Warning: fopen(/home/virtual/enm-kes/journal/upload/ip_log/ip_log_2024-04.txt): failed to open stream: Permission denied in /home/virtual/lib/view_data.php on line 88 Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 89 Bile Acid Nuclear Receptor Farnesoid X Receptor: Therapeutic Target for Nonalcoholic Fatty Liver Disease
Skip Navigation
Skip to contents

Endocrinol Metab : Endocrinology and Metabolism

clarivate
OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > Endocrinol Metab > Volume 31(4); 2016 > Article
Review Article
Bile Acid Nuclear Receptor Farnesoid X Receptor: Therapeutic Target for Nonalcoholic Fatty Liver Disease
Sun-Gi Kim, Byung-Kwon Kim, Kyumin Kim, Sungsoon Fangorcid
Endocrinology and Metabolism 2016;31(4):500-504.
DOI: https://doi.org/10.3803/EnM.2016.31.4.500
Published online: December 20, 2016

Department of Integrative Biosciences and Biotechnology, College of Life Sciences, Sejong University, Seoul, Korea.

Corresponding author: Sungsoon Fang. Department of Integrative Biosciences and Biotechnology, College of Life Sciences, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Korea. Tel: +82-2-6935-2433, Fax: +82-2-3408-4334, sfang@sejong.ac.kr
• Received: October 31, 2016   • Revised: November 16, 2016   • Accepted: November 20, 2016

Copyright © 2016 Korean Endocrine Society

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 5,644 Views
  • 73 Download
  • 33 Web of Science
  • 33 Crossref
  • 32 Scopus
  • Nonalcoholic fatty liver disease (NAFLD) is one of the causes of fatty liver, occurring when fat is accumulated in the liver without alcohol consumption. NAFLD is the most common liver disorder in advanced countries. NAFLD is a spectrum of pathology involving hepatic steatosis with/without inflammation and nonalcoholic steatohepatitis with accumulation of hepatocyte damage and hepatic fibrosis. Recent studies have revealed that NAFLD results in the progression of cryptogenic cirrhosis that leads to hepatocarcinoma and cardiovascular diseases such as heart failure. The main causes of NAFLD have not been revealed yet, metabolic syndromes including obesity and insulin resistance are widely accepted for the critical risk factors for the pathogenesis of NAFLD. Nuclear receptors (NRs) are transcriptional factors that sense environmental or hormonal signals and regulate expression of genes, involved in cellular growth, development, and metabolism. Several NRs have been reported to regulate genes involved in energy and xenobiotic metabolism and inflammation. Among various NRs, farnesoid X receptor (FXR) is abundantly expressed in the liver and a key regulator to control various metabolic processes in the liver. Recent studies have shown that NAFLD is associated with inappropriate function of FXR. The impact of FXR transcriptional activity in NAFLD is likely to be potential therapeutic strategy, but still requires to elucidate underlying potent therapeutic mechanisms of FXR for the treatment of NAFLD. This article will focus the physiological roles of FXR and establish the correlation between FXR transcriptional activity and the pathogenesis of NAFLD.
Nonalcoholic fatty liver disease (NAFLD) is characterized by the accumulation of lipid droplets in the hepatocytes without alcohol consumption. NAFLD is a common liver disorder and affects 15% to 40% of the population in United States [1]. According to National Health and Nutrition Examination Survey, the rate of NAFLD in chronic liver disorder has been substantially increasing 47% in 1988 to 1994, 63% in 1999 to 2004, and 75% in 2005 to 2008 [2]. Quite interestingly, metabolic disorders including obesity, type 2 diabetes, chronic kidney disease, hypertension, and colorectal malignant neoplasm also have been largely increasing in the patients with NAFLD, implying of positive correlation between NAFLD and metabolic disorders [3456]. The spectrum of NAFLD is very wide, covering hepatic steatosis and nonalcoholic steatohepatitis (NASH) which resulting in eventually hepatic cirrhosis and hepatocellular carcinoma. Among the NAFLD patients, at least 10% to 20% would develop to NASH, which is a serious condition of liver disorders [7]. To date, no optimal treatment has been established for NAFLD. Thus, it is required to understand the pathogenesis of NAFLD and to determine potential therapeutic target for effective pharmacological treatment for NAFLD.
Nuclear receptors (NRs) are ligand-activated transcriptional factors that broadly regulate genes involved in metabolism, xenobiotics and cellular growth and cycle. NRs are sensitive to many natural and synthetic ligands including steroid hormones, lipids and fatty acids, vitamins, drugs, and various metabolites. Recently, numerous reports have demonstrated that farnesoid X receptor (FXR) is a bile acid NR and regulates hepatic gluconeogenesis and lipogenesis and hepatic inflammation to maintain metabolic homeostasis in the liver [8].
Though pathogenesis of NAFLD still remains unclear, “two-hit” theory has been proposed to explain the progression [9]. The “first hit” is hepatic steatosis that excessive lipid accumulation in hepatocytes is accompanied by elevated de novo lipogenesis and fatty acid uptake. Subsequently, oxidative stress and hepatic inflammation are critical two factors of the “second hit” which cause remarkable hepatic cellular damage. Besides, multiple hits including genetic mutations and intestinal microbiome also account for the progression of NAFLD [10].
FXR belongs to NR superfamily and is a ligand-activated transcriptional regulator, harbouring DNA binding and ligand binding domains [11]. It has been revealed that bile acids are endogenous ligand for FXR and regulate whole body metabolism, including cholesterol/bile acid metabolism, hepatic gluconeogenesis/lipogenesis, and even inflammation [8]. While FXR is abundantly expressed in the liver, intestine, and kidney, low expression of FXR has also been observed in the lung, adipose tissue, and heart [8]. Upon binding with endogenous bile acids, FXR heterodimerizes with its common binding partner, retinoid X receptor (RXR) to bind to the FXR response element (FXRE), in the target gene promoters. Once FXR heterodimerizes with RXR to bind to the FXRE, FXR is able to regulate expressions of its own target genes involved in various biological processes.
FXR regulates bile acid synthesis by cholesterol catabolism. Bile acid synthesis occurs in the hepatocytes and compose of two pathways, such as classic pathway and alternative pathway [12]. The key enzymes in bile acid synthesis are cholesterol-7α-hydroxylase (CYP7A1) and sterol-27-hydroxylase (CYP27A1), respectively. Converted from cholesterol in the hepatocytes, bile acids are conjugated to taurine and/or glycine and then secreted into the gall bladder. Upon food intake, bile acids are secreted into the small intestine, and 95% of secreted bile acids are recycled and transported back to the liver via portal vein. The circulation of bile acids from the intestine to the liver is called bile acid enterohepatic circulation. Thus, 5% of bile acids excluded in each enterohepatic circulation. To compensate loss of bile acids, the liver synthesizes bile acids from cholesterol to maintain equivalent amount of bile acid pool.
As bile acids have the property of detergent, increasing accumulation of bile acids in the hepatocytes leads to cellular toxicity. To protect bile acids-induced cellular toxicity, bile acids binds to FXR to induces small heterodimer partner, which suppresses CYP7A1 gene expression to reduce the rate of bile acid synthesis in the liver. Besides hepatic regulation of bile acids, enterocytes are also involved in the regulation of bile acid synthesis. Upon bile acid activation in the intestine, FXR induces gene expression of fibroblast growth factor 15/19 (FGF15/19) in the enterocytes. Secreted FGF15/19 from enterocytes travel to the liver via portal vein and bind to FGF receptor 4 in the hepatocytes to activate JNK signaling pathway, which in turn suppresses gene expression of CYP7A1. Therefore, FXR activation in both hepatocytes and enterocytes can negatively regulates bile acid synthesis [12].
Recently, FXR has been revealed to regulate expression of genes involved in glucose and lipid metabolism [13]. Phosphoenolpyruvate carboxykinase (PEPCK) is a well-known vital enzyme that catalyzes a crucial step of gluconeogenesis in the hepatocytes. Animal studies using FXR-null mice have shown that impaired FXR signaling pathway induced insulin resistance and elevated hepatic glucose production. Besides PEPCK, FXR also downregulates expression of glucose-6-phosphatase, a key enzyme to release glucose from the hepatocytes to the circulation [141516]. Altogether, FXR plays a key role to regulate glucose homeostasis via suppression of hepatic gluconeogenesis.
Extended studies have reported that FXR-null mice exhibit impaired lipid metabolism. Previous studies have shown that plasma triglyceride (TG) and cholesterol levels are elevated in FXR-null mice. In addition, treatment of FXR agonist, GW4064 in wild type mice largely reduced plasma TGs [1718]. Furthermore, FXR has been shown to regulate a set of genes involved in lipoprotein metabolism including sterol regulatory element binding transcription factor 1, phospholipid transfer protein, stearoylcoenzyme A desaturase 1, very low density lipoprotein receptor, apolipoprotein C2, and apolipoprotein E, suggesting that FXR plays a key role to regulate hepatic lipid metabolism [17].
In addition to glucose/lipid metabolism, FXR also has anti-inflammatory action in the liver, mainly by supressing nuclear factor-κB signaling pathways. In nurine model of NAFLD, FXR agonist GW4064 treatment reduces hepatic proinflammatory cytokine expressions [19]. Furthermore, FXR activation directly reduces lipopolysaccharide-induced proinflammatory cytokine expressions in macrophage [19], suggesting that FXR directly reduces inflammatory responses in immune cells. Consistent with that FXR directly suppresses inflammation, previous reports have shown that severe intestinal inflammation was observed in bile duct obstruction model with FXR-null mice [20]. Altogether, anti-inflammatory property of FXR proposes that FXR activation would be able to inhibit the progression of NASH from NAFLD by suppression of hepatic inflammation.
To date, no effective treatment has been established to manage the progression of NAFLD. As FXR plays critical roles to regulate various hepatic metabolism and inflammation, there is an emerging idea that FXR is an ideal therapeutic target for the treatment of NAFLD. Consistently, numerous natural and synthetic ligands of FXR have shown protective roles in rodent models of NAFLD.
GW4064 is a nonsteroidal FXR synthetic agonist and has been reported to reduce hepatic gluconeogenesis/lipogenesis and weight gain in rodent obese model. FXR activation by GW4064 represses hepatic steatosis by lowering TG and free fatty acid level in the liver, indicating that GW4064 fatty acid level in the liver, indicating that GW4064 has a therapeutic potential in NAFLD with its property to suppress hepatic lipogenesis [21].
Obeticholic acid (OCA or INT-747, 6α-ethyl-chenodeoxycholic acid) is a semisynthetic derivative of chenodeoxycholic acid found in primary bile acid of human and rodents which is the natural ligand of FXR. Administration of OCA reduces hepatic steatosis and insulin resistance in Zucker obese rats by reducing body weight gain and fat deposit in the liver [22]. OCA treatment largely reduces hepatic gluconeogenesis and lipogenesis, as well as hepatic inflammation. In addition to hepatic inflammation, FXR activation by OCA also reduced intestinal inflammation in experimental colitis rodent model [23]. Recently, administration of OCA has been significantly effective in the patients with type 2 diabetes mellitus and NAFLD (ClinicalTrials. gov, No. NCT00501592). In these patients, OCA treatment significantly improved insulin sensitivity while reduced hepatic damage marker alanine transferase and hepatic fibrosis [24].
Fexaramine is a synthetic FXR agonist and its chemical structure is markedly distinguished from natural bile acids and GW4064. Even target gene expression profile by fexaramine is distinct from those by natural bile acids and GW4064 [25]. Recently, fexaramine has been shown as gut-restricted FXR agonist to activate FXR in the intestine by oral administration [26]. Oral treatment of fexaramine significantly reduced body weight gain and inflammation in rodent obese model. Interestingly, hepatic steatosis and alanine transaminase level were largely decreased by fexaramine treatment, implying that intestinal FXR agonism would be potent therapeutic strategy to treat NAFLD [26].
FXR plays crucial roles in diverse physiological processes associated with cholesterol/bile acid metabolism, glucose/lipid metabolism, and inflammation, and has beneficial effects to reduce pathogenesis of NAFLD. Numerous studies have proved that FXR agonist or FXR agonism may have therapeutic potentials for the prevention and treatment of NAFLD. However, the role of FXR to regulate cholesterol metabolism is still debatable: the plasma high density lipoprotein (HDL) level was elevated in FXR-null mice, whereas inhibition of FXR transcriptional activity decreased plasma low density lipoprotein level and increased HDL level [27]. Consistently, FXR-null mice were protected from diet-induced obesity, glucose tolerance, and hepatic steatosis [28]. Thus, the physiological impact of FXR agonism on cholesterol metabolism requires to be elucidated in detail.
In summary, numerous studies on therapeutic potentials of FXR for the treatment of NAFLD have provided new avenues to develop novel therapeutic strategy for NAFLD. Elucidating and understanding the molecular mechanisms during the pathogenesis of NAFLD would promote development of effective treatment for NAFLD. Although effectiveness of FXR activation on NAFLD is still debatable, FXR is an emerging therapeutic molecular target to manage NAFLD. Further attention and clinical evidence about FXR needs to be paid to avoid undesirable contradictory effects.
Acknowledgements
This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT & Future Planning (NRF-2015R1C1A1A01052195).

CONFLICTS OF INTEREST: No potential conflict of interest relevant to this article was reported.

  • 1. Williams CD, Stengel J, Asike MI, Torres DM, Shaw J, Contreras M, et al. Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle-aged population utilizing ultrasound and liver biopsy: a prospective study. Gastroenterology 2011;140:124–131. ArticlePubMed
  • 2. Lazo M, Hernaez R, Eberhardt MS, Bonekamp S, Kamel I, Guallar E, et al. Prevalence of nonalcoholic fatty liver disease in the United States: the Third National Health and Nutrition Examination Survey, 1988-1994. Am J Epidemiol 2013;178:38–45. ArticlePubMedPMCPDF
  • 3. Bonapace S, Valbusa F, Bertolini L, Pichiri I, Mantovani A, Rossi A, et al. Nonalcoholic fatty liver disease is associated with aortic valve sclerosis in patients with type 2 diabetes mellitus. PLoS One 2014;9:e88371ArticlePubMedPMC
  • 4. El Azeem HA, Khalek el-SA, El-Akabawy H, Naeim H, Khalik HA, Alfifi AA. Association between nonalcoholic fatty liver disease and the incidence of cardiovascular and renal events. J Saudi Heart Assoc 2013;25:239–246. ArticlePubMedPMC
  • 5. Lin XF, Shi KQ, You J, Liu WY, Luo YW, Wu FL, et al. Increased risk of colorectal malignant neoplasm in patients with nonalcoholic fatty liver disease: a large study. Mol Biol Rep 2014;41:2989–2997. ArticlePubMedPDF
  • 6. Targher G, Chonchol M, Zoppini G, Abaterusso C, Bonora E. Risk of chronic kidney disease in patients with non-alcoholic fatty liver disease: is there a link? J Hepatol 2011;54:1020–1029. ArticlePubMed
  • 7. Sanyal AJ. NASH: a global health problem. Hepatol Res 2011;41:670–674. ArticlePubMed
  • 8. Lee FY, Lee H, Hubbert ML, Edwards PA, Zhang Y. FXR, a multipurpose nuclear receptor. Trends Biochem Sci 2006;31:572–580. ArticlePubMed
  • 9. Day CP, James OF. Steatohepatitis: a tale of two “hits”? Gastroenterology 1998;114:842–845. ArticlePubMed
  • 10. Mantena SK, King AL, Andringa KK, Eccleston HB, Bailey SM. Mitochondrial dysfunction and oxidative stress in the pathogenesis of alcohol- and obesity-induced fatty liver diseases. Free Radic Biol Med 2008;44:1259–1272. ArticlePubMedPMC
  • 11. 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. ArticlePubMed
  • 12. Chiang JY. Bile acid regulation of gene expression: roles of nuclear hormone receptors. Endocr Rev 2002;23:443–463. ArticlePubMedPDF
  • 13. Claudel T, Staels B, Kuipers F. The farnesoid X receptor: a molecular link between bile acid and lipid and glucose metabolism. Arterioscler Thromb Vasc Biol 2005;25:2020–2030. ArticlePubMed
  • 14. Stayrook KR, Bramlett KS, Savkur RS, Ficorilli J, Cook T, Christe ME, et al. Regulation of carbohydrate metabolism by the farnesoid X receptor. Endocrinology 2005;146:984–991. ArticlePubMedPDF
  • 15. Duran-Sandoval D, Cariou B, Percevault F, Hennuyer N, Grefhorst A, van Dijk TH, et al. The farnesoid X receptor modulates hepatic carbohydrate metabolism during the fasting-refeeding transition. J Biol Chem 2005;280:29971–29979. ArticlePubMed
  • 16. Ma K, Saha PK, Chan L, Moore DD. Farnesoid X receptor is essential for normal glucose homeostasis. J Clin Invest 2006;116:1102–1109. ArticlePubMedPMC
  • 17. Lambert G, Amar MJ, Guo G, Brewer HB Jr, Gonzalez FJ, Sinal CJ. The farnesoid X-receptor is an essential regulator of cholesterol homeostasis. J Biol Chem 2003;278:2563–2570. ArticlePubMed
  • 18. Zhang Y, Lee FY, Barrera G, Lee H, Vales C, Gonzalez FJ, et al. Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc Natl Acad Sci U S A 2006;103:1006–1011. ArticlePubMedPMC
  • 19. Yao J, Zhou CS, Ma X, Fu BQ, Tao LS, Chen M, et al. FXR agonist GW4064 alleviates endotoxin-induced hepatic inflammation by repressing macrophage activation. World J Gastroenterol 2014;20:14430–14441. ArticlePubMedPMC
  • 20. 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. ArticlePubMedPMC
  • 21. Ma Y, Huang Y, Yan L, Gao M, Liu D. Synthetic FXR agonist GW4064 prevents diet-induced hepatic steatosis and insulin resistance. Pharm Res 2013;30:1447–1457. ArticlePubMedPMCPDF
  • 22. Cipriani S, Mencarelli A, Palladino G, Fiorucci S. FXR activation reverses insulin resistance and lipid abnormalities and protects against liver steatosis in Zucker (fa/fa) obese rats. J Lipid Res 2010;51:771–784. ArticlePubMedPMC
  • 23. Gadaleta RM, van Erpecum KJ, Oldenburg B, Willemsen EC, Renooij W, Murzilli S, et al. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut 2011;60:463–472. ArticlePubMed
  • 24. Mudaliar S, Henry RR, Sanyal AJ, Morrow L, Marschall HU, Kipnes M, et al. Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology 2013;145:574–582.e1. ArticlePubMed
  • 25. Downes M, Verdecia MA, Roecker AJ, Hughes R, Hogenesch JB, Kast-Woelbern HR, et al. A chemical, genetic, and structural analysis of the nuclear bile acid receptor FXR. Mol Cell 2003;11:1079–1092. ArticlePubMedPMC
  • 26. Fang S, Suh JM, Reilly SM, Yu E, Osborn O, Lackey D, et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat Med 2015;21:159–165. ArticlePubMedPMCPDF
  • 27. Urizar NL, Liverman AB, Dodds DT, Silva FV, Ordentlich P, Yan Y, et al. A natural product that lowers cholesterol as an antagonist ligand for FXR. Science 2002;296:1703–1706. ArticlePubMed
  • 28. Prawitt J, Abdelkarim M, Stroeve JH, Popescu I, Duez H, Velagapudi VR, et al. Farnesoid X receptor deficiency improves glucose homeostasis in mouse models of obesity. Diabetes 2011;60:1861–1871. ArticlePubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Geniposide plus chlorogenic acid reverses non-alcoholic steatohepatitis via regulation of gut microbiota and bile acid signaling in a mouse model in vivo
      Hongshan Li, Yingfei Xi, Xin Xin, Qin Feng, Yiyang Hu
      Frontiers in Pharmacology.2023;[Epub]     CrossRef
    • DOT1L Epigenetically Regulates Autophagy and Mitochondria Fusion in Cell Lines of Renal Cancer
      Yanguang Hou, Jiachen Liu, Shiyu Huang, Lei Wang, Juncheng Hu, Xiuheng Liu
      Technology in Cancer Research & Treatment.2023;[Epub]     CrossRef
    • Insights into the molecular targets and emerging pharmacotherapeutic interventions for nonalcoholic fatty liver disease
      Chander K. Negi, Pavel Babica, Lola Bajard, Julie Bienertova-Vasku, Giovanni Tarantino
      Metabolism.2022; 126: 154925.     CrossRef
    • FXR: structures, biology, and drug development for NASH and fibrosis diseases
      Si-yu Tian, Shu-ming Chen, Cheng-xi Pan, Yong Li
      Acta Pharmacologica Sinica.2022; 43(5): 1120.     CrossRef
    • Non-alcoholic fatty liver disease development: A multifactorial pathogenic phenomena
      Aamir Bashir, Ajay Duseja, Arka De, Manu Mehta, Pramil Tiwari
      Liver Research.2022; 6(2): 72.     CrossRef
    • The effects of quercetin on the expression of SREBP-1c mRNA in high-fat diet-induced NAFLD in mice
      Jamal Nasser Saleh Al-maamari, Mahardian Rahmadi, Sisca Melani Panggono, Devita Ardina Prameswari, Eka Dewi Pratiwi, Chrismawan Ardianto, Santhra Segaran Balan, Budi Suprapti
      Journal of Basic and Clinical Physiology and Pharmacology.2021; 32(4): 637.     CrossRef
    • Co‑administration of obeticholic acid and simvastatin protects against high‑fat diet‑induced non‑alcoholic steatohepatitis in mice
      Wen-Cong Li, Su-Xian Zhao, Wei-Guang Ren, Yu-Guo Zhang, Rong-Qi Wang, Ling-Bo Kong, Qing-Shan Zhang, Yue-Min Nan
      Experimental and Therapeutic Medicine.2021;[Epub]     CrossRef
    • Gypenosides regulate farnesoid X receptor-mediated bile acid and lipid metabolism in a mouse model of non-alcoholic steatohepatitis
      Hongshan Li, Yingfei Xi, Xin Xin, Huajie Tian, Yiyang Hu
      Nutrition & Metabolism.2020;[Epub]     CrossRef
    • Molecular mechanisms of hepatic insulin resistance in nonalcoholic fatty liver disease and potential treatment strategies
      Chang-hua Zhang, Bu-gao Zhou, Jun-qing Sheng, Yang Chen, Ying-qian Cao, Chen Chen
      Pharmacological Research.2020; 159: 104984.     CrossRef
    • Farnesoid X Receptor Activation Protects Liver From Ischemia/Reperfusion Injury by Up‐Regulating Small Heterodimer Partner in Kupffer Cells
      Dan Jin, Tianfei Lu, Ming Ni, Han Wang, Jiang Zhang, Chenpeng Zhong, Chuan Shen, Jun Hao, Ronald W. Busuttil, Jerzy W. Kupiec‐Weglinski, Jianjun Zhang, Ning Xu, Yuan Zhai
      Hepatology Communications.2020; 4(4): 540.     CrossRef
    • Salidroside improves high-fat diet-induced non-alcoholic steatohepatitis by regulating the gut microbiota–bile acid–farnesoid X receptor axis
      Hongshan Li, Yingfei Xi, Xin Xin, Huajie Tian, Yiyang Hu
      Biomedicine & Pharmacotherapy.2020; 124: 109915.     CrossRef
    • An Overview of Lipid Metabolism and Nonalcoholic Fatty Liver Disease
      Ke Pei, Ting Gui, Dongfang Kan, Huichao Feng, Yanqiang Jin, Ying Yang, Qian Zhang, Ziwei Du, Zhibo Gai, Jibiao Wu, Yunlun Li
      BioMed Research International.2020; 2020: 1.     CrossRef
    • Intestinal Barrier Function–Non-alcoholic Fatty Liver Disease Interactions and Possible Role of Gut Microbiota
      Yizhe Cui, Qiuju Wang, Renxu Chang, Xiaocui Zhou, Chuang Xu
      Journal of Agricultural and Food Chemistry.2019; 67(10): 2754.     CrossRef
    • A novel ASBT inhibitor, IMB17-15, repressed nonalcoholic fatty liver disease development in high-fat diet-fed Syrian golden hamsters
      Mao-xu Ge, Wei-xiao Niu, Jin-feng Ren, Shi-ying Cai, Dong-ke Yu, Hong-tao Liu, Na Zhang, Yi-xuan Zhang, Yu-cheng Wang, Rong-guang Shao, Ju-xian Wang, Hong-wei He
      Acta Pharmacologica Sinica.2019; 40(7): 895.     CrossRef
    • Consenso mexicano de la enfermedad por hígado graso no alcohólico
      R. Bernal-Reyes, G. Castro-Narro, R. Malé-Velázquez, R. Carmona-Sánchez, M.S. González-Huezo, I. García-Juárez, N. Chávez-Tapia, C. Aguilar-Salinas, I. Aiza-Haddad, M.A. Ballesteros-Amozurrutia, F. Bosques-Padilla, M. Castillo-Barradas, J.A. Chávez-Barrer
      Revista de Gastroenterología de México.2019; 84(1): 69.     CrossRef
    • Fish oil alleviates circadian bile composition dysregulation in male mice with NAFLD
      Yang Liu, Qi Li, Hualin Wang, Xiuju Zhao, Na Li, Hongyu Zhang, Guoxun Chen, Zhiguo Liu
      The Journal of Nutritional Biochemistry.2019; 69: 53.     CrossRef
    • Effects of sanshoamides and capsaicinoids on plasma and liver lipid metabolism in hyperlipidemic rats
      Zhaojun Chen, Yongxiang Liu, Hui Wang, Zhongai Chen, Jia Liu, Hui Liu
      Food Science and Biotechnology.2019; 28(2): 519.     CrossRef
    • The Mexican consensus on nonalcoholic fatty liver disease
      R. Bernal-Reyes, G. Castro-Narro, R. Malé-Velázquez, R. Carmona-Sánchez, M.S. González-Huezo, I. García-Juárez, N. Chávez-Tapia, C. Aguilar-Salinas, I. Aiza-Haddad, M.A. Ballesteros-Amozurrutia, F. Bosques-Padilla, M. Castillo-Barradas, J.A. Chávez-Barrer
      Revista de Gastroenterología de México (English Edition).2019; 84(1): 69.     CrossRef
    • Role of bile acids in the diagnosis and progression of liver cirrhosis: A prospective observational study
      Ning Liu, Jiao Feng, Yang Lv, Qing Liu, Jingfan Deng, Yujing Xia, Chuanyong Guo, Yingqun Zhou
      Experimental and Therapeutic Medicine.2019;[Epub]     CrossRef
    • Essential role of nuclear receptors for the evaluation of the benefits of bioactive herbal extracts on liver function
      Fengling Wang, Yifan Wu, Xiaoting Xie, Jing Sun, Weidong Chen
      Biomedicine & Pharmacotherapy.2018; 99: 798.     CrossRef
    • Maternal obesity has sex‐dependent effects on insulin, glucose and lipid metabolism and the liver transcriptome in young adult rat offspring
      Consuelo Lomas‐Soria, Luis A. Reyes‐Castro, Guadalupe L. Rodríguez‐González, Carlos A. Ibáñez, Claudia J. Bautista, Laura A. Cox, Peter W. Nathanielsz, Elena Zambrano
      The Journal of Physiology.2018; 596(19): 4611.     CrossRef
    • Protective effect of dioscin against thioacetamide-induced acute liver injury via FXR/AMPK signaling pathway in vivo
      Lingli Zheng, Lianhong Yin, Lina Xu, Yan Qi, Hua Li, Youwei Xu, Xu Han, Kexin Liu, Jinyong Peng
      Biomedicine & Pharmacotherapy.2018; 97: 481.     CrossRef
    • New therapeutic perspectives in non-alcoholic steatohepatitis
      Javier Ampuero, Yolanda Sánchez-Torrijos, Virginia Aguilera, Francisco Bellido, Manuel Romero-Gómez
      Gastroenterología y Hepatología (English Edition).2018; 41(2): 128.     CrossRef
    • Hepatic farnesoid X receptor protein level and circulating fibroblast growth factor 19 concentration in children with NAFLD
      Valerio Nobili, Anna Alisi, Antonella Mosca, Claudia Della Corte, Silvio Veraldi, Rita De Vito, Cristiano De Stefanis, Valentina D'Oria, Joerg Jahnel, Evelyn Zohrer, Eleonora Scorletti, Christopher D. Byrne
      Liver International.2018; 38(2): 342.     CrossRef
    • The use of obeticholic acid for the management of non-viral liver disease: current clinical practice and future perspectives
      Stefano Gitto, Valeria Guarneri, Alessandro Sartini, Pietro Andreone
      Expert Review of Gastroenterology & Hepatology.2018; 12(2): 165.     CrossRef
    • Natural products as modulators of the nuclear receptors and metabolic sensors LXR, FXR and RXR
      Verena Hiebl, Angela Ladurner, Simone Latkolik, Verena M. Dirsch
      Biotechnology Advances.2018; 36(6): 1657.     CrossRef
    • Nuevas perspectivas terapéuticas en la esteatohepatitis no alcohólica
      Javier Ampuero, Yolanda Sánchez-Torrijos, Virginia Aguilera, Francisco Bellido, Manuel Romero-Gómez
      Gastroenterología y Hepatología.2018; 41(2): 128.     CrossRef
    • Bile acid regulation: A novel therapeutic strategy in non-alcoholic fatty liver disease
      Qinwei Yu, Zhenzhou Jiang, Luyong Zhang
      Pharmacology & Therapeutics.2018; 190: 81.     CrossRef
    • Hippocampal FXR plays a role in the pathogenesis of depression: A preliminary study based on lentiviral gene modulation
      Wei-Guan Chen, Jia-Xuan Zheng, Xi Xu, Yu-Ming Hu, Yu-Min Ma
      Psychiatry Research.2018; 264: 374.     CrossRef
    • Bile acid receptors and the kidney
      Michal Herman-Edelstein, Talia Weinstein, Moshe Levi
      Current Opinion in Nephrology and Hypertension.2018; 27(1): 56.     CrossRef
    • Effects of resveratrol, exercises and their combination on Farnesoid X receptor, Liver X receptor and Sirtuin 1 gene expression and apoptosis in the liver of elderly rats with nonalcoholic fatty liver
      Amir Hajighasem, Parvin Farzanegi, Zohreh Mazaheri, Marjan Naghizadeh, Ghoncheh Salehi
      PeerJ.2018; 6: e5522.     CrossRef
    • Gut–Liver Axis Derangement in Non-Alcoholic Fatty Liver Disease
      Marco Poeta, Luca Pierri, Pietro Vajro
      Children.2017; 4(8): 66.     CrossRef
    • Farnesoid X Receptor Agonist as a new treatment option for Non-Alcoholic Fatty Liver disease: A Review
      S Singh, KK Kharbanda
      Archives of Hepatitis Research.2017; 3(1): 029.     CrossRef

    • PubReader PubReader
    • Cite
      CITE
      export Copy
      Close
    • XML DownloadXML Download

    Endocrinol Metab : Endocrinology and Metabolism