Combination of retinoic acid and ursodeoxycholic acid attenuates liver injury in bile duct–ligated rats and human hepatic cells

Cholestasis leads to liver cell death, fibrosis, cirrhosis, and eventually liver failure. Despite limited benefits, ursodeoxycholic acid (UDCA) is the only Food and Drug Administration–approved treatment for cholestatic disorders. Retinoic acid (RA) is a ligand for nuclear receptors that modulate bile salt homeostasis. RA also possesses immunomodulatory effects and is used to treat acute promyelocytic leukemia and inflammatory disorders such as psoriasis, acne, and rheumatoid arthritis. To test whether the supplementation of RA with UDCA is superior to UDCA alone for treating cholestasis, male Sprague‐Dawley rats underwent common bile duct ligation (BDL) for 14 days and were treated with phosphate‐buffered saline (PBS), UDCA, all‐trans retinoic acid (atRA), or UDCA and atRA by gavage. Treatment with UDCA and atRA substantially improved animal growth rates, significantly reduced liver fibrosis and bile duct proliferation, and nearly eliminated liver necrosis after BDL. Reductions in the bile salt pool size and liver hydroxyproline content were also seen with treatment with atRA or atRA and UDCA versus PBS and UDCA. Furthermore, atRA and UDCA significantly reduced liver messenger RNA and/or protein expression of transforming growth factor β1 (Tgf‐β1), collagen 1a1 (Col1A1), matrix metalloproteinase 2 (Mmp2), cytokeratin 19, α‐smooth muscle actin (α‐SMA), cytochrome P450 7A1 (Cyp7a1), tumor necrosis factor α, and interleukin‐β1. The molecular mechanisms of this treatment were also assessed in human hepatocytes, hepatic stellate cells, and LX‐2 cells. atRA alone or in combination with UDCA greatly repressed CYP7A1 expression in human hepatocytes and significantly inhibited COL1A1, MMP2, and α‐SMA expression and/or activity in primary human hepatic stellate cells and LX‐2 cells. Furthermore, atRA reduced TGF‐β1–induced Smad2 phosphorylation in LX‐2 cells. Conclusion: Our findings indicate that the addition of RA to UDCA reduces the bile salt pool size and liver fibrosis and might be an effective supplemental therapy with UDCA for cholestatic diseases. (HEPATOLOGY 2011;53:548‐557.)

[1]  Trong Nguyen,et al.  Retinoic acid represses CYP7A1 expression in human hepatocytes and HepG2 cells by FXR/RXR-dependent and independent mechanisms , 2010, Journal of Lipid Research.

[2]  J. Dranoff,et al.  Portal fibroblasts: Underappreciated mediators of biliary fibrosis , 2010, Hepatology.

[3]  G. Gores,et al.  Diagnosis and management of primary sclerosing cholangitis , 2010, Hepatology.

[4]  K. Lindor,et al.  Latest and Emerging Therapies for Primary Biliary Cirrhosis and Primary Sclerosing Cholangitis , 2010, Current gastroenterology reports.

[5]  J. Boyer,et al.  Mouse organic solute transporter α deficiency enhances renal excretion of bile acids and attenuates cholestasis , 2010, Hepatology.

[6]  M. Trojanowska Noncanonical transforming growth factor β signaling in scleroderma fibrosis , 2009, Current opinion in rheumatology.

[7]  J. Chiang,et al.  Bile acids: regulation of synthesis , 2009, Journal of Lipid Research.

[8]  C. Steer,et al.  Bile acids: regulation of apoptosis by ursodeoxycholic acid , 2009, Journal of Lipid Research.

[9]  A. Rosato,et al.  Retinoids as critical modulators of immune functions: new therapeutic perspectives for old compounds. , 2009, Endocrine, metabolic & immune disorders drug targets.

[10]  T. Roskams,et al.  Vinculin and cellular retinol-binding protein-1 are markers for quiescent and activated hepatic stellate cells in formalin-fixed paraffin embedded human liver , 2009, Histochemistry and Cell Biology.

[11]  K. Lindor,et al.  Treatment of primary biliary cirrhosis: therapy with choleretic and immunosuppressive agents. , 2008, Clinics in liver disease.

[12]  F. Marra,et al.  Myofibroblast - like cells and liver fibrogenesis: Emerging concepts in a rapidly moving scenario. , 2008, Molecular aspects of medicine.

[13]  A. Befeler,et al.  High-dose ursodeoxycholic acid for the treatment of primary sclerosing cholangitis , 2007, Current gastroenterology reports.

[14]  J. Reichrath,et al.  Vitamins as hormones. , 2007, Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme.

[15]  T. Pusl,et al.  Ursodeoxycholic acid treatment of vanishing bile duct syndromes. , 2006, World journal of gastroenterology.

[16]  J. Boyer,et al.  Mrp4−/− mice have an impaired cytoprotective response in obstructive cholestasis , 2006, Hepatology.

[17]  G. Svegliati-Baroni,et al.  Bile acids induce hepatic stellate cell proliferation via activation of the epidermal growth factor receptor. , 2005, Gastroenterology.

[18]  S. Bellentani Immunomodulating and anti-apoptotic action of ursodeoxycholic acid: where are we and where should we go? , 2005, European journal of gastroenterology & hepatology.

[19]  D. Schuppan,et al.  Differential modulation of rat hepatic stellate phenotype by natural and synthetic retinoids , 2004, Hepatology.

[20]  M. Reiss,et al.  Smads 2 and 3 are differentially activated by TGF-β in quiescent and activated hepatic stellate cells: Constitutive nuclear localization of Smads in activated cells is TGF-β independent , 2003 .

[21]  F. Lammert,et al.  Ursodeoxycholic acid aggravates bile infarcts in bile duct-ligated and Mdr2 knockout mice via disruption of cholangioles. , 2002, Gastroenterology.

[22]  Jatinder Lamba,et al.  Disrupted Bile Acid Homeostasis Reveals an Unexpected Interaction among Nuclear Hormone Receptors, Transporters, and Cytochrome P450* , 2001, The Journal of Biological Chemistry.

[23]  D. Mangelsdorf,et al.  Regulation of absorption and ABC1-mediated efflux of cholesterol by RXR heterodimers. , 2000, Science.

[24]  H. Schnaper,et al.  The transforming growth factor-βbgr/SMAD signaling pathway is present and functional in human mesangial cells , 1999 .

[25]  W. Stetler-Stevenson,et al.  Quantitative zymography: detection of picogram quantities of gelatinases. , 1994, Analytical biochemistry.

[26]  S. Milani,et al.  Regulation of extracellular matrix synthesis by transforming growth factor beta 1 in human fat-storing cells. , 1993, Gastroenterology.

[27]  H. Abboud,et al.  Phenotypical modulation of liver fat-storing cells by retinoids. Influence on unstimulated and growth factor-induced cell proliferation. , 1992, Journal of hepatology.

[28]  S. Friedman,et al.  Isolated hepatic lipocytes and kupffer cells from normal human liver: Morphological and functional characteristics in primary culture , 1992, Hepatology.

[29]  M. Kaplan,et al.  Primary biliary cirrhosis , 1998, Hepatology.

[30]  N. Davidson,et al.  Retinoic acid modulates rat Ito cell proliferation, collagen, and transforming growth factor beta production. , 1990, The Journal of clinical investigation.

[31]  川田 一仁 Enhanced hepatic Nrf2 activation after ursodeoxycholic acid treatment in patients with primary biliary cirrhosis , 2010 .

[32]  久森 重夫 All-trans-retinoic acid ameliorates carbon tetrachloride-induced liver fibrosis in mice through modulating cytokine production , 2010 .

[33]  Hui Wang 王 晖,et al.  Effect of all-trans retinoic acid on liver fibrosis induced by common bile duct ligation in rats , 2008, Journal of Huazhong University of Science and Technology [Medical Sciences].

[34]  M. Reiss,et al.  Smads 2 and 3 are differentially activated by transforming growth factor-beta (TGF-beta ) in quiescent and activated hepatic stellate cells. Constitutive nuclear localization of Smads in activated cells is TGF-beta-independent. , 2003, The Journal of biological chemistry.

[35]  H. Schnaper,et al.  The transforming growth factor-beta/SMAD signaling pathway is present and functional in human mesangial cells. , 1999, Kidney international.

[36]  K. Lazaridis,et al.  Primary biliary cirrhosis , 1998, Springer Netherlands.