101
|
Jahn D, Kircher S, Hermanns HM, Geier A. Animal models of NAFLD from a hepatologist's point of view. Biochim Biophys Acta Mol Basis Dis 2018; 1865:943-953. [PMID: 29990551 DOI: 10.1016/j.bbadis.2018.06.023] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/22/2018] [Accepted: 06/25/2018] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disorder closely linked to obesity, hyperlipidemia and type 2 diabetes and is increasingly recognized as a major health problem in many parts of the world. While early stages of NAFLD are characterized by a bland accumulation of fat (steatosis) in hepatocytes, the disease can progress to non-alcoholic steatohepatitis (NASH) which involves chronic liver inflammation, tissue damage and fibrosis and can ultimately lead to end-stage liver disease including cirrhosis and cancer. As no approved pharmacological treatment for NAFLD exists today, there is an urgent need to identify promising pharmacological targets and develop future therapies. For this purpose, basic and translational research in NAFLD animal models is indispensable. While a large number of diverse animal models are currently used in the field, there is an ongoing challenge to identify those models that mirror human pathology the closest to allow good translation of obtained results into further clinical development. This review is meant to provide a concise overview of the most relevant NAFLD animal models currently available and will discuss the strengths and weaknesses of these models with regard to their comparability to human disease conditions.
Collapse
Affiliation(s)
- Daniel Jahn
- University Hospital Würzburg, Division of Hepatology, Würzburg, Germany.
| | - Stefan Kircher
- University of Würzburg, Institute of Pathology, Würzburg, Germany; Comprehensive Cancer Center Mainfranken (CCCMF), Würzburg, Germany
| | - Heike M Hermanns
- University Hospital Würzburg, Division of Hepatology, Würzburg, Germany
| | - Andreas Geier
- University Hospital Würzburg, Division of Hepatology, Würzburg, Germany
| |
Collapse
|
102
|
Abstract
Semisynthetic bile acid (BA) obeticholic acid, a potent farnesoid X receptor (FXR) agonist, exhibited beneficial effects on nonalcoholic fatty liver disease (NAFLD). Obeticholic acid, however, did not cause a resolution of nonalcoholic steatohepatitis. Here we discuss several prominent knowledge gaps in BA/FXR biology. Firstly, although many groups reported elevated serum BA levels, there are reports of decreased or normal serum BA levels in NAFLD, underlining the complexity of BA regulation by environmental and genetic factors. Secondly, conflicting data exist in animal studies regarding the effects of FXR signaling on obesity and associated metabolic abnormalities. Thirdly, it remains obscure how the gut microbiome and the BA pool interact and influence the pathogenesis of NAFLD. Lastly, it is not known how FXR-mediated signaling interact with G protein-coupled BA receptor 1-mediated signaling. Answering these questions may lead to an improved pharmaceutical intervention for NAFLD targeting the FXR signaling pathway.
Collapse
|
103
|
Yan T, Wang H, Cao L, Wang Q, Takahashi S, Yagai T, Li G, Krausz KW, Wang G, Gonzalez FJ, Hao H. Glycyrrhizin Alleviates Nonalcoholic Steatohepatitis via Modulating Bile Acids and Meta-Inflammation. Drug Metab Dispos 2018; 46:1310-1319. [PMID: 29959134 DOI: 10.1124/dmd.118.082008] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/27/2018] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the progressive stage of nonalcoholic fatty liver disease that may ultimately lead to cirrhosis and liver cancer, and there are few therapeutic options for its treatment. Glycyrrhizin (GL), extracted from the traditional Chinese medicine liquorice, has potent hepatoprotective effects in both preclinical animal models and in humans. However, little is currently known about its effects and mechanisms in treating NASH. To explore the effects of GL on NASH, GL or its active metabolite glycyrrhetinic acid (GA) was administered to mice treated with a methionine- and choline-deficient (MCD) diet-induced NASH model, and histologic and biochemical analyses were used to measure the degree of lipid disruption, liver inflammation, and fibrosis. GL significantly improved MCD diet-induced hepatic steatosis, inflammation, and fibrosis and inhibited activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome. GL significantly attenuated serum bile acid accumulation in MCD diet-fed mice partially by restoring inflammation-mediated hepatic farnesoid X receptor inhibition. In Raw 264.7 macrophage cells, both GL and GA inhibited deoxycholic acid-induced NLRP3 inflammasome-associated inflammation. Notably, both intraperitoneal injection of GL's active metabolite GA and oral administration of GL prevented NASH in mice, indicating that GL may attenuate NASH via its active metabolite GA. These results reveal that GL, via restoration of bile acid homeostasis and inhibition of inflammatory injury, can be a therapeutic option for treatment of NASH.
Collapse
Affiliation(s)
- Tingting Yan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Hong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Lijuan Cao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Qiong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Shogo Takahashi
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Tomoki Yagai
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Guolin Li
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Kristopher W Krausz
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Frank J Gonzalez
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| |
Collapse
|
104
|
Hypoxic Signaling and Cholesterol Lipotoxicity in Fatty Liver Disease Progression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2548154. [PMID: 29955245 PMCID: PMC6000860 DOI: 10.1155/2018/2548154] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/30/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
Cholesterol is the only lipid whose absorption in the gastrointestinal tract is limited by gate-keeping transporters and efflux mechanisms, preventing its rapid absorption and accumulation in the liver and blood vessels. In this review, I explored the current data regarding cholesterol accumulation in liver cells and key mechanisms in cholesterol-induced fatty liver disease associated with the activation of deleterious hypoxic and nitric oxide signal transduction pathways. Although nonalcoholic fatty liver disease (NAFLD) affects both obese and nonobese individuals, the mechanism of NAFLD progression in lean individuals with healthy metabolism is puzzling. Lean NAFLD individuals exhibit normal metabolic responses, implying that liver damage is not associated with impaired metabolism per se and that direct lipotoxic effects are crucial for disease progression. Several redox and oxidant signaling pathways involving cholesterol are at play in fatty liver disease development. These include impairment of the mitochondrial and lysosomal function by cholesterol loading of the inner-cell membranes; formation of cholesterol crystals and hepatocyte degradation; and crown-like structures surrounding degrading hepatocytes, activating Kupffer cells, and evoking inflammation. The current review focuses on the induction of liver inflammation, fibrosis, and steatosis by free cholesterol via the hypoxia-inducible factor 1α (HIF-1α), a main oxygen-sensing transcription factor involved in all stages of NAFLD. Cholesterol loading in hepatocytes can result in chronic HIF-1α activity because of the decreased oxygen availability and excessive production of nitric oxide and mitochondrial reactive oxygen species.
Collapse
|
105
|
You M, Zhou Z, Daniels M, Jogasuria A. Endocrine Adiponectin-FGF15/19 Axis in Ethanol-Induced Inflammation and Alcoholic Liver Injury. Gene Expr 2018; 18:103-113. [PMID: 29096734 PMCID: PMC5953845 DOI: 10.3727/105221617x15093738210295] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alcoholic liver disease (ALD) is the most prevalent form of liver disease, encompassing a spectrum of progressive pathological changes from steatosis to steatohepatitis to fibrosis/cirrhosis and hepatocellular carcinoma. Alcoholic steatosis/steatohepatitis is the initial stage of ALD and a major risk factor for advanced liver injuries. Adiponectin is a hormone secreted from adipocytes. Fibroblast growth factor (FGF) 15 (human homolog, FGF19) is an ileum-derived hormone. Adipocyte-derived adiponectin and gut-derived FGF15/19 regulate each other, share common signaling cascades, and exert similar beneficial functions. Emerging evidence has revealed that dysregulated adiponectin-FGF15/19 axis and impaired hepatic adiponectin-FGF15/19 signaling are associated with alcoholic liver damage in rodents and humans. More importantly, endocrine adiponectin-FGF15/19 signaling confers protection against ethanol-induced liver damage via fine tuning the adipose-intestine-liver crosstalk, leading to limited hepatic inflammatory responses, and ameliorated alcoholic liver injury. This review is focused on the recently discovered endocrine adiponectin-FGF15/19 axis that is emerging as an essential adipose-gut-liver coordinator involved in the development and progression of alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Min You
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Zhou Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Michael Daniels
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Alvin Jogasuria
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
106
|
Zou A, Magee N, Deng F, Lehn S, Zhong C, Zhang Y. Hepatocyte nuclear receptor SHP suppresses inflammation and fibrosis in a mouse model of nonalcoholic steatohepatitis. J Biol Chem 2018; 293:8656-8671. [PMID: 29666185 DOI: 10.1074/jbc.ra117.001653] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 03/17/2018] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a burgeoning health problem worldwide, ranging from nonalcoholic fatty liver (NAFL, steatosis without hepatocellular injury) to the more aggressive nonalcoholic steatohepatitis (NASH, steatosis with ballooning, inflammation, or fibrosis). Although many studies have greatly contributed to the elucidation of NAFLD pathogenesis, the disease progression from NAFL to NASH remains incompletely understood. Nuclear receptor small heterodimer partner (Nr0b2, SHP) is a transcriptional regulator critical for the regulation of bile acid, glucose, and lipid metabolism. Here, we show that SHP levels are decreased in the livers of patients with NASH and in diet-induced mouse NASH. Exposing primary mouse hepatocytes to palmitic acid and lipopolysaccharide in vitro, we demonstrated that the suppression of Shp expression in hepatocytes is due to c-Jun N-terminal kinase (JNK) activation, which stimulates c-Jun-mediated transcriptional repression of Shp Interestingly, in vivo induction of hepatocyte-specific SHP in steatotic mouse liver ameliorated NASH progression by attenuating liver inflammation and fibrosis, but not steatosis. Moreover, a key mechanism linking the anti-inflammatory role of hepatocyte-specific SHP expression to inflammation involved SHP-induced suppression of NF-κB p65-mediated induction of chemokine (C-C motif) ligand 2 (CCL2), which activates macrophage proinflammatory polarization and migration. In summary, our results indicate that a JNK/SHP/NF-κB/CCL2 regulatory network controls communications between hepatocytes and macrophages and contributes to the disease progression from NAFL to NASH. Our findings may benefit the development of new management or prevention strategies for NASH.
Collapse
Affiliation(s)
- An Zou
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Nancy Magee
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Fengyan Deng
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Sarah Lehn
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Cuncong Zhong
- the Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, Kansas 66045, and
| | - Yuxia Zhang
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, .,the Liver Center, University of Kansas Medical Center, Kansas City, Kansas 66160
| |
Collapse
|
107
|
Luo L, Aubrecht J, Li D, Warner RL, Johnson KJ, Kenny J, Colangelo JL. Assessment of serum bile acid profiles as biomarkers of liver injury and liver disease in humans. PLoS One 2018. [PMID: 29513725 PMCID: PMC5841799 DOI: 10.1371/journal.pone.0193824] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
To assess the potential of individual bile acids (IBA) and their profiles as mechanistic biomarkers of liver injury for humans in real world situations, we interrogated samples collected under minimum controlled conditions (ie subjects were not fasted). Total bile acids (TBA) have been considered to be biomarkers of liver injury for decades, and more recently, monitoring of IBA has been proposed for differentiation of variety of etiologies of liver injury. We established a LC-MS/MS methodology to analyze nine IBA, generated reference ranges, and examined effects of age, gender, and ethnicity for each IBA. Furthermore, we evaluated the ability of IBA and their profiles to detect hepatic injury in subjects with a broad range of liver impairments. To date, our study utilized the largest total cohort of samples (N = 645) that were divided into 2 groups, healthy or liver impaired, to evaluate IBA as biomarkers. The TBA serum levels in the Asian ethnic group trended higher when compared to other ethnic groups, and the serum concentrations of IBA, such as glycocholic acid (GCA), glycochenodeoxycholic acid (GCDCA), chenodeoxycholic acid (CDCA), and taurochenoxycholic acid (TCDCA) were significantly increased. To our knowledge, this report is the first to describe ethnic differences in serum concentrations of IBAs. In patients with hepatic impairments, with the exception of deoxycholic acid (DCA), the concentrations of IBAs were significantly elevated when compared with healthy subjects. The conjugated bile acids displayed greater differences between healthy subjects and subjects with hepatic impairments than non-conjugated bile acids. Furthermore, the subjects with hepatic impairments exhibited distinct profiles (signatures) of IBAs that clustered subjects according the nature of their liver impairments. Although additional studies are needed, our data suggested that the analysis of IBA has the potential to become useful for differentiation of various forms of liver injury.
Collapse
Affiliation(s)
- Lina Luo
- Pharmacokinetics, Dynamics & Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut, United States of America
- * E-mail:
| | - Jiri Aubrecht
- Drug Safety Research & Development, Pfizer Inc., Groton, Connecticut, United States of America
| | - Dingzhou Li
- Drug Safety Research & Development, Pfizer Inc., Groton, Connecticut, United States of America
| | - Roscoe L. Warner
- University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Kent J. Johnson
- University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Julia Kenny
- University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| | - Jennifer L. Colangelo
- Drug Safety Research & Development, Pfizer Inc., Groton, Connecticut, United States of America
| |
Collapse
|
108
|
Kong Y, Gao X, Wang C, Ning C, Liu K, Liu Z, Sun H, Ma X, Sun P, Meng Q. Protective effects of yangonin from an edible botanical Kava against lithocholic acid-induced cholestasis and hepatotoxicity. Eur J Pharmacol 2018; 824:64-71. [PMID: 29427579 DOI: 10.1016/j.ejphar.2018.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/15/2018] [Accepted: 02/06/2018] [Indexed: 02/06/2023]
Abstract
Accumulation of toxic bile acids in liver could cause cholestasis and liver injury. The purpose of the current study is to evaluate the hepatoprotective effect of yangonin, a product isolated from an edible botanical Kava against lithocholic acid (LCA)-induced cholestasis, and further to elucidate the involvement of farnesoid X receptor (FXR) in the anticholestatic effect using in vivo and in vitro experiments. The cholestatic liver injury model was established by intraperitoneal injections of LCA in C57BL/6 mice. Serum biomarkers and H&E staining were used to identify the amelioration of cholestasis after yangonin treatment. Mice hepatocytes culture, gene silencing experiment, real-time PCR and Western blot assay were used to elucidate the mechanisms underlying yangonin hepatoprotection. The results indicated that yangonin promoted bile acid efflux and reduced hepatic uptake via an induction in FXR-target genes Bsep, Mrp2 expression and an inhibition in Ntcp, all of which are responsible for bile acid transport. Furthermore, yangonin reduced bile acid synthesis through repressing FXR-target genes Cyp7a1 and Cyp8b1, and increased bile acid metabolism through an induction in gene expression of Sult2a1, which are involved in bile acid synthesis and metabolism. In addition, yangonin suppressed liver inflammation through repressing inflammation-related gene NF-κB, TNF-α and IL-1β. In vitro evidences showed that the changes in transporters and enzymes induced by yangonin were abrogated when FXR was silenced. In conclusions, yangonin produces protective effect against LCA-induced hepatotoxity and cholestasis due to FXR-mediated regulation. Yangonin may be an effective approach for the prevention against cholestatic liver diseases.
Collapse
Affiliation(s)
- Yulong Kong
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Xiaoguang Gao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Chenqing Ning
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Kexin Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Zhihao Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Xiaodong Ma
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Pengyuan Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
109
|
Puri P, Daita K, Joyce A, Mirshahi F, Santhekadur PK, Cazanave S, Luketic VA, Siddiqui MS, Boyett S, Min HK, Kumar DP, Kohli R, Zhou H, Hylemon PB, Contos MJ, Idowu M, Sanyal AJ. The presence and severity of nonalcoholic steatohepatitis is associated with specific changes in circulating bile acids. Hepatology 2018; 67:534-548. [PMID: 28696585 PMCID: PMC5764808 DOI: 10.1002/hep.29359] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/25/2017] [Accepted: 07/07/2017] [Indexed: 12/12/2022]
Abstract
The histologic spectrum of nonalcoholic fatty liver disease (NAFLD) includes fatty liver (NAFL) and steatohepatitis (NASH), which can progress to cirrhosis in up to 20% of NASH patients. Bile acids (BA) are linked to the pathogenesis and therapy of NASH. We (1) characterized the plasma BA profile in biopsy-proven NAFL and NASH and compared to controls and (2) related the plasma BA profile to liver histologic features, disease activity, and fibrosis. Liquid chromatography/mass spectrometry quantified BAs. Descriptive statistics, paired and multiple group comparisons, and regression analyses were performed. Of 86 patients (24 controls, 25 NAFL, and 37 NASH; mean age 51.8 years and body mass index 31.9 kg/m2 ), 66% were women. Increased total primary BAs and decreased secondary BAs (both P < 0.05) characterized NASH. Total conjugated primary BAs were significantly higher in NASH versus NAFL (P = 0.047) and versus controls (P < 0.0001). NASH had higher conjugated to unconjugated chenodeoxycholate (P = 0.04), cholate (P = 0.0004), and total primary BAs (P < 0.0001). The total cholate to chenodeoxycholate ratio was significantly higher in NAFLD without (P = 0.005) and with (P = 0.02) diabetes. Increased key BAs were associated with higher grades of steatosis (taurocholate), lobular (glycocholate) and portal inflammation (taurolithocholate), and hepatocyte ballooning (taurocholate). Conjugated cholate and taurocholate directly and secondary to primary BA ratio inversely correlated to NAFLD activity score. A higher ratio of total secondary to primary BA decreased (odds ratio, 0.57; P = 0.004) and higher conjugated cholate increased the likelihood of significant fibrosis (F≥2) (P = 0.007). Conclusion: NAFLD is associated with significantly altered circulating BA composition, likely unaffected by type 2 diabetes, and correlated with histological features of NASH; these observations provide the foundation for future hypothesis-driven studies of specific effects of BAs on specific aspects of NASH. (Hepatology 2018;67:534-548).
Collapse
Affiliation(s)
- Puneet Puri
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Kalyani Daita
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | | | - Faridoddin Mirshahi
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Prasanna K. Santhekadur
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Sophie Cazanave
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Velimir A Luketic
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Mohammad S. Siddiqui
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Sherry Boyett
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Hae-Ki Min
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Divya P. Kumar
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Rohit Kohli
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA
| | - Huiping Zhou
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Phillip B. Hylemon
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Melissa J Contos
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Michael Idowu
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Arun J. Sanyal
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
110
|
Jahn D, Geier A. Bile acids in nonalcoholic steatohepatitis: Pathophysiological driving force or innocent bystanders? Hepatology 2018; 67:464-466. [PMID: 28926117 DOI: 10.1002/hep.29543] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/01/2017] [Accepted: 09/18/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Daniel Jahn
- Division of Hepatology, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Geier
- Division of Hepatology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
111
|
Molinaro A, Wahlström A, Marschall HU. Role of Bile Acids in Metabolic Control. Trends Endocrinol Metab 2018; 29:31-41. [PMID: 29195686 DOI: 10.1016/j.tem.2017.11.002] [Citation(s) in RCA: 278] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
Bile acids are endocrine molecules that in addition to facilitating the absorption of fat-soluble nutrients regulate numerous metabolic processes, including glucose, lipid, and energy homeostasis. The signaling actions of bile acids are mediated through specific bile-acid-activated nuclear and membrane-bound receptors. These receptors are not only expressed by tissues within the enterohepatic circulation such as the liver and the intestine, but also in other organs where bile acids mediate their systemic actions. In this review, we discuss bile acid signaling and the interplay with the gut microbiota in the pathophysiology of obesity, type 2 diabetes, and non-alcoholic fatty liver disease, and the role of surgical and pharmacological interventions on bile acid profiles and metabolism.
Collapse
Affiliation(s)
- Antonio Molinaro
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, S-413 45 Gothenburg, Sweden
| | - Annika Wahlström
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, S-413 45 Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, S-413 45 Gothenburg, Sweden.
| |
Collapse
|
112
|
Wong VWS, Chan WK, Chitturi S, Chawla Y, Dan YY, Duseja A, Fan J, Goh KL, Hamaguchi M, Hashimoto E, Kim SU, Lesmana LA, Lin YC, Liu CJ, Ni YH, Sollano J, Wong SKH, Wong GLH, Chan HLY, Farrell G. Asia-Pacific Working Party on Non-alcoholic Fatty Liver Disease guidelines 2017-Part 1: Definition, risk factors and assessment. J Gastroenterol Hepatol 2018; 33:70-85. [PMID: 28670712 DOI: 10.1111/jgh.13857] [Citation(s) in RCA: 326] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 05/30/2017] [Accepted: 06/25/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong.,State Key Laboratory of Digestive Disease and Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Wah-Kheong Chan
- Department of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Shiv Chitturi
- Gastroenterology and Hepatology Unit, The Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Yogesh Chawla
- Department of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Yock Young Dan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ajay Duseja
- Department of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jiangao Fan
- Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Khean-Lee Goh
- Department of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Etsuko Hashimoto
- Departments of Internal Medicine and Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Seung Up Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | | | - Yu-Cheng Lin
- Hepatitis Research Center, National Taiwan University, Taipei, Taiwan
| | - Chun-Jen Liu
- Department of Internal Medicine, Hepatitis Research Center and Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Yen-Hsuan Ni
- Hepatitis Research Center, National Taiwan University, Taipei, Taiwan
| | - Jose Sollano
- University of Santo Tomas, Manila, The Philippines
| | - Simon Kin-Hung Wong
- Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Grace Lai-Hung Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong.,State Key Laboratory of Digestive Disease and Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Henry Lik-Yuen Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong.,State Key Laboratory of Digestive Disease and Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Geoff Farrell
- Gastroenterology and Hepatology Unit, The Canberra Hospital, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
113
|
Changes in Bile Acid Profile After Laparoscopic Sleeve Gastrectomy are Associated with Improvements in Metabolic Profile and Fatty Liver Disease. Obes Surg 2017; 26:1195-202. [PMID: 26337697 DOI: 10.1007/s11695-015-1878-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Bile acids (BA) modulate lipid and glucose metabolism in a feedback loop through production of fibroblast growth factor (FGF) 19 in the terminal ileum. Changes in BA after bariatric surgery may lead to improvements in the metabolic syndrome, including fatty liver disease. This study investigated the relationship between BA and metabolic and inflammatory profiles after laparoscopic sleeve gastrectomy (LSG). METHODS Patients undergoing LSG had fasting blood samples taken pre-operatively and 6 months post-surgery. Liver injury was measured using cytokeratin (CK) 18 fragments. BA were measured using liquid chromatography tandem-mass spectrometry. FGF-19 was measured using enzyme-linked immunosorbent assay. RESULTS The study included 18 patients (12 females), with mean age 46.3 years (SEM ± 2.9) and BMI 60.1 kg/m(2) (±2.6). After 6 months, patients lost 39.8 kg (±3.1; p < 0.001). Fourteen patients (78 %) had steatosis. FGF-19 increased from median 128.1 (IQR 89.4-210.1) to 177.1 (121.8-288.9, p = 0.045) at 6 months. Although total BA did not change, primary glycine- and taurine-conjugated BA, cholic acid decreased, and secondary BA, glycine-conjugated urodeoxycholic acid increased over the study period. These changes are associated with reduction in insulin resistance, pro-inflammatory cytokines and CK-18 levels. CONCLUSIONS The profile of individual BA is altered after LSG. These changes occur in the presence of reductions in inflammatory cytokines and markers of liver injury. This study supports evidence from recent animal models that LSG may have an effect on fatty liver through changes in BA metabolism.
Collapse
|
114
|
Cruz-Ramón V, Chinchilla-López P, Ramírez-Pérez O, Méndez-Sánchez N. Bile Acids in Nonalcoholic Fatty Liver Disease: New Concepts and therapeutic advances. Ann Hepatol 2017; 16:s58-s67. [PMID: 29080343 DOI: 10.5604/01.3001.0010.5498] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 09/09/2017] [Indexed: 02/04/2023]
Abstract
Nonalcoholic liver disease (NAFLD) is a major emerging health burden that is a common cause of illness and death worldwide. NAFLD can progress into nonalcoholic steatohepatitis (NASH) which is a severe form of liver disease characterized by inflammation and fibrosis. Further progression leads to cirrhosis, which predisposes patients to hepatocellular carcinoma or liver failure. The mechanism of the progression from simple steatosis to NASH is unclear. However, there are theories and hypothesis which support the link between disruption of the bile acids homeostasis and the progression of this disorder. Previous studies have been demonstrated that alterations to these pathways can lead to dysregulation of energy balance and increased liver inflammation and fibrosis. In this review, we summarized the current knowledge of the interaction between BA and the process related to the development of NAFLD, besides, the potential targets for novel therapies.
Collapse
Affiliation(s)
- Vania Cruz-Ramón
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | | | | | | |
Collapse
|
115
|
Alvarez-Sola G, Uriarte I, Latasa MU, Jimenez M, Barcena-Varela M, Santamaría E, Urtasun R, Rodriguez-Ortigosa C, Prieto J, Corrales FJ, Baulies A, García-Ruiz C, Fernandez-Checa JC, Berraondo P, Fernandez-Barrena MG, Berasain C, Avila MA. Engineered fibroblast growth factor 19 protects from acetaminophen-induced liver injury and stimulates aged liver regeneration in mice. Cell Death Dis 2017; 8:e3083. [PMID: 28981086 PMCID: PMC5682649 DOI: 10.1038/cddis.2017.480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/31/2017] [Accepted: 08/09/2017] [Indexed: 02/07/2023]
Abstract
The liver displays a remarkable regenerative capacity triggered upon tissue injury or resection. However, liver regeneration can be overwhelmed by excessive parenchymal destruction or diminished by pre-existing conditions hampering repair. Fibroblast growth factor 19 (FGF19, rodent FGF15) is an enterokine that regulates liver bile acid and lipid metabolism, and stimulates hepatocellular protein synthesis and proliferation. FGF19/15 is also important for liver regeneration after partial hepatectomy (PH). Therefore recombinant FGF19 would be an ideal molecule to stimulate liver regeneration, but its applicability may be curtailed by its short half-life. We developed a chimaeric molecule termed Fibapo in which FGF19 is covalently coupled to apolipoprotein A-I. Fibapo retains FGF19 biological activities but has significantly increased half-life and hepatotropism. Here we evaluated the pro-regenerative activity of Fibapo in two clinically relevant models where liver regeneration may be impaired: acetaminophen (APAP) poisoning, and PH in aged mice. The only approved therapy for APAP intoxication is N-acetylcysteine (NAC) and no drugs are available to stimulate liver regeneration. We demonstrate that Fibapo reduced liver injury and boosted regeneration in APAP-intoxicated mice. Fibapo improved survival of APAP-poisoned mice when given at later time points, when NAC is ineffective. Mechanistically, Fibapo accelerated recovery of hepatic glutathione levels, potentiated cell growth-related pathways and increased functional liver mass. When Fibapo was administered to old mice prior to PH, liver regeneration was markedly increased. The exacerbated injury developing in these mice upon PH was attenuated, and the hepatic biosynthetic capacity was enhanced. Fibapo reversed metabolic and molecular alterations that impede regeneration in aged livers. It reduced liver steatosis and downregulated p21 and hepatocyte nuclear factor 4 α (Hnf4α) levels, whereas it stimulated Foxm1b gene expression. Together our findings indicate that FGF19 variants retaining the metabolic and growth-promoting effects of this enterokine may be valuable for the stimulation of liver regeneration.
Collapse
Affiliation(s)
- Gloria Alvarez-Sola
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda, Pio XII, n 36, Pamplona 31008, Spain
| | - Iker Uriarte
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda, Pio XII, n 36, Pamplona 31008, Spain
| | - Maria U Latasa
- Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| | - Maddalen Jimenez
- Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| | - Marina Barcena-Varela
- Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| | - Eva Santamaría
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda, Pio XII, n 36, Pamplona 31008, Spain
| | - Raquel Urtasun
- Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| | - Carlos Rodriguez-Ortigosa
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda, Pio XII, n 36, Pamplona 31008, Spain.,Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| | - Jesús Prieto
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda, Pio XII, n 36, Pamplona 31008, Spain.,Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| | - Fernando J Corrales
- Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain.,CIBERehd, Instituto de Salud Carlos III, Barcelona, Spain
| | - Anna Baulies
- CIBERehd, Instituto de Salud Carlos III, Barcelona, Spain.,Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC and Liver Unit-Hospital Clinic-IDIBAPS, Barcelona, Spain.,Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles 90033, CA, USA
| | - Carmen García-Ruiz
- CIBERehd, Instituto de Salud Carlos III, Barcelona, Spain.,Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC and Liver Unit-Hospital Clinic-IDIBAPS, Barcelona, Spain.,Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles 90033, CA, USA
| | - Jose C Fernandez-Checa
- CIBERehd, Instituto de Salud Carlos III, Barcelona, Spain.,Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC and Liver Unit-Hospital Clinic-IDIBAPS, Barcelona, Spain.,Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles 90033, CA, USA
| | - Pedro Berraondo
- Immunology and Immunotherapy Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| | - Maite G Fernandez-Barrena
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda, Pio XII, n 36, Pamplona 31008, Spain.,Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| | - Carmen Berasain
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda, Pio XII, n 36, Pamplona 31008, Spain.,Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| | - Matías A Avila
- CIBERehd, Instituto de Salud Carlos III, Clinica Universidad de Navarra, Avda, Pio XII, n 36, Pamplona 31008, Spain.,Hepatology Programme, CIMA, Idisna, Universidad de Navarra, Avda, Pio XII, n 55, Pamplona 31008, Spain
| |
Collapse
|
116
|
Legry V, Francque S, Haas JT, Verrijken A, Caron S, Chávez-Talavera O, Vallez E, Vonghia L, Dirinck E, Verhaegen A, Kouach M, Lestavel S, Lefebvre P, Van Gaal L, Tailleux A, Paumelle R, Staels B. Bile Acid Alterations Are Associated With Insulin Resistance, but Not With NASH, in Obese Subjects. J Clin Endocrinol Metab 2017; 102:3783-3794. [PMID: 28938455 DOI: 10.1210/jc.2017-01397] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022]
Abstract
CONTEXT Bile acids (BAs) are signaling molecules controlling energy homeostasis that can be both toxic and protective for the liver. BA alterations have been reported in obesity, insulin resistance (IR), and nonalcoholic steatohepatitis (NASH). However, whether BA alterations contribute to NASH independently of the metabolic status is unclear. OBJECTIVE To assess BA alterations associated with NASH independently of body mass index and IR. DESIGN AND SETTING Patients visiting the obesity clinic of the Antwerp University Hospital (a tertiary referral facility) were recruited from 2006 to 2014. PATIENTS Obese patients with biopsy-proven NASH (n = 32) and healthy livers (n = 26) were matched on body mass index and homeostasis model assessment of IR. MAIN OUTCOME MEASURES Transcriptomic analyses were performed on liver biopsies. Plasma concentrations of 21 BA species and 7α-hydroxy-4-cholesten-3-one, a marker of BA synthesis, were determined by liquid chromatography-tandem mass spectrometry. Plasma fibroblast growth factor 19 was measured by enzyme-linked immunosorbent assay. RESULTS Plasma BA concentrations did not correlate with any hepatic lesions, whereas, as previously reported, primary BA strongly correlated with IR. Transcriptomic analyses showed unaltered hepatic BA metabolism in NASH patients. In line, plasma 7α-hydroxy-4-cholesten-3-one was unchanged in NASH. Moreover, no sign of hepatic BA accumulation or activation of BA receptors-farnesoid X, pregnane X, and vitamin D receptors-was found. Finally, plasma fibroblast growth factor 19, secondary-to-primary BA, and free-to-conjugated BA ratios were similar, suggesting unaltered intestinal BA metabolism and signaling. CONCLUSIONS In obese patients, BA alterations are related to the metabolic phenotype associated with NASH, especially IR, but not liver necroinflammation.
Collapse
Affiliation(s)
- Vanessa Legry
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, 2650 Edegem/Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk/Antwerp, Belgium
| | - Joel T Haas
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| | - An Verrijken
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk/Antwerp, Belgium
- Department of Endocrinology, Diabetes and Metabolism, Antwerp University Hospital, 2650 Egedem/Antwerp, Belgium
| | - Sandrine Caron
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| | - Oscar Chávez-Talavera
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| | - Emmanuelle Vallez
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| | - Luisa Vonghia
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, 2650 Edegem/Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk/Antwerp, Belgium
| | - Eveline Dirinck
- Department of Endocrinology, Diabetes and Metabolism, Antwerp University Hospital, 2650 Egedem/Antwerp, Belgium
| | - Ann Verhaegen
- Department of Endocrinology, Diabetes and Metabolism, Antwerp University Hospital, 2650 Egedem/Antwerp, Belgium
| | - Mostafa Kouach
- Plateau de Spectrométrie de Masse-Groupe de Recherche sur les formes Injectables et les Technologies Associées, (PSM-GRITA), Faculté de Pharmacie, F-59000 Lille, France
| | - Sophie Lestavel
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| | - Philippe Lefebvre
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| | - Luc Van Gaal
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk/Antwerp, Belgium
- Department of Endocrinology, Diabetes and Metabolism, Antwerp University Hospital, 2650 Egedem/Antwerp, Belgium
| | - Anne Tailleux
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| | - Réjane Paumelle
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| | - Bart Staels
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, F-59000 Lille, France
| |
Collapse
|
117
|
Xu L, Sheng T, Liu X, Zhang T, Wang Z, Han H. Analyzing the hepatoprotective effect of the Swertia cincta Burkillextract against ANIT-induced cholestasis in rats by modulating the expression of transporters and metabolic enzymes. JOURNAL OF ETHNOPHARMACOLOGY 2017; 209:91-99. [PMID: 28734962 DOI: 10.1016/j.jep.2017.07.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 07/15/2017] [Accepted: 07/18/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Swertia cincta Burkill was traditionally used for treating jaundice and various types of chronic and acute hepatitis in Yunnan and Tibet in China for hundreds of years. This study aims to investigate the protective effect of S. cincta Burkill (ESC) extract on alpha-naphthylisothiocyanate (ANIT)-induced hepatotoxicity and cholestasis in rats. MATERIALS AND METHODS Crude extracts were prepared using 90% ethanol and by vacuum drying. We utilized an ultra-high-performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight tandem mass spectrometry (UHPLC/Q-TOF-MS) system to conduct a phytochemical analysis of the active components of ESC. Liver function was evaluated by measuring the serum levels of enzymes and components and by analyzing the liver histology. We also measured the expression of bile metabolism-related transporters and metabolic enzymes at both protein and mRNA levels to elucidate the underlying mechanisms. RESULTS ESC analysis using an UHPLC/Q-TOF-MS revealed eight compounds. Oral administration of ESC to ANIT-treated rats can significantly reduce the increases in serum levels of ALT, AST, ALP, TBIL, and TBA. It can also improve liver pathology and bile flow. Western blot and qRT-PCR analyses showed that ESC upregulated the protein and mRNA expression of Fxr, Ntcp, Bsep, Cyp7a1, Mrp2, and Mdr2. CONCLUSION ESC could alleviate liver injury by reducing enzyme activities of serums, improving liver pathology and bile flow. The protective mechanism was associated with regulation of the expression of hepatic transporters and metabolic enzymes.
Collapse
Affiliation(s)
- Lili Xu
- Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China; Institute of Science, Technology and Humanities, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China.
| | - Tingting Sheng
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China.
| | - Xiaolong Liu
- Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China.
| | - Tong Zhang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China.
| | - Zhengtao Wang
- Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China.
| | - Han Han
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China; Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China.
| |
Collapse
|
118
|
Feng R, Luo C, Li C, Du S, Okekunle AP, Li Y, Chen Y, Zi T, Niu Y. Free fatty acids profile among lean, overweight and obese non-alcoholic fatty liver disease patients: a case - control study. Lipids Health Dis 2017; 16:165. [PMID: 28870233 PMCID: PMC5584533 DOI: 10.1186/s12944-017-0551-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 08/15/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) given its association with obesity and diabetes may perhaps exert distinct free fatty acids (FFA) pattern, but the understanding of this phenomenon is limited. To this effect, we evaluated FFA profiles among healthy subjects and NAFLD patients stratified by body weight, to identify FFA valuable for early diagnosis of NAFLD. METHODS Serum FFA profiles of healthy and NAFLD (lean, overweight and obese) subjects was determined using gas chromatography-mass spectrometry (GC-MS) and distinctions in FFA patterns were evaluated using one-way ANOVA while Receiver operating characteristics (ROC) and logistic regression models were used to explore FFA significant for diagnosing NAFLD. RESULTS NAFLD patients presented significantly higher (P < 0.05) serum FFA profiles compared to healthy controls (HC). While total FFA profiles were insignificantly different between lean (2093.33 ± 558.11 μg/ml) and overweight (2420.81 ± 555.18 μg/ml) NAFLD patients, obese NAFLD (2739.01 ± 810.35 μg/ml) presented most significantly elevated (P < 0.05) total FFA profiles compared with HC. Of the four FFA; myristic acid (14:0), palmitoleic acid (16:1), γ-linolenic acid (γ-18:3) and cis-7,10,13,16,19-docosapentaenoic acid (22:5), selected in ROC analysis given their high Youden's index and AUC, only 14:0; 5.58(1.37, 22.76) and 16:1; 4.36(1.34, 14.13) had statistical significant odd ratios. CONCLUSION Our findings suggest 14:0 and 16:1 are promising for early diagnosis of NAFLD.
Collapse
Affiliation(s)
- Rennan Feng
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, Heilongjiang Province, 150081, China.
| | - Chao Luo
- STD & AIDS Center, Harbin Center for Disease Control and Prevention, Harbin, Heilongjiang Province, 150056, China
| | - Chunlong Li
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
| | - Shanshan Du
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Akinkunmi Paul Okekunle
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Yanchuan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Yang Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Tianqi Zi
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Yucun Niu
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Street, Nangang District, Harbin, Heilongjiang Province, 150081, China
| |
Collapse
|
119
|
Nuñez-Garcia M, Gomez-Santos B, Buqué X, García-Rodriguez JL, Romero MR, Marin JJG, Arteta B, García-Monzón C, Castaño L, Syn WK, Fresnedo O, Aspichueta P. Osteopontin regulates the cross-talk between phosphatidylcholine and cholesterol metabolism in mouse liver. J Lipid Res 2017; 58:1903-1915. [PMID: 28754826 DOI: 10.1194/jlr.m078980] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/28/2017] [Indexed: 12/15/2022] Open
Abstract
Osteopontin (OPN) is involved in different liver pathologies in which metabolic dysregulation is a hallmark. Here, we investigated whether OPN could alter liver, and more specifically hepatocyte, lipid metabolism and the mechanism involved. In mice, lack of OPN enhanced cholesterol 7α-hydroxylase (CYP7A1) levels and promoted loss of phosphatidylcholine (PC) content in liver; in vivo treatment with recombinant (r)OPN caused opposite effects. rOPN directly decreased CYP7A1 levels through activation of focal adhesion kinase-AKT signaling in hepatocytes. PC content was also decreased in OPN-deficient (OPN-KO) hepatocytes in which de novo FA and PC synthesis was lower, whereas cholesterol (CHOL) synthesis was higher, than in WT hepatocytes. In vivo inhibition of cholesterogenesis normalized liver PC content in OPN-KO mice, demonstrating that OPN regulates the cross-talk between liver CHOL and PC metabolism. Matched liver and serum samples showed a positive correlation between serum OPN levels and liver PC and CHOL concentration in nonobese patients with nonalcoholic fatty liver. In conclusion, OPN regulates CYP7A1 levels and the metabolic fate of liver acetyl-CoA as a result of CHOL and PC metabolism interplay. The results suggest that CYP7A1 is a main axis and that serum OPN could disrupt liver PC and CHOL metabolism, contributing to nonalcoholic fatty liver disease progression in nonobese patients.
Collapse
Affiliation(s)
- Maitane Nuñez-Garcia
- Departments of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Beatriz Gomez-Santos
- Departments of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Xabier Buqué
- Departments of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Biocruces Health Research Institute, Barakaldo, Spain
| | - Juan L García-Rodriguez
- Departments of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Marta R Romero
- Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca, Salamanca, Spain.,Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Jose J G Marin
- Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca, Salamanca, Spain.,Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Beatriz Arteta
- Cellular Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Carmelo García-Monzón
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain.,Liver Research Unit, Santa Cristina University Hospital, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Luis Castaño
- Biocruces Health Research Institute, Barakaldo, Spain.,Hospital Universitario Cruces, Barakaldo, Spain.,Pediatrics Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,CIBERDEM, CIBERER Carlos III National Institute of Health, Madrid, Spain
| | - Wing-Kin Syn
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, United Kingdom.,Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC.,Section of Gastroenterology, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| | - Olatz Fresnedo
- Departments of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Patricia Aspichueta
- Departments of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain .,Biocruces Health Research Institute, Barakaldo, Spain
| |
Collapse
|
120
|
Krishnan A, Abdullah TS, Mounajjed T, Hartono S, McConico A, White T, LeBrasseur N, Lanza I, Nair S, Gores G, Charlton M. A longitudinal study of whole body, tissue, and cellular physiology in a mouse model of fibrosing NASH with high fidelity to the human condition. Am J Physiol Gastrointest Liver Physiol 2017; 312:G666-G680. [PMID: 28232454 PMCID: PMC6146305 DOI: 10.1152/ajpgi.00213.2016] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 01/31/2023]
Abstract
The sequence of events that lead to inflammation and fibrosing nonalcoholic steatohepatitis (NASH) is incompletely understood. Hence, we investigated the chronology of whole body, tissue, and cellular events that occur during the evolution of diet-induced NASH. Male C57Bl/6 mice were assigned to a fast-food (FF; high calorie, high cholesterol, high fructose) or standard-chow (SC) diet over a period of 36 wk. Liver histology, body composition, mitochondrial respiration, metabolic rate, gene expression, and hepatic lipid content were analyzed. Insulin resistance [homeostasis model assessment-insulin resistance (HOMA-IR)] increased 10-fold after 4 wk. Fibrosing NASH was fully established by 16 wk. Total hepatic lipids increased by 4 wk and remained two- to threefold increased throughout. Hepatic triglycerides declined from sixfold increase at 8 wk to threefold increase by 36 wk. In contrast, hepatic cholesterol levels steadily increased from baseline at 8 wk to twofold by 36 wk. The hepatic immune cell population altered over time with macrophages persisting beyond 16 wk. Mitochondrial oxygen flux rates of FF mice diet were uniformly lower with all the tested substrates (13-276 pmol·s-1·ml-1 per unit citrate synthase) than SC mice (17-394 pmol·s-1·ml-1 per unit citrate synthase) and was accompanied by decreased mitochondrial:nuclear gene copy number ratios after 4 wk. Metabolic rate was lower in FF mice. Mitochondrial glutathione was significantly decreased at 24 wk in FF mice. Expression of dismutases and catalase was also decreased in FF mice. The evolution of NASH in the FF diet-induced model is multiphasic, particularly in terms of hepatic lipid composition. Insulin resistance precedes hepatic inflammation and fibrosis. Mitochondrial dysfunction and depletion occur after the histological features of NASH are apparent. Collectively, these observations provide a unique overview of the sequence of changes that coevolve with the histological evolution of NASH.NEW & NOTEWORTHY This study demonstrates in a first of kind longitudinal analysis, the evolution of nonalcoholic steatohepatitis (NASH) on a fast-food diet-induced model. Key findings include 1) hepatic lipid composition changes in a multiphasic fashion as NASH evolves; 2) insulin resistance precedes hepatic inflammation and fibrosis, answering a longstanding chicken-and-egg question regarding the relationship of insulin resistance to liver histology in NASH; and 3) mitochondrial dysfunction and depletion occur after the histological features of NASH are apparent.
Collapse
Affiliation(s)
| | - Tasduq Sheikh Abdullah
- 2Indian Institute of Integrative Medicine, Council of Scientific and Industrial Research, Jammu and Kashmir, India;
| | - Taofic Mounajjed
- 3Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota;
| | - Stella Hartono
- 4Division of Immunology, Mayo Clinic, Rochester, Minnesota;
| | - Andrea McConico
- 5Division of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota;
| | - Thomas White
- 6Robert and Arlene Kogod Centre for Aging, Mayo Clinic, Rochester, Minnesota;
| | - Nathan LeBrasseur
- 6Robert and Arlene Kogod Centre for Aging, Mayo Clinic, Rochester, Minnesota;
| | - Ian Lanza
- 7Division of Endocrinology, Mayo Clinic, Rochester, Minnesota; and
| | - Sreekumaran Nair
- 7Division of Endocrinology, Mayo Clinic, Rochester, Minnesota; and
| | - Gregory Gores
- 1Division of Gastroenterology, Mayo Clinic, Rochester, Minnesota;
| | - Michael Charlton
- Division of Gastroenterology, Mayo Clinic, Rochester, Minnesota; .,Division of Hepatology, Intermountain Healthcare, Salt Lake City, Utah
| |
Collapse
|
121
|
Chow MD, Lee YH, Guo GL. The role of bile acids in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Mol Aspects Med 2017; 56:34-44. [PMID: 28442273 DOI: 10.1016/j.mam.2017.04.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/11/2017] [Accepted: 04/20/2017] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease is growing in prevalence worldwide. It is marked by the presence of macrosteatosis on liver histology but is often clinically asymptomatic. However, it can progress into nonalcoholic steatohepatitis which is a more severe form of liver disease characterized by inflammation and fibrosis. Further progression leads to cirrhosis, which predisposes patients to hepatocellular carcinoma or liver failure. The mechanism by which simple steatosis progresses to steatohepatitis is not entirely clear. However, multiple pathways have been proposed. A common link amongst many of these pathways is disruption of the homeostasis of bile acids. Other than aiding in the absorption of lipids and lipid-soluble vitamins, bile acids act as ligands. For example, they bind to farnesoid X receptor, which is critically involved in many of the pathways responsible for maintaining bile acid, glucose, and lipid homeostasis. Alterations to these pathways can lead to dysregulation of energy balance and increased inflammation and fibrosis. Repeated insults over time may be the key to development of steatohepatitis. For this reason, current drug therapies target aspects of these pathways to try to reduce and halt inflammation and fibrosis. This review will focus on the role of bile acids in these various pathways and how changes in these pathways may result in steatohepatitis. While there is no approved pharmaceutical treatment for either hepatic steatosis or steatohepatitis, this review will also touch upon the multitude of potential therapies.
Collapse
Affiliation(s)
- Monica D Chow
- Department of Surgery, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Yi-Horng Lee
- Division of Pediatric Surgery, Department of Surgery, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Grace L Guo
- Department of Pharmacy and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
122
|
Dietrich CG, Rau M, Jahn D, Geier A. Changes in drug transport and metabolism and their clinical implications in non-alcoholic fatty liver disease. Expert Opin Drug Metab Toxicol 2017; 13:625-640. [PMID: 28359183 DOI: 10.1080/17425255.2017.1314461] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The incidence of non-alcoholic fatty liver disease (NAFLD) is rising, especially in Western countries. Drug treatment in patients with NAFLD is common since it is linked to other conditions like diabetes, obesity, and cardiovascular disease. Consequently, changes in drug metabolism may have serious clinical implications. Areas covered: A literature search for studies in animal models or patients with obesity, fatty liver, non-alcoholic steatohepatitis (NASH) or NASH cirrhosis published before November 2016 was performed. After discussing epidemiology and animal models for NAFLD, we summarized both basic as well as clinical studies investigating changes in drug transport and metabolism in NAFLD. Important drug groups were assessed separately with emphasis on clinical implications for drug treatment in patients with NAFLD. Expert opinion: Given the frequency of NAFLD even today, a high degree of drug treatment in NAFLD patients appears safe and well-tolerated despite considerable changes in hepatic uptake, distribution, metabolism and transport of drugs in these patients. NASH causes changes in biliary excretion, systemic concentrations, and renal handling of drugs leading to alterations in drug efficacy or toxicity under specific circumstances. Future clinical drug studies should focus on this special patient population in order to avoid serious adverse events in NAFLD patients.
Collapse
Affiliation(s)
- Christoph G Dietrich
- a Bethlehem Center of Health , Department of Medicine , Stolberg/Rhineland , Germany
| | - Monika Rau
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| | - Daniel Jahn
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| | - Andreas Geier
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| |
Collapse
|
123
|
Jasso-Padilla I, Juárez-Flores B, Alvarez-Fuentes G, De la Cruz-Martínez A, González-Ramírez J, Moscosa-Santillán M, González-Chávez M, Oros-Ovalle C, Prell F, Czermak P, Martinez-Gutierrez F. Effect of prebiotics of Agave salmiana fed to healthy Wistar rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2017; 97:556-563. [PMID: 27097820 DOI: 10.1002/jsfa.7764] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/26/2016] [Accepted: 04/16/2016] [Indexed: 05/25/2023]
Abstract
BACKGROUND Inulin and other fructans are synthesized and stored in mezcal agave (Agave salmiana). Fructans provide several health benefits and have excellent technological properties, but only few data report their physiological effect when added in the diet. RESULTS Here, we studied the physiological effects of fructans obtained from A. salmiana when added in the diet of Wistar rats. Results showed favorable changes on Wistar rats when the fructans was added to their diet, including the decrease of the pH in the feces and the increase of the number of lactic acid bacteria (CFU g-1 ) (Lactobacillus spp. and Bifidobacterium spp.), even these changes were enhanced with the synbiotic diet (fructans plus B. animalis subsp. lactis). Synbiotic diet, developed changes in the reduction of cholesterol and triglycerides concentrations in serum, with statistical differences (P < 0.05). Histological analysis of colon sections showed that synbiotic diet promoted colon cells growth suggesting that fructans from A. salmiana confer beneficial health effects through gut microbiota modulation. CONCLUSION Our data underline the advantage of targeting the gut microbiota by colonic nutrients like specific structure of fructans from A. salmiana, with their beneficial effects. More studies are necessary to define the role of fructans to develop more solid therapeutic solutions in humans. © 2016 Society of Chemical Industry.
Collapse
Affiliation(s)
| | - Bertha Juárez-Flores
- Instituto de Investigacion de Zonas Deserticas, Universidad Autonoma de San Luis Potosi, Mexico
| | | | | | | | | | | | - Cuauhtemoc Oros-Ovalle
- Departamento de Patologia, Hospital Central, Dr. Ignacio Morones Prieto, San Luis Potosi, Mexico
| | - Florian Prell
- Justus Liebig University Giessen, Institute of Food Chemistry and Food Biotechnology, Giessen, Germany
| | - Peter Czermak
- Justus Liebig University Giessen, Institute of Food Chemistry and Food Biotechnology, Giessen, Germany
- University of Applied Sciences Mittelhessen, Institute of Bioprocess Engineering and Pharmaceutical Technology, Giessen, Germany
| | | |
Collapse
|
124
|
Myeloid Cell-Specific Lipin-1 Deficiency Stimulates Endocrine Adiponectin-FGF15 Axis and Ameliorates Ethanol-Induced Liver Injury in Mice. Sci Rep 2016; 6:34117. [PMID: 27666676 PMCID: PMC5036185 DOI: 10.1038/srep34117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/07/2016] [Indexed: 02/08/2023] Open
Abstract
Lipin-1 is a phosphatidate phosphohydrolase (PAP) required for the generation of diacylglycerol during glycerolipid synthesis, and exhibits dual functions in the regulation of lipid metabolism. Lipin-1 has been implicated in the pathogenesis of alcoholic liver disease (ALD). In the present study, we assessed lipin-1 function in myeloid cells in ALD using a myeloid cell-specific lipin-1 knockout (mLipin-1KO) mouse model. Utilizing the Gao-binge ethanol feeding protocol, matched mLipin-1KO mice and littermate loxP control (WT) mice were pair-fed with either an ethanol-containing diet or an ethanol-free diet (control). Surprisingly, deletion of lipin-1 in myeloid cells dramatically attenuated liver inflammatory responses and ameliorated liver injury that would normally occur following the ethanol feeding protocol, but slightly exacerbated the ethanol-induced steatosis in mice. Mechanistically, myeloid cell-specific lipin-1 deficiency concomitantly increased the fat-derived adiponectin and ileum-derived fibroblast growth factor (FGF) 15. In concordance with concerted elevation of circulating adiponectin and FGF15, myeloid cell-specific lipin-1 deficiency diminished hepatic nuclear factor kappa B (NF-κB) activity, limited liver inflammatory responses, normalized serum levels of bile acids, and protected mice from liver damage after ethanol challenge. Our novel data demonstrate that myeloid cell-specific deletion of lipin-1 ameliorated inflammation and alcoholic hepatitis in mice via activation of endocrine adiponectin-FGF15 signaling.
Collapse
|
125
|
Hu X, Jogasuria A, Wang J, Kim C, Han Y, Shen H, Wu J, You M. MitoNEET Deficiency Alleviates Experimental Alcoholic Steatohepatitis in Mice by Stimulating Endocrine Adiponectin-Fgf15 Axis. J Biol Chem 2016; 291:22482-22495. [PMID: 27573244 DOI: 10.1074/jbc.m116.737015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 08/25/2016] [Indexed: 12/13/2022] Open
Abstract
MitoNEET (mNT) (CDGSH iron-sulfur domain-containing protein 1 or CISD1) is an outer mitochondrial membrane protein that donates 2Fe-2S clusters to apo-acceptor proteins. In the present study, using a global mNT knock-out (mNTKO) mouse model, we investigated the in vivo functional role of mNT in the development of alcoholic steatohepatitis. Experimental alcoholic steatohepatitis was achieved by pair feeding wild-type (WT) and mNTKO mice with Lieber-DeCarli ethanol-containing diets for 4 weeks. Strikingly, chronically ethanol-fed mNTKO mice were completely resistant to ethanol-induced steatohepatitis as revealed by dramatically reduced hepatic triglycerides, decreased hepatic cholesterol level, diminished liver inflammatory response, and normalized serum ALT levels. Mechanistic studies demonstrated that ethanol administration to mNTKO mice induced two pivotal endocrine hormones, namely, adipose-derived adiponectin and gut-derived fibroblast growth factor 15 (Fgf15). The elevation in circulating levels of adiponectin and Fgf15 led to normalized hepatic and serum levels of bile acids, limited hepatic accumulation of toxic bile, attenuated inflammation, and amelioration of liver injury in the ethanol-fed mNTKO mice. Other potential mechanisms such as reduced oxidative stress, activated Sirt1 signaling, and diminished NF-κB activity also contribute to hepatic improvement in the ethanol-fed mNTKO mice. In conclusion, the present study identified adiponectin and Fgf15 as pivotal adipose-gut-liver metabolic coordinators in mediating the protective action of mNT deficiency against development of alcoholic steatohepatitis in mice. Our findings may help to establish mNT as a novel therapeutic target and pharmacological inhibition of mNT may be beneficial for the prevention and treatment of human alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Xudong Hu
- From the College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio 44272.,the Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China, and
| | - Alvin Jogasuria
- From the College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Jiayou Wang
- From the College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Chunki Kim
- From the College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Yoonhee Han
- From the College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Hong Shen
- From the College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio 44272.,the Department of Liver Diseases, Guangdong Hospital of Traditional Chinese Medicine in Zhuhai, Zhuhai 519015, China
| | - Jiashin Wu
- From the College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Min You
- From the College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio 44272,
| |
Collapse
|
126
|
Simple steatosis sensitizes cholestatic rats to liver injury and dysregulates bile salt synthesis and transport. Sci Rep 2016; 6:31829. [PMID: 27535001 PMCID: PMC4989137 DOI: 10.1038/srep31829] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 07/26/2016] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder. It is uncertain if simple steatosis, the initial and prevailing form of NAFLD, sensitizes the liver to cholestasis. Here, we compared the effects of obstructive cholestasis in rats with a normal liver versus rats with simple steatosis induced by a methionine/choline-deficient diet. We found that plasma liver enzymes were higher and hepatic neutrophil influx, inflammation, and fibrosis were more pronounced in animals with combined steatosis and cholestasis compared to cholestasis alone. Circulating bile salt levels were markedly increased and hepatic bile salt composition shifted from hydrophilic tauro-β-muricholate to hydrophobic taurocholate. This shift was cytotoxic for HepG2 hepatoma cells. Gene expression analysis revealed induction of the rate-limiting enzyme in bile salt synthesis, cytochrome P450 7a1 (CYP7A1), and modulation of the hepatic bile salt transport system. In conclusion, simple steatosis sensitizes the liver to cholestatic injury, inflammation, and fibrosis in part due to a cytotoxic shift in bile salt composition. Plasma bile salt levels were elevated, linked to dysregulation of bile salt synthesis and enhanced trafficking of bile salts from the liver to the systemic circulation.
Collapse
|
127
|
Nuño-Lámbarri N, Barbero-Becerra VJ, Uribe M, Chávez-Tapia NC. Elevated cholesterol levels have a poor prognosis in a cholestasis scenario. J Biochem Mol Toxicol 2016; 31:1-6. [PMID: 27517733 DOI: 10.1002/jbt.21831] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 01/15/2023]
Abstract
Cholestasis results from defective bile flow through the biliary ducts leading to the accumulation of bile acids (BAs) in hepatocytes and serum. It has been seen that cholestasis is associated with hypercholesterolemia, which is a prerequisite for gallstone formation and primary biliary cirrhosis, being some of the most common gastrointestinal disorders in Western societies. Cytotoxic BAs induce proinflammatory mediators, oxidative stress, and apoptosis in hepatocytes, whereas cytoprotective BAs prevent them; they can also modify the plasmatic membrane structure of cells or mitochondrial outer membrane properties as well as the distribution of cholesterol, altering various proteins involved in BAs homeostasis.
Collapse
Affiliation(s)
- Natalia Nuño-Lámbarri
- Traslational Research Unit, Médica Sur Clinic & Foundation, Toriello guerra tlalpan, C.P. 14050, Mexico City, Mexico
| | - Varenka J Barbero-Becerra
- Traslational Research Unit, Médica Sur Clinic & Foundation, Toriello guerra tlalpan, C.P. 14050, Mexico City, Mexico
| | - Misael Uribe
- Obesity and Digestive Diseases Unit, Médica Sur Clinic & Foundation, Toriello guerra tlalpan, C.P. 14050, Mexico City, Mexico
| | - Norberto C Chávez-Tapia
- Traslational Research Unit, Médica Sur Clinic & Foundation, Toriello guerra tlalpan, C.P. 14050, Mexico City, Mexico.,Obesity and Digestive Diseases Unit, Médica Sur Clinic & Foundation, Toriello guerra tlalpan, C.P. 14050, Mexico City, Mexico
| |
Collapse
|
128
|
The intrahepatic expression levels of bile acid transporters are inversely correlated with the histological progression of nonalcoholic fatty liver disease. J Gastroenterol 2016; 51:808-18. [PMID: 26601667 DOI: 10.1007/s00535-015-1148-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/10/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) presents as a spectrum ranging from simple steatosis to nonalcoholic steatohepatitis (NASH). The latter is progressive, and its pathogenesis remains poorly understood. Recently, bile acid (BA) metabolism has become a therapeutic focus in NASH patients. The aim of the present study was to explore changes in bile acid metabolism in NAFLD patients in the context of disease progression. METHODS We prospectively enrolled patients with clinically suspected NAFLD. Patients taking ursodeoxycholic acid were excluded. The intrahepatic expression levels of genes associated with BA metabolism were determined by quantitative PCR and immunohistochemistry. RESULTS Seventy-eight patients (male:female = 49:29) histologically diagnosed with NAFLD were analyzed. The expression levels of farnesoid X receptor, liver receptor homolog 1, and small heterodimer partner, key proteins in BA synthesis, significantly decreased as the NAFLD activity score (NAS) increased in either males or females. The levels of cholesterol 7 alpha-hydroxylase, the rate-limiting enzyme of BA synthesis, were not changed. Notably, the expression levels of a main export transporter, bile salt export pump (BSEP), significantly decreased as the NAS and the each NAS component increased in both genders. The decreases of BSEP levels were also observed by immunohistochemistry, particularly in areas with pronounced fatty changes in cases with high NAS. CONCLUSIONS The expression levels of the BA export transporter BSEP were inversely correlated with NAS in NAFLD patients. Such down-regulation may cause excessive BA levels in hepatocytes, leading to cell injury. Our findings may afford new insights into the pathogenesis of NASH.
Collapse
|
129
|
Mota M, Banini BA, Cazanave SC, Sanyal AJ. Molecular mechanisms of lipotoxicity and glucotoxicity in nonalcoholic fatty liver disease. Metabolism 2016; 65:1049-61. [PMID: 26997538 PMCID: PMC4931958 DOI: 10.1016/j.metabol.2016.02.014] [Citation(s) in RCA: 368] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 02/05/2016] [Accepted: 02/23/2016] [Indexed: 12/21/2022]
Abstract
The exposure of hepatocytes to high concentrations of lipids and carbohydrates and the ensuing hepatocellular injury are termed lipotoxicity and glucotoxicity, respectively. A common denominator is metabolic derangement, especially in regards to intracellular energy homeostasis, which is brought on by glucose intolerance and insulin resistance in tissues. In this review, we highlight the lipids and carbohydrates that provoke hepatocyte injury and the mechanisms involved in lipotoxicity and glucotoxicity, including endoplasmic reticulum stress, oxidative stress and mitochondrial impairment. Through upregulation of proteins involved in various pathways including PKR-like ER kinase (PERK), CCAAT/enhancer-binding homologous protein (CHOP), c-Jun NH2-terminal kinase-1 (JNK), Bcl-2 interacting mediator (BIM), p53 upregulated modulator of apoptosis (PUMA), and eventually caspases, hepatocytes in lipotoxic states ultimately undergo apoptosis. The protective role of certain lipids and possible targets for pharmacological therapy are explored. Finally, we discuss the role of high fructose and glucose diets in contributing to organelle impairment and poor glucose transport mechanisms, which perpetuate hyperglycemia and hyperlipidemia by shunting of excess carbohydrates into lipogenesis.
Collapse
Affiliation(s)
- Manoela Mota
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | - Bubu A Banini
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Sophie C Cazanave
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Arun J Sanyal
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
130
|
McMahan RH, Porsche CE, Edwards MG, Rosen HR. Free Fatty Acids Differentially Downregulate Chemokines in Liver Sinusoidal Endothelial Cells: Insights into Non-Alcoholic Fatty Liver Disease. PLoS One 2016; 11:e0159217. [PMID: 27454769 PMCID: PMC4959750 DOI: 10.1371/journal.pone.0159217] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease is a prevalent problem throughout the western world. Liver sinusoidal endothelial cells (LSEC) have been shown to play important roles in liver injury and repair, but their role in the underlying pathogenetic mechanisms of non-alcoholic fatty liver disease remains undefined. Here, we evaluated the effects of steatosis on LSEC gene expression in a murine model of non-alcoholic fatty liver disease and an immortalized LSEC line. Using microarray we identified distinct gene expression profiles following exposure to free fatty acids. Gene pathway analysis showed a number of differentially expressed genes including those involved in lipid metabolism and signaling and inflammation. Interestingly, in contrast to hepatocytes, fatty acids led to decreased expression of pro-inflammatory chemokines including CCL2 (MCP-1), CXCL10 and CXCL16 in both primary and LSEC cell lines. Chemokine downregulation translated into a significant inhibition of monocyte migration and LSECs isolated from steatotic livers demonstrated a similar shift towards an anti-inflammatory phenotype. Overall, these pathways may represent a compensatory mechanism to reverse the liver damage associated with non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Rachel H. McMahan
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Cara E. Porsche
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Michael G. Edwards
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Hugo R. Rosen
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Denver, Aurora, Colorado, United States of America
- Department of Microbiology and Immunology, University of Colorado Denver, Aurora, Colorado, United States of America
- Denver Veteran’s Affairs Medical Center, Denver, Colorado, United States of America
- * E-mail:
| |
Collapse
|
131
|
Duan XP, Meng Q, Liu KX. Nuclear receptor FXR: A potential therapeutic target for non-alcoholic steatohepatitis. Shijie Huaren Xiaohua Zazhi 2016; 24:2289-2297. [DOI: 10.11569/wcjd.v24.i15.2289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become a very common chronic liver disease all over the world. The high incidence of NAFLD is closely related to obesity, diabetes and metabolic disorders. Insulin resistance and dyslipidemia following the hepatic proinflammatory response and fibrosis are the primary features of NAFLD deterioration. Nuclear receptor farnesoid X receptor (FXR) regulates lipid metabolism and homeostasis. Clarification of FXR function and features can provide a better understanding of the pathophysiological characteristics of non-alcoholic steatohepatitis (NASH) and illuminate the mechanism of NAFLD/NASH potential therapeutic targets. FXR activation can inhibit the de novo hepatic lipogenesis, improve insulin sensitivity and protect against bile acid-induced cytotoxicity. Clinical studies indicated that FXR agonists or modulators are very promising for the clinical treatment of NAFLD and NASH. This review focuses on the important regulatory role of FXR in NASH.
Collapse
|
132
|
Awad ASM, Abd Al Haleem EN, El-Bakly WM, Sherief MA. Thymoquinone alleviates nonalcoholic fatty liver disease in rats via suppression of oxidative stress, inflammation, apoptosis. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:381-91. [PMID: 26753695 DOI: 10.1007/s00210-015-1207-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/21/2015] [Indexed: 02/07/2023]
Abstract
Nonalcoholic steatohepatitis (NAFLD) is a progressive form of liver disease that leads to advanced fibrosis. The present study was designed to assess the hepatoprotective effect of thymoquinone (TQ) on liver functions, insulin resistance, and PPAR-γ expression in NAFLD. Rats were divided into two main groups: one fed with normal rat chow diet and the other with high-fat high-cholesterol diet group for 6 weeks. Every group was subdivided into three subgroups (n = 8): treated with saline, low dose TQ (10 mg/kg), high dose TQ (20 mg/kg). High fat high cholesterol diet caused marked liver damage as noted in histopathology and significant increase in liver index, liver enzymes. There was significant increase in the insulin resistance, serum cholesterol, triglyceride, PPAR-γ gene overexpression with significant decrease in HDL. Additionally, oxidative stress increased by measuring MDA associated with significant decrease in serum total antioxidant capacity. As markers of inflammation, hepatic TNF-α was significantly increased with decrease in IL10. Further, there was increase in BAX protein with decrease in Bcl as compared to control group. This model of 6 weeks high-fat high-cholesterol diet showed minimal fibrosis as noticed by increase MMP2 and Masson trichrome satin. Co-treatment with TQ improved all previous parameters. High dose was more effective, although mostly non-statistically significant. TQ may have a promising agent to improve hepatic steatosis, oxidative stress; inflammatory, apoptotic status, fibrosis and so prevent liver damage in patients with NAFLD. Although PPAR-γ was significantly under-expressed by TQ, insulin resistance was improved significantly suggesting a role of liver damage.
Collapse
Affiliation(s)
- Azza S M Awad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Ekram N Abd Al Haleem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Wesam M El-Bakly
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Mohie A Sherief
- Department of Pharmacology, Faculty of Medicine, Benha University, Al Qalyubia Governorate, Egypt
| |
Collapse
|
133
|
Alexandre PA, Kogelman LJA, Santana MHA, Passarelli D, Pulz LH, Fantinato-Neto P, Silva PL, Leme PR, Strefezzi RF, Coutinho LL, Ferraz JBS, Eler JP, Kadarmideen HN, Fukumasu H. Liver transcriptomic networks reveal main biological processes associated with feed efficiency in beef cattle. BMC Genomics 2015; 16:1073. [PMID: 26678995 PMCID: PMC4683712 DOI: 10.1186/s12864-015-2292-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 12/14/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The selection of beef cattle for feed efficiency (FE) traits is very important not only for productive and economic efficiency but also for reduced environmental impact of livestock. Considering that FE is multifactorial and expensive to measure, the aim of this study was to identify biological functions and regulatory genes associated with this phenotype. RESULTS Eight genes were differentially expressed between high and low feed efficient animals (HFE and LFE, respectively). Co-expression analyses identified 34 gene modules of which 4 were strongly associated with FE traits. They were mainly enriched for inflammatory response or inflammation-related terms. We also identified 463 differentially co-expressed genes which were functionally enriched for immune response and lipid metabolism. A total of 8 key regulators of gene expression profiles affecting FE were found. The LFE animals had higher feed intake and increased subcutaneous and visceral fat deposition. In addition, LFE animals showed higher levels of serum cholesterol and liver injury biomarker GGT. Histopathology of the liver showed higher percentage of periportal inflammation with mononuclear infiltrate. CONCLUSION Liver transcriptomic network analysis coupled with other results demonstrated that LFE animals present altered lipid metabolism and increased hepatic periportal lesions associated with an inflammatory response composed mainly by mononuclear cells. We are now focusing to identify the causes of increased liver lesions in LFE animals.
Collapse
Affiliation(s)
- Pamela A Alexandre
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13635-900, Brazil. .,Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Lisette J A Kogelman
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Miguel H A Santana
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13635-900, Brazil.
| | - Danielle Passarelli
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13635-900, Brazil.
| | - Lidia H Pulz
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13635-900, Brazil.
| | - Paulo Fantinato-Neto
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13635-900, Brazil.
| | - Paulo L Silva
- Department of Animal Sciences, School of Animal Science and Food Engineering, University of São Paulo, Pirassunung, Sao Paulo, Brazil.
| | - Paulo R Leme
- Department of Animal Sciences, School of Animal Science and Food Engineering, University of São Paulo, Pirassunung, Sao Paulo, Brazil.
| | - Ricardo F Strefezzi
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13635-900, Brazil.
| | - Luiz L Coutinho
- Department of Animal Sciences, ESALQ, University of Sao Paulo, Piracicaba, Sao Paulo, Brazil.
| | - José B S Ferraz
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13635-900, Brazil.
| | - Joanie P Eler
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13635-900, Brazil.
| | - Haja N Kadarmideen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Heidge Fukumasu
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of Sao Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, São Paulo, 13635-900, Brazil.
| |
Collapse
|
134
|
Wang CR, Chen LP, Tan C. Liver cell injury caused by bad habits. Shijie Huaren Xiaohua Zazhi 2015; 23:5642-5648. [DOI: 10.11569/wcjd.v23.i35.5642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Apoptosis and necrosis of liver cells induced by environmental or genetic factors are the main features of liver injury. Liver injury is usually caused by apoptosis of liver cells, which is controlled by a complex regulatory system involved in liver damage and secondary inflammation. This article aims to review liver injury caused by bad habits and the underlying molecular mechanisms.
Collapse
|
135
|
ZAGOROVA M, PRASNICKA A, KADOVA Z, DOLEZELOVA E, KAZDOVA L, CERMANOVA J, ROZKYDALOVA L, HROCH M, MOKRY J, MICUDA S. Boldine Attenuates Cholestasis Associated With Nonalcoholic Fatty Liver Disease in Hereditary Hypertriglyceridemic Rats Fed by High-Sucrose Diet. Physiol Res 2015; 64:S467-76. [DOI: 10.33549/physiolres.933206] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of the current study was to clarify the effect of high sucrose diet (HSD) on bile formation (BF) in rats with hereditary hypertriglyceridemia (HHTg). Potentially positive effects were studied for boldine, a natural choleretic agent. Administration of HSD to HHTg rats led to increased triglyceride deposition in the liver. HSD reduced BF as a consequence of decreased biliary secretion of bile acids (BA) and glutathione. Responsible mechanism was down-regulation of hepatic transporters for BA and glutathione, Bsep and Mrp2, respectively. Moreover, gene expressions of transporters for other constituents of bile, namely Abcg5/8 for cholesterol, Abcb4 for phospholipids, and Oatp1a4 for xenobiotics, were also reduced by HSD. Boldine partially attenuated cholestatic effect of HSD by promotion of biliary secretion of BA through up-regulation of Bsep and Ntcp, and by increase in biliary secretion of glutathione as a consequence of its increased hepatic disposition. This study demonstrates mechanisms of impaired BF during nonalcoholic fatty liver disease induced by HSD. Altered function of responsible transporters suggests also potential for changes in kinetics of drugs, which may complicate pharmacotherapy in subjects with high intake of sucrose, and with fatty liver disease. Sucrose induced alterations in BF may be alleviated by administration of boldine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - S. MICUDA
- Department of Pharmacology, Charles University in Prague, Faculty of Medicine in Hradec Kralove, Czech Republic
| |
Collapse
|
136
|
Ferslew BC, Xie G, Johnston CK, Su M, Stewart PW, Jia W, Brouwer KL, Barritt AS. Altered Bile Acid Metabolome in Patients with Nonalcoholic Steatohepatitis. Dig Dis Sci 2015; 60:3318-28. [PMID: 26138654 PMCID: PMC4864493 DOI: 10.1007/s10620-015-3776-8] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/17/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS The prevalence of nonalcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH) is increasing at an alarming rate. The role of bile acids in the development and progression of NAFLD to NASH and cirrhosis is poorly understood. This study aimed to quantify the bile acid metabolome in healthy subjects and patients with non-cirrhotic NASH under fasting conditions and after a standardized meal. METHODS Liquid chromatography tandem mass spectroscopy was used to quantify 30 serum and 16 urinary bile acids from 15 healthy volunteers and 7 patients with biopsy-confirmed NASH. Bile acid concentrations were measured at two fasting and four post-prandial time points following a high-fat meal to induce gallbladder contraction and bile acid reabsorption from the intestine. RESULTS Patients with NASH had significantly higher total serum bile acid concentrations than healthy subjects under fasting conditions (2.2- to 2.4-fold increase in NASH; NASH 2595-3549 µM and healthy 1171-1458 µM) and at all post-prandial time points (1.7- to 2.2-fold increase in NASH; NASH 4444-5898 µM and healthy 2634-2829 µM). These changes were driven by increased taurine- and glycine-conjugated primary and secondary bile acids. Patients with NASH exhibited greater variability in their fasting and post-prandial bile acid profile. CONCLUSIONS Results indicate that patients with NASH have higher fasting and post-prandial exposure to bile acids, including the more hydrophobic and cytotoxic secondary species. Increased bile acid exposure may be involved in liver injury and the pathogenesis of NAFLD and NASH.
Collapse
Affiliation(s)
- Brian C. Ferslew
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, CB #7569 Kerr Hall Chapel Hill, NC 27599,Clinical Pharmacology and Drug Metabolism and Pharmacokinetics, Theravance Biopharma US, Inc., 901 Gateway Blvd, South San Francisco, CA 94080
| | - Guoxiang Xie
- Metabolomics Shared Resource, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813
| | - Curtis K. Johnston
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, CB #7569 Kerr Hall Chapel Hill, NC 27599
| | - Mingming Su
- Metabolomics Shared Resource, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813
| | - Paul W. Stewart
- Department of Biostatistics, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 3105G McGavran-Greenberg Hall, Chapel Hill, NC, 27599
| | - Wei Jia
- Metabolomics Shared Resource, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI 96813
| | - Kim L.R. Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, CB #7569 Kerr Hall Chapel Hill, NC 27599
| | - A. Sidney Barritt
- Division of Gastroenterology and Hepatology, UNC School of Medicine, University of North Carolina at Chapel Hill, 8004 Burnett Womack CB #7584, Chapel Hill, NC 27599
| |
Collapse
|
137
|
Dong Q, Li N, Li Q, Zhang CE, Feng WW, Li GQ, Li RY, Tu C, Han X, Bai ZF, Zhang YM, Niu M, Ma ZJ, Xiao XH, Wang JB. Screening for biomarkers of liver injury induced by Polygonum multiflorum: a targeted metabolomic study. Front Pharmacol 2015; 6:217. [PMID: 26483689 PMCID: PMC4591842 DOI: 10.3389/fphar.2015.00217] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/15/2015] [Indexed: 12/18/2022] Open
Abstract
Heshouwu (HSW), the dry roots of Polygonum multiflorum, a classical traditional Chinese medicine is used as a tonic for a wide range of conditions, particularly those associated with aging. However, it tends to be taken overdose or long term in these years, which has resulted in liver damage reported in many countries. In this study, the indicative roles of nine bile acids (BAs) were evaluated to offer potential biomarkers for HSW induced liver injury. Nine BAs including cholic acid (CA) and chenodeoxycholic acid (CDCA), taurocholic acid (TCA), glycocholic acid (GCA), glycochenodeoxycholic acid (GCDCA), deoxycholic acid (DCA), glycodeoxycholic acid (GDCA), ursodeoxycholic acid (UDCA), and hyodeoxycholic acid (HDCA) in rat bile and serum were detected by a developed LC-MS method after 42 days treatment. Partial least square-discriminate analysis (PLS-DA) was applied to evaluate the indicative roles of the nine BAs, and metabolism of the nine BAs was summarized. Significant change was observed for the concentrations of nine BAs in treatment groups compared with normal control; In the PLS-DA plots of nine BAs in bile, normal control and raw HSW groups were separately clustered and could be clearly distinguished, GDCA was selected as the distinguished components for raw HSW overdose treatment group. In the PLS-DA plots of nine BAs in serum, the normal control and raw HSW overdose treatment group were separately clustered and could be clearly distinguished, and HDCA was selected as the distinguished components for raw HSW overdose treatment group. The results indicated the perturbation of nine BAs was associated with HSW induced liver injury; GDCA in bile, as well as HDCA in serum could be selected as potential biomarkers for HSW induced liver injury; it also laid the foundation for the further search on the mechanisms of liver injury induced by HSW.
Collapse
Affiliation(s)
- Qin Dong
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China ; College of Pharmacy, Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Na Li
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Qi Li
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Cong-En Zhang
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China ; College of Pharmacy, Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Wu-Wen Feng
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China ; College of Pharmacy, Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Guang-Quan Li
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China ; College of Pharmacy, Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Rui-Yu Li
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China ; College of Pharmacy, Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Can Tu
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Xue Han
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China ; College of Pharmacy, Chengdu University of Traditional Chinese Medicine Chengdu, China
| | - Zhao-Fang Bai
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Ya-Ming Zhang
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Zhi-Jie Ma
- Beijing Friendship Hospital, Capital Medical University Beijing, China
| | - Xiao-He Xiao
- Integrative Medicine Center, 302 Military Hospital Beijing, China
| | - Jia-Bo Wang
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| |
Collapse
|
138
|
Abstract
Hepatitis B virus (HBV) infection affects 240 million people worldwide. A liver-specific bile acid transporter named the sodium taurocholate cotransporting polypeptide (NTCP) has been identified as the cellular receptor for HBV and its satellite, the hepatitis D virus (HDV). NTCP likely acts as a major determinant for the liver tropism and species specificity of HBV and HDV at the entry level. NTCP-mediated HBV entry interferes with bile acid transport in cell cultures and has been linked with alterations in bile acid and cholesterol metabolism in vivo. The human liver carcinoma cell line HepG2, complemented with NTCP, now provides a valuable platform for studying the basic biology of the viruses and developing treatments for HBV infection. This review summarizes critical findings regarding NTCP's role as a viral receptor for HBV and HDV and discusses important questions that remain unanswered.
Collapse
Affiliation(s)
- Wenhui Li
- National Institute of Biological Sciences, Zhongguancun Life Science Park, Beijing 102206, China;
| |
Collapse
|
139
|
Rantakokko P, Männistö V, Airaksinen R, Koponen J, Viluksela M, Kiviranta H, Pihlajamäki J. Persistent organic pollutants and non-alcoholic fatty liver disease in morbidly obese patients: a cohort study. Environ Health 2015; 14:79. [PMID: 26420011 PMCID: PMC4588245 DOI: 10.1186/s12940-015-0066-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/17/2015] [Indexed: 05/02/2023]
Abstract
BACKGROUND In animal experiments persistent organic pollutants (POPs) cause hepatosteatosis. In epidemiological studies POPs have positive associations with serum markers of nonalcoholic fatty liver disease (NAFLD) and together with obesity synergistic association with insulin resistance. Because insulin resistance and obesity are critical in NAFLD pathogenesis, we investigated the association of serum pollutant levels with liver histology and alanine aminotransferase (ALT) in morbidly obese. METHODS Liver biopsies were from 161 participants of the Kuopio Obesity Surgery Study (KOBS) who underwent bariatric surgery 2005-2011. Liver histology was categorized as normal, steatosis and non-alcoholic steatohepatitis (NASH). Liver phenotype at baseline and ALT at baseline and 12 months post-surgery were correlated to serum POP concentrations at respective time points. As lipophilic POPs concentrate to smaller fat volume during weight loss, serum levels before and 12 months after bariatric surgery were compared. RESULTS Baseline serum concentration of PCB-118, β-HCH and several PFAAs had an inverse association with lobular inflammation possibly due to changes in bile acid metabolism. ALT had negative associations with many POPs at baseline that turned positive at 12 months after major clinical improvements. There was an interaction between some POPs and sex at 12 months, and in stratified data positive associations were observed mainly in females but not in males. CONCLUSIONS We found a negative association between serum concentrations of PCB-118, β-HCH and several PFAAs with lobular inflammation at baseline. Positive POPs-ATL associations at 12 months among women suggest that increased POP concentrations may decrease the degree of liver recovery.
Collapse
Affiliation(s)
- Panu Rantakokko
- National Institute for Health and Welfare, Department of Health Protection, Chemicals and Health Unit, Kuopio, Finland.
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland.
| | - Riikka Airaksinen
- National Institute for Health and Welfare, Department of Health Protection, Chemicals and Health Unit, Kuopio, Finland.
| | - Jani Koponen
- National Institute for Health and Welfare, Department of Health Protection, Chemicals and Health Unit, Kuopio, Finland.
| | - Matti Viluksela
- National Institute for Health and Welfare, Department of Health Protection, Chemicals and Health Unit, Kuopio, Finland.
- Department of Environmental Science, University of Eastern Finland, Kuopio, Finland.
| | - Hannu Kiviranta
- National Institute for Health and Welfare, Department of Health Protection, Chemicals and Health Unit, Kuopio, Finland.
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.
- Clinical Nutrition and Obesity Center, Kuopio University Hospital, Kuopio, Finland.
| |
Collapse
|
140
|
Watanabe S, Fujita K, Tsuneyama K, Nose M. Changes in liver lipidomics associated with sodium cholate-induced liver injury and its prevention by boiogito, a Japanese herbal medicine, in mice. ACTA ACUST UNITED AC 2015. [DOI: 10.1002/tkm2.1032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shiro Watanabe
- Division of Nutritional Biochemistry; Institute of Natural Medicine, University of Toyama; Toyama Japan
| | - Kyosuke Fujita
- Division of Nutritional Biochemistry; Institute of Natural Medicine, University of Toyama; Toyama Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine; Tokushima University Graduate School; Tokushima Japan
| | - Mitsuhiko Nose
- Department of Pharmacognosy; Faculty of Pharmacy, Meijo University; Nagoya Japan
| |
Collapse
|
141
|
Kälsch J, Bechmann LP, Heider D, Best J, Manka P, Kälsch H, Sowa JP, Moebus S, Slomiany U, Jöckel KH, Erbel R, Gerken G, Canbay A. Normal liver enzymes are correlated with severity of metabolic syndrome in a large population based cohort. Sci Rep 2015; 5:13058. [PMID: 26269425 PMCID: PMC4535035 DOI: 10.1038/srep13058] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023] Open
Abstract
Key features of the metabolic syndrome are insulin resistance and diabetes. The liver as central metabolic organ is not only affected by the metabolic syndrome as non-alcoholic fatty liver disease (NAFLD), but may contribute to insulin resistance and metabolic alterations. We aimed to identify potential associations between liver injury markers and diabetes in the population-based Heinz Nixdorf RECALL Study. Demographic and laboratory data were analyzed in participants (n = 4814, age 45 to 75y). ALT and AST values were significantly higher in males than in females. Mean BMI was 27.9 kg/m2 and type-2-diabetes (known and unkown) was present in 656 participants (13.7%). Adiponectin and vitamin D both correlated inversely with BMI. ALT, AST, and GGT correlated with BMI, CRP and HbA1c and inversely correlated with adiponectin levels. Logistic regression models using HbA1c and adiponectin or HbA1c and BMI were able to predict diabetes with high accuracy. Transaminase levels within normal ranges were closely associated with the BMI and diabetes risk. Transaminase levels and adiponectin were inversely associated. Re-assessment of current normal range limits should be considered, to provide a more exact indicator for chronic metabolic liver injury, in particular to reflect the situation in diabetic or obese individuals.
Collapse
Affiliation(s)
- Julia Kälsch
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen
| | - Lars P Bechmann
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen
| | - Dominik Heider
- Department of Bioinformatics, Straubing Center of Science, University of Applied Science Weihenstephan-Triesdorf
| | - Jan Best
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen
| | - Paul Manka
- 1] Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen [2] Regeneration and Repair Group, The Institute of Hepatology, Foundation for Liver Research, London, UK
| | - Hagen Kälsch
- Department of Cardiology, West-German Heart Center, University Hospital, University Duisburg-Essen
| | - Jan-Peter Sowa
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen
| | - Susanne Moebus
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital, University Duisburg-Essen
| | - Uta Slomiany
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital, University Duisburg-Essen
| | - Karl-Heinz Jöckel
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital, University Duisburg-Essen
| | - Raimund Erbel
- Department of Cardiology, West-German Heart Center, University Hospital, University Duisburg-Essen
| | - Guido Gerken
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen
| | - Ali Canbay
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen
| |
Collapse
|
142
|
The nuclear receptor FXR, but not LXR, up-regulates bile acid transporter expression in non-alcoholic fatty liver disease. Ann Hepatol 2015. [DOI: 10.1016/s1665-2681(19)31170-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
143
|
Liu Y, Chen H, Wang J, Zhou W, Sun R, Xia M. Association of serum retinoic acid with hepatic steatosis and liver injury in nonalcoholic fatty liver disease. Am J Clin Nutr 2015; 102:130-7. [PMID: 25948673 DOI: 10.3945/ajcn.114.105155] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/25/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Retinoic acid (RA), an active metabolite of vitamin A (retinol), has been implicated in the regulation of lipid metabolism and hepatic steatosis in animal models. However, the relation between RA and liver histology in patients with nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) is unknown. OBJECTIVE This study aimed at examining the association of RA with NAFLD and NASH in Chinese subjects. DESIGN Serum RA concentration was determined by ELISA in 41 control subjects, 45 patients with NAFLD, and 38 patients with NASH. The associations of RA with adiposity, serum glucose, lipid profiles, and markers of liver damage were studied. Moreover, both mRNA and protein levels of retinoic X receptor α (RXRα) in the liver were analyzed in subjects with different degrees of hepatic steatosis. RESULTS Serum RA concentrations in patients with NAFLD (1.42 ± 0.47 ng/mL) and NASH (1.14 ± 0.26 ng/mL) were significantly lower than those in control subjects (2.70 ± 0.52 ng/mL) (P < 0.01). Furthermore, serum RA concentrations were significantly different between subjects with normal glucose tolerance and those with type 2 diabetes in control [2.87 ± 0.52 (n = 28) vs. 2.32 ± 0.44 ng/mL (n = 13)], NAFLD [1.61 ± 0.37 (n = 29) vs. 1.28 ± 0.41 ng/mL (n = 16)], and NASH [1.35 ± 0.34 (n = 24) vs. 1.07 ± 0.29 ng/mL (n = 14)] groups. In human liver tissue, RXRα mRNA expression was inversely correlated with the exacerbation of hepatic steatosis. Both serum RA concentrations and RXRα mRNA levels were inversely correlated with intrahepatic triglyceride content (r = -0.700, P < 0.001, and r = -0.611, P = 0.002, respectively). Compared with grade 0 severity, the concentration of RXRα protein was lower in more severe grades in patients with NAFLD. CONCLUSION These results show that circulating RA concentrations were lower in subjects with NAFLD and were associated with hepatic lipid metabolism and insulin resistance. This trial was registered at clinicaltrials.gov as NCT01940263.
Collapse
Affiliation(s)
- Yan Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hongen Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jingjing Wang
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wenjing Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ruifang Sun
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
144
|
Bile Acids as Biomarkers: Liver and Gut Cross Talk in NAFLD. J Pediatr Gastroenterol Nutr 2015; 61:2-3. [PMID: 25844712 DOI: 10.1097/mpg.0000000000000809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
145
|
Männistö VT, Simonen M, Hyysalo J, Soininen P, Kangas AJ, Kaminska D, Matte AK, Venesmaa S, Käkelä P, Kärjä V, Arola J, Gylling H, Cederberg H, Kuusisto J, Laakso M, Yki-Järvinen H, Ala-Korpela M, Pihlajamäki J. Ketone body production is differentially altered in steatosis and non-alcoholic steatohepatitis in obese humans. Liver Int 2015; 35:1853-61. [PMID: 25533197 DOI: 10.1111/liv.12769] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/13/2014] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Levels of ketone bodies have been reported to be both increased and decreased in individuals with non-alcoholic fatty liver disease. We investigated whether the metabolism of ketone bodies is different in simple steatosis and in non-alcoholic steatohepatitis (NASH). METHODS Serum low molecular weight molecules including ketone bodies were measured using high-throughput proton (1H) nuclear magnetic resonance in 116 (76 categorized unequivocally to those with normal liver, simple steatosis or NASH) morbidly obese individuals [age 47.3 ± 8.7 (mean ± SD) years, body mass index 45.1 ± 6.1 kg/m(2) , 39 men and 77 women] with histological assessment of NASH and analysis of gene expression in the liver. Finally, we correlated β-hydroxybutyrate (β-OHB) levels with NASH predicting score in Metabolic Syndrome in Men Study (METSIM) population study (n = 8749 non-diabetic men). RESULTS Levels of ketone bodies were lower in individuals with NASH compared to individuals with simple steatosis (P = 0.004 and P = 0.018 for β-OHB and acetoacetate respectively). Lower levels of β-OHB were associated with the NASH predicting score in the METSIM study (P = 0.001). Liver inflammation correlated with mRNA expression of genes regulating ketolysis in the liver (Spearman correlation 0.379-0.388, P < 0.0006 for ACAT1, ACSS2 and BDH1). CONCLUSION Lower levels of ketone bodies in individuals with NASH compared to individuals with simple steatosis suggest a decrease in ketone body metabolism in NASH.
Collapse
Affiliation(s)
- Ville T Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Marko Simonen
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jenni Hyysalo
- Department of Medicine, University of Helsinki, Helsinki and Minerva Medical Research Institute, Helsinki, Finland
| | - Pasi Soininen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.,Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Antti J Kangas
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.,Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Dorota Kaminska
- Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Ananda K Matte
- Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Sari Venesmaa
- Department of Surgery, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Pirjo Käkelä
- Department of Surgery, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Vesa Kärjä
- Department of Pathology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Johanna Arola
- Department of Pathology, The Laboratory of Helsinki University Central Hospital, Helinski, Finland
| | - Helena Gylling
- Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Division of Internal Medicine, University of Helsinki, Helsinki, Finland
| | - Henna Cederberg
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki, Helsinki and Minerva Medical Research Institute, Helsinki, Finland
| | - Mika Ala-Korpela
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.,Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland.,Computational Medicine, School of Social and Community Medicine & Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Jussi Pihlajamäki
- Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Clinical Nutrition and Obesity Center, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
146
|
Sydor S, Canbay A, Bechmann LP. Identifying soluble mediators of nuclear receptor and insulin signaling may enhance noninvasive diagnosis of fibrosis in Fatty liver disease. Digestion 2015; 90:33-4. [PMID: 25139186 DOI: 10.1159/000365886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Svenja Sydor
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | | | | |
Collapse
|
147
|
González-Reimers E, Quintero-Platt G, Rodríguez-Gaspar M, Alemán-Valls R, Pérez-Hernández O, Santolaria-Fernández F. Liver steatosis in hepatitis C patients. World J Hepatol 2015; 7:1337-1346. [PMID: 26052379 PMCID: PMC4450197 DOI: 10.4254/wjh.v7.i10.1337] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 01/31/2015] [Accepted: 03/09/2015] [Indexed: 02/06/2023] Open
Abstract
There is controversy regarding some aspects of hepatitis C virus (HCV) infection-associated liver steatosis, and their relationship with body fat stores. It has classically been found that HCV, especially genotype 3, exerts direct metabolic effects which lead to liver steatosis. This supports the existence of a so called viral steatosis and a metabolic steatosis, which would affect HCV patients who are also obese or diabetics. In fact, several genotypes exert metabolic effects which overlap with some of those observed in the metabolic syndrome. In this review we will analyse the pathogenic pathways involved in the development of steatosis in HCV patients. Several cytokines and adipokines also become activated and are involved in “pure” steatosic effects, in addition to inflammation. They are probably responsible for the evolution of simple steatosis to steatohepatitis, making it difficult to explain why such alterations only affect a proportion of steatosic patients.
Collapse
|
148
|
Beilfuss A, Sowa JP, Sydor S, Beste M, Bechmann LP, Schlattjan M, Syn WK, Wedemeyer I, Mathé Z, Jochum C, Gerken G, Gieseler RK, Canbay A. Vitamin D counteracts fibrogenic TGF-β signalling in human hepatic stellate cells both receptor-dependently and independently. Gut 2015; 64:791-9. [PMID: 25134788 DOI: 10.1136/gutjnl-2014-307024] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 07/31/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is closely linked to obesity and constitutes part of the metabolic syndrome, which have been associated with low serum vitamin D (VD). Due to known crosstalk between VD and transforming growth factor (TGF)-β signalling, VD has been proposed as an antifibrotic treatment. DESIGN We evaluated the association between VD, the vitamin D receptor (VDR) and liver fibrosis in primary human hepatic stellate cells (phHSC) and 106 morbidly obese patients with NAFLD. RESULTS Treating phHSC with VD ameliorated TGF-β-induced fibrogenesis via both VDR-dependent and VDR-independent mechanisms. Reduction of fibrogenic response was abolished in cells homozygous for GG at the A1012G single nucleotide polymorphisms within the VDR gene. Compared with healthy livers, NAFLD livers expressed higher levels of VDR mRNA and VDR fragments. VDR mRNA was lower in patients homozygous for GG at A1012G and expression of pro-fibrogenic genes was higher in patients carrying the G allele. CONCLUSIONS VD may be an antifibrotic treatment option early in the onset of fibrosis in specific genotypes for VDR. Known polymorphisms of the VDR may influence the response to VD treatment.
Collapse
Affiliation(s)
- Anja Beilfuss
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Jan-Peter Sowa
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Svenja Sydor
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Mechthild Beste
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Lars P Bechmann
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Martin Schlattjan
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Wing-Kin Syn
- The Institute of Hepatology, Regeneration and Repair Group, London, UK
| | - Inga Wedemeyer
- Institute for Pathology, University Hospital Cologne, Cologne, Germany
| | - Zoltan Mathé
- Department of General, Visceral and Transplantation Surgery, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Christoph Jochum
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Guido Gerken
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Robert K Gieseler
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany Rodos BioTarget GmbH, Medical Park Hannover, Hannover, Germany
| | - Ali Canbay
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
149
|
Yang Q, Yang F, Tang X, Ding L, Xu Y, Xiong Y, Wang Z, Yang L. Chlorpromazine-induced perturbations of bile acids and free fatty acids in cholestatic liver injury prevented by the Chinese herbal compound Yin-Chen-Hao-Tang. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 15:122. [PMID: 25887351 PMCID: PMC4410582 DOI: 10.1186/s12906-015-0627-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 03/20/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUNDS Yin-Chen-Hao-Tang (YCHT), a commonly used as a traditional chinese medicine for liver disease. Several studies indicated that YCHT may improving hepatic triglyceride metabolism and anti-apoptotic response as well as decreasing oxidative stress .However, little is known about the role of YCHT in chlorpromazine (CPZ) -induced chlolestatic liver injury. Therefore, we aimed to facilitate the understanding of the pathogenesis of cholestatic liver injury and evaluate the effect of Yin-Chen-Hao-Tang (YCHT) on chlorpromazine (CPZ)-induced cholestatic liver injury in rats based on the change of bile acids (BAs) and free fatty acids (FFAs) alone with the biochemical indicators and histological examination. METHODS We conducted an experiment on CPZ-induced cholestatic liver injury in Wistar rats with and without YCHT for nine consecutive days. Serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin (ALB), total bilirubin (TBIL), total cholesterol (TC), triglycerides (TG), low density lipoprotein-cholesterol (LDL-C) were measured to evaluate the protective effect of YCHT against chlorpromazine (CPZ)-induced cholestatic liver injury. Histopathology of the liver tissue showed that pathological injuries were relieved after YCHT pretreatment. In addition, ultra-performance lipid chromatography coupled with quadrupole mass spectrometry (UPLC-MS) and gas chromatography coupled with mass spectrometry (GC-MS) was applied to determine the content of bile acids, free fatty acids, respectively. RESULTS Obtained data showed that YCHT attenuated the effect of CPZ-induced cholestatic liver injury, which was manifested by the serum biochemical parameters and histopathology of the liver tissue. YCHT regulated the lipid levels as indicated by the reversed serum levels of TC, TG, and LDL-C. YCHT also regulated the disorder of BA and FFA metabolism by CPZ induction. CONCLUSIONS Results indicated that YCHT exerted a protective effect on CPZ-induced cholestasis liver injury. The variance of BA and FFA concentrations can be used to evaluate the cholestatic liver injury caused by CPZ and the hepatoprotective effect of YCHT.
Collapse
Affiliation(s)
- Qiaoling Yang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines and the State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 201210, Shanghai, China.
| | - Fan Yang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines and the State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 201210, Shanghai, China.
| | - Xiaowen Tang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines and the State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 201210, Shanghai, China.
| | - Lili Ding
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines and the State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 201210, Shanghai, China.
| | - Ying Xu
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines and the State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 201210, Shanghai, China.
| | - Yinhua Xiong
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines and the State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 201210, Shanghai, China.
| | - Zhengtao Wang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines and the State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 201210, Shanghai, China.
| | - Li Yang
- The Ministry of Education Key Laboratory for Standardization of Chinese Medicines and the State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 201210, Shanghai, China.
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Institute of Traditional Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201210, China.
| |
Collapse
|
150
|
Pisonero-Vaquero S, Martínez-Ferreras Á, García-Mediavilla MV, Martínez-Flórez S, Fernández A, Benet M, Olcoz JL, Jover R, González-Gallego J, Sánchez-Campos S. Quercetin ameliorates dysregulation of lipid metabolism genes via the PI3K/AKT pathway in a diet-induced mouse model of nonalcoholic fatty liver disease. Mol Nutr Food Res 2015; 59:879-93. [PMID: 25712622 DOI: 10.1002/mnfr.201400913] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/03/2015] [Accepted: 02/09/2015] [Indexed: 12/12/2022]
Abstract
SCOPE Flavonoids and related compounds seem to have favorable effects on nonalcoholic fatty liver disease (NAFLD) progression, although the exact mechanisms implicated are poorly understood. In this study, we aimed to investigate the effect of the flanovol quercetin on gene expression deregulation involved in the development of NAFLD, as well as the possible implication of phosphatidylinositol 3-kinase (PI3K)/AKT pathway modulation. METHODS AND RESULTS We used an in vivo model based on methionine- and choline-deficient (MCD) diet-fed mice and an in vitro model consisting of Huh7 cells incubated with MCD medium. MCD-fed mice showed classical pathophysiological characteristics of nonalcoholic steatohepatitis, associated with altered transcriptional regulation of fatty acid uptake- and trafficking-related gene expression, with increased lipoperoxidation. PI3K/AKT pathway was activated by MCD and triggered gene deregulation causing either activation or inhibition of all studied genes as demonstrated through cell incubation with the PI3K-inhibitor LY294002. Treatment with quercetin reduced AKT phosphorylation, and oxidative/nitrosative stress, inflammation and lipid metabolism-related genes displayed a tendency to normalize in both in vivo and in vitro models. CONCLUSION These results place quercetin as a potential therapeutic strategy for preventing NAFLD progression by attenuating gene expression deregulation, at least in part through PI3K/AKT pathway inactivation.
Collapse
|