Manley, S.1    Ni, H.M.2    Kong, B.3    Apte, U.4    Guo, G.5    Ding, W.X.6
  • 128
    • 33645509321 scopus 로고    scopus 로고
    • Farnesoid X receptor is essential for normal glucose homeostasis
    • Ma K, Saha PK, Chan L, Moore DD. Farnesoid X receptor is essential for normal glucose homeostasis. J Clin Invest 2006, 116:1102-1109.
    • (2006) J Clin Invest , vol.116 , pp. 1102-1109
    • Ma, K.1    Saha, P.K.2    Chan, L.3    Moore, D.D.4
  • 129
    • 84858796689 scopus 로고    scopus 로고
    • Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR
    • Calkin AC, Tontonoz P. Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR. Nat Rev Mol Cell Biol 2012, 13:213-224.
    • (2012) Nat Rev Mol Cell Biol , vol.13 , pp. 213-224
    • Calkin, A.C.1    Tontonoz, P.2
  • 131
    • 1942518840 scopus 로고    scopus 로고
    • PPARs and the complex journey to obesity
    • Evans RM, Barish GD, Wang YX. PPARs and the complex journey to obesity. Nat Med 2004, 10:355-361.
    • (2004) Nat Med , vol.10 , pp. 355-361
    • Evans, R.M.1    Barish, G.D.2    Wang, Y.X.3
  • 132
    • 0026551309 scopus 로고
    • Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor
    • Göttlicher M, Widmark E, Li Q, Gustafsson JA. Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor. Proc Natl Acad Sci U S A 1992, 89:4653-4657.
    • (1992) Proc Natl Acad Sci U S A , vol.89 , pp. 4653-4657
    • Göttlicher, M.1    Widmark, E.2    Li, Q.3    Gustafsson, J.A.4
  • 133
    • 0027447461 scopus 로고
    • Fatty acids and retinoids control lipid metabolism through activation of peroxisome proliferator-activated receptor-retinoid X receptor heterodimers
    • Keller H, Dreyer C, Medin J, Mahfoudi A, Ozato K, Wahli W. Fatty acids and retinoids control lipid metabolism through activation of peroxisome proliferator-activated receptor-retinoid X receptor heterodimers. Proc Natl Acad Sci U S A 1993, 90:2160-2164.
    • (1993) Proc Natl Acad Sci U S A , vol.90 , pp. 2160-2164
    • Keller, H.1    Dreyer, C.2    Medin, J.3    Mahfoudi, A.4    Ozato, K.5    Wahli, W.6
  • 134
    • 84922968506 scopus 로고    scopus 로고
    • Transcriptional regulation of autophagy by an FXR-CREB axis
    • Seok S, Fu T, Choi SE, Li Y, Zhu R, Kumar S, et al. Transcriptional regulation of autophagy by an FXR-CREB axis. Nature 2014, 516:108-111.
    • (2014) Nature , vol.516 , pp. 108-111
    • Seok, S.1    Fu, T.2    Choi, S.E.3    Li, Y.4    Zhu, R.5    Kumar, S.6
  • 135
    • 84923031534 scopus 로고    scopus 로고
    • Nutrient-sensing nuclear receptors coordinate autophagy
    • Lee JM, Wagner M, Xiao R, Kim KH, Feng D, Lazar MA, et al. Nutrient-sensing nuclear receptors coordinate autophagy. Nature 2014, 516:112-115.
    • (2014) Nature , vol.516 , pp. 112-115
    • Lee, J.M.1    Wagner, M.2    Xiao, R.3    Kim, K.H.4    Feng, D.5    Lazar, M.A.6
  • 137
    • 33847044019 scopus 로고    scopus 로고
    • Spontaneous development of liver tumors in the absence of the bile acid receptor farnesoid X receptor
    • Yang F, Huang X, Yi T, Yen Y, Moore DD, Huang W. Spontaneous development of liver tumors in the absence of the bile acid receptor farnesoid X receptor. Cancer Res 2007, 67:863-867.
    • (2007) Cancer Res , vol.67 , pp. 863-867
    • Yang, F.1    Huang, X.2    Yi, T.3    Yen, Y.4    Moore, D.D.5    Huang, W.6
  • 138
    • 79955492012 scopus 로고    scopus 로고
    • Persistent activation of NRF2 through p62 in hepatocellular carcinoma cells
    • Inami Y, Waguri S, Sakamoto A, Kouno T, Nakada K, Hino O, et al. Persistent activation of NRF2 through p62 in hepatocellular carcinoma cells. J Cell Biol 2011, 193:275-284.
    • (2011) J Cell Biol , vol.193 , pp. 275-284
    • Inami, Y.1    Waguri, S.2    Sakamoto, A.3    Kouno, T.4    Nakada, K.5    Hino, O.6
  • 140
    • 84906315540 scopus 로고    scopus 로고
    • NRF2 promotes the development of fibrosis and tumorigenesis in mice with defective hepatic autophagy
    • Ni HM, Woolbright BL, Williams J, Copple B, Cui W, Luyendyk JP, et al. NRF2 promotes the development of fibrosis and tumorigenesis in mice with defective hepatic autophagy. J Hepatol 2014, 61:617-625.
    • (2014) J Hepatol , vol.61 , pp. 617-625
    • Ni, H.M.1    Woolbright, B.L.2    Williams, J.3    Copple, B.4    Cui, W.5    Luyendyk, J.P.6
  • 141
    • 4644353364 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptor α protects against alcohol-induced liver damage
    • Nakajima T, Kamijo Y, Tanaka N, Sugiyama E, Tanaka E, Kiyosawa K, et al. Peroxisome proliferator-activated receptor α protects against alcohol-induced liver damage. Hepatology 2004, 40:972-980.
    • (2004) Hepatology , vol.40 , pp. 972-980
    • Nakajima, T.1    Kamijo, Y.2    Tanaka, N.3    Sugiyama, E.4    Tanaka, E.5    Kiyosawa, K.6
  • 142
    • 0042847330 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptor alpha (PPARα) agonist treatment reverses PPARα dysfunction and abnormalities in hepatic lipid metabolism in ethanol-fed mice
    • Fischer M, You M, Matsumoto M, Crabb DW. Peroxisome proliferator-activated receptor alpha (PPARα) agonist treatment reverses PPARα dysfunction and abnormalities in hepatic lipid metabolism in ethanol-fed mice. J Biol Chem 2003, 278:27997-28004.
    • (2003) J Biol Chem , vol.278 , pp. 27997-28004
    • Fischer, M.1    You, M.2    Matsumoto, M.3    Crabb, D.W.4
  • 143
    • 84855177721 scopus 로고    scopus 로고
    • Activation of peroxisome proliferator activated receptor alpha ameliorates ethanol induced steatohepatitis in mice
    • Kong L, Ren W, Li W, Zhao S, Mi H, Wang R, et al. Activation of peroxisome proliferator activated receptor alpha ameliorates ethanol induced steatohepatitis in mice. Lipids Health Dis 2011, 10:246.
    • (2011) Lipids Health Dis , vol.10 , pp. 246
    • Kong, L.1    Ren, W.2    Li, W.3    Zhao, S.4    Mi, H.5    Wang, R.6
  • 144
    • 84878606239 scopus 로고    scopus 로고
    • TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop
    • Settembre C, De Cegli R, Mansueto G, Saha PK, Vetrini F, Visvikis O, et al. TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop. Nat Cell Biol 2013, 15:647-658.
    • (2013) Nat Cell Biol , vol.15 , pp. 647-658
    • Settembre, C.1    De Cegli, R.2    Mansueto, G.3    Saha, P.K.4    Vetrini, F.5    Visvikis, O.6
  • 145
    • 84857781617 scopus 로고    scopus 로고
    • Dysregulated phosphorylation and nuclear translocation of cyclic AMP response element binding protein (CREB) in rat liver after chronic ethanol binge
    • Aroor AR, Jackson DE, Shukla SD. Dysregulated phosphorylation and nuclear translocation of cyclic AMP response element binding protein (CREB) in rat liver after chronic ethanol binge. Eur J Pharmacol 2012, 679:101-108.
    • (2012) Eur J Pharmacol , vol.679 , pp. 101-108
    • Aroor, A.R.1    Jackson, D.E.2    Shukla, S.D.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.