101
|
Huang F, Wang J, Yu F, Tang Y, Ding G, Yang Z, Sun Y. Protective Effect of Meretrix meretrix Oligopeptides on High-Fat-Diet-Induced Non-Alcoholic Fatty Liver Disease in Mice. Mar Drugs 2018; 16:md16020039. [PMID: 29360762 PMCID: PMC5852467 DOI: 10.3390/md16020039] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/09/2018] [Accepted: 01/20/2018] [Indexed: 12/15/2022] Open
Abstract
The present study investigated the effects of MMO (Meretrix meretrix oligopeptides) on mice fed a high-fat diet. Mice were fed either a normal control diet (NC) or a high-fat diet (HFD) without or with MMO (50 mg/kg or 250 mg/kg) for four weeks. Levels of ALT, AST, liver tissue GSH-Px, and SOD activities, MDA levels were measured using commercially available kits; HE staining was performed to analyze pathologic changes of the liver; a TEM assay was performed to measure the ultrastructural alterations of the mitochondria, and Western blotting was performed to detect the expression of gene proteins related to lipid metabolism, inflammation, and liver apoptosis. After six weeks, body weight, ALT, AST, and MDA levels were significantly increased, and GSH-Px levels and SOD activities were significantly decreased in the HFD control group compared with the NC group. Consumption of the HFD compared with the NC caused fatty liver abnormal mitochondria with loss of cristae, intramitochondrial granules, and a swollen and rarefied matrix. Administration of MMO significantly decreased body weight gain, and ALT, AST, and MDA levels; increased SOD activity and GSH-Px levels; alleviated fatty liver steatosis; decreased the early apoptosis population; downregulated SREBP-1c, Bax, Caspase-9, Caspase-3, TNF-α, and NF-κB protein levels; and upregulated PPAR-α, Bcl-2, and AMPK-α, compared with the HFD control group. MMO exhibited protective effects in mice with NAFLD by regulating the NF-κB anti-inflammation signaling pathways to inhibit inflammation, regulate AMPK-α, PPAR-α and SREBP-1c to improve lipid metabolism disorder, and regulate Bcl-2/Bax anti-apoptosis signaling pathways to prevent liver cell apoptosis. These results suggest that dietary supplementation with MMO ameliorates high-fat-diet-induced NAFLD.
Collapse
Affiliation(s)
- Fangfang Huang
- School of Food Science and Pharmacy, Zhejiang Provincial Key Engineering Technology Research Center of Marine Biomedical Products, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Jiajia Wang
- School of Food Science and Pharmacy, Zhejiang Provincial Key Engineering Technology Research Center of Marine Biomedical Products, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Fangmiao Yu
- School of Food Science and Pharmacy, Zhejiang Provincial Key Engineering Technology Research Center of Marine Biomedical Products, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Yunping Tang
- School of Food Science and Pharmacy, Zhejiang Provincial Key Engineering Technology Research Center of Marine Biomedical Products, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Guofang Ding
- School of Food Science and Pharmacy, Zhejiang Provincial Key Engineering Technology Research Center of Marine Biomedical Products, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Zuisu Yang
- School of Food Science and Pharmacy, Zhejiang Provincial Key Engineering Technology Research Center of Marine Biomedical Products, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Yu Sun
- Zhejiang Provincial Key Engineering Technology Research Center of Marine Biomedical Products, Zhejiang Ocean University Donghai Science and Technology College, Zhoushan 316000, China.
| |
Collapse
|
102
|
Tølbøl KS, Kristiansen MNB, Hansen HH, Veidal SS, Rigbolt KTG, Gillum MP, Jelsing J, Vrang N, Feigh M. Metabolic and hepatic effects of liraglutide, obeticholic acid and elafibranor in diet-induced obese mouse models of biopsy-confirmed nonalcoholic steatohepatitis. World J Gastroenterol 2018; 24:179-194. [PMID: 29375204 PMCID: PMC5768937 DOI: 10.3748/wjg.v24.i2.179] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/24/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the pharmacodynamics of compounds in clinical development for nonalcoholic steatohepatitis (NASH) in obese mouse models of biopsy-confirmed NASH.
METHODS Male wild-type C57BL/6J mice (DIO-NASH) and Lepob/ob (ob/ob-NASH) mice were fed a diet high in trans-fat (40%), fructose (20%) and cholesterol (2%) for 30 and 21 wk, respectively. Prior to treatment, all mice underwent liver biopsy for confirmation and stratification of liver steatosis and fibrosis, using the nonalcoholic fatty liver disease activity score (NAS) and fibrosis staging system. The mice were kept on the diet and received vehicle, liraglutide (0.2 mg/kg, SC, BID), obeticholic acid (OCA, 30 mg/kg PO, QD), or elafibranor (30 mg/kg PO, QD) for eight weeks. Within-subject comparisons were performed on changes in steatosis, inflammation, ballooning degeneration, and fibrosis scores. In addition, compound effects were evaluated by quantitative liver histology, including percent fractional area of liver fat, galectin-3, and collagen 1a1.
RESULTS Liraglutide and elafibranor, but not OCA, reduced body weight in both models. Liraglutide improved steatosis scores in DIO-NASH mice only. Elafibranor and OCA reduced histopathological scores of hepatic steatosis and inflammation in both models, but only elafibranor reduced fibrosis severity. Liraglutide and OCA reduced total liver fat, collagen 1a1, and galectin-3 content, driven by significant reductions in liver weight. The individual drug effects on NASH histological endpoints were supported by global gene expression (RNA sequencing) and liver lipid biochemistry.
CONCLUSION DIO-NASH and ob/ob-NASH mouse models show distinct treatment effects of liraglutide, OCA, and elafibranor, being in general agreement with corresponding findings in clinical trials for NASH. The present data therefore further supports the clinical translatability and utility of DIO-NASH and ob/ob-NASH mouse models of NASH for probing the therapeutic efficacy of compounds in preclinical drug development for NASH.
Collapse
Affiliation(s)
- Kirstine S Tølbøl
- Gubra Aps, Hørsholm DK-2970, Denmark
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
- Section for Metabolic Imaging and Liver Metabolism, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Maria NB Kristiansen
- Gubra Aps, Hørsholm DK-2970, Denmark
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | | | | | | | - Matthew P Gillum
- Section for Metabolic Imaging and Liver Metabolism, The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | | | - Niels Vrang
- Gubra Aps, Hørsholm DK-2970, Denmark
- Department of Chemistry, Faculty of Science, University of Copenhagen, Copenhagen DK-2200, Denmark
| | | |
Collapse
|
103
|
Khavasi N, Somi MH, Khadem E, Faramarzi E, Ayati MH, Fazljou SMB, Torbati M. Effect of Daily Caper Fruit Pickle Consumption on Disease Regression in Patients with Non-Alcoholic Fatty Liver Disease: a Double-Blinded Randomized Clinical Trial. Adv Pharm Bull 2017; 7:645-650. [PMID: 29399555 PMCID: PMC5788220 DOI: 10.15171/apb.2017.077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 12/31/2022] Open
Abstract
Purpose: Despite numerous studies on the effects of complementary medicine, to our knowledge, there is no study on the effects of Capparis spinosa on disease regression in non-alcoholic fatty liver disease (NAFLD) patients. We compared the effects of caper fruit pickle consumption, as an Iranian traditional medicine product, on the anthropometric measures and biochemical parameters in different NAFLD patients. Methods: A 12-weeks randomized, controlled, double-blind trial was designed in 44 NAFLD patients randomly categorized for the control (n=22) or caper (n=22). The caper group received 40-50 gr of caper fruit pickles with meals daily. Before and after treatment, we assessed anthropometric measures, grade of fatty liver, serum lipoproteins and liver enzymes. Results: Weight and BMI were significantly decreased in the caper (p<0.001 and p<0.001) and control group (p=0.001 and p=0.001), respectively. Serum TG, TC and LDL.C just were significantly decreased in the control group (p=0.01, p<0.001 and p<0.001, respectively). Adjusted to the baseline measures, serum ALT and AST reduction were significantly higher in the caper than control group from baseline up to the end of the study (p<0.001 and p=0.02, respectively). After weeks 12, disease severity was significantly decreased in the caper group (p <0.001). Conclusion: Our results suggest that daily caper fruit pickle consumption for 12 weeks may be potentially effective on improving the biochemical parameters in NAFLD patients. Further, additional larger controlled trials are needed for the verification of these results.
Collapse
Affiliation(s)
- Narjes Khavasi
- Department of Traditional Medicine, Faculty of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hosein Somi
- Department of liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical sciences, Tabriz, Iran
| | - Ebrahim Khadem
- Department of Traditional Medicine, School of Traditional Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elnaz Faramarzi
- Department of liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical sciences, Tabriz, Iran
| | - Mohammad Hossein Ayati
- Department of Traditional Medicine, School of Traditional Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyyed Muhammad Bagher Fazljou
- Department of Traditional Medicine, Faculty of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadali Torbati
- Department of Food Science and Technology, Faculty of nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
104
|
Izdebska M, Piątkowska-Chmiel I, Korolczuk A, Herbet M, Gawrońska-Grzywacz M, Gieroba R, Sysa M, Czajkowska-Bania K, Cygal M, Korga A, Dudka J. The beneficial effects of resveratrol on steatosis and mitochondrial oxidative stress in HepG2 cells. Can J Physiol Pharmacol 2017; 95:1442-1453. [DOI: 10.1139/cjpp-2016-0561] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is currently one of the most common chronic liver diseases, especially in developed countries. One group of substances with a potential use in the treatment of NAFLD are plant polyphenols, represented by resveratrol. The aim of this study was to evaluate the effect of resveratrol on steatosis and oxidative stress in HepG2 cells. The steatosis of cells was carried out using free fatty acids: oleic or palmitic acid and their mixtures. Steatosis was visualized using the intracellular lipid staining by Nile Red dye with a fluorescence microscope. This study also determined the viability of cells and mitochondrial membrane potential. The current study showed that fatty acids and their mixtures induced fat overloading in HepG2 cells. In the group of cells incubated with oleic acid (OA), observed changes were moderate with prevailing micro-vesicular steatosis. In case of cells incubated with palmitic acid (PA) and the mixtures of fatty acids, micro- and macro-vacuolar steatosis occurred in most of the cells. Resveratrol decreased steatosis in HepG2 cells induced by OA, PA, as well as their mixtures, and in most of experimental groups did not reduce cells viability. Resveratrol reduced the oxidative stress in HepG2 cells treated with fatty acids mixtures.
Collapse
Affiliation(s)
- Magdalena Izdebska
- Department of Toxicology, Medical University of Lublin, Chodzki 8, Lublin 20-093, Poland
| | | | - Agnieszka Korolczuk
- Department of Clinical Pathomorphology, Medical University of Lublin, Jaczewskiego 8b, Lublin 20-090, Poland
| | - Mariola Herbet
- Department of Toxicology, Medical University of Lublin, Chodzki 8, Lublin 20-093, Poland
| | | | - Renata Gieroba
- Medical Biology Unit, Medical University of Lublin, Jaczewskiego 8b, Lublin 20-090, Poland
| | - Marcin Sysa
- Department of Toxicology, Medical University of Lublin, Chodzki 8, Lublin 20-093, Poland
| | | | - Marcelina Cygal
- Department of Toxicology, Medical University of Lublin, Chodzki 8, Lublin 20-093, Poland
| | - Agnieszka Korga
- Department of Toxicology, Medical University of Lublin, Chodzki 8, Lublin 20-093, Poland
| | - Jarosław Dudka
- Department of Toxicology, Medical University of Lublin, Chodzki 8, Lublin 20-093, Poland
| |
Collapse
|
105
|
Auguet T, Aragonès G, Berlanga A, Martínez S, Sabench F, Binetti J, Aguilar C, Porras JA, Molina A, Del Castillo D, Richart C. Hepcidin in morbidly obese women with non-alcoholic fatty liver disease. PLoS One 2017; 12:e0187065. [PMID: 29065180 PMCID: PMC5655438 DOI: 10.1371/journal.pone.0187065] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/12/2017] [Indexed: 02/07/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in Western countries. Both iron and lipid metabolism seem to be involved in its pathogenesis. We aimed to assess the relationship between levels of hepcidin, the master iron-regulatory protein, in plasma and the presence of NAFLD in morbidly obese (MO) patients, and to investigate the association between the hepatic expression of the main iron and lipid metabolism -related genes. Materials and methods Enzyme-linked immunosorbent assay was used to measure plasma hepcidin levels in 49 normal-weight control women, 23 MO women with normal liver (NL) histology and 46 MO women with NAFLD. The mRNA expression of hepcidin, the main iron metabolism-related genes, and the main lipid-metabolism genes was quantified by qRT-PCR in liver biopsies from members of the MO group undergoing bariatric surgery. Results Circulating hepcidin levels were significantly greater in MO than in normal-weight control women. However, there were no significant differences between MO women with NL and those with NAFLD. PCR analysis showed increased expression of hepcidin, FPN1, TfR1 and TfR2 in the liver of MO NAFLD women compared to those with NL. Moreover, a positive association of hepatic hepcidin mRNA expression and the iron metabolism-related genes was found with some key genes involved in the lipid metabolism. Conclusion These findings suggest that circulating hepcidin levels are associated with obesity but not with the presence of NAFLD. However, the hepatic expression of hepcidin and the iron metabolism-related genes seem to play a role in regulating lipid metabolism pathways in liver, which has implications for NAFLD pathogenesis.
Collapse
Affiliation(s)
- 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), Tarragona, Spain
- Servei de Medicina Interna, Hospital Universitari Joan XXIII, Tarragona, Spain
- * E-mail: (TA); (CR)
| | - 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), 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), Tarragona, Spain
| | - Salomé Martínez
- Servei d’ Anatomia Patològica, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Fàtima Sabench
- Servei de Cirurgia, Hospital Universitari Sant Joan, Reus, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili (URV), IISPV, Reus, Spain
| | - Jessica Binetti
- Servei de Medicina Interna, Hospital Universitari Joan XXIII, Tarragona, 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), Tarragona, Spain
| | - José Antonio Porras
- Servei de Medicina Interna, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Alicia Molina
- Servei de Cirurgia, Hospital Universitari Sant Joan, Reus, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili (URV), IISPV, Reus, Spain
| | - Daniel Del Castillo
- Servei de Cirurgia, Hospital Universitari Sant Joan, Reus, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili (URV), IISPV, 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), Tarragona, Spain
- Servei de Medicina Interna, Hospital Universitari Joan XXIII, Tarragona, Spain
- * E-mail: (TA); (CR)
| |
Collapse
|
106
|
Arguin G, Bourzac JF, Placet M, Molle CM, Paquette M, Beaudoin JF, Rousseau JA, Lecomte R, Plourde M, Gendron FP. The loss of P2X7 receptor expression leads to increase intestinal glucose transit and hepatic steatosis. Sci Rep 2017; 7:12917. [PMID: 29018292 PMCID: PMC5635021 DOI: 10.1038/s41598-017-13300-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/22/2017] [Indexed: 12/25/2022] Open
Abstract
In intestinal epithelial cells (IEC), it was reported that the activation of the P2X7 receptor leads to the internalization of the glucose transporter GLUT2, which is accompanied by a reduction of IEC capacity to transport glucose. In this study, we used P2rx7 -/- mice to decipher P2X7 functions in intestinal glucose transport and to evaluate the impacts on metabolism. Immunohistochemistry analyses revealed the presence of GLUT2 at the apical domain of P2rx7 -/- jejunum enterocytes. Positron emission tomography and biodistribution studies demonstrated that glucose was more efficiently delivered to the circulation of knockout animals. These findings correlated with increase blood glucose, insulin, triglycerides and cholesterol levels. In fact, P2rx7 -/- mice had increased serum triglyceride and cholesterol levels and displayed glucose intolerance and resistance to insulin. Finally, P2rx7 -/- mice developed a hepatic steatosis characterized by a reduction of Acaca, Acacb, Fasn and Acox1 mRNA expression, as well as for ACC and FAS protein expression. Our study suggests that P2X7 could play a central role in metabolic diseases.
Collapse
Affiliation(s)
- Guillaume Arguin
- Department of Anatomy and Cell Biology, Pavillon de Recherche Appliquée sur le Cancer, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-François Bourzac
- Department of Anatomy and Cell Biology, Pavillon de Recherche Appliquée sur le Cancer, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Morgane Placet
- Department of Anatomy and Cell Biology, Pavillon de Recherche Appliquée sur le Cancer, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Caroline M Molle
- Department of Anatomy and Cell Biology, Pavillon de Recherche Appliquée sur le Cancer, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Michel Paquette
- Sherbrooke Molecular Imaging Centre of CRCHUS and Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-François Beaudoin
- Sherbrooke Molecular Imaging Centre of CRCHUS and Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jacques A Rousseau
- Sherbrooke Molecular Imaging Centre of CRCHUS and Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Roger Lecomte
- Sherbrooke Molecular Imaging Centre of CRCHUS and Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mélanie Plourde
- Department of Medicine, Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Fernand-Pierre Gendron
- Department of Anatomy and Cell Biology, Pavillon de Recherche Appliquée sur le Cancer, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
107
|
Vecchione G, Grasselli E, Cioffi F, Baldini F, Oliveira PJ, Sardão VA, Cortese K, Lanni A, Voci A, Portincasa P, Vergani L. The Nutraceutic Silybin Counteracts Excess Lipid Accumulation and Ongoing Oxidative Stress in an In Vitro Model of Non-Alcoholic Fatty Liver Disease Progression. Front Nutr 2017; 4:42. [PMID: 28971098 PMCID: PMC5609553 DOI: 10.3389/fnut.2017.00042] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/30/2017] [Indexed: 01/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major cause of liver-related morbidity and mortality. Oxidative stress and release of pro-inflammatory cytokines, such as tumor necrosis factor α (TNFα), are major consequences of hepatic lipid overload, which can contribute to progression of NAFLD to non-alcoholic steatohepatitis (NASH). Also, mitochondria are involved in the NAFLD pathogenesis for their role in hepatic lipid metabolism. Definitive treatments for NAFLD/NASH are lacking so far. Silybin, the extract of the milk thistle seeds, has previously shown beneficial effects in NAFLD. Sequential exposure of hepatocytes to high concentrations of fatty acids (FAs) and TNFα resulted in fat overload and oxidative stress, which mimic in vitro the progression of NAFLD from simple steatosis (SS) to steatohepatitis (SH). The exposure to 50 µM silybin for 24 h reduced fat accumulation in the model of NAFLD progression. The in vitro progression of NAFLD from SS to SH resulted in reduced hepatocyte viability, increased apoptosis and oxidative stress, reduction in lipid droplet size, and up-regulation of IκB kinase β-interacting protein and adipose triglyceride lipase expressions. The direct action of silybin on SS or SH cells and the underlying mechanisms were assessed. Beneficial action of silybin was sustained by changes in expression/activity of peroxisome proliferator-activated receptors and enzymes for FA oxidation. Moreover, silybin counteracted the FA-induced mitochondrial damage by acting on complementary pathways: (i) increased the mitochondrial size and improved the mitochondrial cristae organization; (ii) stimulated mitochondrial FA oxidation; (iii) reduced basal and maximal respiration and ATP production in SH cells; (iv) stimulated ATP production in SS cells; and (v) rescued the FA-induced apoptotic signals and oxidative stress in SH cells. We provide new insights about the direct protective effects of the nutraceutic silybin on hepatocytes mimicking in vitro NAFLD progression.
Collapse
Affiliation(s)
- Giulia Vecchione
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Elena Grasselli
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Federica Cioffi
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Francesca Baldini
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Paulo J Oliveira
- Center for Neuroscience and Cellular Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Vilma A Sardão
- Center for Neuroscience and Cellular Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Katia Cortese
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy
| | - Antonia Lanni
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Adriana Voci
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Laura Vergani
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| |
Collapse
|
108
|
Senese R, Cioffi F, de Lange P, Leanza C, Iannucci LF, Silvestri E, Moreno M, Lombardi A, Goglia F, Lanni A. Both 3,5-Diiodo-L-Thyronine and 3,5,3'-Triiodo-L-Thyronine Prevent Short-term Hepatic Lipid Accumulation via Distinct Mechanisms in Rats Being Fed a High-Fat Diet. Front Physiol 2017; 8:706. [PMID: 28959215 PMCID: PMC5603695 DOI: 10.3389/fphys.2017.00706] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/31/2017] [Indexed: 12/14/2022] Open
Abstract
3,3′,5-triiodo-L-thyronine (T3) improves hepatic lipid accumulation by increasing lipid catabolism but it also increases lipogenesis, which at first glance appears contradictory. Recent studies have shown that 3,5-diiodothyronine (T2), a natural thyroid hormone derivative, also has the capacity to stimulate hepatic lipid catabolism, however, little is known about its possible effects on lipogenic gene expression. Because genes classically involved in hepatic lipogenesis such as SPOT14, acetyl-CoA-carboxylase (ACC), and fatty acid synthase (FAS) contain thyroid hormone response elements (TREs), we studied their transcriptional regulation, focusing on TRE-mediated effects of T3 compared to T2 in rats receiving high-fat diet (HFD) for 1 week. HFD rats showed a marked lipid accumulation in the liver, which was significantly reduced upon simultaneous administration of either T3 or T2 with the diet. When administered to HFD rats, T2, in contrast with T3, markedly downregulated the expression of the above-mentioned genes. T2 downregulated expression of the transcription factors carbohydrate-response element-binding protein (ChREBP) and sterol regulatory element binding protein-1c (SREBP-1c) involved in activation of transcription of these genes, which explains the suppressed expression of their target genes involved in lipogenesis. T3, however, did not repress expression of the TRE-containing ChREBP gene but repressed SREBP-1c expression. Despite suppression of SREBP-1c expression by T3 (which can be explained by the presence of nTRE in its promoter), the target genes were not suppressed, but normalized to HFD reference levels or even upregulated (ACC), partly due to the presence of TREs on the promoters of these genes and partly to the lack of suppression of ChREBP. Thus, T2 and T3 probably act by different molecular mechanisms to achieve inhibition of hepatic lipid accumulation.
Collapse
Affiliation(s)
- Rosalba Senese
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| | - Federica Cioffi
- Dipartimento di Scienze e Tecnologie, Università degli Studi del SannioBenevento, Italy
| | - Pieter de Lange
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| | - Cristina Leanza
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| | - Liliana F Iannucci
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| | - Elena Silvestri
- Dipartimento di Scienze e Tecnologie, Università degli Studi del SannioBenevento, Italy
| | - Maria Moreno
- Dipartimento di Scienze e Tecnologie, Università degli Studi del SannioBenevento, Italy
| | - Assunta Lombardi
- Dipartimento di Biologia, Università degli Studi di Napoli Federico IINaples, Italy
| | - Fernando Goglia
- Dipartimento di Scienze e Tecnologie, Università degli Studi del SannioBenevento, Italy
| | - Antonia Lanni
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli" CasertaCaserta, Italy
| |
Collapse
|
109
|
Li J, Verhaar AP, Pan Q, de Knegt RJ, Peppelenbosch MP. Serum levels of caspase-cleaved cytokeratin 18 (CK18-Asp396) predict severity of liver disease in chronic hepatitis B. Clin Exp Gastroenterol 2017; 10:203-209. [PMID: 28860836 PMCID: PMC5560566 DOI: 10.2147/ceg.s135526] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background and aim Caspase-cleaved cytokeratin 18 (CK18-Asp396) is a potential clinically useful biomarker in liver disease as it is released from hepatocytes during apoptosis. In this study, we investigated serum CK18-Asp396 levels in chronic hepatitis B (CHB). Patients and methods Overall, 163 patients with CHB were included. Serum CK18-Asp396 levels were determined by enzyme-linked immunosorbent assay (ELISA), and results were related to steatosis grade, histological activity index, inflammation score, and METAVIR fibrosis grade as well as to viral load, serum levels of liver enzymes, and albumin. Receiver operating characteristic analysis was used to evaluate the diagnostic performance of serum CK18-Asp396 levels for assessing disease activity. Results A higher level of serum CK 18 concentrations was found in patients with significant inflammation vs no significant inflammation (378.5 [interquartile range {IQR}: 173.2–629.6] vs 137.3 [87.5–197.7], P < 0.05; approximately threefold increase) and in patients with significant fibrosis vs no significant fibrosis (177.8 [IQR: 120.8–519.1] vs 142.7 [IQR: 88.8–214.4], P < 0.05; 1.25-fold increase). There was no differential CK 18 level by degree of steatosis. CK 18 was an independent predictor of significant inflammation with an 82% specificity and a 94% negative predictive value. We found the strongest correlation of CK 18 with alanine aminotransferase and aspartate aminotransferase (both r = 0.52; P < 0.001), but less with albumin (r = −0.24; P < 0.05) and viral load (log) (r = 0.19; P < 0.05). Conclusion CHB appears to be accompanied by continuous high levels of hepatocyte apoptosis as judged from serum CK 18, suggesting that elimination of the infected compartment constitutes a defensive strategy against disease. Accordingly, CK 18 works as an independent predictor of significant inflammation with a high specificity.
Collapse
Affiliation(s)
- Juan Li
- Erasmus MC Cancer Institute, Erasmus MC - University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Auke P Verhaar
- Erasmus MC Cancer Institute, Erasmus MC - University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Qiuwei Pan
- Erasmus MC Cancer Institute, Erasmus MC - University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Robert Jacobus de Knegt
- Erasmus MC Cancer Institute, Erasmus MC - University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maikel P Peppelenbosch
- Erasmus MC Cancer Institute, Erasmus MC - University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
110
|
Komada H, Sakaguchi K, Hirota Y, Sou A, Nakamura T, Kyotani K, Kawamitsu H, Sugimura K, Okuno Y, Ogawa W. Pancreatic fat content assessed by 1 H magnetic resonance spectroscopy is correlated with insulin resistance, but not with insulin secretion, in Japanese individuals with normal glucose tolerance. J Diabetes Investig 2017; 9:505-511. [PMID: 28766895 PMCID: PMC5934259 DOI: 10.1111/jdi.12720] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/23/2017] [Accepted: 07/26/2017] [Indexed: 12/18/2022] Open
Abstract
AIMS/INTRODUCTION Whereas some clinical studies have shown that excessive fat accumulation in the pancreas is associated with impairment of insulin secretion, others have not found such an association. 1 H magnetic resonance spectroscopy allows quantitative fat analysis in various tissues including the pancreas. The pathological relevance of pancreatic fat content (PFC) in Japanese individuals remains unclear, however. MATERIALS AND METHODS We analyzed PFC in 30 Japanese individuals with normal glucose tolerance by 1 H magnetic resonance spectroscopy, and then investigated the relationships between PFC and indexes of insulin secretion and insulin sensitivity-resistance determined by an oral glucose tolerance test. We also measured hepatic fat content and intramyocellular lipid content by 1 H magnetic resonance spectroscopy, as well as visceral fat area and subcutaneous fat area by magnetic resonance imaging, and we examined the relationships between these fat content measures and oral glucose tolerance test-derived parameters. RESULTS PFC was correlated with indexes of insulin sensitivity-resistance, but not with those of insulin secretion. Hepatic fat content and visceral fat area were correlated with similar sets of parameters as was PFC, whereas subcutaneous fat area was correlated with parameters of insulin secretion, and intramyocellular lipid content was not correlated with any of the measured parameters. The correlation between PFC and homeostasis model assessment of insulin resistance remained significant after adjustment for age, body mass index and sex. Among fat content measures, PFC was most highly correlated with hepatic fat content and visceral fat area. CONCLUSIONS PFC was correlated with indexes of insulin resistance, but not with those of insulin secretion in non-obese Japanese individuals with normal glucose tolerance.
Collapse
Affiliation(s)
- Hisako Komada
- Division of Diabetes and EndocrinologyDepartment of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Kazuhiko Sakaguchi
- Division of Diabetes and EndocrinologyDepartment of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Yushi Hirota
- Division of Diabetes and EndocrinologyDepartment of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Anna Sou
- Division of Diabetes and EndocrinologyDepartment of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Tomoaki Nakamura
- Division of Diabetes and EndocrinologyDepartment of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | | | | | - Kazuro Sugimura
- Department of RadiologyKobe University Graduate School of MedicineKobeJapan
| | - Yoko Okuno
- Division of Diabetes and EndocrinologyDepartment of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| | - Wataru Ogawa
- Division of Diabetes and EndocrinologyDepartment of Internal MedicineKobe University Graduate School of MedicineKobeJapan
| |
Collapse
|
111
|
Willy JA, Young SK, Mosley AL, Gawrieh S, Stevens JL, Masuoka HC, Wek RC. Function of inhibitor of Bruton's tyrosine kinase isoform α (IBTKα) in nonalcoholic steatohepatitis links autophagy and the unfolded protein response. J Biol Chem 2017; 292:14050-14065. [PMID: 28710282 DOI: 10.1074/jbc.m117.799304] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/03/2017] [Indexed: 01/15/2023] Open
Abstract
Nonalcoholic fatty liver disease (steatosis) is the most prevalent liver disease in the Western world. One of the advanced pathologies is nonalcoholic steatohepatitis (NASH), which is associated with induction of the unfolded protein response (UPR) and disruption of autophagic flux. However, the mechanisms by which these processes contribute to the pathogenesis of human diseases are unclear. Herein, we identify the α isoform of the inhibitor of Bruton's tyrosine kinase (IBTKα) as a member of the UPR, whose expression is preferentially translated during endoplasmic reticulum (ER) stress. We found that IBTKα is located in the ER and associates with proteins LC3b, SEC16A, and SEC31A and plays a previously unrecognized role in phagophore initiation from ER exit sites. Depletion of IBTKα helps prevent accumulation of autophagosome intermediates stemming from exposure to saturated free fatty acids and rescues hepatocytes from death. Of note, induction of IBTKα and the UPR, along with inhibition of autophagic flux, was associated with progression from steatosis to NASH in liver biopsies. These results indicate a function for IBTKα in NASH that links autophagy with activation of the UPR.
Collapse
Affiliation(s)
- Jeffrey A Willy
- From the Departments of Biochemistry and Molecular Biology and Indianapolis, Indiana 46202-5126
| | - Sara K Young
- From the Departments of Biochemistry and Molecular Biology and Indianapolis, Indiana 46202-5126
| | - Amber L Mosley
- From the Departments of Biochemistry and Molecular Biology and Indianapolis, Indiana 46202-5126
| | - Samer Gawrieh
- Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202-5126
| | - James L Stevens
- From the Departments of Biochemistry and Molecular Biology and Indianapolis, Indiana 46202-5126
| | - Howard C Masuoka
- From the Departments of Biochemistry and Molecular Biology and Indianapolis, Indiana 46202-5126
| | - Ronald C Wek
- From the Departments of Biochemistry and Molecular Biology and Indianapolis, Indiana 46202-5126.
| |
Collapse
|
112
|
Engin A. Non-Alcoholic Fatty Liver Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:443-467. [DOI: 10.1007/978-3-319-48382-5_19] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
113
|
Han J, Dzierlenga AL, Lu Z, Billheimer DD, Torabzadehkhorasani E, Lake AD, Li H, Novak P, Shipkova P, Aranibar N, Robertson D, Reily MD, Lehman-McKeeman LD, Cherrington NJ. Metabolomic profiling distinction of human nonalcoholic fatty liver disease progression from a common rat model. Obesity (Silver Spring) 2017; 25:1069-1076. [PMID: 28452429 PMCID: PMC5513172 DOI: 10.1002/oby.21855] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 03/24/2017] [Accepted: 03/25/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Characteristic pathological changes define the progression of steatosis to nonalcoholic steatohepatitis (NASH) and are correlated to metabolic pathways. A common rodent model of NASH is the methionine and choline deficient (MCD) diet. The objective of this study was to perform full metabolomic analyses on liver samples to determine which pathways are altered most pronouncedly in this condition in humans, and to compare these changes to rodent models of nonalcoholic fatty liver disease (NAFLD). METHODS A principal component analysis for all 91 metabolites measured indicated that metabolome perturbation is greater and less varied for humans than for rodents. RESULTS Metabolome changes in human and rat NAFLD were greatest for the amino acid and bile acid metabolite families (e.g., asparagine, citrulline, gamma-aminobutyric acid, lysine); although, in many cases, the trends were reversed when compared between species (cholic acid, betaine). CONCLUSIONS Overall, these results indicate that metabolites of specific pathways may be useful biomarkers for NASH progression, although these markers may not correspond to rodent NASH models. The MCD model may be useful when studying certain end points of NASH; however, the metabolomics results indicate important differences between humans and rodents in the biochemical pathogenesis of the disease.
Collapse
Affiliation(s)
- JianHua Han
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ, USA
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Department of Clinical Laboratory, Beijing, China
| | - Anika L. Dzierlenga
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ, USA
| | - Zhengqiang Lu
- The Arizona Statistical Consulting Laboratory, University of Arizona, Tucson, AZ, USA
| | - Dean D. Billheimer
- The Arizona Statistical Consulting Laboratory, University of Arizona, Tucson, AZ, USA
| | | | - April D. Lake
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ, USA
| | - Hui Li
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ, USA
| | - Petr Novak
- Biology Centre CAS, Institute of Plant Molecular Biology, Ceske Budejovice, Czech Republic
| | - Petia Shipkova
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Princeton, NJ, USA
| | - Nelly Aranibar
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Princeton, NJ, USA
| | - Donald Robertson
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Princeton, NJ, USA
| | - Michael D. Reily
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Princeton, NJ, USA
| | | | | |
Collapse
|
114
|
Amirkalali B, Sohrabi MR, Rahimi-Moghaddam P, Esrafily A, Gholami A, Hosseinzadeh P, Keyvani H, Shidfar F, Zamani F. The Association of Nicotinamide Phosphoribosyltransferase Polymorphism with Markers of Hepatic Injury and De Novo Lipogenesis in Nonalcoholic Fatty Liver Disease. HEPATITIS MONTHLY 2017; 17. [DOI: 10.5812/hepatmon.13570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
115
|
Janssens S, Ciapaite J, Wolters JC, van Riel NA, Nicolay K, Prompers JJ. An In Vivo Magnetic Resonance Spectroscopy Study of the Effects of Caloric and Non-Caloric Sweeteners on Liver Lipid Metabolism in Rats. Nutrients 2017; 9:nu9050476. [PMID: 28489050 PMCID: PMC5452206 DOI: 10.3390/nu9050476] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/26/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022] Open
Abstract
We aimed to elucidate the effects of caloric and non-caloric sweeteners on liver lipid metabolism in rats using in vivo magnetic resonance spectroscopy (MRS) and to determine their roles in the development of liver steatosis. Wistar rats received normal chow and either normal drinking water, or solutions containing 13% (w/v) glucose, 13% fructose, or 0.4% aspartame. After 7 weeks, in vivo hepatic dietary lipid uptake and de novo lipogenesis were assessed with proton-observed, carbon-13-edited MRS combined with 13C-labeled lipids and 13C-labeled glucose, respectively. The molecular basis of alterations in hepatic liver metabolism was analyzed in detail ex vivo using immunoblotting and targeted quantitative proteomics. Both glucose and fructose feeding increased adiposity, but only fructose induced hepatic lipid accumulation. In vivo MRS showed that this was not caused by increased hepatic uptake of dietary lipids, but could be attributed to an increase in de novo lipogenesis. Stimulation of lipogenesis by fructose was confirmed by a strong upregulation of lipogenic enzymes, which was more potent than with glucose. The non-caloric sweetener aspartame did not significantly affect liver lipid content or metabolism. In conclusion, liquid fructose more severely affected liver lipid metabolism in rats than glucose, while aspartame had no effect.
Collapse
Affiliation(s)
- Sharon Janssens
- Biomedical Nuclear Magnetic Resonance (NMR), Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Jolita Ciapaite
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Justina C Wolters
- Systems Biology Centre for Energy Metabolism and Ageing, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
- Department of Pharmacy, Analytical Biochemistry, University of Groningen, Antonius Deusinglaan, 9713 AV Groningen, The Netherlands.
| | - Natal A van Riel
- Computational Biology, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Klaas Nicolay
- Biomedical Nuclear Magnetic Resonance (NMR), Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Jeanine J Prompers
- Biomedical Nuclear Magnetic Resonance (NMR), Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.
| |
Collapse
|
116
|
Pakravan H, Ahmadian M, Fani A, Aghaee D, Brumanad S, Pakzad B. The Effects of Melatonin in Patients with Nonalcoholic Fatty Liver Disease: A Randomized Controlled Trial. Adv Biomed Res 2017; 6:40. [PMID: 28503495 PMCID: PMC5414412 DOI: 10.4103/2277-9175.204593] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: This study was designed to evaluate the effect of melatonin on nonalcoholic fatty liver disease (NAFLD) in compared to placebo. Materials and Methods: A total of 100 patients with histopathological diagnosis NAFLD in two groups of case and control received oral melatonin or placebo thrice daily for 3 months. Collected data were weight, waist, systolic blood pressure (SBP), diastolic blood pressure (DBP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), high sensitive C-reactive protein (hsCRP), fatty liver grade, and side effects which were measured at baseline and after treatment period using standard clinical chemistry techniques. Results: Before treatment the mean of weight, waist, SBP, DBP, ALT, AST, and hsCRP between cases and controls were similar (P > 0.5). After treatment, only the differences in the mean of hsCRP in cases was significantly lower than controls (P = 0.003). In case group, all variables after treatment were significantly decreased compare to baseline (P > 0.5) and only AST after treatment was similar to before treatment (P > 0.5). The mean of a decrease in the level of weight, waist, SBP, and ALT were not statistically significant between groups (P > 0.5). In the case group in compare to control group the level of DBP, AST, and hsCRP significantly more decreased. After treatment fatty, liver grade was statistically improved in more cases than controls (P = 0.001). Side effects were similar between the two groups. Conclusion: Melatonin significantly decreases liver enzymes, so the use of melatonin in patients with NAFLD can be effective.
Collapse
Affiliation(s)
- Hassan Pakravan
- Department of Internal Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Ahmadian
- Department of Gastroenterology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Fani
- Department of Internal Medicine, Arak University of Medical, Arak, Iran
| | - Davood Aghaee
- Department of Internal Medicine, Arak University of Medical, Arak, Iran
| | - Sareh Brumanad
- Department of Radiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahram Pakzad
- Department of Internal Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
117
|
Choi Y, Abdelmegeed MA, Song BJ. Diet high in fructose promotes liver steatosis and hepatocyte apoptosis in C57BL/6J female mice: Role of disturbed lipid homeostasis and increased oxidative stress. Food Chem Toxicol 2017; 103:111-121. [PMID: 28257781 DOI: 10.1016/j.fct.2017.02.039] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/17/2017] [Accepted: 02/28/2017] [Indexed: 02/06/2023]
Abstract
The effects of high (H)-fructose (FR) diet (D) (HFRD) on hepatic lipid homeostasis, oxidative stress, inflammation and hepatocyte apoptosis were investigated in 6-week old female C57BL/6J mice fed a regular chow (ContD) or HFRD (35% fructose-derived calories) for 3 weeks. HFRD-fed mice exhibited increased levels of hepatic steatosis with a significant elevation of serum levels of triglyceride, cholesterol and TNFα compared to ContD-fed mice (P<0.05). HFRD-fed mice exhibited ∼2.7- fold higher levels FAS along with significantly decreased protein levels of adiponection-R2 (∼30%), P-AMPK (∼60%), P-ACC (∼70%) and RXR-α (∼55%), suggesting decreased hepatic fat oxidation compared to controls. Interestingly, hepatic fatty acid uptake into hepatocytes and lipolysis were significantly increased in HFRD-fed mice, as shown by decreased CD36 and fatty acid transporter protein-2, and increased adipose triglyceride lipase, respectively (P<0.05). Increased hepatic levels of iNOS and GSSG/GSH suggest elevated oxidative stress with a higher number of macrophages in the adipose tissue in HFRD-fed mice (P<0.05). Significantly elevated rates of hepatocyte apoptosis (∼2.4-fold), as determined by TUNEL analysis with increased Bax/Bcl2 ratio and PARP-1 levels (∼2- and 1.5-fold, respectively), were observed in HFRD-fed mice. Thus, HFRD exposure increased hepatic steatosis accompanied by oxidative stress and inflammation, leading to hepatocyte apoptosis.
Collapse
Affiliation(s)
- Youngshim Choi
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Mohamed A Abdelmegeed
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| |
Collapse
|
118
|
Souza-Mello V. Hepatic structural enhancement and insulin resistance amelioration due to AT1 receptor blockade. World J Hepatol 2017; 9:74-79. [PMID: 28144388 PMCID: PMC5241531 DOI: 10.4254/wjh.v9.i2.74] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/18/2016] [Accepted: 11/22/2016] [Indexed: 02/06/2023] Open
Abstract
Over the last decade, the role of renin-angiotensin system (RAS) on the development of obesity and its comorbidities has been extensively addressed. Both circulating and local RAS components are up-regulated in obesity and involved in non-alcoholic fatty liver disease onset. Pharmacological manipulations of RAS are viable strategies to tackle metabolic impairments caused by the excessive body fat mass. Renin inhibitors rescue insulin resistance, but do not have marked effects on hepatic steatosis. However, angiotensin-converting enzyme inhibitors and angiotensin receptor blockers (ARB) yield beneficial hepatic remodeling. ARBs elicit body mass loss and normalize insulin levels, tackling insulin resistance. Also, this drug class increases adiponectin levels, besides countering interleukin-6, tumoral necrosis factor-alpha, and transforming growth factor-beta 1. The latter is essential to prevent from liver fibrosis. When conjugated with peroxisome proliferator-activated receptor (PPAR)-alpha activation, ARB fully rescues fatty liver. These effects might be orchestrated by an indirect up-regulation of MAS receptor due to angiotensin II receptor type 1 (AT1R) blockade. These associations of ARB with PPAR activation and ACE2-angiotensin (ANG) (1-7)-MAS receptor axis deserve a better understanding. This editorial provides a brief overview of the current knowledge regarding AT1R blockade effects on sensitivity to insulin and hepatic structural alterations as well as the intersections of AT1R blockade with peroxisome proliferator-activated receptor activation and ACE2-ANG (1-7) - MAS receptor axis.
Collapse
|
119
|
Gu JJ, Yao M, Yang J, Cai Y, Zheng WJ, Wang L, Yao DB, Yao DF. Mitochondrial carnitine palmitoyl transferase-II inactivity aggravates lipid accumulation in rat hepatocarcinogenesis. World J Gastroenterol 2017; 23:256-264. [PMID: 28127199 PMCID: PMC5236505 DOI: 10.3748/wjg.v23.i2.256] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/29/2016] [Accepted: 10/31/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the dynamic alteration of mitochondrial carnitine palmitoyl transferase II (CPT-II) expression during malignant transformation of rat hepatocytes.
METHODS Sprague-Dawley male rats were fed with normal, high fat (HF), and HF containing 2-fluorenylacetamide (2-FAA) diet, respectively. According to the Hematoxylin and Eosin staining of livers, rats were divided into control, fatty liver, degeneration, precancerous, and cancerous groups. Liver lipids were dyed with Oil Red O, CPT-II alterations were analyzed by immunohistochemistry, and compared with CPT-II specific concentration (μg/mg protein). Levels of total cholesterol (Tch), triglyceride (TG), and amino-transferases [alanine aminotransferase (ALT), aspartate aminotransferase (AST)] were determined by the routine methods.
RESULTS After intake of HF and/or HF+2-FAA diets, the rat livers showed mass lipid accumulation. The lipid level in the control group was significantly lower than that in other groups. The changes of serum TG and Tch levels were abnormally increasing, 2-3 times more than those in the controls (P < 0.05). During the rat liver morphological changes from normal to cancer development process with hepatocyte injury, serum AST and ALT levels were significantly higher (4-8 times, P < 0.05) than those in the control group. The specific concentration of CPT-II in liver tissues progressively decreased during hepatocyte malignant transformation, with the lowest CPT-II levels in the cancer group than in any of the other groups (P < 0.05).
CONCLUSION Low CPT-II expression might lead to abnormal hepatic lipid accumulation, which should promote the malignant transformation of hepatocytes.
Collapse
|
120
|
Park CH, Kim JH, Lee EB, Hur W, Kwon OJ, Park HJ, Yoon SK. Aronia melanocarpa Extract Ameliorates Hepatic Lipid Metabolism through PPARγ2 Downregulation. PLoS One 2017; 12:e0169685. [PMID: 28081181 PMCID: PMC5230775 DOI: 10.1371/journal.pone.0169685] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 12/19/2016] [Indexed: 12/30/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a hepatic manifestation of metabolic syndrome. Studies have demonstrated that anthocyanin-rich foods may improve hyperlipidemia and ameliorate hepatic steatosis. Here, effects of Aronia melanocarpa (AM), known to be rich of anthocyanins, on hepatic lipid metabolism and adipogenic genes were determined. AM was treated to C57BL/6N mice fed with high fat diet (HFD) or to FL83B cells treated with free fatty acid (FFA). Changes in levels of lipids, enzymes and hormones were observed, and expressions of adipogenic genes involved in hepatic lipid metabolism were detected by PCR, Western blotting and luciferase assay. In mice, AM significantly reduced the body and liver weight, lipid accumulation in the liver, and levels of biochemical markers such as fatty acid synthase, hepatic triglyceride and leptin. Serum transaminases, indicators for hepatocyte injury, were also suppressed, while superoxide dismutase activity and liver antioxidant capacity were significantly increased. In FL83B cells, AM significantly reduced FFA-induced lipid droplet accumulation. Protein synthesis of an adipogenic transcription factor, peroxisome proliferator-activated receptor γ2 (PPARγ2) was inhibited in vivo. Furthermore, transcriptional activity of PPARγ2 was down-regulated in vitro, and mRNA expression of PPARγ2 and its downstream target genes, adipocyte protein 2 and lipoprotein lipase were down-regulated by AM both in vitro and in vivo. These results show beneficial effects of AM against hepatic lipid accumulation through the inhibition of PPARγ2 expression along with improvements in body weight, liver functions, lipid profiles and antioxidant capacity suggesting the potential therapeutic efficacy of AM on NAFLD.
Collapse
Affiliation(s)
- Chung-Hwa Park
- Division of Gastroenterology & Hepatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center (CULRC), The Catholic University of Korea, Seoul, Korea
| | - Jung-Hee Kim
- The Catholic University Liver Research Center (CULRC), The Catholic University of Korea, Seoul, Korea
- WHO Collaborating Center of Viral Hepatitis, The Catholic University of Korea, Seoul, Korea
| | - Eun Byul Lee
- The Catholic University Liver Research Center (CULRC), The Catholic University of Korea, Seoul, Korea
- WHO Collaborating Center of Viral Hepatitis, The Catholic University of Korea, Seoul, Korea
| | - Wonhee Hur
- The Catholic University Liver Research Center (CULRC), The Catholic University of Korea, Seoul, Korea
- WHO Collaborating Center of Viral Hepatitis, The Catholic University of Korea, Seoul, Korea
| | - Oh-Joo Kwon
- Department of Medical Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | - Seung Kew Yoon
- Division of Gastroenterology & Hepatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center (CULRC), The Catholic University of Korea, Seoul, Korea
- WHO Collaborating Center of Viral Hepatitis, The Catholic University of Korea, Seoul, Korea
- * E-mail:
| |
Collapse
|
121
|
Amirkalali B, Sohrabi MR, Esrafily A, Jalali M, Gholami A, Hosseinzadeh P, Keyvani H, Shidfar F, Zamani F. Association between Nicotinamide Phosphoribosyltransferase and de novo Lipogenesis in Nonalcoholic Fatty Liver Disease. Med Princ Pract 2017; 26:251-257. [PMID: 28092906 PMCID: PMC5588391 DOI: 10.1159/000455862] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 01/09/2017] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE This study explored the association between serum nicotinamide phosphoribosyltransferase (NAMPT) and hepatic de novo lipogenesis (DNL) in nonalcoholic fatty liver disease (NAFLD) and determined whether or not this association is sex dependent. SUBJECTS AND METHODS In this cross-sectional study, 62 consecutive patients (32 males, 30 females) with NAFLD were recruited. Serum NAMPT (by ELISA), palmitic acid, and the DNL index of erythrocyte membranes as markers of hepatic DNL (by gas chromatography) were analyzed. The controlled attenuation parameter (CAP) and body impedance analyzer were used to assess hepatic and body fat, respectively. Univariate and multiple linear regressions (to adjust for confounders) were used to analyze the association of serum NAMPT with palmitic acid, DNL index, CAP, and body fat. RESULTS The respective values of serum NAMPT (2.44 ± 1.03 vs. 2.45 ± 1.13 ng/mL, p = 0.98), DNL index (3.11 [2.60-3.71] vs. 3.05 [2.40-3.59], p = 0.90), and palmitic acid (20.55% [15.34-24.04] vs. 22.64% [21.15-25.95], p = 0.07) were not significantly different between men and women, but those of CAP (326 [300-340] vs. 300 [261.25-329], p = 0.002) and body fat (37.71 ± 3.80 vs. 26.60 ± 5.70, p < 0.001) were significantly higher in women. In women, serum NAMPT had a significant negative association with the DNL index (β = -0.56, p = 0.01). The DNL index also had a significant negative association with body fat (β = -0.46, p = 0.02). In men, the only significant association was the positive association between serum NAMPT and CAP (β = 0.35, p = 0.035). CONCLUSION Higher serum NAMPT in women was associated with a lower hepatic DNL index, while in men it was associated with higher hepatic fat and had no association with the DNL index. Therefore, the serum NAMPT level interpretation for NAFLD prognosis is probably sex dependent.
Collapse
Affiliation(s)
- Bahareh Amirkalali
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Reza Sohrabi
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences, Tehran, Iran
| | - Ali Esrafily
- Department of Environmental Health Engineering, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Jalali
- Department of Nutrition and Biochemistry, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Gholami
- Department of Public Health, School of Public Health, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Payam Hosseinzadeh
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Keyvani
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Farhad Zamani
- Gastrointestinal and Liver Disease Research Center (GILDRC), Iran University of Medical Sciences, Tehran, Iran
- *Farhad Zamani, Gastrointestinal and Liver Disease Research Center, Karim khan Boulevard, Behafarin Street, Firoozgar Hospital, Tehran 1593748711 (Iran), E-Mail
| |
Collapse
|
122
|
Banini BA, Sanyal AJ. Nonalcoholic Fatty Liver Disease: Epidemiology, Pathogenesis, Natural History, Diagnosis, and Current Treatment Options. ACTA ACUST UNITED AC 2016; 8:75-84. [PMID: 28670148 DOI: 10.4137/cmt.s18885] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is on the rise and has become a major etiology for chronic liver disease. It is frequently associated with obesity, insulin resistance, hypertension, and dyslipidemia and is considered the hepatic manifestation of metabolic syndrome. In this review, we present a summary of the epidemiology and pathogenesis of NAFLD, and discuss the clinical evaluation and stratification of NAFLD patients into low, intermediate, and high risk with respect to liver-related outcomes. While diet and exercise are the cornerstone of treatment in all patients, the low rate of adherence and inadequacy of these recommendations necessitate pharmacologic intervention, especially in intermediate- and high-risk patients. We discuss vitamin E and pioglitazone which are often used as first-line therapy by many practitioners, with pentoxifylline and liraglutide as backup agents. Several drugs are in advanced-phase clinical trials and will likely change the landscape for management of NAFLD in the very near future.
Collapse
Affiliation(s)
- Bubu A Banini
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| |
Collapse
|
123
|
Glycycoumarin inhibits hepatocyte lipoapoptosis through activation of autophagy and inhibition of ER stress/GSK-3-mediated mitochondrial pathway. Sci Rep 2016; 6:38138. [PMID: 27901086 PMCID: PMC5128870 DOI: 10.1038/srep38138] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/07/2016] [Indexed: 01/04/2023] Open
Abstract
Herbal medicine as an alternative approach in the treatment of disease has drawn growing attention. Identification of the active ingredient is needed for effective utilization of the herbal medicine. Licorice is a popular herbal plant that is widely used to treat various diseases including liver diseases. Glycycoumarin (GCM) is a representative of courmarin compounds isolated from licorice. In the present study, the protective effect of GCM on hepatocyte lipoapoptosis has been evaluated using both cell culture model of palmitate-induced lipoapoptosis and animal model of non-alcoholic steatohepatitis (NASH). The results demonstrated for the first time that GCM was highly effective in suppressing hepatocyte lipoapoptosis in both in vitro and in vivo. Mechanistically, GCM was able to re-activate the impaired autophagy by lipid metabolic disorders. In line with the activation of autophagy, ER stress-mediated JNK and mitochondrial apoptotic pathway activation was inhibited by GCM both in vitro and in vivo. In addition, inactivation of GSK-3 might also contribute to the protective effect of GCM on hepatocyte lipoapoptosis. Our findings supported GCM as a novel active component of licorice against non-alcoholic fatty liver disease (NAFLD).
Collapse
|
124
|
Serviddio G, Bellanti F, Villani R, Tamborra R, Zerbinati C, Blonda M, Ciacciarelli M, Poli G, Vendemiale G, Iuliano L. Effects of dietary fatty acids and cholesterol excess on liver injury: A lipidomic approach. Redox Biol 2016; 9:296-305. [PMID: 27639112 PMCID: PMC5026694 DOI: 10.1016/j.redox.2016.09.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/07/2016] [Accepted: 09/09/2016] [Indexed: 02/05/2023] Open
Abstract
Lipid accumulation is the hallmark of Non-alcoholic Fatty Liver Disease (NAFLD) and has been suggested to play a role in promoting fatty liver inflammation. Previous findings indicate that during oxidative stress conditions excess cholesterol autoxidizes to oxysterols. To date, the role of oxysterols and their potential interaction with fatty acids accumulation in NASH pathogenesis remains little investigated. We used the nutritional model of high fatty acids (HFA), high cholesterol (HCh) or high fat and high cholesterol (HFA+FCh) diets and explored by a lipidomic approach, the blood and liver distribution of fatty acids and oxysterols in response to dietary manipulation. We observed that HFA or HCh diets induced fatty liver without inflammation, which was otherwise observed only after supplementation of HFA+HCh. Very interestingly, the combination model was associated with a specific oxysterol fingerprint. The present work provides a complete analysis of the change in lipids and oxysterols profile induced by different lipid dietary model and their association with histological alteration of the liver. This study allows the generation of interesting hypotheses on the role of interaction of lipid and cholesterol metabolites in the liver injury during NAFLD development and progression. Moreover, the changes in the concentration and quality of oxysterols induced by a combination diet suggest a novel potential pathogenic mechanism in the progression from simple steatosis to steatohepatitis.
Collapse
Affiliation(s)
- Gaetano Serviddio
- CURE University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy.
| | - Francesco Bellanti
- CURE University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Rosanna Villani
- CURE University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Rosanna Tamborra
- CURE University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Chiara Zerbinati
- Laboratory of Vascular Biology and Mass Spectrometry, Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Maria Blonda
- CURE University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Marco Ciacciarelli
- Laboratory of Vascular Biology and Mass Spectrometry, Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Torino at San Luigi Gonzaga Hospital, 10043 Torino, Orbassano, Italy
| | - Gianluigi Vendemiale
- CURE University Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Luigi Iuliano
- Laboratory of Vascular Biology and Mass Spectrometry, Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| |
Collapse
|
125
|
Giles DA, Moreno-Fernandez ME, Divanovic S. IL-17 Axis Driven Inflammation in Non-Alcoholic Fatty Liver Disease Progression. Curr Drug Targets 2016; 16:1315-23. [PMID: 26028039 DOI: 10.2174/1389450116666150531153627] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/25/2015] [Indexed: 02/08/2023]
Abstract
Obesity is a primary risk factor for the development of non-alcoholic fatty liver disease (NAFLD). NAFLD, the most common chronic liver disease in the world, represents a spectrum of disorders that range from steatosis (NAFL) to steatohepatitis (NASH) to cirrhosis. It is anticipated that NAFLD will soon surpass chronic hepatitis C infection as the leading cause for needing liver transplantation. Despite its clinical and public health significance no specific therapies are available. Although the etiology of NAFLD is multifactorial and remains largely enigmatic, it is well accepted that inflammation is a central component of NAFLD pathogenesis. Despite the significance, critical immune mediators, loci of immune activation, the immune signaling pathways and the mechanism(s) underlying disease progression remain incompletely understood. Recent findings have focused on the role of Interleukin 17 (IL-17) family of proinflammatory cytokines in obesity and pathogenesis of obesity-associated sequelae. Notably, obesity favors a Th17 bias and is associated with increased IL-17A expression in both humans and mice. Further, in mice, IL-17 axis has been implicated in regulation of both obesity and NAFLD pathogenesis. However, despite these recent advances several important questions require further evaluation including: the relevant cellular source of IL-17A production; the critical IL- 17RA-expressing cell type; the critical liver infiltrating immune cells; and the underlying cellular effector mechanisms. Addressing these questions may aid in the identification and development of novel therapeutic targets for prevention of inflammation- driven NAFLD progression.
Collapse
Affiliation(s)
| | | | - Senad Divanovic
- Division of Immunobiology Cincinnati Children's Hospital Medical Center TCHRF - Location S, Room #S.5.409 3333 Burnet Avenue Cincinnati, Ohio 45229-3039 U.S.A.
| |
Collapse
|
126
|
Zhou J, Gao S, Chen J, Zhao R, Yang X. Maternal sodium butyrate supplement elevates the lipolysis in adipose tissue and leads to lipid accumulation in offspring liver of weaning-age rats. Lipids Health Dis 2016; 15:119. [PMID: 27449927 PMCID: PMC4957328 DOI: 10.1186/s12944-016-0289-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 07/12/2016] [Indexed: 01/22/2023] Open
Abstract
Background Sodium butyrate (SB) is reported to regulate lipid metabolism in mammals, and the relationship between maternal nutrition and offspring growth has drawn much attention in the last several years. Methods To elucidate the effects of maternal dietary SB supplementation on hepatic lipid metabolism in weaning rats, we fed 16 primiparous purebred female SD rats either a chow-diet or a 1 % sodium butyrate diet throughout pregnancy and lactation. At weaning age, samples of the maternal subcutaneous adipose tissue and offspring liver were taken. The serum indexes and expressions of proteins related to lipid metabolism were detected in the mother and offspring, respectively. Results The results showed that the maternal SB supplement increased the concentration of non-esterified fatty acid (NEFA) in the maternal and offspring serum (P < 0.05). Total cholesterol (Tch) increased significantly in the weaning-rat serum (P < 0.05). Maternal adipose tissue from the SB-supplemented rats showed higher content of protein G-coupled protein (GPR43) and protein kinase A (PKA) (P < 0.05). The expression of protein adipose triglyceride lipase (ATGL), and of total and phosphorylated hormone sensitive lipase (HSL), in the maternal adipose tissue increased significantly (P < 0.05) compared to the control group. However the proteins related to lipogenesis showed no significant changes. Moreover, the concentration of triglyceride in the offspring liver increased significantly, and this likely resulted from an increase in the levels of fatty acids binding protein (FABP) and fatty acid translocase (CD36) protein (P < 0.05). SB exposure during pregnancy and lactation increased the hepatic total cholesterol (Tch) content (P < 0.01), which was related to a significantly up-regulated offspring hepatic expression of low density lipoprotein receptor (LDLR) protein (P < 0.05). Conclusion These results indicate that a maternal SB supplement during pregnancy and the lactation period promotes maternal fat mobilization, which may result in fatty acid uptake and lipid accumulation in the liver of the offspring.
Collapse
Affiliation(s)
- Jiabin Zhou
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Shixing Gao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Jinglong Chen
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.,Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing, 210095, People's Republic of China
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
127
|
Gu J, Yao M, Yao D, Wang L, Yang X, Yao D. Nonalcoholic Lipid Accumulation and Hepatocyte Malignant Transformation. J Clin Transl Hepatol 2016; 4:123-30. [PMID: 27350942 PMCID: PMC4913080 DOI: 10.14218/jcth.2016.00010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/13/2016] [Accepted: 05/14/2016] [Indexed: 02/07/2023] Open
Abstract
Worldwide incidence of hepatocellular carcinoma (HCC) is steadily increasing, highlighting its status as a public health concern, particularly due to its significant association with other comorbidities, such as diabetes. However, nonalcoholic fatty liver disease (NAFLD) has emerged as a primary risk factor, with its own prevalence increasing in recent years, and it has gradually caught up with the historical primary etiological factors of infection with hepatitis B virus and hepatitis C virus, exposure to aflatoxin, or alcohol liver disease. The deeply worrisome aspects of all of these high risk factors, however, are their remarkable presence within populations. Systemic and genetic mechanisms involved in the malignant transformation of liver cells, as well as useful biomarkers of early stage HCC are being investigated. However, the exact mechanisms underlying the interrelation of NAFLD and HCC remain largely unknown. In this review, some of the recent advances in our understanding of liver lipid accumulation are summarized and discussed to provide insights into the relationship between NAFLD and hepatocyte malignant transformation.
Collapse
Affiliation(s)
- Juanjuan Gu
- Research Center of Clinical Medicine and Department of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Min Yao
- Department of Immunology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Dengbing Yao
- School of Life Sciences, Nantong University, Nantong, Jiangsu, China
| | - Li Wang
- Department of Medical Informatics, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xuli Yang
- Research Center of Clinical Medicine and Department of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Dengfu Yao
- Research Center of Clinical Medicine and Department of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- *Correspondence to: Professor Dengfu Yao, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China. Tel: +86-513-85052297, Fax: +86-513-85052554, E-mail:
| |
Collapse
|
128
|
Araújo S, Soares E Silva A, Gomes F, Ribeiro E, Oliveira W, Oliveira A, Lima I, Lima MDC, Pitta I, Peixoto C. Effects of the new thiazolidine derivative LPSF/GQ-02 on hepatic lipid metabolism pathways in non-alcoholic fatty liver disease (NAFLD). Eur J Pharmacol 2016; 788:306-314. [PMID: 27349145 DOI: 10.1016/j.ejphar.2016.06.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the most common manifestation of metabolic syndrome. One of its most important features is the accumulation of triglycerides in the hepatocyte cells. Thiazolidinediones (TZDs) act as insulin sensitizers and are used to treat patients with type 2 diabetes and other conditions that are resistant to insulin, such as hepatic steatosis. Controversially, TZDs are also associated with the development of cardiovascular events and liver problems. For this reason, new therapeutic strategies are necessary to improve liver function in patients with chronic liver diseases. The aim of the present study was to evaluate the effects of LPSF/GQ-02 on the liver lipid metabolism in a murine model of NAFLD. Eighty male LDLR-/- mice were divided into 3 groups: 1-fed with a high-fat diet (HFD); 2-HFD+Pioglitazone (20mg/kg/day); 3-HFD+LPSF/GQ-02 (30mg/kg/day). The experiments lasted 12 weeks and drugs were administered daily by gavage in the final four weeks. The liver was processed for optical microscopy, Oil Red O, immunohistochemistry, immunofluorescence and western blot analysis. LPSF/GQ-02 effectively decreased fat accumulation, increased the hepatic levels of p-AMPK, FoxO1, ATGL, p-ACC and PPARα, and reduced the expression of LXRα, SREBP-1c and ACC. These results suggest that LPSF/GQ-02 acts directly on the hepatic lipid metabolism through the activation of the PPAR-α/AMPK/FoxO1/ATGL lipolytic pathway, and the inhibition of the AMPK/LXR/SREBP-1c/ACC/FAS lipogenic pathway.
Collapse
Affiliation(s)
- Shyrlene Araújo
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Recife, Pernambuco, Brasil; Universidade Federal de Pernambuco, Recife, Pernambuco, Brasil.
| | - Amanda Soares E Silva
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Recife, Pernambuco, Brasil; Universidade Federal de Pernambuco, Recife, Pernambuco, Brasil
| | - Fabiana Gomes
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Recife, Pernambuco, Brasil; Universidade Federal de Pernambuco, Recife, Pernambuco, Brasil
| | - Edlene Ribeiro
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Recife, Pernambuco, Brasil; Universidade Federal de Pernambuco, Recife, Pernambuco, Brasil
| | - Wilma Oliveira
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Recife, Pernambuco, Brasil; Universidade Federal de Pernambuco, Recife, Pernambuco, Brasil
| | - Amanda Oliveira
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Recife, Pernambuco, Brasil; Universidade Federal de Pernambuco, Recife, Pernambuco, Brasil
| | - Ingrid Lima
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Recife, Pernambuco, Brasil; Universidade Federal de Pernambuco, Recife, Pernambuco, Brasil
| | - Maria do Carmo Lima
- Laboratório de Planejamento e Síntese de Fármacos, Universidade Federal de Pernambuco, Recife, Brasil
| | - Ivan Pitta
- Laboratório de Planejamento e Síntese de Fármacos, Universidade Federal de Pernambuco, Recife, Brasil
| | - Christina Peixoto
- Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), Recife, Pernambuco, Brasil.
| |
Collapse
|
129
|
Rial SA, Karelis AD, Bergeron KF, Mounier C. Gut Microbiota and Metabolic Health: The Potential Beneficial Effects of a Medium Chain Triglyceride Diet in Obese Individuals. Nutrients 2016; 8:nu8050281. [PMID: 27187452 PMCID: PMC4882694 DOI: 10.3390/nu8050281] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/05/2016] [Accepted: 05/09/2016] [Indexed: 02/07/2023] Open
Abstract
Obesity and associated metabolic complications, such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D), are in constant increase around the world. While most obese patients show several metabolic and biometric abnormalities and comorbidities, a subgroup of patients representing 3% to 57% of obese adults, depending on the diagnosis criteria, remains metabolically healthy. Among many other factors, the gut microbiota is now identified as a determining factor in the pathogenesis of metabolically unhealthy obese (MUHO) individuals and in obesity-related diseases such as endotoxemia, intestinal and systemic inflammation, as well as insulin resistance. Interestingly, recent studies suggest that an optimal healthy-like gut microbiota structure may contribute to the metabolically healthy obese (MHO) phenotype. Here, we describe how dietary medium chain triglycerides (MCT), previously found to promote lipid catabolism, energy expenditure and weight loss, can ameliorate metabolic health via their capacity to improve both intestinal ecosystem and permeability. MCT-enriched diets could therefore be used to manage metabolic diseases through modification of gut microbiota.
Collapse
Affiliation(s)
- Sabri Ahmed Rial
- BioMed Research Center, Biological Sciences Department, University of Quebec at Montreal, Montreal, QC H2X 1Y4, Canada.
| | - Antony D Karelis
- Department of Exercise Science, University of Quebec at Montreal, Montreal, QC H2X 1Y4, Canada.
| | - Karl-F Bergeron
- BioMed Research Center, Biological Sciences Department, University of Quebec at Montreal, Montreal, QC H2X 1Y4, Canada.
| | - Catherine Mounier
- BioMed Research Center, Biological Sciences Department, University of Quebec at Montreal, Montreal, QC H2X 1Y4, Canada.
| |
Collapse
|
130
|
Xiao J, Lv D, Zhao Y, Chen X, Song M, Liu J, Bei Y, Wang F, Yang W, Yang C. miR-149 controls non-alcoholic fatty liver by targeting FGF-21. J Cell Mol Med 2016; 20:1603-8. [PMID: 27061435 PMCID: PMC4956949 DOI: 10.1111/jcmm.12848] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/24/2016] [Indexed: 12/14/2022] Open
Abstract
Non‐alcoholic fatty liver disease (NAFLD), a lipid metabolism disorder characterized by the accumulation of intrahepatic fat, has emerged as a global public health problem. However, its underlying molecular mechanism remains unclear. We previously have found that miR‐149 was elevated in NAFLD induced by high‐fat diet mice model, whereas decreased by a 16‐week running programme. Here, we reported that miR‐149 was increased in HepG2 cells treated with long‐chain fatty acid (FFA). In addition, miR‐149 was able to promote lipogenesis in HepG2 cells in the absence of FFA treatment. Moreover, inhibition of miR‐149 was capable of inhibiting lipogenesis in HepG2 cells in the presence of FFA treatment. Meanwhile, fibroblast growth factor‐21 (FGF‐21) was identified as a target gene of miR‐149, which was demonstrated by the fact that miR‐149 could negatively regulate the protein expression level of FGF‐21, and FGF‐21 was also responsible for the effect of miR‐149 inhibitor in decreasing lipogenesis in HepG2 cells in the presence of FFA treatment. These data implicate that miR‐149 might be a novel therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Junjie Xiao
- Regeneration and Ageing Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai, China.,Innovative Drug Research Center of Shanghai University, Shanghai, China
| | - Dongchao Lv
- Regeneration and Ageing Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai, China.,Innovative Drug Research Center of Shanghai University, Shanghai, China
| | - Yingying Zhao
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyu Chen
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Meiyi Song
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingqi Liu
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yihua Bei
- Regeneration and Ageing Lab, Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai, China.,Innovative Drug Research Center of Shanghai University, Shanghai, China
| | - Fei Wang
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenzhuo Yang
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Changqing Yang
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
131
|
Xiong J, Wang K, He J, Zhang G, Zhang D, Chen F. TFE3 Alleviates Hepatic Steatosis through Autophagy-Induced Lipophagy and PGC1α-Mediated Fatty Acid β-Oxidation. Int J Mol Sci 2016; 17:387. [PMID: 26999124 PMCID: PMC4813243 DOI: 10.3390/ijms17030387] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 03/06/2016] [Accepted: 03/07/2016] [Indexed: 02/06/2023] Open
Abstract
Autophagy flux deficiency is closely related to the development of hepatic steatosis. Transcription factor E3 (TFE3) is reported to be a crucial gene that regulates autophagy flux and lysosome function. Therefore, we investigated the role of TFE3 in a cell model of hepatic steatosis. We constructed L02 hepatocyte lines that stably over-expressed or knocked down the expression of TFE3. Subsequently, the effects of TFE3 on hepatocellular lipid metabolism were determined by autophagy flux assay, lipid oil red O (ORO) staining, immunofluorescence staining, and mitochondrial β-oxidation assessment. Finally, we analyzed whether peroxisome proliferative activated receptor gamma coactivator 1α (PGC1α) was the potential target gene of TFE3 in the regulation of hepatic steatosis using a chromatin immunoprecipitation (CHIP) assay and a luciferase reporter system. We found that overexpression of TFE3 markedly alleviated hepatocellular steatosis. On the contrary, downregulation of TFE3 resulted in an aggravated steatosis. The mechanistic studies revealed that the TFE3-manipulated regulatory effects on hepatocellular steatosis are dependent on autophagy-induced lipophagy and PGC1α-mediated fatty acid β-oxidation because blocking these pathways with an Atg5 small interfering RNA (siRNA) or PGC1α siRNA dramatically blunted the TFE3-mediated regulation of steatosis. In conclusion, TFE3 gene provides a novel insight into the treatment of hepatic steatosis and other metabolic disease.
Collapse
Affiliation(s)
- Jie Xiong
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China.
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Kezhou Wang
- Department of Pathology and Pathophysiology, Dalian Medical University, Dalian 116044, China.
| | - Jiangping He
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China.
| | - Guangya Zhang
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China.
| | - Dandan Zhang
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China.
| | - Fengling Chen
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China.
| |
Collapse
|
132
|
Nati M, Haddad D, Birkenfeld AL, Koch CA, Chavakis T, Chatzigeorgiou A. The role of immune cells in metabolism-related liver inflammation and development of non-alcoholic steatohepatitis (NASH). Rev Endocr Metab Disord 2016; 17:29-39. [PMID: 26847547 DOI: 10.1007/s11154-016-9339-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The low grade inflammatory state present in obesity promotes the progression of Non-Alcoholic Fatty Liver Disease (NAFLD). In Non-Alcoholic Steatohepatitis (NASH), augmented hepatic steatosis is accompanied by aberrant intrahepatic inflammation and exacerbated hepatocellular injury. NASH is an important disorder and can lead to fibrosis, cirrhosis and even neoplasia. The pathology of NASH involves a complex network of mechanisms, including increased infiltration of different subsets of immune cells, such as monocytes, T-lymphocytes and neutrophils, to the liver, as well as activation and in situ expansion of liver resident cells such as Kupffer cells or stellate cells. In this review, we summarize recent advances regarding understanding the role of the various cells of the innate and adaptive immunity in NASH development and progression, and discuss possible future therapeutic options and tools to interfere with disease progression.
Collapse
Affiliation(s)
- Marina Nati
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, MTZ, Fiedlerstrasse 42, 01307, Dresden, Germany
| | - David Haddad
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, MTZ, Fiedlerstrasse 42, 01307, Dresden, Germany
| | - Andreas L Birkenfeld
- Section of Metabolic Vascular Medicine, Medical Clinic III, Faculty of Medicine, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Division of Diabetes and Nutritional Sciences, Rayne Institute, King's College London, London, UK
| | - Christian A Koch
- Division of Endocrinology, Endocrine Tumor Program, Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, MTZ, Fiedlerstrasse 42, 01307, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Antonios Chatzigeorgiou
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, MTZ, Fiedlerstrasse 42, 01307, Dresden, Germany.
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany.
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, Dresden, Germany.
| |
Collapse
|
133
|
Mateos-Muñoz B, García-Martín E, Torrejón MJ, Devesa-Medina MJ, Esguevillas G, Cárdenas MC, Fernández C, Carballo M, Agúndez JA, Ladero JM. GC Gene Polymorphism and Unbound Serum Retinol-Binding Protein 4 Are Related to the Risk of Insulin Resistance in Patients With Chronic Hepatitis C: A Prospective Cross-Sectional Study. Medicine (Baltimore) 2016; 95:e3019. [PMID: 26962819 PMCID: PMC4998900 DOI: 10.1097/md.0000000000003019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Insulin resistance (IR) is found in chronic hepatitis C (CHC) more frequently than in other chronic liver diseases.Prospective cross-sectional study to evaluate a wide multitest panel to identify factors related with IR in CHC and their possible interactions.In 76 patients with CHC we performed a series of routine laboratory analysis as well as specifically designed serum biochemical tests [retinol, retinol-binding protein 4 (RBP4), 25-OH vitamin D, Vitamin E, lipopolysaccharide-binding protein (LBP), interleukin-6 (IL-6), and cystatin C]. The single nucleotide polymorphisms rs7041 and rs4588 GC-DBP (group-specific component-Vitamin D-binding protein), rs738409 PNPLA3 (patatin-like phospholipase domain containing 3), and rs12979860 IL28B (interleukin-28 B) genes were determined. Insulin sensitivity was established with the HOMA-IR and IR was diagnosed when HOMA-IR > 3. Fibrosis staging was assessed with liver biopsy or transient elastography.After backward logistic regression analysis, independent variables associated with IR were Gc1s/Gc1s DBP phenotype, that results from the homozygous carriage of the rs7041G/rs4588C haplotype (P = 0.033); low retinol/RBP4 ratio, reflecting a greater rate of unbound RBP4 (P = 0.005); older age (P = 0.01); high serum tryglicerides (P = 0.026); and advanced (F3-F4) fibrosis stage. The AUROC provided by the multivariate model was 0.950 (95% CI = 0.906-0.993).In addition to previously known ones, the Gc1s/Gc1s phenotype variant of DBP and the unbound fraction of plasma RBP4 may be considered as factors related with the incidence, and possibly the risk, of IR in CHC patients.
Collapse
Affiliation(s)
- Beatriz Mateos-Muñoz
- From the Services of Gastroenterology (BM-M, MJD-M, JML), Clinical Laboratory (MJT, MCC), and Clinical Epidemiology (CE), Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain (CF), Department of Pharmacology, Universidad de Extremadura (EG-M, GEN, JAA), Laboratory of Molecular Genetics, Hospital de Terrassa, Terrassa, Barcelona, Spain (MC), and Department of Medicine, Universidad Complutense, Madrid, Spain (MJD-M, JML)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Tarry-Adkins JL, Fernandez-Twinn DS, Hargreaves IP, Neergheen V, Aiken CE, Martin-Gronert MS, McConnell JM, Ozanne SE. Coenzyme Q10 prevents hepatic fibrosis, inflammation, and oxidative stress in a male rat model of poor maternal nutrition and accelerated postnatal growth. Am J Clin Nutr 2016; 103:579-88. [PMID: 26718412 PMCID: PMC4733260 DOI: 10.3945/ajcn.115.119834] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/11/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND It is well established that low birth weight and accelerated postnatal growth increase the risk of liver dysfunction in later life. However, molecular mechanisms underlying such developmental programming are not well characterized, and potential intervention strategies are poorly defined. OBJECTIVES We tested the hypotheses that poor maternal nutrition and accelerated postnatal growth would lead to increased hepatic fibrosis (a pathological marker of liver dysfunction) and that postnatal supplementation with the antioxidant coenzyme Q10 (CoQ10) would prevent this programmed phenotype. DESIGN A rat model of maternal protein restriction was used to generate low-birth-weight offspring that underwent accelerated postnatal growth (termed "recuperated"). These were compared with control rats. Offspring were weaned onto standard feed pellets with or without dietary CoQ10 (1 mg/kg body weight per day) supplementation. At 12 mo, hepatic fibrosis, indexes of inflammation, oxidative stress, and insulin signaling were measured by histology, Western blot, ELISA, and reverse transcriptase-polymerase chain reaction. RESULTS Hepatic collagen deposition (diameter of deposit) was greater in recuperated offspring (mean ± SEM: 12 ± 2 μm) than in controls (5 ± 0.5 μm) (P < 0.001). This was associated with greater inflammation (interleukin 6: 38% ± 24% increase; P < 0.05; tumor necrosis factor α: 64% ± 24% increase; P < 0.05), lipid peroxidation (4-hydroxynonenal, measured by ELISA: 0.30 ± 0.02 compared with 0.19 ± 0.05 μg/mL per μg protein; P < 0.05), and hyperinsulinemia (P < 0.05). CoQ10 supplementation increased (P < 0.01) hepatic CoQ10 concentrations and ameliorated liver fibrosis (P < 0.001), inflammation (P < 0.001), some measures of oxidative stress (P < 0.001), and hyperinsulinemia (P < 0.01). CONCLUSIONS Suboptimal in utero nutrition combined with accelerated postnatal catch-up growth caused more hepatic fibrosis in adulthood, which was associated with higher indexes of oxidative stress and inflammation and hyperinsulinemia. CoQ10 supplementation prevented liver fibrosis accompanied by downregulation of oxidative stress, inflammation, and hyperinsulinemia.
Collapse
Affiliation(s)
- Jane L Tarry-Adkins
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Iain P Hargreaves
- Neurometabolic Unit, National Hospital, University College London, London, United Kingdom
| | - Viruna Neergheen
- Neurometabolic Unit, National Hospital, University College London, London, United Kingdom
| | - Catherine E Aiken
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Malgorzata S Martin-Gronert
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Josie M McConnell
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| |
Collapse
|
135
|
Longo N, Frigeni M, Pasquali M. Carnitine transport and fatty acid oxidation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2422-35. [PMID: 26828774 DOI: 10.1016/j.bbamcr.2016.01.023] [Citation(s) in RCA: 480] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/27/2016] [Accepted: 01/28/2016] [Indexed: 12/14/2022]
Abstract
Carnitine is essential for the transfer of long-chain fatty acids across the inner mitochondrial membrane for subsequent β-oxidation. It can be synthesized by the body or assumed with the diet from meat and dairy products. Defects in carnitine biosynthesis do not routinely result in low plasma carnitine levels. Carnitine is accumulated by the cells and retained by kidneys using OCTN2, a high affinity organic cation transporter specific for carnitine. Defects in the OCTN2 carnitine transporter results in autosomal recessive primary carnitine deficiency characterized by decreased intracellular carnitine accumulation, increased losses of carnitine in the urine, and low serum carnitine levels. Patients can present early in life with hypoketotic hypoglycemia and hepatic encephalopathy, or later in life with skeletal and cardiac myopathy or sudden death from cardiac arrhythmia, usually triggered by fasting or catabolic state. This disease responds to oral carnitine that, in pharmacological doses, enters cells using the amino acid transporter B(0,+). Primary carnitine deficiency can be suspected from the clinical presentation or identified by low levels of free carnitine (C0) in the newborn screening. Some adult patients have been diagnosed following the birth of an unaffected child with very low carnitine levels in the newborn screening. The diagnosis is confirmed by measuring low carnitine uptake in the patients' fibroblasts or by DNA sequencing of the SLC22A5 gene encoding the OCTN2 carnitine transporter. Some mutations are specific for certain ethnic backgrounds, but the majority are private and identified only in individual families. Although the genotype usually does not correlate with metabolic or cardiac involvement in primary carnitine deficiency, patients presenting as adults tend to have at least one missense mutation retaining residual activity. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.
Collapse
Affiliation(s)
- Nicola Longo
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA; Department of Pathology, University of Utah, and ARUP Laboratories, 500 Chipeta Way, Salt Lake City, UT, USA.
| | - Marta Frigeni
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Marzia Pasquali
- Department of Pathology, University of Utah, and ARUP Laboratories, 500 Chipeta Way, Salt Lake City, UT, USA
| |
Collapse
|
136
|
Mann JP, Semple RK, Armstrong MJ. How Useful Are Monogenic Rodent Models for the Study of Human Non-Alcoholic Fatty Liver Disease? Front Endocrinol (Lausanne) 2016; 7:145. [PMID: 27899914 PMCID: PMC5110950 DOI: 10.3389/fendo.2016.00145] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 11/01/2016] [Indexed: 12/22/2022] Open
Abstract
Improving understanding of the genetic basis of human non-alcoholic fatty liver disease (NAFLD) has the potential to facilitate risk stratification of affected patients, permit personalized treatment, and inform development of new therapeutic strategies. Animal models have been widely used to interrogate the pathophysiology of, and genetic predisposition to, NAFLD. Nevertheless, considerable interspecies differences in intermediary metabolism potentially limit the extent to which results can be extrapolated to humans. For example, human genome-wide association studies have identified polymorphisms in PNPLA3 and TM6SF2 as the two most prevalent determinants of susceptibility to NAFLD and its inflammatory component (NASH), but animal models of these mutations have had only variable success in recapitulating this link. In this review, we critically appraise selected murine monogenic models of NAFLD, NASH, and hepatocellular carcinoma (HCC) with a focus on how closely they mirror human disease.
Collapse
Affiliation(s)
- Jake P. Mann
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Robert K. Semple
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
- The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- *Correspondence: Robert K. Semple,
| | - Matthew J. Armstrong
- Centre for Liver Research, National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, Birmingham, UK
- Liver Unit, Queen Elizabeth University Hospital Birmingham, Birmingham, UK
| |
Collapse
|
137
|
Janssens S, Heemskerk MM, van den Berg SA, van Riel NA, Nicolay K, Willems van Dijk K, Prompers JJ. Effects of low-stearate palm oil and high-stearate lard high-fat diets on rat liver lipid metabolism and glucose tolerance. Nutr Metab (Lond) 2015; 12:57. [PMID: 26691906 PMCID: PMC4683731 DOI: 10.1186/s12986-015-0053-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/11/2015] [Indexed: 01/01/2023] Open
Abstract
Background Excess consumption of energy-dense, high-fat Western diets contributes to the development of obesity and obesity-related disorders, such as fatty liver disease. However, not only the quantity but also the composition of dietary fat may play a role in the development of liver steatosis. The aim of this study was to determine the effects of low-stearate palm oil and high-stearate lard high-fat diets on in vivo liver lipid metabolism. Methods Wistar rats were fed with either normal chow (CON), a high-fat diet based on palm oil (HFP), or a high-fat diet based on lard (HFL). After 10 weeks of diet, magnetic resonance spectroscopy was applied for the in vivo determination of intrahepatocellular lipid content and the uptake and turnover of dietary fat after oral administration of 13C-labeled lipids. Derangements in liver lipid metabolism were further assessed by measuring hepatic very-low density lipoprotein (VLDL) secretion and ex vivo respiratory capacity of liver mitochondria using fat-derived substrates. In addition, whole-body and hepatic glucose tolerance were determined with an intraperitoneal glucose tolerance test. Results Both high-fat diets induced liver lipid accumulation (p < 0.001), which was accompanied by a delayed uptake and/or slower turnover of dietary fat in the liver (p < 0.01), but without any change in VLDL secretion rates. Surprisingly, liver lipid content was higher in HFP than in HFL (p < 0.05), despite the increased fatty acid oxidative capacity in isolated liver mitochondria of HFP animals (p < 0.05). In contrast, while both high-fat diets induced whole-body glucose intolerance, only HFL impaired hepatic glucose tolerance. Conclusion High-fat diets based on palm oil and lard similarly impair the handling of dietary lipids in the liver, but only the high-fat lard diet induces hepatic glucose intolerance.
Collapse
Affiliation(s)
- Sharon Janssens
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Mattijs M Heemskerk
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd A van den Berg
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands ; Present address: Amphia Hospital, Breda, The Netherlands
| | - Natal A van Riel
- Computational Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Klaas Nicolay
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands ; Department of Medicine, division Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeanine J Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
138
|
Wang G, Wang H, Singh S, Zhou P, Yang S, Wang Y, Zhu Z, Zhang J, Chen A, Billiar T, Monga SP, Wang Q. ADAR1 Prevents Liver Injury from Inflammation and Suppresses Interferon Production in Hepatocytes. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:3224-37. [PMID: 26453800 PMCID: PMC4729276 DOI: 10.1016/j.ajpath.2015.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 07/15/2015] [Accepted: 08/11/2015] [Indexed: 12/18/2022]
Abstract
Adenosine deaminase acting on RNA 1 (ADAR1) is an essential protein for embryonic liver development. ADAR1 loss is embryonically lethal because of severe liver damage. Although ADAR1 is required in adult livers to prevent liver cell death, as demonstrated by liver-specific conditional knockout (Alb-ADAR1(KO)) mice, the mechanism remains elusive. We systematically analyzed Alb-ADAR1(KO) mice for liver damage. Differentiation genes and inflammatory pathways were examined in hepatic tissues from Alb-ADAR1(KO) and littermate controls. Inducible ADAR1 KO mice were used to validate regulatory effects of ADAR1 on inflammatory cytokines. We found that Alb-ADAR1(KO) mice showed dramatic growth retardation and high mortality because of severe structural and functional damage to the liver, which showed overwhelming inflammation, cell death, fibrosis, fatty change, and compensatory regeneration. Simultaneously, Alb-ADAR1(KO) showed altered expression of key differentiation genes and significantly higher levels of hepatic inflammatory cytokines, especially type I interferons, which was also verified by inducible ADAR1 knockdown in primary hepatocyte cultures. We conclude that ADAR1 is an essential molecule for maintaining adult liver homeostasis and, in turn, morphological and functional integrity. It inhibits the production of type I interferons and other inflammatory cytokines. Our findings may provide novel insight in the pathogenesis of liver diseases caused by excessive inflammatory responses, including autoimmune hepatitis.
Collapse
Affiliation(s)
- Guoliang Wang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of General Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sucha Singh
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Pei Zhou
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shengyong Yang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yujuan Wang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zhaowei Zhu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jinxiang Zhang
- Department of General Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Alex Chen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Cardiology, Center for Vascular Disease and Translational Medicine, Third Xiangya Hospital, Central South University, Changsha, China
| | - Timothy Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Satdarshan P Monga
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Cardiology, Center for Vascular Disease and Translational Medicine, Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
139
|
Xu C, Qiao Z, Lu Y, Zhang D, Jia Z, Zhuang X, Shi Y, Xu T, Xing L, Shen J. Influence of Fatty Liver on the Severity and Clinical Outcome in Acute Pancreatitis. PLoS One 2015; 10:e0142278. [PMID: 26571385 PMCID: PMC4646451 DOI: 10.1371/journal.pone.0142278] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 10/20/2015] [Indexed: 02/08/2023] Open
Abstract
Acute pancreatitis (AP) is a common disease in the department of gastroenterology with variable severity, from being mild and self-limited to severe and fatal. The early diagnosis and accurate prediction of AP severity are of great importance. Our primary observation showed that fatty liver (FL) was frequently detected in patients with AP. In this retrospective study, we aimed to evaluate the relation between FL and the severity and outcomes of AP. The medical records of 2671 patients with AP were reviewed retrospectively, and characteristics of AP patients were recorded. FL was assessed by abdominal CT scan, and AP patients were categorized by the occurrence of FL for the analysis. The variation of mortality, clinical severity and the appearance of CT were analyzed between the non-FL group and FL groups. Compared with patients without FL, an obviously higher rate of death and higher frequency of severe AP (SAP) and necrotizing AP (ANP) were observed in patients with FL, as well as the incidence of local complications and systemic complications. Taking obesity into consideration, a higher rate of death and more severe AP were found in patients with FL, no matter whether they were obese or not. Alcoholic fatty liver (AFL) and non-alcoholic fatty liver (NAFL) were also separated for comparison in this study; the incidence of ANP and the clinical severity had no significant difference between the AFL and NAFL groups. In conclusion, FL could influence the severity and clinical outcome and may play a prognostic role in AP. This study is of clinical significance, because few reports have been previously issued on FL and AP.
Collapse
Affiliation(s)
- Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Zhenguo Qiao
- Department of Gastroenterology, Affiliated Wujiang Hospital of Nantong University, Jiangsu, China
| | - Yongda Lu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Deqing Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Zhenyu Jia
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Xiaohui Zhuang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Yuqi Shi
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Ting Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Lihua Xing
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Jiaqing Shen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Jiangsu, China
| |
Collapse
|
140
|
Siddiqui RA, Xu Z, Harvey KA, Pavlina TM, Becker MJ, Zaloga GP. Comparative study of the modulation of fructose/sucrose-induced hepatic steatosis by mixed lipid formulations varying in unsaturated fatty acid content. Nutr Metab (Lond) 2015; 12:41. [PMID: 26583036 PMCID: PMC4650347 DOI: 10.1186/s12986-015-0038-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/08/2015] [Indexed: 12/18/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in developed countries. NAFLD encompasses a spectrum of diseases, ranging from hepatic steatosis to non-alcoholic steatohepatitis (NASH), cirrhosis, and liver failure. The etiology of NAFLD remains unclear but is thought to relate to increased fatty acid flux within the liver that results in toxic fatty acid metabolite production. One source of increased fatty acid flux is fructose/sucrose-induced hepatic lipogenesis. Current treatment for NAFLD encompasses dietary modifications. However, little scientific evidence exists on which to base many dietary recommendations, especially the intake of different types of carbohydrates and fats. We hypothesized that lipid mixtures of unsaturated fatty acids would inhibit lipogenesis and subsequent hepatic steatosis induced by high carbohydrate diets. The aim of this study was to examine the effects of different complex mixtures of fatty acids upon the development of fructose/sucrose-induced hepatic steatosis. Methods C57BL/6 mice were randomized to normocaloric chow-based diets that varied in the type of carbohydrate (starch, sucrose, fructose). Animals in each carbohydrate group were further randomized to diets that varied in lipid type (no additional lipid, soybean oil, fish oil, olive/soybean oil, macadamia nut oil). These oils were chosen based upon their content of omega-6 polyunsaturated fatty acids, omega-3 polyunsaturated fatty acids, omega-9 monounsaturated fatty acids, or omega-7 monounsaturated fatty acids. Fatty acid flux in the liver was determine by assessing hepatic lipid content (steatosis). We also assessed fatty acid levels in the plasma and liver of the animals, hepatic lipogenesis activity, hepatic stearoyl-CoA-1 desaturase activity, and hepatic elongase activity. Results Animals consumed similar amounts of the diets and maintained normal body weights throughout the study. Both sucrose and fructose induced hepatic lipogenesis and steatosis, with fructose being more potent. All mixed lipids similarly inhibited steatosis, limiting lipid content to levels found in the control (starch) animals. Lipogenesis and stearoyl-CoA-1 desaturase activity were increased in the sucrose and fructose groups. Levels of these enzymatic processes remained at baseline in all of the lipid groups. Conclusion This is the first study to compare various complex lipid mixtures, based upon dietary oils with different types of long-chain fatty acids, upon development of sucrose/fructose-induced steatosis. Both carbohydrate source and lipid content appear important for the modulation of steatosis. Moderate intake of complex lipids with high unsaturated to saturated fatty acid ratios inhibited both lipogenesis and steatosis.
Collapse
Affiliation(s)
- Rafat A Siddiqui
- Methodist Research Institute, Indiana University Health, 1800 N. Capitol Ave, Indianapolis, IN 46202 USA
| | - Zhidong Xu
- Methodist Research Institute, Indiana University Health, 1800 N. Capitol Ave, Indianapolis, IN 46202 USA
| | - Kevin A Harvey
- Methodist Research Institute, Indiana University Health, 1800 N. Capitol Ave, Indianapolis, IN 46202 USA
| | | | | | - Gary P Zaloga
- Baxter Healthcare Corporation, Deerfield, IL 60015 USA
| |
Collapse
|
141
|
Ramos-Lopez O, Martinez-Lopez E, Roman S, Fierro NA, Panduro A. Genetic, metabolic and environmental factors involved in the development of liver cirrhosis in Mexico. World J Gastroenterol 2015; 21:11552-11566. [PMID: 26556986 PMCID: PMC4631960 DOI: 10.3748/wjg.v21.i41.11552] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/29/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
Liver cirrhosis (LC) is a chronic illness caused by inflammatory responses and progressive fibrosis. Globally, the most common causes of chronic liver disease include persistent alcohol abuse, followed by viral hepatitis infections and nonalcoholic fatty liver disease. However, regardless of the etiological factors, the susceptibility and degree of liver damage may be influenced by genetic polymorphisms that are associated with distinct ethnic and cultural backgrounds. Consequently, metabolic genes are influenced by variable environmental lifestyle factors, such as diet, physical inactivity, and emotional stress, which are associated with regional differences among populations. This Topic Highlight will focus on the genetic and environmental factors that may influence the metabolism of alcohol and nutrients in the setting of distinct etiologies of liver disease. The interaction between genes and environment in the current-day admixed population, Mestizo and Native Mexican, will be described. Additionally, genes involved in immune regulation, insulin sensitivity, oxidative stress and extracellular matrix deposition may modulate the degree of severity. In conclusion, LC is a complex disease. The onset, progression, and clinical outcome of LC among the Mexican population are influenced by specific genetic and environmental factors. Among these are an admixed genome with a heterogenic distribution of European, Amerindian and African ancestry; a high score of alcohol consumption; viral infections; a hepatopathogenic diet; and a high prevalence of obesity. The variance in risk factors among populations suggests that intervention strategies directed towards the prevention and management of LC should be tailored according to such population-based features.
Collapse
|
142
|
Alashry SE, Gaballah MA, Malek HA, Elsalam AIA. Effect of Sildenafil on Non-Alcoholic Fatty Liver. INT J PHARMACOL 2015. [DOI: 10.3923/ijp.2015.814.820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
143
|
Molecular mechanisms of fatty liver in obesity. Front Med 2015; 9:275-87. [PMID: 26290284 DOI: 10.1007/s11684-015-0410-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 05/25/2015] [Indexed: 12/17/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) covers a spectrum of liver disorders ranging from simple steatosis to advanced pathologies, including nonalcoholic steatohepatitis and cirrhosis. NAFLD significantly contributes to morbidity and mortality in developed societies. Insulin resistance associated with central obesity is the major cause of hepatic steatosis, which is characterized by excessive accumulation of triglyceride-rich lipid droplets in the liver. Accumulating evidence supports that dysregulation of adipose lipolysis and liver de novo lipogenesis (DNL) plays a key role in driving hepatic steatosis. In this work, we reviewed the molecular mechanisms responsible for enhanced adipose lipolysis and increased hepatic DNL that lead to hepatic lipid accumulation in the context of obesity. Delineation of these mechanisms holds promise for developing novel avenues against NAFLD.
Collapse
|
144
|
Shan W, Gao L, Zeng W, Hu Y, Wang G, Li M, Zhou J, Ma X, Tian X, Yao J. Activation of the SIRT1/p66shc antiapoptosis pathway via carnosic acid-induced inhibition of miR-34a protects rats against nonalcoholic fatty liver disease. Cell Death Dis 2015. [PMID: 26203862 PMCID: PMC4650741 DOI: 10.1038/cddis.2015.196] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent studies have demonstrated that miR-34a expression is significantly upregulated and associated with apoptosis in nonalcoholic fatty liver disease (NAFLD). Carnosic acid (CA) is a novel antioxidant and a potential inhibitor of apoptosis in organ injury, including liver injury. This study aimed to investigate the signaling mechanisms underlying miR-34a expression and the antiapoptotic effect of CA in NAFLD. CA treatment significantly reduced the high-fat diet (HFD)-induced elevations in aminotransferase activity as well as in serum triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) and malondialdehyde (MDA) levels but increased serum high-density lipoprotein cholesterol (HDL-C) and hepatic superoxide dismutase (SOD) levels. Moreover, CA treatment ameliorated the increase in cleaved caspase-3 caused by HFD exposure and completely reversed the HFD-induced decreases in manganese superoxide dismutase (MnSOD) and B-cell lymphoma-extra large expression. CA also counteracted the HFD- or palmitic acid (PA)-induced increases in caspase-3 and caspase-9 activity. Mechanistically, CA reversed the HFD- or PA-induced upregulation of miR-34a, which is the best-characterized regulator of SIRT1. Importantly, the decrease in miR-34a expression was closely associated with the activation of the SIRT1/p66shc pathway, which attenuates hepatocyte apoptosis in liver ischemia/reperfusion injury. A dual luciferase assay in L02 cells validated the modulation of SIRT1 by CA, which occurs at least partly via miR-34a. In addition, miR-34a overexpression was significantly counteracted by CA, which prevented the miR-34a-dependent repression of the SIRT1/p66shc pathway and apoptosis. Collectively, our results support a link between liver cell apoptosis and the miR-34a/SIRT1/p66shc pathway, which can be modulated by CA in NAFLD.
Collapse
Affiliation(s)
- W Shan
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - L Gao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - W Zeng
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Y Hu
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - G Wang
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - M Li
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - J Zhou
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - X Ma
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - X Tian
- Department of General Surgery, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - J Yao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| |
Collapse
|
145
|
Roman S, Panduro A. Genomic medicine in gastroenterology: A new approach or a new specialty? World J Gastroenterol 2015; 21:8227-37. [PMID: 26217074 PMCID: PMC4507092 DOI: 10.3748/wjg.v21.i27.8227] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/24/2015] [Accepted: 05/04/2015] [Indexed: 02/06/2023] Open
Abstract
Throughout history, many medical milestones have been achieved to prevent and treat human diseases. Man's early conception of illness was naturally holistic or integrative. However, scientific knowledge was atomized into quantitative and qualitative research. In the field of medicine, the main trade-off was the creation of many medical specialties that commonly treat patients in advanced stages of disease. However, now that we are immersed in the post-genomic era, how should we reevaluate medicine? Genomic medicine has evoked a medical paradigm shift based on the plausibility to predict the genetic susceptibility to disease. Additionally, the development of chronic diseases should be viewed as a continuum of interactions between the individual's genetic make-up and environmental factors such as diet, physical activity, and emotions. Thus, personalized medicine is aimed at preventing or reversing clinical symptoms, and providing a better quality of life by integrating the genetic, environmental and cultural factors of diseases. Whether using genomic medicine in the field of gastroenterology is a new approach or a new medical specialty remains an open question. To address this issue, it will require the mutual work of educational and governmental authorities with public health professionals, with the goal of translating genomic medicine into better health policies.
Collapse
|
146
|
Shimada Y, Kuninaga S, Ariyoshi M, Zhang B, Shiina Y, Takahashi Y, Umemoto N, Nishimura Y, Enari H, Tanaka T. E2F8 promotes hepatic steatosis through FABP3 expression in diet-induced obesity in zebrafish. Nutr Metab (Lond) 2015; 12:17. [PMID: 26052340 PMCID: PMC4456805 DOI: 10.1186/s12986-015-0012-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 05/04/2015] [Indexed: 12/13/2022] Open
Abstract
Background Diet-induced hepatic steatosis is highly associated with nonalcoholic fatty liver disease, which is related to the development of metabolic syndrome. While advanced stage nonalcoholic hepatic steatosis and steatohepatitis (NASH) result ultimately in fibrosis and cirrhosis, the molecular basis for lipid droplet formation is poorly understood. Common pathways underlie the pathology of mammalian obesity and the zebrafish diet-induced obesity model (DIO-zebrafish) used in this study. Methods Our analysis involved a combination of transcriptome (DNA microarray) and proteome (two-dimensional electrophoresis) methods using liver tissue from DIO-zebrafish to find candidate genes involved in hepatic steatosis. We conducted intraperitoneal injection (i.p.) of morpholino antisense oligonucleotides (MOs) for each gene into DIO-zebrafish. We also conducted in vitro overexpression in human cells. Additionally, we examined gene expression during feeding experiments involving anti-obesity compounds, creatine and anserine. Results We found that fatty acid binding protein 3 (fabp3) and E2F transcription factors were upregulated in hepatic steatosis. E2f8 MO i.p. suppressed fabp3 expression in liver, and ameliorated hepatic steatosis. In human cells (HepG2), E2F8 overexpression promoted FABP3 expression. Additionally, co-administration of creatine and anserine suppressed obesity associated phenotypes including hepatic steatosis as indicated by e2f8 and fabp3 down regulation. Conclusion We discovered that the e2f8–fabp3 axis is important in the promotion of hepatic steatosis in DIO-zebrafish. The combination of transcriptome and proteome analyses using the disease model zebrafish allow identification of novel pathways involved in human diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12986-015-0012-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yasuhito Shimada
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie Japan ; Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan ; Mie University Medical Zebrafish Research Center, Mie, Japan ; Department of Bioinformatics, Mie University Life Science Research Center, Mie, Japan ; Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie, Japan
| | - Shisei Kuninaga
- Central Research Institute, Maruha Nichiro Corporation, Ibaraki, Japan
| | - Michiko Ariyoshi
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie Japan
| | - Beibei Zhang
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie Japan
| | - Yasuhiko Shiina
- Central Research Institute, Maruha Nichiro Corporation, Ibaraki, Japan
| | | | - Noriko Umemoto
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie Japan ; Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan
| | - Yuhei Nishimura
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie Japan ; Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan ; Mie University Medical Zebrafish Research Center, Mie, Japan ; Department of Bioinformatics, Mie University Life Science Research Center, Mie, Japan ; Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie, Japan
| | - Hiroyuki Enari
- Central Research Institute, Maruha Nichiro Corporation, Ibaraki, Japan
| | - Toshio Tanaka
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie Japan ; Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie, Japan ; Mie University Medical Zebrafish Research Center, Mie, Japan ; Department of Bioinformatics, Mie University Life Science Research Center, Mie, Japan ; Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie, Japan
| |
Collapse
|
147
|
Trzepizur W, Gaceb A, Arnaud C, Ribuot C, Levy P, Martinez MC, Gagnadoux F, Andriantsitohaina R. Vascular and hepatic impact of short-term intermittent hypoxia in a mouse model of metabolic syndrome. PLoS One 2015; 10:e0124637. [PMID: 25993257 PMCID: PMC4436258 DOI: 10.1371/journal.pone.0124637] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 03/17/2015] [Indexed: 01/25/2023] Open
Abstract
Background Experimental models of intermittent hypoxia (IH) have been developed during the last decade to investigate the consequences of obstructive sleep apnea. IH is usually associated with detrimental metabolic and vascular outcomes. However, paradoxical protective effects have also been described depending of IH patterns and durations applied in studies. We evaluated the impact of short-term IH on vascular and metabolic function in a diet-induced model of metabolic syndrome (MS). Methods Mice were fed either a standard diet or a high fat diet (HFD) for 8 weeks. During the final 14 days of each diet, animals were exposed to either IH (1 min cycle, FiO2 5% for 30s, FiO2 21% for 30s; 8 h/day) or intermittent air (FiO2 21%). Ex-vivo vascular reactivity in response to acetylcholine was assessed in aorta rings by myography. Glucose, insulin and leptin levels were assessed, as well as serum lipid profile, hepatic mitochondrial activity and tissue nitric oxide (NO) release. Results Mice fed with HFD developed moderate markers of dysmetabolism mimicking MS, including increased epididymal fat, dyslipidemia, hepatic steatosis and endothelial dysfunction. HFD decreased mitochondrial complex I, II and IV activities and increased lactate dehydrogenase (LDH) activity in liver. IH applied to HFD mice induced a major increase in insulin and leptin levels and prevented endothelial dysfunction by restoring NO production. IH also restored mitochondrial complex I and IV activities, moderated the increase in LDH activity and liver triglyceride accumulation in HFD mice. Conclusion In a mouse model of MS, short-term IH increases insulin and leptin levels, restores endothelial function and mitochondrial activity and limits liver lipid accumulation.
Collapse
Affiliation(s)
- Wojciech Trzepizur
- INSERM U1063, Sopam, Angers University, F-49045, Angers, France
- Department of Respiratory Diseases, Angers University hospital, Angers, France
- * E-mail:
| | - Abderahim Gaceb
- INSERM U1063, Sopam, Angers University, F-49045, Angers, France
| | - Claire Arnaud
- INSERM U1042, HP2 laboratory, Joseph Fourier University, Grenoble, France
| | - Christophe Ribuot
- INSERM U1042, HP2 laboratory, Joseph Fourier University, Grenoble, France
| | - Patrick Levy
- INSERM U1042, HP2 laboratory, Joseph Fourier University, Grenoble, France
- Laboratoires du Sommeil et EFCR, A. Michallon University Hospital, Grenoble, France
| | | | - Frédéric Gagnadoux
- INSERM U1063, Sopam, Angers University, F-49045, Angers, France
- Department of Respiratory Diseases, Angers University hospital, Angers, France
| | | |
Collapse
|
148
|
Gorman S, Black LJ, Feelisch M, Hart PH, Weller R. Can skin exposure to sunlight prevent liver inflammation? Nutrients 2015; 7:3219-39. [PMID: 25951129 PMCID: PMC4446748 DOI: 10.3390/nu7053219] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 04/23/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023] Open
Abstract
Liver inflammation contributes towards the pathology of non-alcoholic fatty liver disease (NAFLD). Here we discuss how skin exposure to sunlight may suppress liver inflammation and the severity of NAFLD. Following exposure to sunlight-derived ultraviolet radiation (UVR), the skin releases anti-inflammatory mediators such as vitamin D and nitric oxide. Animal modeling studies suggest that exposure to UVR can prevent the development of NAFLD. Association studies also support a negative link between circulating 25-hydroxyvitamin D and NAFLD incidence or severity. Clinical trials are in their infancy and are yet to demonstrate a clear beneficial effect of vitamin D supplementation. There are a number of potentially interdependent mechanisms whereby vitamin D could dampen liver inflammation, by inhibiting hepatocyte apoptosis and liver fibrosis, modulating the gut microbiome and through altered production and transport of bile acids. While there has been a focus on vitamin D, other mediators induced by sun exposure, such as nitric oxide may also play important roles in curtailing liver inflammation.
Collapse
Affiliation(s)
- Shelley Gorman
- Telethon Kids Institute, University of Western Australia, 100 Roberts Rd, Subiaco, Western Australia 6008, Australia.
| | - Lucinda J Black
- Telethon Kids Institute, University of Western Australia, 100 Roberts Rd, Subiaco, Western Australia 6008, Australia.
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK.
| | - Prue H Hart
- Telethon Kids Institute, University of Western Australia, 100 Roberts Rd, Subiaco, Western Australia 6008, Australia.
| | - Richard Weller
- University of Edinburgh, MRC Centre for Inflammation Research, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
149
|
Willy JA, Young SK, Stevens JL, Masuoka HC, Wek RC. CHOP links endoplasmic reticulum stress to NF-κB activation in the pathogenesis of nonalcoholic steatohepatitis. Mol Biol Cell 2015; 26:2190-204. [PMID: 25904325 PMCID: PMC4462938 DOI: 10.1091/mbc.e15-01-0036] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/16/2015] [Indexed: 01/10/2023] Open
Abstract
During metabolic stress, the UPR transcription factor CHOP activates NF-κB through a pathway involving IRAK2 expression, resulting in hepatocyte secretion of cytokines IL-8 and TNFα, which trigger inflammation and hepatocellular death. Free fatty acid induction of inflammation and cell death is an important feature of nonalcoholic steatohepatitis (NASH) and has been associated with disruption of the endoplasmic reticulum and activation of the unfolded protein response (UPR). After chronic UPR activation, the transcription factor CHOP (GADD153/DDIT3) triggers cell death; however, the mechanisms linking the UPR or CHOP to hepatoceullular injury and inflammation in the pathogenesis of NASH are not well understood. Using HepG2 and primary human hepatocytes, we found that CHOP induces cell death and inflammatory responses after saturated free fatty acid exposure by activating NF-κB through a pathway involving IRAK2 expression, resulting in secretion of cytokines IL-8 and TNFα directly from hepatocytes. TNFα facilitates hepatocyte death upon exposure to saturated free fatty acids, and secretion of both IL-8 and TNFα contribute to inflammation. Of interest, CHOP/NF-κB signaling is not conserved in primary rodent hepatocytes. Our studies suggest that CHOP plays a vital role in the pathophysiology of NASH by induction of secreted factors that trigger inflammation and hepatocellular death via a signaling pathway specific to human hepatocytes.
Collapse
Affiliation(s)
- Jeffrey A Willy
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Sara K Young
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - James L Stevens
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Howard C Masuoka
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Ronald C Wek
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
150
|
Arias N, Macarulla MT, Aguirre L, Miranda J, Portillo MP. Liver delipidating effect of a combination of resveratrol and quercetin in rats fed an obesogenic diet. J Physiol Biochem 2015; 71:569-76. [PMID: 25827944 DOI: 10.1007/s13105-015-0403-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/18/2015] [Indexed: 01/14/2023]
Abstract
Liver steatosis is characterized by an abnormal accumulation of triacylglycerols in this organ. This metabolic disorder is closely associated with obesity. In the present study, we aimed to analyse the effect of a combination of resveratrol and quercetin on liver steatosis in an animal model of dietetic obesity, and to compare it with one induced by the administration of each polyphenol separately. Rats were divided into four dietary groups of nine animals each and fed a high-fat, high-sucrose diet: an untreated control group and three groups treated either with resveratrol (RSV; 15 mg/kg/day), with quercetin (Q; 30 mg/kg/day), or with both (RSV + Q; 15 mg resveratrol/kg/day and 30 mg quercetin/kg/day) for 6 weeks. Liver weight and triacylglycerol content decreased only in the RSV + Q group. A significant reduction in acetyl-CoA carboxylase activity was observed in RSV and RSV + Q groups, without changes in fatty acid synthase activity. A significant increase in carnitine palmitoyltransferase-1a activity was observed only in rats treated with the combination of resveratrol and quercetin, suggesting increased fatty acid oxidation. Citrate synthase, a marker of mitochondrial density, remained unchanged in all groups. No significant changes were observed in the expression of peroxisome proliferator-activated receptor α (PPARα), nuclear respiratory factor 1 (NRF-1) and transcription factor A mitochondrial (TFAM). In conclusion, resveratrol and quercetin together, combining two doses which were shown to be ineffective singly, is an interesting tool to prevent liver steatosis associated with high-fat high-sucrose feeding. The delipidating effect seems to be mediated by increased fatty acid oxidation not associated with increased mitochondriogenesis, and by reduced de novo lipogenesis.
Collapse
Affiliation(s)
- Noemí Arias
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Faculty of Pharmacy and Lucio Lascaray Research Center, University of the Basque Country (UPV/EHU), Vitoria, Spain
| | | | | | | | | |
Collapse
|