1
|
Marino L, Kim A, Ni B, Celi FS. Thyroid hormone action and liver disease, a complex interplay. Hepatology 2025; 81:651-669. [PMID: 37535802 PMCID: PMC11737129 DOI: 10.1097/hep.0000000000000551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/05/2023] [Indexed: 08/05/2023]
Abstract
Thyroid hormone action is involved in virtually all physiological processes. It is well known that the liver and thyroid are intimately linked, with thyroid hormone playing important roles in de novo lipogenesis, beta-oxidation (fatty acid oxidation), cholesterol metabolism, and carbohydrate metabolism. Clinical and mechanistic research studies have shown that thyroid hormone can be involved in chronic liver diseases, including alcohol-associated or NAFLD and HCC. Thyroid hormone action and synthetic thyroid hormone analogs can exert beneficial actions in terms of lowering lipids, preventing chronic liver disease and as liver anticancer agents. More recently, preclinical and clinical studies have indicated that some analogs of thyroid hormone could also play a role in the treatment of liver disease. These synthetic molecules, thyromimetics, can modulate lipid metabolism, particularly in NAFLD/NASH. In this review, we first summarize the thyroid hormone signaling axis in the context of liver biology, then we describe the changes in thyroid hormone signaling in liver disease and how liver diseases affect the thyroid hormone homeostasis, and finally we discuss the use of thyroid hormone-analog for the treatment of liver disease.
Collapse
Affiliation(s)
- Luigi Marino
- Department of Medicine, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - Adam Kim
- Division of Gastroenterology and Hepatology, Department of Medicine, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - Bin Ni
- Alliance Pharma, Philadelphia, Pennsylvania, USA
| | - Francesco S. Celi
- Department of Medicine, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| |
Collapse
|
2
|
Cheng SY, Jiang L, Wang Y, Cai W. Emerging role of regulated cell death in intestinal failure-associated liver disease. Hepatobiliary Pancreat Dis Int 2024; 23:228-233. [PMID: 36621400 DOI: 10.1016/j.hbpd.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/08/2022] [Indexed: 01/10/2023]
Abstract
Intestinal failure-associated liver disease (IFALD) is a common complication of long-term parenteral nutrition that is associated with significant morbidity and mortality. It is mainly characterized by cholestasis in children and steatohepatitis in adults. Unfortunately, there is no effective approach to prevent or reverse the disease. Regulated cell death (RCD) represents a fundamental biological paradigm that determines the outcome of a variety of liver diseases. Nowadays cell death is reclassified into several types, based on the mechanisms and morphological phenotypes. Emerging evidence has linked different modes of RCD, such as apoptosis, necroptosis, ferroptosis, and pyroptosis to the pathogenesis of liver diseases. Recent studies have shown that different modes of RCD are present in animal models and patients with IFALD. Understanding the pathogenic roles of cell death may help uncover the underlying mechanisms and develop novel therapeutic strategies in IFALD. In this review, we discuss the current knowledge on how RCD may link to the pathogenesis of IFALD. We highlight examples of cell death-targeted interventions aiming to attenuate the disease, and provide perspectives for future basic and translational research in the field.
Collapse
Affiliation(s)
- Si-Yang Cheng
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Lu Jiang
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Ying Wang
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Wei Cai
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China.
| |
Collapse
|
3
|
Peleman C, Francque S, Berghe TV. Emerging role of ferroptosis in metabolic dysfunction-associated steatotic liver disease: revisiting hepatic lipid peroxidation. EBioMedicine 2024; 102:105088. [PMID: 38537604 PMCID: PMC11026979 DOI: 10.1016/j.ebiom.2024.105088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/22/2024] [Accepted: 03/12/2024] [Indexed: 04/14/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is characterised by cell death of parenchymal liver cells which interact with their microenvironment to drive disease activity and liver fibrosis. The identification of the major death type could pave the way towards pharmacotherapy for MASH. To date, increasing evidence suggest a type of regulated cell death, named ferroptosis, which occurs through iron-catalysed peroxidation of polyunsaturated fatty acids (PUFA) in membrane phospholipids. Lipid peroxidation enjoys renewed interest in the light of ferroptosis, as druggable target in MASH. This review recapitulates the molecular mechanisms of ferroptosis in liver physiology, evidence for ferroptosis in human MASH and critically appraises the results of ferroptosis targeting in preclinical MASH models. Rewiring of redox, iron and PUFA metabolism in MASH creates a proferroptotic environment involved in MASH-related hepatocellular carcinoma (HCC) development. Ferroptosis induction might be a promising novel approach to eradicate HCC, while its inhibition might ameliorate MASH disease progression.
Collapse
Affiliation(s)
- Cédric Peleman
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium; Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Sven Francque
- Laboratory of Experimental Medicine and Paediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium; Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium.
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
4
|
Gupta AC, Bhat A, Maras JS. Early hepatic proteomic signatures reveal metabolic changes in high-fat-induced obesity in rats. Br J Nutr 2024; 131:773-785. [PMID: 37886840 DOI: 10.1017/s0007114523002453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The prevalence of diet-related obesity is increasing dramatically worldwide, making it important to understand the associated metabolic alterations in the liver. It is well known that obesity is a multifactorial condition that is the result of complex integration between many gene expressions and dietary factors. Obesity alone or in conjunction with other chronic diseases such as diabetes and insulin resistance causes many health problems and is considered a major risk factor for developing non-alcoholic steatohepatitis (NASH) and cirrhosis. In this study, we aimed to understand the molecular mechanisms underlying early hepatic changes in the pathophysiology of high-fat diet (HFD)-induced abdominal obesity in rats. Hepatic protein profiles of normal diet and HFD-induced obesity for 24 weeks were analysed using two-dimensional differential gel electrophoresis (DIGE) and protein identification by MS. Fifty-two proteins were identified by matrix-assisted laser desorption ionization-time of flight (MALDI-TOF), and computer-assisted DIGE image software analysis showed that eighteen major proteins were significantly differentially expressed between comparable groups, with 2·0–4·0-fold change/more (P < 0·01). These proteins are regulated in response to a HFD, and differentially expressed proteins are involved in key metabolic pathways such as lipid metabolism, energy metabolism, detoxification, urea cycle and hepatic Ca homoeostasis. In addition, Western blot and immunohistochemistry of liver-specific arginase-1 (Arg-1) showed significant increased expression in the liver of high-fat-fed rats (P < 0·01). Further, Arg-1 expression was correlated with NASH patients with obesity-related fibrosis (F0–F4). It is concluded that high-fat content may affect changes in liver pathways and may be a therapeutic target for obesity-related liver disease. Arg-1 expressions may be a potential pathological marker for assessing the progression of the disease.
Collapse
Affiliation(s)
- Abhishak C Gupta
- Department of Education and Research, Artemis Hospitals, Gurugram, Haryana, India
- Department of Molecular and Cellular Medicine (MCM), Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
| | - Adil Bhat
- Department of Molecular and Cellular Medicine (MCM), Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
| | - Jaswinder S Maras
- Department of Molecular and Cellular Medicine (MCM), Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
| |
Collapse
|
5
|
Gîlcă-Blanariu GE, Budur DS, Mitrică DE, Gologan E, Timofte O, Bălan GG, Olteanu VA, Ștefănescu G. Advances in Noninvasive Biomarkers for Nonalcoholic Fatty Liver Disease. Metabolites 2023; 13:1115. [PMID: 37999211 PMCID: PMC10672868 DOI: 10.3390/metabo13111115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/15/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) currently represents one of the most common liver diseases worldwide. Early diagnosis and disease staging is crucial, since it is mainly asymptomatic, but can progress to nonalcoholic steatohepatitis (NASH) or cirrhosis or even lead to the development of hepatocellular carcinoma. Over time, efforts have been put into developing noninvasive diagnostic and staging methods in order to replace the use of a liver biopsy. The noninvasive methods used include imaging techniques that measure liver stiffness and biological markers, with a focus on serum biomarkers. Due to the impressive complexity of the NAFLD's pathophysiology, biomarkers are able to assay different processes involved, such as apoptosis, fibrogenesis, and inflammation, or even address the genetic background and "omics" technologies. This article reviews not only the currently validated noninvasive methods to investigate NAFLD but also the promising results regarding recently discovered biomarkers, including biomarker panels and the combination of the currently validated evaluation methods and serum markers.
Collapse
Affiliation(s)
- Georgiana-Emmanuela Gîlcă-Blanariu
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Daniela Simona Budur
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
| | - Dana Elena Mitrică
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Elena Gologan
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
| | - Oana Timofte
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Gheorghe Gh Bălan
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Vasile Andrei Olteanu
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Gabriela Ștefănescu
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| |
Collapse
|
6
|
Powell NR, Liang T, Ipe J, Cao S, Skaar TC, Desta Z, Qian HR, Ebert PJ, Chen Y, Thomas MK, Chalasani N. Clinically important alterations in pharmacogene expression in histologically severe nonalcoholic fatty liver disease. Nat Commun 2023; 14:1474. [PMID: 36927865 PMCID: PMC10020163 DOI: 10.1038/s41467-023-37209-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Polypharmacy is common in patients with nonalcoholic fatty liver disease (NAFLD) and previous reports suggest that NAFLD is associated with altered drug disposition. This study aims to determine if patients with NAFLD are at risk for altered drug response by characterizing changes in hepatic mRNA expression of genes mediating drug disposition (pharmacogenes) across the histological NAFLD severity spectrum. We utilize RNA-seq for 93 liver biopsies with histologically staged NAFLD Activity Score (NAS), fibrosis stage, and steatohepatitis (NASH). We identify 37 significant pharmacogene-NAFLD severity associations including CYP2C19 downregulation. We chose to validate CYP2C19 due to its actionability in drug prescribing. Meta-analysis of 16 independent studies demonstrate that CYP2C19 is significantly downregulated to 46% in NASH, to 58% in high NAS, and to 43% in severe fibrosis. Our data demonstrate the downregulation of CYP2C19 in NAFLD which supports developing personalized medicine approaches for drugs sensitive to metabolism by the CYP2C19 enzyme.
Collapse
Affiliation(s)
- Nicholas R Powell
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis, IN, USA
| | - Tiebing Liang
- Indiana University School of Medicine, Department of Medicine, Division of Gastroenterology Hepatology, Indianapolis, IN, USA
| | - Joseph Ipe
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis, IN, USA
| | - Sha Cao
- Indiana University School of Medicine, Department of Medicine, Division of Gastroenterology Hepatology, Indianapolis, IN, USA
| | - Todd C Skaar
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis, IN, USA
| | - Zeruesenay Desta
- Indiana University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Indianapolis, IN, USA
| | | | | | - Yu Chen
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Naga Chalasani
- Indiana University School of Medicine, Department of Medicine, Division of Gastroenterology Hepatology, Indianapolis, IN, USA.
| |
Collapse
|
7
|
Fahlbusch P, Nikolic A, Hartwig S, Jacob S, Kettel U, Köllmer C, Al-Hasani H, Lehr S, Müller-Wieland D, Knebel B, Kotzka J. Adaptation of Oxidative Phosphorylation Machinery Compensates for Hepatic Lipotoxicity in Early Stages of MAFLD. Int J Mol Sci 2022; 23:ijms23126873. [PMID: 35743314 PMCID: PMC9224893 DOI: 10.3390/ijms23126873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/18/2022] [Accepted: 06/18/2022] [Indexed: 12/10/2022] Open
Abstract
Alterations in mitochondrial function are an important control variable in the progression of metabolic dysfunction-associated fatty liver disease (MAFLD), while also noted by increased de novo lipogenesis (DNL) and hepatic insulin resistance. We hypothesized that the organization and function of a mitochondrial electron transport chain (ETC) in this pathologic condition is a consequence of shifted substrate availability. We addressed this question using a transgenic mouse model with increased hepatic insulin resistance and DNL due to constitutively active human SREBP-1c. The abundance of ETC complex subunits and components of key metabolic pathways are regulated in the liver of these animals. Further omics approaches combined with functional assays in isolated liver mitochondria and primary hepatocytes revealed that the SREBP-1c-forced fatty liver induced a substrate limitation for oxidative phosphorylation, inducing enhanced complex II activity. The observed increased expression of mitochondrial genes may have indicated a counteraction. In conclusion, a shift of available substrates directed toward activated DNL results in increased electron flows, mainly through complex II, to compensate for the increased energy demand of the cell. The reorganization of key compounds in energy metabolism observed in the SREBP-1c animal model might explain the initial increase in mitochondrial function observed in the early stages of human MAFLD.
Collapse
Affiliation(s)
- Pia Fahlbusch
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| | - Aleksandra Nikolic
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| | - Sonja Hartwig
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| | - Sylvia Jacob
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
| | - Ulrike Kettel
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
| | - Cornelia Köllmer
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
| | - Hadi Al-Hasani
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
- Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Stefan Lehr
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| | - Dirk Müller-Wieland
- Clinical Research Centre, Department of Internal Medicine I, University Hospital Aachen, 52074 Aachen, Germany;
| | - Birgit Knebel
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
- Correspondence: ; Tel.: +49-211-3382-536
| | - Jörg Kotzka
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, 40225 Duesseldorf, Germany; (P.F.); (A.N.); (S.H.); (S.J.); (U.K.); (C.K.); (H.A.-H.); (S.L.); (J.K.)
- German Center for Diabetes Research (DZD), Partner Duesseldorf, 40225 Duesseldorf, Germany
| |
Collapse
|
8
|
Nakao Y, Fukushima M, Mauer AS, Liao CY, Ferris A, Dasgupta D, Heppelmann CJ, Vanderboom PM, Saraswat M, Pandey A, Nair KS, Allen AM, Nakao K, Malhi H. A Comparative Proteomic Analysis of Extracellular Vesicles Associated With Lipotoxicity. Front Cell Dev Biol 2021; 9:735001. [PMID: 34805145 PMCID: PMC8600144 DOI: 10.3389/fcell.2021.735001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are emerging mediators of intercellular communication in nonalcoholic steatohepatitis (NASH). Palmitate, a lipotoxic saturated fatty acid, activates hepatocellular endoplasmic reticulum stress, which has been demonstrated to be important in NASH pathogenesis, including in the release of EVs. We have previously demonstrated that the release of palmitate-stimulated EVs is dependent on the de novo synthesis of ceramide, which is trafficked by the ceramide transport protein, STARD11. The trafficking of ceramide is a critical step in the release of lipotoxic EVs, as cells deficient in STARD11 do not release palmitate-stimulated EVs. Here, we examined the hypothesis that protein cargoes are trafficked to lipotoxic EVs in a ceramide-dependent manner. We performed quantitative proteomic analysis of palmitate-stimulated EVs in control and STARD11 knockout hepatocyte cell lines. Proteomics was performed on EVs isolated by size exclusion chromatography, ultracentrifugation, and density gradient separation, and EV proteins were measured by mass spectrometry. We also performed human EV proteomics from a control and a NASH plasma sample, for comparative analyses with hepatocyte-derived lipotoxic EVs. Size exclusion chromatography yielded most unique EV proteins. Ceramide-dependent lipotoxic EVs contain damage-associated molecular patterns and adhesion molecules. Haptoglobin, vascular non-inflammatory molecule-1, and insulin-like growth factor-binding protein complex acid labile subunit were commonly detected in NASH and hepatocyte-derived ceramide-dependent EVs. Lipotoxic EV proteomics provides novel candidate proteins to investigate in NASH pathogenesis and as diagnostic biomarkers for hepatocyte-derived EVs in NASH patients.
Collapse
Affiliation(s)
- Yasuhiko Nakao
- Division of Gastroenterology and Hepatology, Rochester, MN, United States.,Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Masanori Fukushima
- Division of Gastroenterology and Hepatology, Rochester, MN, United States.,Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Amy S Mauer
- Division of Gastroenterology and Hepatology, Rochester, MN, United States
| | - Chieh-Yu Liao
- Division of Gastroenterology and Hepatology, Rochester, MN, United States
| | - Anya Ferris
- Division of Gastroenterology and Hepatology, Rochester, MN, United States.,California Polytechnic State University, San Luis Obispo, CA, United States
| | - Debanjali Dasgupta
- Division of Gastroenterology and Hepatology, Rochester, MN, United States.,Department of Physiology and Biomedical Engineering, Manipal, India
| | | | - Patrick M Vanderboom
- Mayo Clinic Medical Genome Facility-Proteomics Core, Manipal, India.,Mayo Endocrine Research Unit, Manipal, India
| | - Mayank Saraswat
- Department of Laboratory Medicine and Pathology, Rochester, MN, United States.,Institute of Bioinformatics, Bangalore, India.,Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Rochester, MN, United States.,Institute of Bioinformatics, Bangalore, India.,Manipal Academy of Higher Education (MAHE), Manipal, India.,Center for Individualized Medicine, Rochester, MN, United States
| | | | - Alina M Allen
- Division of Gastroenterology and Hepatology, Rochester, MN, United States
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Rochester, MN, United States
| |
Collapse
|
9
|
Subudhi S, Drescher HK, Dichtel LE, Bartsch LM, Chung RT, Hutter MM, Gee DW, Meireles OR, Witkowski ER, Gelrud L, Masia R, Osganian SA, Gustafson JL, Rwema S, Bredella MA, Bhatia SN, Warren A, Miller KK, Lauer GM, Corey KE. Distinct Hepatic Gene-Expression Patterns of NAFLD in Patients With Obesity. Hepatol Commun 2021; 6:77-89. [PMID: 34558849 PMCID: PMC8710788 DOI: 10.1002/hep4.1789] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/13/2021] [Indexed: 02/06/2023] Open
Abstract
Approaches to manage nonalcoholic fatty liver disease (NAFLD) are limited by an incomplete understanding of disease pathogenesis. The aim of this study was to identify hepatic gene‐expression patterns associated with different patterns of liver injury in a high‐risk cohort of adults with obesity. Using the NanoString Technologies (Seattle, WA) nCounter assay, we quantified expression of 795 genes, hypothesized to be involved in hepatic fibrosis, inflammation, and steatosis, in liver tissue from 318 adults with obesity. Liver specimens were categorized into four distinct NAFLD phenotypes: normal liver histology (NLH), steatosis only (steatosis), nonalcoholic steatohepatitis without fibrosis (NASH F0), and NASH with fibrosis stage 1‐4 (NASH F1‐F4). One hundred twenty‐five genes were significantly increasing or decreasing as NAFLD pathology progressed. Compared with NLH, NASH F0 was characterized by increased inflammatory gene expression, such as gamma‐interferon‐inducible lysosomal thiol reductase (IFI30) and chemokine (C‐X‐C motif) ligand 9 (CXCL9), while complement and coagulation related genes, such as C9 and complement component 4 binding protein beta (C4BPB), were reduced. In the presence of NASH F1‐F4, extracellular matrix degrading proteinases and profibrotic/scar deposition genes, such as collagens and transforming growth factor beta 1 (TGFB1), were simultaneously increased, suggesting a dynamic state of tissue remodeling. Conclusion: In adults with obesity, distinct states of NAFLD are associated with intrahepatic perturbations in genes related to inflammation, complement and coagulation pathways, and tissue remodeling. These data provide insights into the dynamic pathogenesis of NAFLD in high‐risk individuals.
Collapse
Affiliation(s)
- Sonu Subudhi
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hannah K Drescher
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laura E Dichtel
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lea M Bartsch
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raymond T Chung
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthew M Hutter
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Denise W Gee
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ozanan R Meireles
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Elan R Witkowski
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Louis Gelrud
- Department of Medicine, St. Mary's Hospital Bon Secours, Richmond, VA, USA
| | - Ricard Masia
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephanie A Osganian
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jenna L Gustafson
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Steve Rwema
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Miriam A Bredella
- Division of Musculoskeletal Radiology and Interventions, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sangeeta N Bhatia
- Ludwig Center for Molecular Oncology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew Warren
- Ludwig Center for Molecular Oncology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Karen K Miller
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Georg M Lauer
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kathleen E Corey
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Wernberg CW, Ravnskjaer K, Lauridsen MM, Thiele M. The Role of Diagnostic Biomarkers, Omics Strategies, and Single-Cell Sequencing for Nonalcoholic Fatty Liver Disease in Severely Obese Patients. J Clin Med 2021; 10:930. [PMID: 33804302 PMCID: PMC7957539 DOI: 10.3390/jcm10050930] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/29/2022] Open
Abstract
Liver disease due to metabolic dysfunction constitute a worldwide growing health issue. Severe obesity is a particularly strong risk factor for non-alcoholic fatty liver disease, which affects up to 93% of these patients. Current diagnostic markers focus on the detection of advanced fibrosis as the major predictor of liver-related morbidity and mortality. The most accurate diagnostic tools use elastography to measure liver stiffness, with diagnostic accuracies similar in normal-weight and severely obese patients. The effectiveness of elastography tools are however hampered by limitations to equipment and measurement quality in patients with very large abdominal circumference and subcutaneous fat. Blood-based biomarkers are therefore attractive, but those available to date have only moderate diagnostic accuracy. Ongoing technological advances in omics technologies such as genomics, transcriptomics, and proteomics hold great promise for discovery of biomarkers and increased pathophysiological understanding of non-alcoholic liver disease and steatohepatitis. Very recent developments have allowed for single-cell sequencing and cell-type resolution of gene expression and function. In the near future, we will therefore likely see a multitude of breakthrough biomarkers, developed from a deepened understanding of the biological function of individual cell types in the healthy and injured liver.
Collapse
Affiliation(s)
- Charlotte W. Wernberg
- Department of Gastroenterology and Hepatology, Hospital Southwest of Jutland, 6700 Esbjerg, Denmark; (C.W.W.); (M.M.L.)
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense, Denmark;
| | - Kim Ravnskjaer
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense, Denmark;
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Mette M. Lauridsen
- Department of Gastroenterology and Hepatology, Hospital Southwest of Jutland, 6700 Esbjerg, Denmark; (C.W.W.); (M.M.L.)
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense, Denmark;
| | - Maja Thiele
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense, Denmark;
- Center for Liver Research, Department of Hepatology and Gastroenterology, Odense University Hospital, 5000 Odense, Denmark
- Institute for Clinical Research, University of Southern Denmark, 5230 Odense, Denmark
| |
Collapse
|
11
|
Morral N, Liu S, Conteh AM, Chu X, Wang Y, Dong XC, Liu Y, Linnemann AK, Wan J. Aberrant gene expression induced by a high fat diet is linked to H3K9 acetylation in the promoter-proximal region. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2021; 1864:194691. [PMID: 33556624 DOI: 10.1016/j.bbagrm.2021.194691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/22/2021] [Accepted: 01/30/2021] [Indexed: 12/13/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, with an estimated global prevalence of 1 in 4 individuals. Aberrant transcriptional control of gene expression is central to the pathophysiology of metabolic diseases. However, the molecular mechanisms leading to gene dysregulation are not well understood. Histone modifications play important roles in the control of transcription. Acetylation of histone 3 at lysine 9 (H3K9ac) is associated with transcriptional activity and is implicated in transcript elongation by controlling RNA polymerase II (RNAPII) pause-release. Hence, changes in this histone modification may shed information on novel pathways linking transcription control and metabolic dysfunction. Here, we carried out genome-wide analysis of H3K9ac in the liver of mice fed a control or a high-fat diet (an animal model of NAFLD), and asked whether this histone mark associates with changes in gene expression. We found that over 70% of RNAPII peaks in promoter-proximal regions overlapped with H3K9ac, consistent with a role of H3K9ac in the regulation of transcription. When comparing high-fat with control diet, approximately 17% of the differentially expressed genes were associated with changes in H3K9ac in their promoters, showing a strong correlation between changes in H3K9ac signal and gene expression. Overall, our data indicate that in response to a high-fat diet, dysregulated gene expression of a subset of genes may be attributable to changes in transcription elongation driven by H3K9ac. Our results point at an added mechanism of gene regulation that may be important in the development of metabolic diseases.
Collapse
Affiliation(s)
- Núria Morral
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States of America; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America.
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Abass M Conteh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Xiaona Chu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Yue Wang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - X Charlie Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States of America; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Amelia K Linnemann
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States of America; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| |
Collapse
|
12
|
Perakakis N, Stefanakis K, Mantzoros CS. The role of omics in the pathophysiology, diagnosis and treatment of non-alcoholic fatty liver disease. Metabolism 2020; 111S:154320. [PMID: 32712221 PMCID: PMC7377759 DOI: 10.1016/j.metabol.2020.154320] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifaceted metabolic disorder, whose spectrum covers clinical, histological and pathophysiological developments ranging from simple steatosis to non-alcoholic steatohepatitis (NASH) and liver fibrosis, potentially evolving into cirrhosis, hepatocellular carcinoma and liver failure. Liver biopsy remains the gold standard for diagnosing NAFLD, while there are no specific treatments. An ever-increasing number of high-throughput Omics investigations on the molecular pathobiology of NAFLD at the cellular, tissue and system levels produce comprehensive biochemical patient snapshots. In the clinical setting, these applications are considerably enhancing our efforts towards obtaining a holistic insight on NAFLD pathophysiology. Omics are also generating non-invasive diagnostic modalities for the distinct stages of NAFLD, that remain though to be validated in multiple, large, heterogenous and independent cohorts, both cross-sectionally as well as prospectively. Finally, they aid in developing novel therapies. By tracing the flow of information from genomics to epigenomics, transcriptomics, proteomics, metabolomics, lipidomics and glycomics, the chief contributions of these techniques in understanding, diagnosing and treating NAFLD are summarized herein.
Collapse
Affiliation(s)
- Nikolaos Perakakis
- Department of Internal Medicine, Boston VA Healthcare system and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA..
| | - Konstantinos Stefanakis
- Department of Internal Medicine, Boston VA Healthcare system and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christos S Mantzoros
- Department of Internal Medicine, Boston VA Healthcare system and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
13
|
Lonardo A, Leoni S, Alswat KA, Fouad Y. History of Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2020; 21:E5888. [PMID: 32824337 PMCID: PMC7460697 DOI: 10.3390/ijms21165888] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/15/2022] Open
Abstract
Based on the assumption that characterizing the history of a disease will help in improving practice while offering a clue to research, this article aims at reviewing the history of nonalcoholic fatty liver disease (NAFLD) in adults and children. To this end, we address the history of NAFLD histopathology, which begins in 1980 with Ludwig's seminal studies, although previous studies date back to the 19th century. Moreover, the principal milestones in the definition of genetic NAFLD are summarized. Next, a specific account is given of the evolution, over time, of our understanding of the association of NAFLD with metabolic syndrome, spanning from the outdated concept of "NAFLD as a manifestation of the Metabolic Syndrome", to the more appropriate consideration that NAFLD has, with metabolic syndrome, a mutual and bi-directional relationship. In addition, we also report on the evolution from first intuitions to more recent studies, supporting NAFLD as an independent risk factor for cardiovascular disease. This association probably has deep roots, going back to ancient Middle Eastern cultures, wherein the liver had a significance similar to that which the heart holds in contemporary society. Conversely, the notions that NAFLD is a forerunner of hepatocellular carcinoma and extra-hepatic cancers is definitely more modern. Interestingly, guidelines issued by hepatological societies have lagged behind the identification of NAFLD by decades. A comparative analysis of these documents defines both shared attitudes (e.g., ultrasonography and lifestyle changes as the first approaches) and diverging key points (e.g., the threshold of alcohol consumption, screening methods, optimal non-invasive assessment of liver fibrosis and drug treatment options). Finally, the principal historical steps in the general, cellular and molecular pathogenesis of NAFLD are reviewed. We conclude that an in-depth understanding of the history of the disease permits us to better comprehend the disease itself, as well as to anticipate the lines of development of future NAFLD research.
Collapse
Affiliation(s)
- Amedeo Lonardo
- Ospedale Civile di Baggiovara, UOC Medicina Metabolica, Dipartimento di Medicina Interna Generale, d’Urgenza e post Acuzie, Azienda Ospedaliero-Universitaria di Modena, Via Giardini 1135, 41125 Modena, Italy
| | - Simona Leoni
- Internal Medicine Unit, Department of Digestive Diseases, S.Orsola-Malpighi Hospital, Via Massarenti 9, 40136 Bologna, Italy;
| | - Khalid A. Alswat
- Liver Research Center, Department of Medicine, College of Medicine, King Saud University, Riyadh 11322, Saudi Arabia;
| | - Yasser Fouad
- Department of Gastroenterology, Hepatology and Endemic Medicine, Faculty of Medicine, Minia University, Minya 19111, Egypt;
| |
Collapse
|
14
|
Moroldo M, Munyaka PM, Lecardonnel J, Lemonnier G, Venturi E, Chevaleyre C, Oswald IP, Estellé J, Rogel-Gaillard C. Integrative analysis of blood and gut microbiota data suggests a non-alcoholic fatty liver disease (NAFLD)-related disorder in French SLA dd minipigs. Sci Rep 2020; 10:234. [PMID: 31937803 PMCID: PMC6959234 DOI: 10.1038/s41598-019-57127-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 12/18/2019] [Indexed: 11/29/2022] Open
Abstract
Minipigs are a group of small-sized swine lines, which show a broad range of phenotype variation and which often tend to be obese. The SLAdd (DD) minipig line was created by the NIH and selected as homozygous at the SLA locus. It was brought to France more than 30 years ago and maintained inbred ever since. In this report, we characterized the physiological status of a herd of French DD pigs by measuring intermediate phenotypes from blood and faeces and by using Large White (LW) pigs as controls. Three datasets were produced, i.e. complete blood counts (CBCs), microarray-based blood transcriptome, and faecal microbiota obtained by 16S rRNA sequencing. CBCs and expression profiles suggested a non-alcoholic fatty liver disease (NAFLD)-related pathology associated to comorbid cardiac diseases. The characterization of 16S sequencing data was less straightforward, suggesting only a potential weak link to obesity. The integration of the datasets identified several fine-scale associations between CBCs, gene expression, and faecal microbiota composition. NAFLD is a common cause of chronic liver disease in Western countries and is linked to obesity, type 2 diabetes mellitus and cardiac pathologies. Here we show that the French DD herd is potentially affected by this syndrome.
Collapse
Affiliation(s)
- Marco Moroldo
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.
| | - Peris Mumbi Munyaka
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Canada
| | - Jérôme Lecardonnel
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Gaëtan Lemonnier
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | | | | | - Isabelle P Oswald
- Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toxalim, 31027, Toulouse, France
| | - Jordi Estellé
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | | |
Collapse
|
15
|
Suppli MP, Rigbolt KTG, Veidal SS, Heebøll S, Eriksen PL, Demant M, Bagger JI, Nielsen JC, Oró D, Thrane SW, Lund A, Strandberg C, Kønig MJ, Vilsbøll T, Vrang N, Thomsen KL, Grønbæk H, Jelsing J, Hansen HH, Knop FK. Hepatic transcriptome signatures in patients with varying degrees of nonalcoholic fatty liver disease compared with healthy normal-weight individuals. Am J Physiol Gastrointest Liver Physiol 2019; 316:G462-G472. [PMID: 30653341 DOI: 10.1152/ajpgi.00358.2018] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents a spectrum of conditions ranging from simple steatosis (NAFL), over nonalcoholic steatohepatitis (NASH) with or without fibrosis, to cirrhosis with end-stage disease. The hepatic molecular events underlying the development of NAFLD and transition to NASH are poorly understood. The present study aimed to determine hepatic transcriptome dynamics in patients with NAFL or NASH compared with healthy normal-weight and obese individuals. RNA sequencing and quantitative histomorphometry of liver fat, inflammation and fibrosis were performed on liver biopsies obtained from healthy normal-weight ( n = 14) and obese ( n = 12) individuals, NAFL ( n = 15) and NASH ( n = 16) patients. Normal-weight and obese subjects showed normal liver histology and comparable gene expression profiles. Liver transcriptome signatures were largely overlapping in NAFL and NASH patients, however, clearly separated from healthy normal-weight and obese controls. Most marked pathway perturbations identified in both NAFL and NASH were associated with markers of lipid metabolism, immunomodulation, extracellular matrix remodeling, and cell cycle control. Interestingly, NASH patients with positive Sonic hedgehog hepatocyte staining showed distinct transcriptome and histomorphometric changes compared with NAFL. In conclusion, application of immunohistochemical markers of hepatocyte injury may serve as a more objective tool for distinguishing NASH from NAFL, facilitating improved resolution of hepatic molecular changes associated with progression of NAFLD. NEW & NOTEWORTHY Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease in Western countries. NAFLD is associated with the metabolic syndrome and can progress to the more serious form, nonalcoholic steatohepatitis (NASH), and ultimately lead to irreversible liver damage. Using gold standard molecular and histological techniques, this study demonstrates that the currently used diagnostic tools are problematic for differentiating mild NAFLD from NASH and emphasizes the marked need for developing improved histological markers of NAFLD progression.
Collapse
Affiliation(s)
- Malte P Suppli
- Department of Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | | | - Sara Heebøll
- Department of Hepatology and Gastroenterology, Aarhus University Hospital , Aarhus , Denmark
| | - Peter Lykke Eriksen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital , Aarhus , Denmark
| | - Mia Demant
- Department of Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Jonatan I Bagger
- Department of Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | | | | | - Asger Lund
- Department of Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Charlotte Strandberg
- Department of Radiology, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Merete J Kønig
- Department of Radiology, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Tina Vilsbøll
- Department of Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | | | - Karen L Thomsen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital , Aarhus , Denmark
| | - Henning Grønbæk
- Department of Hepatology and Gastroenterology, Aarhus University Hospital , Aarhus , Denmark
| | | | | | - Filip K Knop
- Department of Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
16
|
Singh S, McDonough CW, Gong Y, Alghamdi WA, Arwood MJ, Bargal SA, Dumeny L, Li WY, Mehanna M, Stockard B, Yang G, de Oliveira FA, Fredette NC, Shahin MH, Bailey KR, Beitelshees AL, Boerwinkle E, Chapman AB, Gums JG, Turner ST, Cooper-DeHoff RM, Johnson JA. Genome Wide Association Study Identifies the HMGCS2 Locus to be Associated With Chlorthalidone Induced Glucose Increase in Hypertensive Patients. J Am Heart Assoc 2018. [PMID: 29523524 PMCID: PMC5907544 DOI: 10.1161/jaha.117.007339] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Background Thiazide and thiazide‐like diuretics are first‐line medications for treating uncomplicated hypertension. However, their use has been associated with adverse metabolic events, including hyperglycemia and incident diabetes mellitus, with incompletely understood mechanisms. Our goal was to identify genomic variants associated with thiazide‐like diuretic/chlorthalidone‐induced glucose change. Methods and Results Genome‐wide analysis of glucose change after treatment with chlorthalidone was performed by race among the white (n=175) and black (n=135) participants from the PEAR‐2 (Pharmacogenomic Evaluation of Antihypertensive Responses‐2). Single‐nucleotide polymorphisms with P<5×10−8 were further prioritized using in silico analysis based on their expression quantitative trait loci function. Among blacks, an intronic single‐nucleotide polymorphism (rs9943291) in the HMGCS2 was associated with increase in glucose levels following chlorthalidone treatment (ß=12.5; P=4.17×10−8). G‐allele carriers of HMGCS2 had higher glucose levels (glucose change=+16.29 mg/dL) post chlorthalidone treatment compared with noncarriers of G allele (glucose change=+2.80 mg/dL). This association was successfully replicated in an independent replication cohort of hydrochlorothiazide‐treated participants from the PEAR study (ß=5.54; P=0.023). A meta‐analysis of the 2 studies was performed by race in Meta‐Analysis Helper, where this single‐nucleotide polymorphism, rs9943291, was genome‐wide significant with a meta‐analysis P value of 3.71×10−8. HMGCS2, a part of the HMG‐CoA synthase family, is important for ketogenesis and cholesterol synthesis pathways that are essential in glucose homeostasis. Conclusions These results suggest that HMGCS2 is a promising candidate gene involved in chlorthalidone and Hydrochlorothiazide (HCTZ)‐induced glucose change. This may provide insights into the mechanisms involved in thiazide‐induced hyperglycemia that may ultimately facilitate personalized approaches to antihypertensive selection for hypertension treatment. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifiers: NCT00246519 and NCT01203852.
Collapse
Affiliation(s)
- Sonal Singh
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Caitrin W McDonough
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Wael A Alghamdi
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Meghan J Arwood
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Salma A Bargal
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Leanne Dumeny
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Wen-Yi Li
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Mai Mehanna
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Bradley Stockard
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Guang Yang
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Felipe A de Oliveira
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Natalie C Fredette
- Department of Pathology, Center of Regenerative Medicine, University of Florida, Gainesville, FL
| | - Mohamed H Shahin
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Kent R Bailey
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | - Eric Boerwinkle
- Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | | | - John G Gums
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Rhonda M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL .,Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL.,Division of Cardiovascular Medicine, Department of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
17
|
Hotta K, Kikuchi M, Kitamoto T, Kitamoto A, Ogawa Y, Honda Y, Kessoku T, Kobayashi K, Yoneda M, Imajo K, Tomeno W, Nakaya A, Suzuki Y, Saito S, Nakajima A. Identification of core gene networks and hub genes associated with progression of non-alcoholic fatty liver disease by RNA sequencing. Hepatol Res 2017; 47:1445-1458. [PMID: 28219123 DOI: 10.1111/hepr.12877] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/07/2017] [Accepted: 02/17/2017] [Indexed: 12/11/2022]
Abstract
AIM Non-alcoholic fatty liver disease (NAFLD) progresses because of the interaction between numerous genes. Thus, we carried out a weighted gene coexpression network analysis to identify core gene networks and key genes associated with NAFLD progression. METHODS We enrolled 39 patients with mild NAFLD (fibrosis stages 0-2) and 21 with advanced NAFLD (fibrosis stages 3-4). Total RNA was extracted from frozen liver biopsies, and sequenced to capture a large dynamic range of expression levels. RESULTS A total of 1777 genes differentially expressed between mild and advanced NAFLD (q-value <0.05) clustered into four modules. One module was enriched for genes that encode cell surface or extracellular matrix proteins, and are involved in cell adhesion, proliferation, and signaling. This module formed a scale-free network containing four hub genes (PAPLN, LBH, DPYSL3, and JAG1) overexpressed in advanced NAFLD. PAPLN is a component of the extracellular matrix, LBH and DPYSL3 are reported to be tumor suppressors, and JAG1 is tumorigenic. Another module formed a random network, and was enriched for genes that accumulate in the mitochondria. These genes were downregulated in advanced NAFLD, reflecting impaired mitochondrial function. However, the other two modules did not form unambiguous networks. KEGG analysis indicated that 71 differentially expressed genes were involved in "pathways in cancer". Strikingly, expression of half of all differentially expressed genes was inversely correlated with methylation of CpG sites (q-value <0.05). Among clinical parameters, serum type IV collagen 7 s was most strongly associated with the epigenetic status in NAFLD. CONCLUSIONS Newly identified core gene networks suggest that the NAFLD liver undergoes mitochondrial dysfunction and fibrosis, and acquires tumorigenic potential epigenetically. Our data provide novel insights into the pathology and etiology of NAFLD progression, and identify potential targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Kikuko Hotta
- Department of Medical Innovation, Osaka University Hospital, Suita, Japan
| | - Masataka Kikuchi
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takuya Kitamoto
- Pharmacogenomics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Laboratory of Radiation Safety, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Aya Kitamoto
- Pharmacogenomics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Advanced Research Facilities and Services, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuji Ogawa
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yasushi Honda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takaomi Kessoku
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kaori Kobayashi
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Suita, Japan.,Medical Solutions Division, NEC Corporation, Tokyo, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kento Imajo
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Wataru Tomeno
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akihiro Nakaya
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, Japan
| | - Satoru Saito
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
18
|
Atanasovska B, Rensen SS, van der Sijde MR, Marsman G, Kumar V, Jonkers I, Withoff S, Shiri-Sverdlov R, Greve JWM, Faber KN, Moshage H, Wijmenga C, van de Sluis B, Hofker MH, Fu J. A liver-specific long noncoding RNA with a role in cell viability is elevated in human nonalcoholic steatohepatitis. Hepatology 2017; 66:794-808. [PMID: 28073183 DOI: 10.1002/hep.29034] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 12/12/2016] [Accepted: 01/06/2017] [Indexed: 12/28/2022]
Abstract
UNLABELLED Hepatocyte apoptosis in nonalcoholic steatohepatitis (NASH) can lead to fibrosis and cirrhosis, which permanently damage the liver. Understanding the regulation of hepatocyte apoptosis is therefore important to identify therapeutic targets that may prevent the progression of NASH to fibrosis. Recently, increasing evidence has shown that long noncoding (lnc) RNAs are involved in various biological processes and that their dysregulation underlies a number of complex human diseases. By performing gene expression profiling of 4,383 lncRNAs in 82 liver samples from individuals with NASH (n = 48), simple steatosis but no NASH (n = 11), and healthy controls (n = 23), we discovered a liver-specific lncRNA (RP11-484N16.1) on chromosome 18 that showed significantly elevated expression in the liver tissue of NASH patients. This lncRNA, which we named lnc18q22.2 based on its chromosomal location, correlated with NASH grade (r = 0.51, P = 8.11 × 10-7 ), lobular inflammation (r = 0.49, P = 2.35 × 10-6 ), and nonalcoholic fatty liver disease activity score (r = 0.48, P = 4.69 × 10-6 ). The association of lnc18q22.2 to liver steatosis and steatohepatitis was replicated in 44 independent liver biopsies (r = 0.47, P = 0.0013). We provided a genetic structure of lnc18q22.2 showing an extended exon 2 in liver. Knockdown of lnc18q22.2 in four different hepatocyte cell lines resulted in severe phenotypes ranging from reduced cell growth to lethality. This observation was consistent with pathway analyses of genes coexpressed with lnc18q22.2 in human liver or affected by lnc18q22.2 knockdown. CONCLUSION We identified an lncRNA that can play an important regulatory role in liver function and provide new insights into the regulation of hepatocyte viability in NASH. (Hepatology 2017;66:794-808).
Collapse
Affiliation(s)
- Biljana Atanasovska
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sander S Rensen
- Department of Surgery, University Hospital Maastricht, and Nutrition and Toxicology Research Institute, University of Maastricht, Maastricht, The Netherlands
| | - Marijke R van der Sijde
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Glenn Marsman
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Vinod Kumar
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iris Jonkers
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ronit Shiri-Sverdlov
- Departments of Molecular Genetics, Molecular Cell Biology, and Population Genetics, Nutrition and Toxicology Research Institute, University of Maastricht, Maastricht, The Netherlands
| | - Jan Willem M Greve
- Department of Surgery, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Marten H Hofker
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Pediatrics, Molecular Genetics, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
19
|
Wang C, Gong J, Wu H. Development of gene polymorphisms in meditators of nonalcoholic fatty liver disease. Biomed Rep 2017; 7:95-104. [PMID: 28804621 DOI: 10.3892/br.2017.926] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/12/2017] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent liver disease worldwide, the morbidity of which closely correlates with diversity of ethnicity, minority, family and location. Its histology spans from simple steatosis, to nonalcoholic steatohepatitis, which ultimately results in fibrosis, cirrhosis and hepatocellular carcinoma. The accelerating prevalence of NAFLD is due to an incremental incidence of metabolic syndrome that is distinguished by dyslipidemia, glucose impairment, obesity, excessive oxidative stress and adipocytokine impairment. Additionally, the pathogenesis of NAFLD is thought to be a multifactorial and complicated disease associated with lifestyle habits, nutritional factors and genetics. However, the pathogenesis and underlying mechanism in the development of NAFLD caused by genetics remains unclear. People have been increasingly emphasizing on the relationship between NAFLD and gene polymorphisms in recent years, with the aim of having a comprehensive elucidation of associated gene polymorphisms influencing the pathogenesis of the disease. In the current article, the authors attempted to critically summarize the most recently identified gene polymorphisms from the facets of glucose metabolism, fatty acid metabolism, oxidative stress and related cytokines in NAFLD that contribute to promoting the progression of the disease.
Collapse
Affiliation(s)
- Chun Wang
- Department of General Surgery, Yongchuan Hospital of Traditional Chinese Medicine, Chongqing 402161, P.R. China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Hao Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
20
|
In silico search for modifier genes associated with pancreatic and liver disease in Cystic Fibrosis. PLoS One 2017; 12:e0173822. [PMID: 28339466 PMCID: PMC5365109 DOI: 10.1371/journal.pone.0173822] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/27/2017] [Indexed: 12/15/2022] Open
Abstract
Cystic Fibrosis is the most common lethal autosomal recessive disorder in the white population, affecting among other organs, the lung, the pancreas and the liver. Whereas Cystic Fibrosis is a monogenic disease, many studies reveal a very complex relationship between genotype and clinical phenotype. Indeed, the broad phenotypic spectrum observed in Cystic Fibrosis is far from being explained by obvious genotype-phenotype correlations and it is admitted that Cystic Fibrosis disease is the result of multiple factors, including effects of the environment as well as modifier genes. Our objective was to highlight new modifier genes with potential implications in the lung, pancreatic and liver outcomes of the disease. For this purpose we performed a system biology approach which combined, database mining, literature mining, gene expression study and network analysis as well as pathway enrichment analysis and protein-protein interactions. We found that IFI16, CCNE2 and IGFBP2 are potential modifiers in the altered lung function in Cystic Fibrosis. We also found that EPHX1, HLA-DQA1, HLA-DQB1, DSP and SLC33A1, GPNMB, NCF2, RASGRP1, LGALS3 and PTPN13, are potential modifiers in pancreas and liver, respectively. Associated pathways indicate that immune system is likely involved and that Ubiquitin C is probably a central node, linking Cystic Fibrosis to liver and pancreatic disease. We highlight here new modifier genes with potential implications in Cystic Fibrosis. Nevertheless, our in silico analysis requires functional analysis to give our results a physiological relevance.
Collapse
|
21
|
Walenbergh SMA, Shiri-Sverdlov R. Cholesterol is a significant risk factor for non-alcoholic steatohepatitis. Expert Rev Gastroenterol Hepatol 2016; 9:1343-6. [PMID: 26395315 DOI: 10.1586/17474124.2015.1092382] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is characterized by hepatic lipid accumulation (steatosis) and inflammation (steatohepatitis). Currently, the exact underlying mechanisms leading to hepatic inflammation remain incompletely understood and therefore therapy options are poor. Analogous to the predominant metabolic risk factor for the metabolic syndrome, NASH patients often display diet-induced dyslipidemia and are therefore also at high risk for cardiovascular disease. Higher lipid levels, in general, are also widely associated with the production of reactive oxygen species during oxidation. However, the exact contribution of the specific type of lipids to hepatic inflammation still remains unclear. In this editorial, we aim to show that cholesterol, in addition to triglycerides and free fatty acids, is an important risk factor in NASH disease pathogenesis. Developing a better understanding of the contribution of lipids underlying NASH pathogenesis is essential for creating effective therapies against this prevalent disease.
Collapse
Affiliation(s)
- Sofie M A Walenbergh
- a Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Ronit Shiri-Sverdlov
- a Department of Molecular Genetics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
22
|
Alizai PH, Wendl J, Roeth AA, Klink CD, Luedde T, Steinhoff I, Neumann UP, Schmeding M, Ulmer F. Functional Liver Recovery After Bariatric Surgery--a Prospective Cohort Study with the LiMAx Test. Obes Surg 2016; 25:2047-53. [PMID: 25869925 DOI: 10.1007/s11695-015-1664-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Bariatric surgery provides long-term weight loss and improvement of obesity-associated diseases such as nonalcoholic steatohepatitis (NASH). Histologic improvement of NASH has been reported in some studies after bariatric surgery. This study was designed to assess the liver function in obese patients as well as its recovery after bariatric surgery with a noninvasive test method. METHODS In a prospective cohort study from October 2011 to May 2014, morbidly obese individuals receiving bariatric surgery were investigated for functional liver recovery (n = 34). Liver function was determined by the LiMAx test (enzymatic capacity of cytochrome P450 1A2) preoperatively, 6 and 12 months postoperatively. Liver biopsy specimens were obtained from 18 participants and classified according to the nonalcoholic fatty liver disease (NAFLD) activity score (NAS). RESULTS The mean age of participants was 44 years, and the mean body mass index (BMI) was 52 kg/m(2). The mean percent excess BMI loss (%EBMIL) was 53 % after 6 months and 68 % after 1 year. Mean liver function capacity increased significantly from 255 μg/kg/h preoperative to 324 μg/kg/h after 6 months and 342 μg/kg/h after 12 months. A negative correlation was observed between %EBMIL and alteration of liver function capacity in the first 6 months. Finally, the median NAS showed a negative correlation with liver function capacity. CONCLUSIONS Bariatric surgery leads to a significant functional recovery of the liver. An initial marked weight loss may negatively influence functional liver recovery.
Collapse
Affiliation(s)
- Patrick H Alizai
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Janica Wendl
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Anjali A Roeth
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Christian D Klink
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Tom Luedde
- Department of Gastroenterology, Digestive Diseases and Intensive Care Medicine, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Inga Steinhoff
- Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Ulf P Neumann
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Maximilian Schmeding
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Florian Ulmer
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
23
|
Aragonès G, Auguet T, Berlanga A, Guiu-Jurado E, Martinez S, Armengol S, Sabench F, Ras R, Hernandez M, Aguilar C, Colom J, Sirvent JJ, Del Castillo D, Richart C. Increased Circulating Levels of Alpha-Ketoglutarate in Morbidly Obese Women with Non-Alcoholic Fatty Liver Disease. PLoS One 2016; 11:e0154601. [PMID: 27123846 PMCID: PMC4849715 DOI: 10.1371/journal.pone.0154601] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/16/2016] [Indexed: 01/06/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) causes a wide spectrum of liver damage, ranging from simple steatosis to cirrhosis. However, simple steatosis (SS) and steatohepatitis (NASH) cannot yet be distinguished by clinical or laboratory features. The aim of this study was to assess the relationship between alpha-ketoglutarate and the degrees of NAFLD in morbidly obese patients. Materials and Methods We used a gas chromatography-quadruple time-of-flight-mass spectrometry analysis to quantify alpha-ketoglutarate in serum from normal-weight subjects (n = 30) and morbidly obese women (n = 97) with or without NAFLD. Results We found that serum levels of alpha-ketoglutarate were significantly higher in morbidly obese women than in normal-weight women. We showed that circulating levels of alpha-ketoglutarate were lower in lean controls and morbidly obese patients without NAFLD. We also found that alpha-ketoglutarate serum levels were higher in both SS and NASH than in normal liver of morbidly obese patients. However, there was no difference between SS and NASH. Moreover, we observed that circulating levels of alpha-ketoglutarate were associated with glucose metabolism parameters, lipid profile, hepatic enzymes and steatosis degree. In addition, diagnostic performance of alpha-ketoglutarate has been analyzed in NAFLD patients. The AUROC curves from patients with liver steatosis exhibited an acceptable clinical utility. Finally, we showed that the combination of biomarkers (AST, ALT and alpha-ketoglutarate) had the highest accuracy in diagnosing liver steatosis. Conclusion These findings suggest that alpha-ketoglutarate can determine the presence of non-alcoholic fatty liver in morbidly obese patients but it is not valid a biomarker for NASH.
Collapse
Affiliation(s)
- Gemma Aragonès
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Teresa Auguet
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
- Servei Medicina Interna, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
| | - Alba Berlanga
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Esther Guiu-Jurado
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Salomé Martinez
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
| | - Sandra Armengol
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Fàtima Sabench
- Servei de Cirurgia. Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Rosa Ras
- Group of Research on Omic Methodologies (GROM). Centre for Omic Sciences (COS), Reus, Spain
| | - Mercè Hernandez
- Servei de Cirurgia. Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Carmen Aguilar
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Josep Colom
- Servei Medicina Interna, Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Joan Josep Sirvent
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
| | - Daniel Del Castillo
- Servei de Cirurgia. Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Cristóbal Richart
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
- Servei Medicina Interna, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
- * E-mail:
| |
Collapse
|
24
|
Zhu R, Baker SS, Moylan CA, Abdelmalek MF, Guy CD, Zamboni F, Wu D, Lin W, Liu W, Baker RD, Govindarajan S, Cao Z, Farci P, Diehl AM, Zhu L. Systematic transcriptome analysis reveals elevated expression of alcohol-metabolizing genes in NAFLD livers. J Pathol 2016; 238:531-542. [DOI: 10.1002/path.4650] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Ruixin Zhu
- Department of Bioinformatics; Tongji University; Shanghai China
| | - Susan S Baker
- Digestive Diseases and Nutrition Center, Department of Pediatrics; The State University of New York at Buffalo; Buffalo New York USA
| | - Cynthia A Moylan
- Division of Gastroenterology and Hepatology, Department of Medicine; Duke University; Durham North Carolina USA
- Division of Gastroenterology and Hepatology, Department of Medicine; Durham Veterans Affairs Medical Center; Durham North Carolina USA
| | - Manal F Abdelmalek
- Division of Gastroenterology and Hepatology, Department of Medicine; Duke University; Durham North Carolina USA
| | - Cynthia D Guy
- Department of Pathology; Duke University; Durham North Carolina USA
| | - Fausto Zamboni
- Liver Transplantation Center; Brotzu Hospital; 09134 Cagliari Italy
| | - Dingfeng Wu
- Department of Bioinformatics; Tongji University; Shanghai China
| | - Weili Lin
- Department of Bioinformatics; Tongji University; Shanghai China
| | - Wensheng Liu
- Digestive Diseases and Nutrition Center, Department of Pediatrics; The State University of New York at Buffalo; Buffalo New York USA
| | - Robert D Baker
- Digestive Diseases and Nutrition Center, Department of Pediatrics; The State University of New York at Buffalo; Buffalo New York USA
| | - Sugantha Govindarajan
- Department of Pathology; University of Southern California; Los Angeles California USA
| | - Zhiwei Cao
- Department of Bioinformatics; Tongji University; Shanghai China
| | - Patrizia Farci
- Laboratory of Infectious Diseases; National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda Maryland USA
| | - Anna Mae Diehl
- Division of Gastroenterology and Hepatology, Department of Medicine; Duke University; Durham North Carolina USA
| | - Lixin Zhu
- Digestive Diseases and Nutrition Center, Department of Pediatrics; The State University of New York at Buffalo; Buffalo New York USA
| |
Collapse
|
25
|
Versican: a novel modulator of hepatic fibrosis. J Transl Med 2016; 96:361-74. [PMID: 26752747 DOI: 10.1038/labinvest.2015.152] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/29/2015] [Accepted: 11/02/2015] [Indexed: 01/17/2023] Open
Abstract
Little is known about the deposition and turnover of proteoglycans in liver fibrosis, despite their abundance in the extracellular matrix. Versican plays diverse roles in modulating cell behavior in other fibroproliferative diseases, but remains poorly described in the liver. Hepatic fibrosis was induced by carbon tetrachloride treatment of C57BL/6 mice over 4 weeks followed by recovery over a 28-day period. Primary mouse hepatic stellate cells (HSCs) were activated in culture and versican was transiently knocked down in human (LX2) and mouse HSCs. Expression of versican, A Disintegrin-like and Metalloproteinase with Thrombospondin-1 motifs (ADAMTS)-1, -4, -5, -8, -9, -15, and -20, and markers of fibrogenesis were studied using immunohistochemistry, real-time quantitative PCR, and western blotting. Immunohistochemistry showed increased expression of versican in cirrhotic human livers and the mouse model of fibrosis. Carbon tetrachloride treatment led to significant increases in versican expression and the proteoglycanases ADAMTS-5, -9, -15, and -20, alongside TNF-α, α-smooth muscle actin (α-SMA), collagen-1, and TGF-β expression. During recovery, expression of many of these genes returned to control levels. However, expression of ADAMTS-5, -8, -9, and -15 showed delayed increases in expression at 28 days of recovery, which corresponded with decreases in versican V0 and V1 cleavage products (G1-DPEAAE(1401) and G1-DPEAAE(441)). Activation of primary HSCs in vitro significantly increased versican, α-SMA, and collagen-1 expression. Transient knockdown of versican in HSCs led to decreases in markers of fibrogenesis and reduced cell proliferation, without inducing apoptosis. Versican expression increases during HSC activation and liver fibrosis, and proteolytic processing occurs during the resolution of fibrosis. Knockdown studies in vitro suggest a possible role of versican in modulating hepatic fibrogenesis.
Collapse
|
26
|
Mayoral Monibas R, Johnson AMF, Osborn O, Traves PG, Mahata SK. Distinct Hepatic Macrophage Populations in Lean and Obese Mice. Front Endocrinol (Lausanne) 2016; 7:152. [PMID: 27999564 PMCID: PMC5138231 DOI: 10.3389/fendo.2016.00152] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/22/2016] [Indexed: 12/17/2022] Open
Abstract
Obesity is a complex metabolic disorder associated with the development of non-communicable diseases such as cirrhosis, non-alcoholic fatty liver disease, and type 2 diabetes. In humans and rodents, obesity promotes hepatic steatosis and inflammation, which leads to increased production of pro-inflammatory cytokines and acute-phase proteins. Liver macrophages (resident as well as recruited) play a significant role in hepatic inflammation and insulin resistance (IR). Interestingly, depletion of hepatic macrophages protects against the development of high-fat-induced steatosis, inflammation, and IR. Kupffer cells (KCs), liver-resident macrophages, are the first-line defense against invading pathogens, clear toxic or immunogenic molecules, and help to maintain the liver in a tolerogenic immune environment. During high fat diet feeding and steatosis, there is an increased number of recruited hepatic macrophages (RHMs) in the liver and activation of KCs to a more inflammatory or M1 state. In this review, we will focus on the role of liver macrophages (KCs and RHMs) during obesity.
Collapse
Affiliation(s)
- Rafael Mayoral Monibas
- Merck Research Laboratories, Kenilworth, NJ, USA
- CIBERehd – Networked Biomedical Research Center, Hepatic and Digestive Diseases, Madrid, Spain
- *Correspondence: Rafael Mayoral Monibas, ; Sushil K. Mahata,
| | - Andrew M. F. Johnson
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| | - Olivia Osborn
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| | - Paqui G. Traves
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA, USA
| | - Sushil K. Mahata
- Metabolic Physiology & Ultrastructural Biology Laboratory, Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
- Metabolic Physiology & Ultrastructural Biology Laboratory, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- *Correspondence: Rafael Mayoral Monibas, ; Sushil K. Mahata,
| |
Collapse
|
27
|
Arendt BM, Comelli EM, Ma DWL, Lou W, Teterina A, Kim T, Fung SK, Wong DKH, McGilvray I, Fischer SE, Allard JP. Altered hepatic gene expression in nonalcoholic fatty liver disease is associated with lower hepatic n-3 and n-6 polyunsaturated fatty acids. Hepatology 2015; 61:1565-78. [PMID: 25581263 DOI: 10.1002/hep.27695] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 12/31/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED In nonalcoholic fatty liver disease, hepatic gene expression and fatty acid (FA) composition have been reported independently, but a comprehensive gene expression profiling in relation to FA composition is lacking. The aim was to assess this relationship. In a cross-sectional study, hepatic gene expression (Illumina Microarray) was first compared among 20 patients with simple steatosis (SS), 19 with nonalcoholic steatohepatitis (NASH), and 24 healthy controls. The FA composition in hepatic total lipids was compared between SS and NASH, and associations between gene expression and FAs were examined. Gene expression differed mainly between healthy controls and patients (SS and NASH), including genes related to unsaturated FA metabolism. Twenty-two genes were differentially expressed between NASH and SS; most of them correlated with disease severity and related more to cancer progression than to lipid metabolism. Biologically active long-chain polyunsaturated FAs (PUFAs; eicosapentaenoic acid + docosahexaenoic acid, arachidonic acid) in hepatic total lipids were lower in NASH than in SS. This may be related to overexpression of FADS1, FADS2, and PNPLA3. The degree and direction of correlations between PUFAs and gene expression were different among SS and NASH, which may suggest that low PUFA content in NASH modulates gene expression in a different way compared with SS or, alternatively, that gene expression influences PUFA content differently depending on disease severity (SS versus NASH). CONCLUSION Well-defined subjects with either healthy liver, SS, or NASH showed distinct hepatic gene expression profiles including genes involved in unsaturated FA metabolism. In patients with NASH, hepatic PUFAs were lower and associations with gene expression were different compared to SS.
Collapse
Affiliation(s)
- Bianca M Arendt
- Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ye H, Liu W. Transcriptional networks implicated in human nonalcoholic fatty liver disease. Mol Genet Genomics 2015; 290:1793-804. [PMID: 25851235 DOI: 10.1007/s00438-015-1037-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/27/2015] [Indexed: 02/06/2023]
Abstract
The transcriptome of nonalcoholic fatty liver disease (NAFLD) was investigated in several studies. However, the implications of transcriptional networks in progressive NAFLD are not clear and mechanisms inducing transition from nonalcoholic simple fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) are still elusive. The aims of this study were to (1) construct networks for progressive NAFLD, (2) identify hub genes and functional modules in these networks and (3) infer potential linkages among hub genes, transcription factors and microRNAs (miRNA) for NAFLD progression. A systems biology approach by combining differential expression analysis and weighted gene co-expression network analysis (WGCNA) was utilized to dissect transcriptional profiles in 19 normal, 10 NAFL and 16 NASH patients. Based on this framework, 3 modules related to chromosome organization, proteasomal ubiquitin-dependent protein degradation and immune response were identified in NASH network. Furthermore, 9 modules of co-expressed genes associated with NAFL/NASH transition were found. Further characterization of these modules defined 13 highly connected hub genes in NAFLD progression network. Interestingly, 11 significantly changed miRNAs were predicted to target 10 of the 13 hub genes. Characterization of modules and hub genes that may be regulated by miRNAs could facilitate the identification of candidate genes and pathways responsible for NAFL/NASH transition and lead to a better understanding of NAFLD pathogenesis. The identified modules and hub genes may point to potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Hua Ye
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Ningbo, 315040, China
| | - Wei Liu
- School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
- Department of Pathology, Human Centrifuge Medical Training Center, Institute of Aviation Medicine of Chinese PLA Air Force, Beijing, 100089, China.
| |
Collapse
|
29
|
Clarke JD, Cherrington NJ. Nonalcoholic steatohepatitis in precision medicine: Unraveling the factors that contribute to individual variability. Pharmacol Ther 2015; 151:99-106. [PMID: 25805597 DOI: 10.1016/j.pharmthera.2015.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/17/2015] [Indexed: 01/14/2023]
Abstract
There are numerous factors in individual variability that make the development and implementation of precision medicine a challenge in the clinic. One of the main goals of precision medicine is to identify the correct dose for each individual in order to maximize therapeutic effect and minimize the occurrence of adverse drug reactions. Many promising advances have been made in identifying and understanding how factors such as genetic polymorphisms can influence drug pharmacokinetics (PK) and contribute to variable drug response (VDR), but it is clear that there remain many unidentified variables. Underlying liver diseases such as nonalcoholic steatohepatitis (NASH) alter absorption, distribution, metabolism, and excretion (ADME) processes and must be considered in the implementation of precision medicine. There is still a profound need for clinical investigation into how NASH-associated changes in ADME mediators, such as metabolism enzymes and transporters, affect the pharmacokinetics of individual drugs known to rely on these pathways for elimination. This review summarizes the key PK factors in individual variability and VDR and highlights NASH as an essential underlying factor that must be considered as the development of precision medicine advances. A multifactorial approach to precision medicine that considers the combination of two or more risk factors (e.g. genetics and NASH) will be required in our effort to provide a new era of benefit for patients.
Collapse
Affiliation(s)
- John D Clarke
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, United States
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, United States.
| |
Collapse
|
30
|
Gorden DL, Myers DS, Ivanova PT, Fahy E, Maurya MR, Gupta S, Min J, Spann NJ, McDonald JG, Kelly SL, Duan J, Sullards MC, Leiker TJ, Barkley RM, Quehenberger O, Armando AM, Milne SB, Mathews TP, Armstrong MD, Li C, Melvin WV, Clements RH, Washington MK, Mendonsa AM, Witztum JL, Guan Z, Glass CK, Murphy RC, Dennis EA, Merrill AH, Russell DW, Subramaniam S, Brown HA. Biomarkers of NAFLD progression: a lipidomics approach to an epidemic. J Lipid Res 2015; 56:722-736. [PMID: 25598080 DOI: 10.1194/jlr.p056002] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The spectrum of nonalcoholic fatty liver disease (NAFLD) includes steatosis, nonalcoholic steatohepatitis (NASH), and cirrhosis. Recognition and timely diagnosis of these different stages, particularly NASH, is important for both potential reversibility and limitation of complications. Liver biopsy remains the clinical standard for definitive diagnosis. Diagnostic tools minimizing the need for invasive procedures or that add information to histologic data are important in novel management strategies for the growing epidemic of NAFLD. We describe an "omics" approach to detecting a reproducible signature of lipid metabolites, aqueous intracellular metabolites, SNPs, and mRNA transcripts in a double-blinded study of patients with different stages of NAFLD that involves profiling liver biopsies, plasma, and urine samples. Using linear discriminant analysis, a panel of 20 plasma metabolites that includes glycerophospholipids, sphingolipids, sterols, and various aqueous small molecular weight components involved in cellular metabolic pathways, can be used to differentiate between NASH and steatosis. This identification of differential biomolecular signatures has the potential to improve clinical diagnosis and facilitate therapeutic intervention of NAFLD.
Collapse
Affiliation(s)
- D Lee Gorden
- Departments of Surgery, Vanderbilt University Medical Center, Nashville, TN; Cancer Biology, Vanderbilt University Medical Center, Nashville, TN
| | - David S Myers
- Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | | | - Eoin Fahy
- Department of Bioengineering, School of Engineering, University of California, San Diego, La Jolla, CA
| | - Mano R Maurya
- Department of Bioengineering, School of Engineering, University of California, San Diego, La Jolla, CA
| | - Shakti Gupta
- Department of Bioengineering, School of Engineering, University of California, San Diego, La Jolla, CA
| | - Jun Min
- Department of Bioengineering, School of Engineering, University of California, San Diego, La Jolla, CA
| | - Nathanael J Spann
- Departments of Cellular and Molecular Medicine and Medicine, University of California, San Diego, La Jolla, CA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Samuel L Kelly
- Schools of Biology, Chemistry, and Biochemistry, and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - Jingjing Duan
- Schools of Biology, Chemistry, and Biochemistry, and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - M Cameron Sullards
- Schools of Biology, Chemistry, and Biochemistry, and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - Thomas J Leiker
- Department of Pharmacology, University of Colorado at Denver, Aurora, CO
| | - Robert M Barkley
- Department of Pharmacology, University of Colorado at Denver, Aurora, CO
| | - Oswald Quehenberger
- Departments of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA; Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA
| | - Aaron M Armando
- Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA
| | - Stephen B Milne
- Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Thomas P Mathews
- Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | | | - Chijun Li
- Department of Biochemistry, Duke University Medical Center, Durham, NC
| | - Willie V Melvin
- Departments of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Ronald H Clements
- Departments of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - M Kay Washington
- Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | | | - Joseph L Witztum
- Departments of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA
| | - Ziqiang Guan
- Department of Biochemistry, Duke University Medical Center, Durham, NC
| | - Christopher K Glass
- Departments of Cellular and Molecular Medicine and Medicine, University of California, San Diego, La Jolla, CA
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado at Denver, Aurora, CO
| | - Edward A Dennis
- Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA; Chemistry and Biochemistry, School of Medicine, University of California, San Diego, La Jolla, CA
| | - Alfred H Merrill
- Schools of Biology, Chemistry, and Biochemistry, and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA
| | - David W Russell
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Shankar Subramaniam
- Department of Bioengineering, School of Engineering, University of California, San Diego, La Jolla, CA; Chemistry and Biochemistry, School of Medicine, University of California, San Diego, La Jolla, CA.
| | - H Alex Brown
- Pharmacology, Vanderbilt University Medical Center, Nashville, TN; Biochemistry, and the Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
31
|
Fitzpatrick E, Dhawan A. Noninvasive biomarkers in non-alcoholic fatty liver disease: Current status and a glimpse of the future. World J Gastroenterol 2014; 20:10851-10863. [PMID: 25152587 PMCID: PMC4138464 DOI: 10.3748/wjg.v20.i31.10851] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 04/23/2014] [Indexed: 02/06/2023] Open
Abstract
The development of non invasive biomarkers of disease has become a major focus of interest in nonalcoholic fatty liver disease (NAFLD). The large prevalence of the disease and the invasive nature of the investigation means that screening with liver biopsy is impractical. In addition to screening, the differentiation of those with simple steatosis vs steatohepatitis and fibrosis is clinically important as the prognosis of each differs. Serum biomarkers may be a combination of simple markers derived from large data sets or direct markers of disease activity. Serum markers of inflammation, apoptosis and oxidative stress in addition to fibrosis have been extensively studied in patients with NAFLD. Other techniques such as transient elastography, magnetic resonance elastography and acoustic radiation force imaging are becoming more established as noninvasive methods of detecting fibrosis in a variety of chronic liver conditions in addition to NAFLD. Newer high throughput methods such as proteomics and glycomics allow the nonhypothesis-driven identification of novel markers and may also potentially contribute to our understanding of the pathogenesis of the condition. This review addresses some of the methodological issues which need to be considered in the search for the ideal biomarker. It is likely that a combination of serum biomarkers and techniques such as transient elastography may provide the optimal diagnostic discrimination however this remains to be proven in large studies.
Collapse
|
32
|
Moya D, Baker SS, Liu W, Garrick M, Kozielski R, Baker RD, Zhu L. Novel pathway for iron deficiency in pediatric non-alcoholic steatohepatitis. Clin Nutr 2014; 34:549-56. [PMID: 25000850 DOI: 10.1016/j.clnu.2014.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 06/17/2014] [Accepted: 06/18/2014] [Indexed: 01/11/2023]
Abstract
BACKGROUND & AIMS Iron may be an important factor in the pathogenesis of non-alcoholic steatohepatitis (NASH) as it catalyzes the production of potent reactive oxygen species. We aim to examine iron status in pediatric NASH. METHODS Serum indices of NASH patients (N = 36) were compared to those in the U.S. National Health and Nutrition Examination Survey database (N = 802). Iron related gene expression was examined in NASH livers and normal livers, using microarray and quantitative real-time PCR (10 NASH livers and 6 controls). Transferrin and catalase expression were also examined in hydrogen peroxide treated HepG2 cells. RESULTS Serum iron concentration (P < 0.01) and soluble transferrin receptor 1 (P < 0.0001) were decreased while serum ferritin was elevated in NASH patients (P < 0.01). No detectable iron was observed in NASH liver by Perls' Prussian blue staining. Transferrin (P < 0.01) and transferrin receptor 2 (P < 0.01) mRNA were elevated in NASH patients. Of particular interest, transferrin mRNA was positively correlated with catalase mRNA (r = 0.9338, P < 0.0001). H2O2 treatment of HepG2 cells induced mRNA expression of transferrin and catalase. CONCLUSIONS Pediatric NASH patients exhibited decreased serum iron concentration and no detectable iron was observed in any NASH liver by Perls' Prussian blue staining. These changes are consistent with the facts that most NASH patients are obese and exhibit chronic inflammation. In line with a status of iron deficiency, gene expression studies suggested decreased expression of transferrin and transferrin receptor 2 in NASH livers. Induction of transferrin by H2O2, and consequently, decreased iron absorption, suggests a novel mechanism for iron deficiency in NASH patients.
Collapse
Affiliation(s)
- Diana Moya
- Department of Pediatrics, SUNY at Buffalo, United States; Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, United States
| | - Susan S Baker
- Department of Pediatrics, SUNY at Buffalo, United States; Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, United States.
| | - Wensheng Liu
- Department of Pediatrics, SUNY at Buffalo, United States; Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, United States
| | - Michael Garrick
- Department of Pediatrics, SUNY at Buffalo, United States; Department of Biochemistry, SUNY at Buffalo, United States
| | - Rafal Kozielski
- Department of Pathology, SUNY at Buffalo, Women and Children's Hospital of Buffalo, Buffalo, NY 14214, United States
| | - Robert D Baker
- Department of Pediatrics, SUNY at Buffalo, United States; Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, United States
| | - Lixin Zhu
- Department of Pediatrics, SUNY at Buffalo, United States; Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, United States.
| |
Collapse
|
33
|
Hennig EE, Mikula M, Goryca K, Paziewska A, Ledwon J, Nesteruk M, Woszczynski M, Walewska-Zielecka B, Pysniak K, Ostrowski J. Extracellular matrix and cytochrome P450 gene expression can distinguish steatohepatitis from steatosis in mice. J Cell Mol Med 2014; 18:1762-72. [PMID: 24913135 PMCID: PMC4196652 DOI: 10.1111/jcmm.12328] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/15/2014] [Indexed: 12/22/2022] Open
Abstract
One of the main questions regarding nonalcoholic fatty liver disease is the molecular background of the transition from simple steatosis (SS) to the inflammatory and fibrogenic condition of steatohepatitis (NASH). We examined the gene expression changes during progression from histologically normal liver to SS and NASH in models of obesity caused by hyperphagia or a high-fat diet. Microarray-based analysis revealed that the expression of 1445 and 264 probe sets was changed exclusively in SS and NASH samples, respectively, and 1577 probe sets were commonly altered in SS and NASH samples. Functional annotations indicated that transcriptome alterations that were common for NASH and SS, as well as exclusive for NASH, involved extracellular matrix (ECM)-related processes, although they differed in the type of matrix structure change. The expression of 80 genes was significantly changed in all three comparisons: SS versus control, NASH versus control and NASH versus SS. Of these genes, epithelial membrane protein 1, IKBKB interacting protein and decorin were progressively up-regulated in both SS and NASH compared to normal tissue. The molecular context of interactions of encoded 80 proteins revealed that they are highly interconnected and significantly enriched for processes involving metabolism by cytochrome P450. Validation of 10 selected mRNAs encoding genes related to ECM and cytochrome P450 with quantitative RT-PCR analysis showed consistent changes in their expression during NASH development. The expression profile of these genes has the potential to distinguish NASH from SS and normal tissue and may possibly be beneficial in the clinical diagnosis of NASH.
Collapse
Affiliation(s)
- Ewa E Hennig
- Department of Gastroenterology and Hepatology, Medical Center for Postgraduate Education, Warsaw, Poland; Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Clarke JD, Novak P, Lake AD, Shipkova P, Aranibar N, Robertson D, Severson PL, Reily MD, Futscher BW, Lehman-McKeeman LD, Cherrington NJ. Characterization of hepatocellular carcinoma related genes and metabolites in human nonalcoholic fatty liver disease. Dig Dis Sci 2014; 59:365-74. [PMID: 24048683 PMCID: PMC3945102 DOI: 10.1007/s10620-013-2873-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/02/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND The worldwide prevalences of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are estimated to range from 30 to 40 % and 5-17 %, respectively. Hepatocellular carcinoma (HCC) is primarily caused by hepatitis B infection, but retrospective data suggest that 4-29 % of NASH cases will progress to HCC. Currently the connection between NASH and HCC is unclear. AIMS The purpose of this study was to identify changes in expression of HCC-related genes and metabolite profiles in NAFLD progression. METHODS Transcriptomic and metabolomic datasets from human liver tissue representing NAFLD progression (normal, steatosis, NASH) were utilized and compared to published data for HCC. RESULTS Genes involved in Wnt signaling were downregulated in NASH but have been reported to be upregulated in HCC. Extracellular matrix/angiogenesis genes were upregulated in NASH, similar to reports in HCC. Iron homeostasis is known to be perturbed in HCC and we observed downregulation of genes in this pathway. In the metabolomics analysis of hepatic NAFLD samples, several changes were opposite to what has been reported in plasma of HCC patients (lysine, phenylalanine, citrulline, creatine, creatinine, glycodeoxycholic acid, inosine, and alpha-ketoglutarate). In contrast, multiple acyl-lyso-phosphatidylcholine metabolites were downregulated in NASH livers, consistent with observations in HCC patient plasma. CONCLUSIONS These data indicate an overlap in the pathogenesis of NAFLD and HCC where several classes of HCC related genes and metabolites are altered in NAFLD. Importantly, Wnt signaling and several metabolites are different, thus implicating these genes and metabolites as mediators in the transition from NASH to HCC.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cluster Analysis
- Databases, Genetic
- Fatty Liver/genetics
- Fatty Liver/metabolism
- Fatty Liver/pathology
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Metabolomics
- Non-alcoholic Fatty Liver Disease
- Signal Transduction/genetics
Collapse
Affiliation(s)
- John D. Clarke
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ
| | - Petr Novak
- Southwest Environmental Health Sciences Center, College of Pharmacy, University of Arizona, Tucson AZ
- Biology Centre ASCR, Institute of Plant Molecular Biology, Ceske Budejovice, Czech Republic
| | - April D. Lake
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ
| | | | | | | | - Paul L. Severson
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ
| | | | - Bernard W. Futscher
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ
| | | | | |
Collapse
|
35
|
Nutrigenomics of high fat diet induced obesity in mice suggests relationships between susceptibility to fatty liver disease and the proteasome. PLoS One 2013; 8:e82825. [PMID: 24324835 PMCID: PMC3855786 DOI: 10.1371/journal.pone.0082825] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 10/28/2013] [Indexed: 01/22/2023] Open
Abstract
Nutritional factors play important roles in the etiology of obesity, type 2 diabetes mellitus and their complications through genotype x environment interactions. We have characterised molecular adaptation to high fat diet (HFD) feeding in inbred mouse strains widely used in genetic and physiological studies. We carried out physiological tests, plasma lipid assays, obesity measures, liver histology, hepatic lipid measurements and liver genome-wide gene transcription profiling in C57BL/6J and BALB/c mice fed either a control or a high fat diet. The two strains showed marked susceptibility (C57BL/6J) and relative resistance (BALB/c) to HFD-induced insulin resistance and non alcoholic fatty liver disease (NAFLD). Global gene set enrichment analysis (GSEA) of transcriptome data identified consistent patterns of expression of key genes (Srebf1, Stard4, Pnpla2, Ccnd1) and molecular pathways in the two strains, which may underlie homeostatic adaptations to dietary fat. Differential regulation of pathways, including the proteasome, the ubiquitin mediated proteolysis and PPAR signalling in fat fed C57BL/6J and BALB/c suggests that altered expression of underlying diet-responsive genes may be involved in contrasting nutrigenomic predisposition and resistance to insulin resistance and NAFLD in these models. Collectively, these data, which further demonstrate the impact of gene x environment interactions on gene expression regulations, contribute to improved knowledge of natural and pathogenic adaptive genomic regulations and molecular mechanisms associated with genetically determined susceptibility and resistance to metabolic diseases.
Collapse
|
36
|
Sanyal AJ. An integrated view of liver injury and disease progression in nonalcoholic steatohepatitis. Hepatol Int 2013. [PMID: 26202294 DOI: 10.1007/s12072-013-9479-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is rapidly becoming the most common cause of chronic liver disease globally. NAFLD represents a host of pathophysiologic mechanisms that culminate in the accumulation of fat, in a predominantly macrovesicular pattern, in the liver along with varying degrees of inflammation, hepatocellular injury, apoptosis and fibrosis. The most common mechanism for the development of NAFLD is insulin resistance. Insulin resistance is commonly associated with obesity, although it can develop in individuals who do not have obesity. A consequence of insulin resistance is increased peripheral lipolysis and increased delivery of free fatty acids to the liver. The concept of lipotoxicity emerged as the mechanisms by which fatty acids produce cell injury, promote apoptosis and activate inflammatory pathways were elucidated. While much work has been done mainly in cell culture models, the free fatty acid concentration in the liver is not significantly changed in NAFLD. Recently, the focus has shifted to alterations in other lipid metabolic pathways that appear to play an important role in the genesis of nonalcoholic steatohepatitis, the aggressive form of NAFLD. The innate immune system and the intestinal microbiota have been implicated in the development of NAFLD. These mechanisms are reviewed in this article.
Collapse
Affiliation(s)
- Arun J Sanyal
- Division of Gastroenterology, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, MCV Box 980342, Richmond, VA, 23298-0342, USA.
| |
Collapse
|
37
|
Wang G, Li B, Hao Y, Zhi J, He C, Xu C. Correlation analysis between gene expression profile of high-fat emulsion-induced non-alcoholic fatty liver and liver regeneration in rat. Cell Biol Int 2013; 37:917-28. [PMID: 23619824 DOI: 10.1002/cbin.10118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/29/2013] [Indexed: 12/21/2022]
Abstract
To explore the relevance of non-alcoholic fatty liver disease (NAFLD) to liver regeneration (LR), rat models of non-alcoholic steatohepatitis (NASH) and LR were established, respectively, then Rat Genome 230 2.0 Array was used to detect the gene expression abundance of them, and the reliabilities of the array data were confirmed by real-time RT-PCR. As a result, the expression of 93 genes was significantly changed during NAFLD occurrence and 948 genes in LR. Hierarchical clustering indicated that the expression profiles of the above two events were quite different. K-means cluster classified their expression patterns into four clusters, and gene expression trends of clusters 1, 2 were similar in NAFLD and LR, while clusters 3, 4 were contrary with the gene expression changes of LR more abundant. DAVID classifications and functional enrichment analysis found that lipid metabolism and carbohydrate metabolism were stronger in NAFLD than in LR, but some other physiological activities including inflammation/immune response, cell adhesion, and migration, cell proliferation and differentiation in NAFLD were weaker than in LR. IPA further indicated that lipid metabolism, inflammation response, and cellular development were highly associated with NAFLD, and thus identified some potential biomarkers for NAFLD.
Collapse
Affiliation(s)
- Gaiping Wang
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, Henan Province, China
| | | | | | | | | | | |
Collapse
|
38
|
Sales V, Patti ME. The Ups and Downs of Insulin Resistance and Type 2 Diabetes: Lessons from Genomic Analyses in Humans. CURRENT CARDIOVASCULAR RISK REPORTS 2012; 7:46-59. [PMID: 23459395 DOI: 10.1007/s12170-012-0283-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We are in the midst of a worldwide epidemic of type 2 diabetes (T2D) and obesity. Understanding the mechanisms underlying these diseases is critical if we are to halt their progression and ultimately prevent their development. The advent and widespread implementation of microarray technology has allowed analysis of small samples of human skeletal muscle, adipose, liver, pancreas and blood. While patterns differ in each tissue, several dominant themes have emerged from these studies, including altered expression of genes indicating increased inflammation and altered lipid and mitochondrial oxidative metabolism and insulin signaling in patients with T2D, and in some cases, in those at risk for disease. Unraveling which changes in gene expression are primary, and which are secondary to an insulin resistant or diabetes metabolic milieu remains a scientific challenge but we are one step closer.
Collapse
Affiliation(s)
- Vicencia Sales
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School ; Department of Biophysics, Federal University of São Paulo, UNIFESP/EPM, São Paulo, SP, Brazil
| | | |
Collapse
|
39
|
Rahman W, Huang P, Belov L, Chrisp JS, Christopherson RI, Stapelberg PM, Warner FJ, George J, Bowen DG, Strasser SI, Koorey D, Sharland AF, McCaughan GW, Shackel NA. Analysis of human liver disease using a cluster of differentiation (CD) antibody microarray. Liver Int 2012; 32:1527-34. [PMID: 22863037 DOI: 10.1111/j.1478-3231.2012.02854.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 06/17/2012] [Accepted: 06/27/2012] [Indexed: 02/13/2023]
Abstract
BACKGROUND A CD antibody microarray has been previously developed allowing semi-quantitative identification of greater than 80 CD antigens on circulating leucocytes from peripheral blood samples. This assay, which uses a live cell-capture technique, enables an extensive leucocyte immunophenotype determination in a single analysis and to date this has been used successfully to characterise diseases including human leukaemias and HIV infection. AIMS To determine CD antigen expression profiles for patients with various liver diseases and to look for preserved disease-specific signatures. METHODS Three liver disease groups including hepatitis C (HCV) (n = 35), non-alcoholic steatohepatitis (NASH) (n = 21) and alcohol-related liver disease (n = 14) were compared with a normal group (n = 23). Hierarchal Clustering (HCL) and Principal Component Analysis (PCA) of the data revealed distinct binding patterns for patients with and without cirrhosis. RESULTS Patients with cirrhosis and portal hypertension compared with those without cirrhosis had significantly reduced expression of several markers of T-cell function including CD45, CD8, CD28 and TCR α/β. Disease prediction algorithms based on the expression data were able to discriminate cirrhotics from non-cirrhotics with 71% overall success, which improved to 77% when only patients with HCV were considered. CONCLUSIONS These results demonstrate disease-specific consensus patterns of expression of CD antigens for patients with chronic liver disease, suggesting that the CD antibody array is a promising tool in the analysis of human liver disease, and with further refinement may have future research and clinical utility.
Collapse
Affiliation(s)
- Wassim Rahman
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Starmann J, Fälth M, Spindelböck W, Lanz KL, Lackner C, Zatloukal K, Trauner M, Sültmann H. Gene expression profiling unravels cancer-related hepatic molecular signatures in steatohepatitis but not in steatosis. PLoS One 2012; 7:e46584. [PMID: 23071592 PMCID: PMC3468618 DOI: 10.1371/journal.pone.0046584] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 09/02/2012] [Indexed: 02/07/2023] Open
Abstract
Background Pathogenesis and factors for determining progression of alcoholic and non-alcoholic steatosis to steatohepatitis with risk of further progression to liver cirrhosis and cancer are poorly understood. In the present study, we aimed to identify potential molecular signatures for discrimination of steatohepatitis from steatosis. Methodology and Results Global microarray gene expression analysis was applied to unravel differentially expressed genes between steatohepatitis compared to steatosis and control samples. For functional annotation as well as the identification of disease-relevant biological processes of the differentially expressed genes the gene ontology (GO) database was used. Selected candidate genes (n = 46) were validated in 87 human liver samples from two sample cohorts by quantitative real-time PCR (qRT-PCR). The GO analysis revealed that genes down-regulated in steatohepatitis were mainly involved in metabolic processes. Genes up-regulated in steatohepatitis samples were associated with cancer progression and proliferation. In surgical liver resection samples, 39 genes and in percutaneous liver biopsies, 30 genes were significantly up-regulated in steatohepatitis. Furthermore, immunohistochemical investigation of human liver tissue revealed a significant increase of AKR1B10 protein expression in steatohepatitis. Conclusions The development of steatohepatitis is characterized by distinct molecular changes. The most striking examples in this respect were KRT23 and AKR1B10, which we found to be highly differentially expressed in steatohepatitis compared to steatosis and normal liver. We propose that KRT23 and AKR1B10 may serve as future potential biomarkers for steatohepatitis as well as markers for progression to HCC.
Collapse
Affiliation(s)
- Julia Starmann
- Unit Cancer Genome Research, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
| | - Maria Fälth
- Unit Cancer Genome Research, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
- Cellzome/GlaxoSmithKline, Heidelberg, Germany
| | - Walter Spindelböck
- Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Katja-Lauren Lanz
- Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | | | - Kurt Zatloukal
- Institute of Pathology, Medical University of Graz, Austria
| | - Michael Trauner
- Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Holger Sültmann
- Unit Cancer Genome Research, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
41
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the Western world. It is closely associated with metabolic syndrome. The alarming epidemics of diabetes and obesity have fueled an increasing prevalence of NAFLD, particularly among these high-risk groups. Histologically, NAFLD encompasses a disease spectrum ranging from simple steatosis to non-alcoholic steatohepatitis (NASH), which is characterized by hepatocyte injury, inflammation, and variable degrees of fibrosis on liver biopsy. Non-alcoholic steatohepatitis can progress to cirrhosis in a fraction of patients. There is currently little understanding of risk factors for disease progression and the disease pathogenesis has not been fully defined. Liver biopsy remains the gold standard for diagnosis. Weight loss, dietary modification, and the treatment of underlying metabolic syndrome remain the mainstays of therapy once the diagnosis is established. There are no well-established pharmacological agents for treatment of NASH, although this is a subject of ongoing research.
Collapse
|
42
|
Athinarayanan S, Liu W. Non-Alcoholic Fatty Liver Disease: Current Perspectives and Future Direction in Disease pathogenesis, Treatment and Diagnosis. Med Chem 2012; 2:e104. [PMID: 24660114 PMCID: PMC3959643 DOI: 10.4172/2161-0444.1000e104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases in the world. An important implication of this disease is the progression of the disease to a more complicated condition called non-alcoholic steatohepatitis (NASH) and the wide variety of clinical presentations. Over the past 5 years, remarkable progresses have been made in understanding the genetic basis for the disease. Recent clinical trials in pharmacotherapy for the disease have been encouraging as well. It is anticipated that the integration of the wide spectrum information retrieved from genomics, transcriptomics and proteomics studies conducted in NAFLD and NASH will mediate a better understanding of the disease pathogenesis and facilitate the postulation of disease pathobiology pathways. Genetic and biological markers identified from the omics studies may hold promise for diagnosis, personalized treatment, early prevention and new drug development.
Collapse
Affiliation(s)
- Shaminie Athinarayanan
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. West Lafayette, IN 47907, USA
| | - Wanqing Liu
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. West Lafayette, IN 47907, USA
| |
Collapse
|
43
|
Valle A, Catalán V, Rodríguez A, Rotellar F, Valentí V, Silva C, Salvador J, Frühbeck G, Gómez-Ambrosi J, Roca P, Oliver J. Identification of liver proteins altered by type 2 diabetes mellitus in obese subjects. Liver Int 2012; 32:951-61. [PMID: 22340678 DOI: 10.1111/j.1478-3231.2012.02765.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 01/15/2012] [Indexed: 02/13/2023]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a well-known factor risk for non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) in obese patients. AIMS To better understand the association between T2DM and NAFLD, global changes in protein expression in diabetic and non-diabetic obese subjects were assessed by a proteomic approach. METHODS Liver samples were obtained from diabetic and non-diabetic morbid obese subjects (BMI>40 kg/m(2) ). Histological analysis was used to evaluate hepatic steatosis and the degree of anatomopathological alteration. Changes in protein expression were analysed by two-dimentional electrophoresis combined with MALDI-TOF mass spectrometry. Levels of glutathione, carbonyl and 4-HNE protein adducts were used to assess oxidative stress status. RESULTS Of 850 proteins analysed, 33 were differentially expressed in T2DM obese subjects. Of these, 27 were unequivocally identified by mass spectrometry. Analysis of protein sets revealed patterns of decreased abundance in mitochondrial enzymes, proteins involved in methione metabolism, and oxidative stress response. Accordingly, T2DM subjects showed decreased levels of glutathione, the antioxidant byproduct of methionine metabolism via the transsulfuration pathway, and higher levels of protein and lipid oxidative damage. Changes in detoxyfing enzymes, carbohydrate metabolism, proteasome subunits and retinoic acid synthesis were also found. CONCLUSIONS The results suggest alterations in mitochondrial function and methionine metabolism as potential contributing factors to increased oxidative stress in liver of obese diabetic patients which may be influencing the development of NAFLD and NASH.
Collapse
Affiliation(s)
- Adamo Valle
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d'Investigació en Ciències de la Salut, Universitat de les Illes Balears, Palma de Mallorca, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Szalowska E, Dijkstra M, Elferink MGL, Weening D, de Vries M, Bruinenberg M, Hoek A, Roelofsen H, Groothuis GMM, Vonk RJ. Comparative analysis of the human hepatic and adipose tissue transcriptomes during LPS-induced inflammation leads to the identification of differential biological pathways and candidate biomarkers. BMC Med Genomics 2011; 4:71. [PMID: 21978410 PMCID: PMC3196688 DOI: 10.1186/1755-8794-4-71] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Accepted: 10/06/2011] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Insulin resistance (IR) is accompanied by chronic low grade systemic inflammation, obesity, and deregulation of total body energy homeostasis. We induced inflammation in adipose and liver tissues in vitro in order to mimic inflammation in vivo with the aim to identify tissue-specific processes implicated in IR and to find biomarkers indicative for tissue-specific IR. METHODS Human adipose and liver tissues were cultured in the absence or presence of LPS and DNA Microarray Technology was applied for their transcriptome analysis. Gene Ontology (GO), gene functional analysis, and prediction of genes encoding for secretome were performed using publicly available bioinformatics tools (DAVID, STRING, SecretomeP). The transcriptome data were validated by proteomics analysis of the inflamed adipose tissue secretome. RESULTS LPS treatment significantly affected 667 and 483 genes in adipose and liver tissues respectively. The GO analysis revealed that during inflammation adipose tissue, compared to liver tissue, had more significantly upregulated genes, GO terms, and functional clusters related to inflammation and angiogenesis. The secretome prediction led to identification of 399 and 236 genes in adipose and liver tissue respectively. The secretomes of both tissues shared 66 genes and the remaining genes were the differential candidate biomarkers indicative for inflamed adipose or liver tissue. The transcriptome data of the inflamed adipose tissue secretome showed excellent correlation with the proteomics data. CONCLUSIONS The higher number of altered proinflammatory genes, GO processes, and genes encoding for secretome during inflammation in adipose tissue compared to liver tissue, suggests that adipose tissue is the major organ contributing to the development of systemic inflammation observed in IR. The identified tissue-specific functional clusters and biomarkers might be used in a strategy for the development of tissue-targeted treatment of insulin resistance in patients.
Collapse
Affiliation(s)
- Ewa Szalowska
- Centre for Medical Biomics, University Medical Centre Groningen (UMCG), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lake AD, Novak P, Fisher CD, Jackson JP, Hardwick RN, Billheimer DD, Klimecki WT, Cherrington NJ. Analysis of global and absorption, distribution, metabolism, and elimination gene expression in the progressive stages of human nonalcoholic fatty liver disease. Drug Metab Dispos 2011; 39:1954-60. [PMID: 21737566 PMCID: PMC3186211 DOI: 10.1124/dmd.111.040592] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 07/07/2011] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by a series of pathological changes that range from simple fatty liver to nonalcoholic steatohepatitis (NASH). The objective of this study is to describe changes in global gene expression associated with the progression of human NAFLD. This study is focused on the expression levels of genes responsible for the absorption, distribution, metabolism, and elimination (ADME) of drugs. Differential gene expression between three clinically defined pathological groups-normal, steatosis, and NASH-was analyzed. Genome-wide mRNA levels in samples of human liver tissue were assayed with Affymetrix GeneChip Human 1.0ST arrays. A total of 11,633 genes exhibited altered expression out of 33,252 genes at a 5% false discovery rate. Most gene expression changes occurred in the progression from steatosis to NASH. Principal component analysis revealed that hepatic disease status was the major determinant of differential ADME gene expression rather than age or sex of sample donors. Among the 515 drug transporters and 258 drug-metabolizing enzymes (DMEs) examined, uptake transporters but not efflux transporters or DMEs were significantly over-represented in the number of genes down-regulated. These results suggest that uptake transporter genes are coordinately targeted for down-regulation at the global level during the pathological development of NASH and that these patients may have decreased drug uptake capacity. This coordinated regulation of uptake transporter genes is indicative of a hepatoprotective mechanism acting to prevent accumulation of toxic intermediates in disease-compromised hepatocytes.
Collapse
Affiliation(s)
- April D Lake
- Department of Pharmacology and Toxicology, Tucson, AZ 85721, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Promrat K, Longato L, Wands JR, de la Monte SM. Weight loss amelioration of non-alcoholic steatohepatitis linked to shifts in hepatic ceramide expression and serum ceramide levels. Hepatol Res 2011; 41:754-62. [PMID: 21794038 PMCID: PMC4550290 DOI: 10.1111/j.1872-034x.2011.00815.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIM Non-alcoholic steatohepatitis (NASH) is associated with increased hepatic insulin resistance. Ceramides and other toxic sphingolipids promote inflammation, lipotoxicity and insulin resistance; however, the role of ceramides in the pathogenesis of NASH has not been determined. This study characterizes expression of ceramide-related genes in human livers with NASH and examines the effects of weight loss on NASH and pro-ceramide gene expression in liver. METHODS Liver biopsies were obtained to assess the histopathological status of non-alcoholic fatty liver disease/NASH prior to and following completion of a 1-year course of implementing either lifestyle changes or a standard enrichment protocol designed to encourage weight loss. Liver biopsy samples were used to measure pro-ceramide gene expression by quantitative reverse transcriptase polymerase chain reaction analysis (qRT-PCR), and serum was used to measure ceramide immunoreactivity. RESULTS At baseline, serine palmitoyltransferase (SPTLC)2 (P = 0.02) and ceramide synthase (CER)1 (P = 0.001) mRNA transcripts were less abundantly expressed in livers with NASH relative to normal controls. After weight loss (average 9.3%), SPTLC1 (P = 0.005) and uridine diphosphate glucose ceramide glucosyltransferase (UGCG) (P = 0.001) expression significantly declined while CER1 increased (P = 0.001) among subjects randomized to the lifestyle change subgroup. Reductions in calorie and fat consumption were significantly correlated with changes in ceramide-related gene expression. Finally, both net and relative reductions in serum ceramide levels were significantly greater in the lifestyles compared with the standard enrichment (control) protocol group (both P < 0.005). CONCLUSION NASH is associated with increased insulin resistance and altered ceramide gene expression in liver. Weight loss-mediated reversal of NASH is associated with reduced pro-ceramide gene expression in liver.
Collapse
Affiliation(s)
- Kittichai Promrat
- Division of Gastroenterology and Hepatology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA,Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
| | - Lisa Longato
- Liver Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Jack R. Wands
- Division of Gastroenterology and Hepatology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA,Liver Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Suzanne M. de la Monte
- Liver Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA,Department of Pathology, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
47
|
Younossi ZM, Page S, Rafiq N, Birerdinc A, Stepanova M, Hossain N, Afendy A, Younoszai Z, Goodman Z, Baranova A. A biomarker panel for non-alcoholic steatohepatitis (NASH) and NASH-related fibrosis. Obes Surg 2011; 21:431-9. [PMID: 20532833 DOI: 10.1007/s11695-010-0204-1] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Patients with biopsy-proven NASH and especially those with fibrosis are at risk for progressive liver disease, emphasizing the clinical importance of developing non-invasive biomarkers for NASH and NASH-related fibrosis. AIM This study examines the performance of a new biomarker panel for NASH and NASH-related fibrosis with a combination of clinical and laboratory variables. METHODS Enrolled patients had biopsy-proven NAFLD. Clinical data, laboratory data, and serum samples were collected at the time of biopsy. Fasting serum was assayed for adiponectin, resistin, glucose, M30, M65, Tissue inhibitor of metalloproteinases-1 (Timp-1), ProCollagen 3 N-terminal peptide (PIIINP), and hyaluronic acid (HA). Regression models predictive of NASH, NASH-related fibrosis, and NASH-related advanced fibrosis were designed and cross-validated. RESULTS Of the 79 enrolled NAFLD patients, 40 had biopsy-proven NASH and 39 had non-NASH NAFLD. Clinical and laboratory data were from this cohort were used to develop a NAFLD Diagnostic Panel that includes three models (models for NASH, NASH-related fibrosis, and NASH-related advanced fibrosis). The model for predicting NASH includes diabetes, gender, BMI, triglycerides, M30 (apoptosis), and M65-M30 (necrosis) [AUC: 0.81, 95% CI, 0.70-0.89, 300 p value <9E 301 (-06)]. The NASH-related fibrosis prediction model includes the same predictors [AUC: 0.80, 95% CI 0.68-0.88, 307 p value <0.00014]. Finally, the NASH-related advanced fibrosis model includes type 2 diabetes, serum triglycerides, Timp-1, and AST [AUC: 0.81, 95% CI, 0.70-0.89; p value, 0.000062]. CONCLUSIONS This NAFLD Diagnostic Panel based on a clinical and laboratory data has good performance characteristics and is easy to use. This biomarker panel could become useful in the management of patients with NAFLD.
Collapse
Affiliation(s)
- Zobair M Younossi
- Center for Liver Diseases, Inova Fairfax Hospital, 3300 Gallows Road, Falls Church, VA 22042, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Merrell MD, Cherrington NJ. Drug metabolism alterations in nonalcoholic fatty liver disease. Drug Metab Rev 2011; 43:317-34. [PMID: 21612324 DOI: 10.3109/03602532.2011.577781] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug-metabolizing enzymes play a vital role in the elimination of the majority of therapeutic drugs. The major organ involved in drug metabolism is the liver. Chronic liver diseases have been identified as a potential source of significant interindividual variation in metabolism. Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the United States, affecting between 60 and 90 million Americans, yet the vast majority of NAFLD patients are undiagnosed. NAFLD encompasses a spectrum of pathologies, ranging from steatosis to nonalcoholic steatohepatitis and fibrosis. Numerous animal studies have investigated the effects of NAFLD on hepatic gene expression, observing significant alterations in mRNA, protein, and activity levels. Information on the effects of NAFLD in human patients is limited, though several significant investigations have recently been published. Significant alterations in the activity of drug-metabolizing enzymes may affect the clearance of therapeutic drugs, with the potential to result in adverse drug reactions. With the enormous prevalence of NAFLD, it is conceivable that every drug currently on the market is being given to patients with NAFLD. The current review is intended to present the results from both animal models and human patients, summarizing the observed alterations in the expression and activity of the phase I and II drug-metabolizing enzymes.
Collapse
Affiliation(s)
- Matthew D Merrell
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, USA
| | | |
Collapse
|
49
|
Miller MH, Ferguson MAJ, Dillon JF. Systematic review of performance of non-invasive biomarkers in the evaluation of non-alcoholic fatty liver disease. Liver Int 2011; 31:461-73. [PMID: 21382157 DOI: 10.1111/j.1478-3231.2011.02451.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
This systematic review evaluates the many studies carried out to discover and evaluate non-invasive markers of non-alcoholic fatty liver disease (NAFLD). Many different strategies and methods have been used in this task, from the discovery of new markers by global 'shotgun' studies to hypothesis-driven approaches, to the development of algorithm tests based on routinely available clinical and biochemical parameters. We examined the various different approaches, summarising the findings in an attempt to give an overview of the field of non-invasive markers in NAFLD, encompassing markers of steatosis, necro-inflammation and fibrosis. The body of literature surrounding this topic is complex and varied, encompassing not only different methodologies but also different patient characteristics, different disease definitions, as well as different end points. This reflects the heterogeneity of NAFLD, which, however, introduces considerably difficulty when trying to draw a conclusion between studies. We have divided this review into three main chapters based on the characteristics of the studies. The Genomics/Proteomics chapter reviews studies using a non-hypothesis-driven approach to biomarker discovery. Thereafter, we evaluate studies of association - studies that target-specific markers, comparing levels between disease and control groups. Finally, we examine the algorithm tests - mathematical systems developed on the basis of previously described markers and assessed, usually, by receiver operator curve analysis. While radiological examination and investigations offer important diagnostic information, such studies are not discussed in this review - the body of literature surrounding blood and anthropological markers is complex and varied, demanding close attention.
Collapse
Affiliation(s)
- Michael H Miller
- Biomedical Research Institute, University of Dundee, Ninewells Hospital, Dundee, UK.
| | | | | |
Collapse
|
50
|
Tateno C, Kataoka M, Utoh R, Tachibana A, Itamoto T, Asahara T, Miya F, Tsunoda T, Yoshizato K. Growth hormone-dependent pathogenesis of human hepatic steatosis in a novel mouse model bearing a human hepatocyte-repopulated liver. Endocrinology 2011; 152:1479-91. [PMID: 21303949 DOI: 10.1210/en.2010-0953] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Clinical studies have shown a close association between nonalcoholic fatty liver disease and adult-onset GH deficiency, but the relevant molecular mechanisms are still unclear. No mouse model has been suitable to study the etiological relationship of human nonalcoholic fatty liver disease and human adult-onset GH deficiency under conditions similar to the human liver in vivo. We generated human (h-)hepatocyte chimeric mice with livers that were predominantly repopulated with h-hepatocytes in a h-GH-deficient state. The chimeric mouse liver was mostly repopulated with h-hepatocytes about 50 d after transplantation and spontaneously became fatty in the h-hepatocyte regions after about 70 d. Infusion of the chimeric mouse with h-GH drastically decreased steatosis, showing the direct cause of h-GH deficiency in the generation of hepatic steatosis. Using microarray profiles aided by real-time quantitative RT-PCR, comparison between h-hepatocytes from h-GH-untreated and -treated mice identified 14 GH-up-regulated and four GH-down-regulated genes, including IGF-I, SOCS2, NNMT, IGFLS, P4AH1, SLC16A1, SRD5A1, FADS1, and AKR1B10, respectively. These GH-up- and -down-regulated genes were expressed in the chimeric mouse liver at lower and higher levels than in human livers, respectively. Treatment of the chimeric mice with h-GH ameliorated their altered expression. h-Hepatocytes were separated from chimeric mouse livers for testing in vitro effects of h-GH or h-IGF-I on gene expression, and results showed that GH directly regulated the expression of IGF-I, SOCS2, NNMT, IGFALS, P4AH1, FADS1, and AKR1B10. In conclusion, the chimeric mouse is a novel h-GH-deficient animal model for studying in vivo h-GH-dependent human liver dysfunctions.
Collapse
Affiliation(s)
- Chise Tateno
- Yoshizato Project, Hiroshima Prefectural Institute of Industrial Science and Technology, Cooperative Link of Unique Science and Technology for Economy Revitalization, Higashihirosima, Hiroshima 739-0046, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|