101
|
Rozman J, Rathkolb B, Oestereicher MA, Schütt C, Ravindranath AC, Leuchtenberger S, Sharma S, Kistler M, Willershäuser M, Brommage R, Meehan TF, Mason J, Haselimashhadi H, Hough T, Mallon AM, Wells S, Santos L, Lelliott CJ, White JK, Sorg T, Champy MF, Bower LR, Reynolds CL, Flenniken AM, Murray SA, Nutter LMJ, Svenson KL, West D, Tocchini-Valentini GP, Beaudet AL, Bosch F, Braun RB, Dobbie MS, Gao X, Herault Y, Moshiri A, Moore BA, Kent Lloyd KC, McKerlie C, Masuya H, Tanaka N, Flicek P, Parkinson HE, Sedlacek R, Seong JK, Wang CKL, Moore M, Brown SD, Tschöp MH, Wurst W, Klingenspor M, Wolf E, Beckers J, Machicao F, Peter A, Staiger H, Häring HU, Grallert H, Campillos M, Maier H, Fuchs H, Gailus-Durner V, Werner T, Hrabe de Angelis M. Identification of genetic elements in metabolism by high-throughput mouse phenotyping. Nat Commun 2018; 9:288. [PMID: 29348434 PMCID: PMC5773596 DOI: 10.1038/s41467-017-01995-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 10/30/2017] [Indexed: 12/20/2022] Open
Abstract
Metabolic diseases are a worldwide problem but the underlying genetic factors and their relevance to metabolic disease remain incompletely understood. Genome-wide research is needed to characterize so-far unannotated mammalian metabolic genes. Here, we generate and analyze metabolic phenotypic data of 2016 knockout mouse strains under the aegis of the International Mouse Phenotyping Consortium (IMPC) and find 974 gene knockouts with strong metabolic phenotypes. 429 of those had no previous link to metabolism and 51 genes remain functionally completely unannotated. We compared human orthologues of these uncharacterized genes in five GWAS consortia and indeed 23 candidate genes are associated with metabolic disease. We further identify common regulatory elements in promoters of candidate genes. As each regulatory element is composed of several transcription factor binding sites, our data reveal an extensive metabolic phenotype-associated network of co-regulated genes. Our systematic mouse phenotype analysis thus paves the way for full functional annotation of the genome.
Collapse
Affiliation(s)
- Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Ludwig-Maximilians-Universität München, Gene Center, Institute of Molecular Animal Breeding and Biotechnology, Feodor-Lynen Strasse 25, 81377, Munich, Germany
| | - Manuela A Oestereicher
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Christine Schütt
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Aakash Chavan Ravindranath
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Stefanie Leuchtenberger
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Sapna Sharma
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Martin Kistler
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Monja Willershäuser
- Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, 85354, Freising, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, 85354, Freising, Germany
- ZIEL - Institute for Food & Health, Technical University of Munich, 85354, Freising, Germany
| | - Robert Brommage
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Terrence F Meehan
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Jeremy Mason
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Hamed Haselimashhadi
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Tertius Hough
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Ann-Marie Mallon
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Sara Wells
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Luis Santos
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Christopher J Lelliott
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Jacqueline K White
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Tania Sorg
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), 1 Rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Parc d'innovation, 1 Rue Laurent Fries - BP 10142, 67404, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67404, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67404, Illkirch, France
- Université de Strasbourg, 67404, Illkirch, France
| | - Marie-France Champy
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), 1 Rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Parc d'innovation, 1 Rue Laurent Fries - BP 10142, 67404, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67404, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67404, Illkirch, France
- Université de Strasbourg, 67404, Illkirch, France
| | - Lynette R Bower
- Mouse Biology Program, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Corey L Reynolds
- Department of Molecular and Human Genetics, Baylor College of Medicine, 7702 Main St, Houston Medical Center, Houston, TX, 77030-4406, USA
| | - Ann M Flenniken
- The Centre for Phenogenomics, 25 Orde St, Toronto, M5T 3H7, ON, Canada
- The Hospital for Sick Children, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Stephen A Murray
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, 25 Orde St, Toronto, M5T 3H7, ON, Canada
- The Hospital for Sick Children, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Karen L Svenson
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - David West
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA, 94609, USA
| | - Glauco P Tocchini-Valentini
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Institute of Cell Biology and Neurobiology, Adriano Buzzati-Traverso Campus, Via E. Ramarini 32, Monterotondo Scalo, RM, 00015, Italy
| | - Arthur L Beaudet
- The Centre for Phenogenomics, 25 Orde St, Toronto, M5T 3H7, ON, Canada
- The Hospital for Sick Children, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Robert B Braun
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Michael S Dobbie
- Australian Phenomics Network, John Curtin School of Medical Research, Australian National University, 131 Garran Road, Canberra, ACT, 2601, Australia
| | - Xiang Gao
- SKL of Pharmaceutical Biotechnology and Model Animal Research Center, Collaborative Innovation Center for Genetics and Development, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Yann Herault
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), 1 Rue Laurent Fries, 67404, Illkirch-Graffenstaden, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Parc d'innovation, 1 Rue Laurent Fries - BP 10142, 67404, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, 67404, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, 67404, Illkirch, France
- Université de Strasbourg, 67404, Illkirch, France
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, 77 Cadillac Drive, Sacramento, 95825, CA, USA
| | - Bret A Moore
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, U.C. Davis, One Shields Avenue, Davis, 95616, CA, USA
| | - K C Kent Lloyd
- Mouse Biology Program, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Colin McKerlie
- The Centre for Phenogenomics, 25 Orde St, Toronto, M5T 3H7, ON, Canada
- The Hospital for Sick Children, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Hiroshi Masuya
- RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Nobuhiko Tanaka
- RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Helen E Parkinson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Je Kyung Seong
- Korea Mouse Phenotyping Consortium (KMPC) and BK21 Program for Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 599 Gwanangno, Gwanak-gu, Seoul, 151-742, South Korea
| | - Chi-Kuang Leo Wang
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), 128 Yen-Chiou-Yuan Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan
| | | | - Steve D Brown
- Medical Research Council Harwell (Mammalian Genetics Unit and Mary Lyon Centre), Oxfordshire, OX11 0RD, UK
| | - Matthias H Tschöp
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333, Munich, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Chair of Developmental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Deutsches Institut für Neurodegenerative Erkrankungen (DZNE) Site Munich, Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 17, 81377, Munich, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences Weihenstephan, 85354, Freising, Germany
- EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, 85354, Freising, Germany
- ZIEL - Institute for Food & Health, Technical University of Munich, 85354, Freising, Germany
| | - Eckhard Wolf
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Ludwig-Maximilians-Universität München, Gene Center, Institute of Molecular Animal Breeding and Biotechnology, Feodor-Lynen Strasse 25, 81377, Munich, Germany
| | - Johannes Beckers
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany
| | - Fausto Machicao
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen, 72076, Tübingen, Germany
| | - Andreas Peter
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen, 72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard-Karls-University of Tuebingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Harald Staiger
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard-Karls-University of Tuebingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen, 72076, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard-Karls-University of Tuebingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Harald Grallert
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München and Ludwig-Maximilians Universität München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Monica Campillos
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Holger Maier
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Thomas Werner
- Internal Medicine Nephrology and Center for Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354, Freising, Germany.
| |
Collapse
|
102
|
Wang H, Zhang H, Huang B, Miao G, Yan X, Gao G, Luo Y, Chen H, Chen W, Yang L. Mesenchymal stem cells reverse high‑fat diet‑induced non‑alcoholic fatty liver disease through suppression of CD4+ T lymphocytes in mice. Mol Med Rep 2017; 17:3769-3774. [PMID: 29286155 DOI: 10.3892/mmr.2017.8326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 03/27/2017] [Indexed: 12/14/2022] Open
Abstract
Although the multipotency of mesenchymal stem cells (MSCs) makes them an attractive choice for clinical applications, immune modulation is an important factor affecting MSC transplantation. At present, the effect of treatment with MSCs on non‑alcoholic fatty liver disease (NAFLD) has received little attention. In the present study, a compact bone‑derived method was used to isolate mouse MSCs (mMSCs) and a high‑fat diet was used to establish a mouse model of NAFLD. Immunophenotypic features of mMSCs were analyzed using flow cytometry. Paraffin sections were stained with hematoxylin and eosin to assess inflammation and steatosis, and with picrosirius red to assess fibrosis. Spleen leukocytes were analyzed by flow cytometry. The results demonstrated that compact bone‑derived MSC transplantation decreased high‑fat diet‑induced weight gain, expansion of subcutaneous adipose tissue, steatosis, lobular inflammation and liver fibrogenesis. Flow cytometry analysis of spleen leukocytes demonstrated that compact bone‑derived MSC transplantation suppressed the proliferation of cluster of differentiation (CD) 4+ T lymphocytes in the spleen, which had been induced by the high‑fat diet. In conclusion, compact bone‑derived MSCs may exhibit clinical value in the treatment of NAFLD through their capacity to suppress the activation of CD4+ T cells.
Collapse
Affiliation(s)
- Huafeng Wang
- Modern College of Arts and Science and School of Life Science, Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| | - Huan Zhang
- Clinical Laboratory, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, P.R. China
| | - Biao Huang
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Guolin Miao
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xiaoyan Yan
- Modern College of Arts and Science and School of Life Science, Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| | - Gang Gao
- Modern College of Arts and Science and School of Life Science, Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| | - Yongping Luo
- Modern College of Arts and Science and School of Life Science, Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| | - Huize Chen
- Modern College of Arts and Science and School of Life Science, Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| | - Wei Chen
- Modern College of Arts and Science and School of Life Science, Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| | - Luhong Yang
- Modern College of Arts and Science and School of Life Science, Shanxi Normal University, Linfen, Shanxi 041004, P.R. China
| |
Collapse
|
103
|
Gouda W, Ashour E, Shaker Y, Ezzat W. MTP genetic variants associated with non-alcoholic fatty liver in metabolic syndrome patients. Genes Dis 2017; 4:222-228. [PMID: 30258926 PMCID: PMC6147179 DOI: 10.1016/j.gendis.2017.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/27/2017] [Indexed: 02/08/2023] Open
Abstract
This study was performed for investigation the relationship between variants of MTP gene polymorphism and the development of NAFLD in patients with and without MS. The study was included 174 NAFLD patients (106 with MS and 68 without MS), and 141 healthy control subjects. The 493 G/T polymorphism of MTP gene was evaluated by PCR-RFLP method. The frequency of MTP TT genotype and T allele were significantly higher in NAFLD patients when compared to healthy controls. Moreover, a significant association in MTP gene polymorphism was observed in NAFLD patients with MS compared to NAFLD patients without MS and controls. Our study suggested that MTP 493 G/T gene polymorphism may act as susceptibility biomarker for NAFLD and MS.
Collapse
Affiliation(s)
- Weaam Gouda
- Biochemistry Dept., National Research Center, Dokki, Giza, Egypt
| | - Esmat Ashour
- Biochemistry Dept., National Research Center, Dokki, Giza, Egypt
| | - Yehia Shaker
- Biochemistry Dept., National Research Center, Dokki, Giza, Egypt
| | - Wafaa Ezzat
- Internal Medicine Dept., National Research Center, Dokki, Giza, Egypt
| |
Collapse
|
104
|
Prevalence of Pre-Diabetes across Ethnicities: A Review of Impaired Fasting Glucose (IFG) and Impaired Glucose Tolerance (IGT) for Classification of Dysglycaemia. Nutrients 2017; 9:nu9111273. [PMID: 29165385 PMCID: PMC5707745 DOI: 10.3390/nu9111273] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/15/2017] [Accepted: 11/18/2017] [Indexed: 12/11/2022] Open
Abstract
Prediabetes can be defined by the presence of impaired fasting glucose (IFG) and/or impaired glucose tolerance (IGT), or glycated haemoglobin (HbA1c) to identify individuals at increased risk of developing type 2 diabetes (T2D). The World Health Organization (WHO, 1999) and the American Diabetes Association (ADA, 2003) utilise different cut-off values for IFG (WHO: 6.1–6.9 mmol/L; ADA: 5.6–6.9 mmol/L) but the same cut-off values for IGT (7.8–11.0 mmol/L). This review investigates whether there are differences in prevalence of IFG, IGT, and combined IFG&IGT between ethnicities, in particular Asian Chinese and European Caucasians. In total, we identified 19 studies using the WHO1999 classification, for which the average proportional prevalence for isolated (i)-IFG, i-IGT, and combined IFG&IGT were 43.9%, 41.0%, and 13.5%, respectively, for Caucasian and 29.2%, 49.4%, and 18.2%, respectively, for Asian. For the 14 studies using ADA2003 classification, the average proportional i-IFG, i-IGT, and combined IFG&IGT prevalences were 58.0%, 20.3%, and 19.8%, respectively, for Caucasian; 48.1%, 27.7%, and 20.5%, respectively, for Asian. Whilst not statistically different, there may be clinically relevant differences in the two populations, with our observations for both classifications indicating that prevalence of i-IFG is higher in Caucasian cohorts whilst i-IGT and combined IFG&IGT are both higher in Asian cohorts.
Collapse
|
105
|
Coudriet GM, Delmastro-Greenwood MM, Previte DM, Marré ML, O'Connor EC, Novak EA, Vincent G, Mollen KP, Lee S, Dong HH, Piganelli JD. Treatment with a Catalytic Superoxide Dismutase (SOD) Mimetic Improves Liver Steatosis, Insulin Sensitivity, and Inflammation in Obesity-Induced Type 2 Diabetes. Antioxidants (Basel) 2017; 6:antiox6040085. [PMID: 29104232 PMCID: PMC5745495 DOI: 10.3390/antiox6040085] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress and persistent inflammation are exaggerated through chronic over-nutrition and a sedentary lifestyle, resulting in insulin resistance. In type 2 diabetes (T2D), impaired insulin signaling leads to hyperglycemia and long-term complications, including metabolic liver dysfunction, resulting in non-alcoholic fatty liver disease (NAFLD). The manganese metalloporphyrin superoxide dismustase (SOD) mimetic, manganese (III) meso-tetrakis (N-ethylpyridinium-2-yl) porphyrin (MnP), is an oxidoreductase known to scavenge reactive oxygen species (ROS) and decrease pro-inflammatory cytokine production, by inhibiting nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. We hypothesized that targeting oxidative stress-induced inflammation with MnP would assuage liver complications and enhance insulin sensitivity and glucose tolerance in a high-fat diet (HFD)-induced mouse model of T2D. During 12 weeks of feeding, we saw significant improvements in weight, hepatic steatosis, and biomarkers of liver dysfunction with redox modulation by MnP treatment in HFD-fed mice. Additionally, MnP treatment improved insulin sensitivity and glucose tolerance, while reducing serum insulin and leptin levels. We attribute these effects to redox modulation and inhibition of hepatic NF-κB activation, resulting in diminished ROS and pro-inflammatory cytokine production. This study highlights the importance of controlling oxidative stress and secondary inflammation in obesity-mediated insulin resistance and T2D. Our data confirm the role of NF-κB-mediated inflammation in the development of T2D, and demonstrate the efficacy of MnP in preventing the progression to disease by specifically improving liver pathology and hepatic insulin resistance in obesity.
Collapse
Affiliation(s)
- Gina M Coudriet
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Meghan M Delmastro-Greenwood
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Dana M Previte
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Meghan L Marré
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Erin C O'Connor
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Elizabeth A Novak
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Garret Vincent
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Kevin P Mollen
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Sojin Lee
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - H Henry Dong
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Jon D Piganelli
- Department of Surgery, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| |
Collapse
|
106
|
Zhu M, Hao S, Liu T, Yang L, Zheng P, Zhang L, Ji G. Lingguizhugan decoction improves non-alcoholic fatty liver disease by altering insulin resistance and lipid metabolism related genes: a whole trancriptome study by RNA-Seq. Oncotarget 2017; 8:82621-82631. [PMID: 29137289 PMCID: PMC5669915 DOI: 10.18632/oncotarget.19734] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 06/29/2017] [Indexed: 12/23/2022] Open
Abstract
Lingguizhugan decoction, a classic traditional Chinese medicine formula, has been used to treat non-alcoholic fatty liver disease (NAFLD), however, the underlying mechanisms remains unclear. In the present study, we compared the phenotype of the normal rats (fed with chow diet), high-fat-diet (HFD) induced NAFLD rats and Lingguizhugan decoction (LGZG, comprises four Chinese herbs: Poria, Ramulus Cinnamomi, Rhizoma Atractylodis Macrocephalae, and Radix Glycyrrhizae.) intervened rats, and detected whole genome gene expression by RNA-Seq. Our results demonstrated that LGZG decoction attenuated phenotypic characteristics of NAFLD rats. RNA-Seq data analysis revealed that gene expression profiles exerted differential patterns between different groups. 2690 (1445 up-regulated, 1245 down-regulated) genes in NAFLD versus (vs) normal group, 69 (16 up-regulated, 53 down-regulated) genes in LGZG vs NAFLD group, and 42 overlapped (12 up- regulated, 30 down-regulated) genes between NAFLDvs normal group and LGZG vs NAFLD group were identified as differentially expressed. GO, pathway enrichment and PPI networks analysis of the overlapped genes revealed that LGZG decoction might attenuate NAFLD possibly by affecting insulin resistance and lipid metabolism related pathways (e.g., PI3K-Akt, AMPK). Differentially expressed genes involved in these pathways such as Pik3r1, Foxo1, Foxo3, Scd1, Col3a1 and Fn1 might be candidate targets for treating NAFLD.
Collapse
Affiliation(s)
- Mingzhe Zhu
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Public Health College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shijun Hao
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Liu
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Yang
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peiyong Zheng
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Zhang
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
107
|
Dietary Flaxseed Oil Prevents Western-Type Diet-Induced Nonalcoholic Fatty Liver Disease in Apolipoprotein-E Knockout Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3256241. [PMID: 29081885 PMCID: PMC5610846 DOI: 10.1155/2017/3256241] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 06/28/2017] [Accepted: 07/05/2017] [Indexed: 02/08/2023]
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) has dramatically increased globally during recent decades. Intake of n-3 polyunsaturated fatty acids (PUFAs), mainly eicosapentaenoic acid (EPA, C20:5n-3) and docosahexaenoic acid (DHA, C22:6n-3), is believed to be beneficial to the development of NAFLD. However, little information is available with regard to the effect of flaxseed oil rich in α-linolenic acid (ALA, C18:3n-3), a plant-derived n-3 PUFA, in improving NAFLD. This study was to gain the effect of flaxseed oil on NAFLD and further investigate the underlying mechanisms. Apolipoprotein-E knockout (apoE-KO) mice were given a normal chow diet, a western-type high-fat and high-cholesterol diet (WTD), or a WTD diet containing 10% flaxseed oil (WTD + FO) for 12 weeks. Our data showed that consumption of flaxseed oil significantly improved WTD-induced NAFLD, as well as ameliorated impaired lipid homeostasis, attenuated oxidative stress, and inhibited inflammation. These data were associated with the modification effects on expression levels of genes involved in de novo fat synthesis (SREBP-1c, ACC), triacylglycerol catabolism (PPARα, CPT1A, and ACOX1), inflammation (NF-κB, IL-6, TNF-α, and MCP-1), and oxidative stress (ROS, MDA, GSH, and SOD).
Collapse
|
108
|
Yousef MH, Al Juboori A, Albarrak AA, Ibdah JA, Tahan V. Fatty liver without a large "belly": Magnified review of non-alcoholic fatty liver disease in non-obese patients. World J Gastrointest Pathophysiol 2017; 8:100-107. [PMID: 28868179 PMCID: PMC5561430 DOI: 10.4291/wjgp.v8.i3.100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 05/22/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is well described as a common cause of chronic liver disease, mostly in the obese population. It refers to a spectrum of chronic liver disease that starts with simple steatosis than progresses to nonalcoholic steatohepatitis and cirrhosis in patients without significant alcohol consumption. NAFLD in the non-obese population has been increasingly reported and studied recently. The pathogenesis of nonobese NAFLD is poorly understood and is related to genetic predisposition, most notably patatin-like phospholipase domain-containing 33 G allele polymorphism that leads to intrahepatic triglyceride accumulation and insulin resistance. Non-obese NAFLD is associated with components of metabolic syndrome and, especially, visceral obesity which seems to be an important etiological factor in this group. Dietary factors and, specifically, a high fructose diet seem to play a role. Cardiovascular events remain the main cause of mortality and morbidity in NAFLD, including in the non-obese population. There is not enough data regarding treatment in non-obese NAFLD patients, but similar to NAFLD in obese subjects, lifestyle changes that include dietary modification, physical activity, and weight loss remain the mainstay of treatment.
Collapse
|
109
|
Doulberis M, Kotronis G, Gialamprinou D, Kountouras J, Katsinelos P. Non-alcoholic fatty liver disease: An update with special focus on the role of gut microbiota. Metabolism 2017; 71:182-197. [PMID: 28521872 DOI: 10.1016/j.metabol.2017.03.013] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 03/19/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a significant global health burden in children, adolescents and adults with substantial rise in prevalence over the last decades. Accumulating data from manifold studies support the idea of NAFLD as a hepatic manifestation of metabolic syndrome, being rather a systemic metabolic disease than a liver confined pathology. Emerging data support that the gut microbiome represents a significant environmental factor contributing to NAFLD development and progression. Apart from other regimens, probiotics may have a positive role in the management of NAFLD through a plethora of possible mechanisms. The current review focuses on the NAFLD multifactorial pathogenesis, including mainly the role of intestinal microbiome and all relevant issues are raised. Furthermore, the clinical manifestations and appropriate diagnostic approach of the disease are discussed, with all possible therapeutic measures that can be taken, also including the potential beneficial effect of probiotics.
Collapse
Affiliation(s)
- Michael Doulberis
- Bürgerspital Hospital, Department of Internal Medicine, Solothurn 4500, Switzerland.
| | - Georgios Kotronis
- Agios Pavlos Hospital, Department of Internal Medicine, Thessaloniki, Macedonia, 55134, Greece
| | - Dimitra Gialamprinou
- Papageorgiou General Hospital, Department of Pediatrics, Aristotle University of Thessaloniki, Macedonia, 56403, Greece
| | - Jannis Kountouras
- Ippokration Hospital, Department of Internal Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, 54642, Greece
| | - Panagiotis Katsinelos
- Ippokration Hospital, Department of Internal Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, 54642, Greece
| |
Collapse
|
110
|
Bai X, Hong W, Cai P, Chen Y, Xu C, Cao D, Yu W, Zhao Z, Huang M, Jin J. Valproate induced hepatic steatosis by enhanced fatty acid uptake and triglyceride synthesis. Toxicol Appl Pharmacol 2017; 324:12-25. [PMID: 28366540 DOI: 10.1016/j.taap.2017.03.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/06/2017] [Accepted: 03/28/2017] [Indexed: 02/07/2023]
Abstract
Steatosis is the characteristic type of VPA-induced hepatotoxicity and may result in life-threatening hepatic lesion. Approximately 61% of patients treated with VPA have been diagnosed with hepatic steatosis through ultrasound examination. However, the mechanisms underlying VPA-induced intracellular fat accumulation are not yet fully understood. Here we demonstrated the involvement of fatty acid uptake and lipogenesis in VPA-induced hepatic steatosis in vitro and in vivo by using quantitative real-time PCR (qRT-PCR) analysis, western blotting analysis, fatty acid uptake assays, Nile Red staining assays, and Oil Red O staining assays. Specifically, we found that the expression of cluster of differentiation 36 (CD36), an important fatty acid transport, and diacylglycerol acyltransferase 2 (DGAT2) were significantly up-regulated in HepG2 cells and livers of C57B/6J mice after treatment with VPA. Furthermore, VPA treatment remarkably enhanced the efficiency of fatty acid uptake mediated by CD36, while this effect was abolished by the interference with CD36-specific siRNA. Also, VPA treatment significantly increased DGAT2 expression as a result of the inhibition of mitogen-activated protein kinase kinase (MEK) - extracellular regulated kinase (ERK) pathway; however, DGAT2 knockdown significantly alleviated VPA-induced intracellular lipid accumulation. Additionally, we also found that sterol regulatory element binding protein-1c (SREBP-1c)-mediated fatty acid synthesis may be not involved in VPA-induced hepatic steatosis. Overall, VPA-triggered over-regulation of CD36 and DGAT2 could be helpful for a better understanding of the mechanisms underlying VPA-induced hepatic steatosis and may offer novel therapeutic strategies to combat VPA-induced hepatotoxicity.
Collapse
Affiliation(s)
- Xupeng Bai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Weipeng Hong
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peiheng Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yibei Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Chuncao Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Di Cao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weibang Yu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhongxiang Zhao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jing Jin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
111
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a spectrum of chronic liver conditions that are characterized by steatosis, inflammation, fibrosis, and liver injury. The global prevalence of NAFLD is rapidly increasing in proportion to the rising incidence of obesity and type 2 diabetes. Because NAFLD is a multifaceted disorder with many underlying metabolic abnormalities, currently, there is no pharmacological agent that is therapeutically approved for the treatment of this disease. Folate is a water-soluble B vitamin that plays an essential role in one-carbon transfer reactions involved in nucleic acid biosynthesis, methylation reactions, and sulfur-containing amino acid metabolism. The liver is the primary organ responsible for storage and metabolism of folates. Low serum folate levels have been observed in patients with obesity and diabetes. It has been reported that a low level of endogenous folates in rodents perturbs folate-dependent one-carbon metabolism, and may be associated with development of metabolic diseases such as NAFLD. This review highlights the biological role of folate in the progression of NAFLD and its associated metabolic complications including obesity and type 2 diabetes. Understanding the role of folate in metabolic disease may position this vitamin as a potential therapeutic for NAFLD.
Collapse
Affiliation(s)
- Victoria Sid
- a St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Yaw L Siow
- a St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.,c Agriculture and Agri-Food Canada, Winnipeg, MB R3C 1B2, Canada
| | - Karmin O
- a St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.,d Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
112
|
Xue L, He J, Gao N, Lu X, Li M, Wu X, Liu Z, Jin Y, Liu J, Xu J, Geng Y. Probiotics may delay the progression of nonalcoholic fatty liver disease by restoring the gut microbiota structure and improving intestinal endotoxemia. Sci Rep 2017; 7:45176. [PMID: 28349964 PMCID: PMC5368635 DOI: 10.1038/srep45176] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/20/2017] [Indexed: 02/07/2023] Open
Abstract
Gut-derived bacterial lipopolysaccharide (LPS) and subsequent hepatic toll-like receptor 4 (TLR4) activation have been recognized to be involved in the onset of diet-induced nonalcoholic fatty liver disease (NAFLD), but little is known about the variation of LPS and TLR4 during the progression of NAFLD. Probiotics were able to inhibit proliferation of harmful bacteria and improve gastrointestinal barrier function. However, it's unclear whether LPS/TLR4 is involved in the protection effect of probiotics on NAFLD. In this study, we described characteristic of gut microbiota structure in the progression of NAFLD, and we also analyzed the relationship between gut microbiota and LPS/TLR4 in this process. Furthermore, we applied probiotics intervention to investigate the effect of probiotics on gut flora structure, intestinal integrity, serum LPS, liver TLR4 and liver pathology. Our results showed that serum LPS and liver TLR4 were highly increased during progression of NAFLD, with gut flora diversity and gut mircobiological colonization resistance (B/E) declining. Furthermore, probiotics could improve gut microbiota structure and liver pathology. Probiotics could also downregulate serum LPS and liver TLR4. Our results suggested that both gut flora alteration and endotoxemia may be involved in the progression of NAFLD. Probiotics may delay the progression of NAFLD via LPS/TLR4 signaling.
Collapse
Affiliation(s)
- Li Xue
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Juntao He
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Ning Gao
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xiaolan Lu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Ming Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaokang Wu
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zeshi Liu
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yaofeng Jin
- Department of Pathology, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jiali Liu
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jiru Xu
- Department of Immunology and Pathogenic Biology, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Geng
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
113
|
Liu C, Liao JZ, Li PY. Traditional Chinese herbal extracts inducing autophagy as a novel approach in therapy of nonalcoholic fatty liver disease. World J Gastroenterol 2017; 23:1964-1973. [PMID: 28373762 PMCID: PMC5360637 DOI: 10.3748/wjg.v23.i11.1964] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/23/2016] [Accepted: 01/18/2017] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the leading causes of chronic liver diseases around the world due to the modern sedentary and food-abundant lifestyle, which is characterized by excessive fat accumulation in the liver related with causes other than alcohol abuse. It is widely acknowledged that insulin resistance, dysfunctional lipid metabolism, endoplasmic reticulum stress, oxidative stress, inflammation, and apoptosis/necrosis may all contribute to NAFLD. Autophagy is a protective self-digestion of intracellular organelles, including lipid droplets (lipophagy), in response to stress to maintain homeostasis. Lipophagy is another pathway for lipid degradation besides lipolysis. It is reported that impaired autophagy also contributes to NAFLD. Some studies have suggested that the histological characteristics of NAFLD (steatosis, lobular inflammation, and peri-sinusoid fibrosis) might be improved by treatment with traditional Chinese herbal extracts, while autophagy may be induced. This review will provide insights into the characteristics of autophagy in NAFLD and the related role/mechanisms of autophagy induced by traditional Chinese herbal extracts such as resveratrol, Lycium barbarum polysaccharides, dioscin, bergamot polyphenol fraction, capsaicin, and garlic-derived S-allylmercaptocysteine, which may inhibit the progression of NAFLD. Regulation of autophagy/lipophagy with traditional Chinese herbal extracts may be a novel approach for treating NAFLD, and the molecular mechanisms should be elucidated further in the near future.
Collapse
|
114
|
Motamed N, Rabiee B, Poustchi H, Dehestani B, Hemasi GR, Khonsari MR, Maadi M, Saeedian FS, Zamani F. Non-alcoholic fatty liver disease (NAFLD) and 10-year risk of cardiovascular diseases. Clin Res Hepatol Gastroenterol 2017; 41:31-38. [PMID: 27597641 DOI: 10.1016/j.clinre.2016.07.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/15/2016] [Accepted: 07/18/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS The association between cardiovascular diseases (CVD) and non-alcoholic fatty liver disease (NAFLD) was confirmed by a large body of evidence. This study was conducted to determine the association between NAFLD and 10-year CVD risk. METHODS This study utilized the data of 2804 subjects aged 40-74 years from a cohort study of northern Iran. Two CVD risk assessment tools, American College of Cardiology/American Heart Association and Framingham general cardiovascular risk profile for use in primary care, were utilized to determine the 10-year CVD risk in patients with NAFLD and the individuals without this condition. The mean risks were compared between these two groups. RESULTS Using ACC/AHA approach, the mean risk in male participants suffering NAFLD was 14.2%, while in men without NAFLD was 11.7% (P-value < 0.0001). Using Framingham approach, the mean risks were 16.0 and 12.7% in men with and without NAFLD, respectively (P-value < 0.0001). Using ACC/AHA approach, the mean risks in female participants with and without NAFLD were 6.7 and 4.6%, respectively (P-value < 0.0001). Applying Framingham approach, the mean risk was 8.2% in women with NAFLD and 5.4% in women without NAFLD (P-value < 0.0001). CONCLUSION The individuals with NAFLD had a higher risk of 10-year CVD events than individuals without NAFLD, according to both ACC/AHA tool and primary care version of Framingham tool. A large proportion of NAFLD patients fulfill the criteria of statin therapy recommendation, suggesting that statin therapy could reduce 10-year CVD risk in NAFLD patients.
Collapse
Affiliation(s)
- Nima Motamed
- Department of Social Medicine, Zanjan University of Medical Sciences, Gavazang Road, Zanjan 45139-56184, Iran.
| | - Behnam Rabiee
- Department of Gastroenterology and Liver Disease, Gastrointestinal and Liver Disease Research Center (GILDRC), Tehran Firoozgar Hospital, Iran University of Medical Sciences, Beh Afarin St., Karim Khan Zand Ave, Tehran 15900, Iran.
| | - Hossein Poustchi
- Digestive Diseases Research Institute (DDRI), Shariati Hospital, Tehran University of Medical Sciences, Kargar Shomali Ave, Tehran 14117-13135, Iran.
| | - Babak Dehestani
- Center for Cardiovascular Research, Washington University in Saint Louis School of Medicine, 660 South Euclid Avenue, Campus Box 8086, Saint Louis, MO 63110, USA.
| | - Gholam Reza Hemasi
- Department of Gastroenterology and Liver Disease, Gastrointestinal and Liver Disease Research Center (GILDRC), Tehran Firoozgar Hospital, Iran University of Medical Sciences, Beh Afarin St., Karim Khan Zand Ave, Tehran 15900, Iran.
| | - Mahmood Reza Khonsari
- Department of Gastroenterology and Liver Disease, Gastrointestinal and Liver Disease Research Center (GILDRC), Tehran Firoozgar Hospital, Iran University of Medical Sciences, Beh Afarin St., Karim Khan Zand Ave, Tehran 15900, Iran.
| | - Mansooreh Maadi
- Department of Gastroenterology and Liver Disease, Gastrointestinal and Liver Disease Research Center (GILDRC), Tehran Firoozgar Hospital, Iran University of Medical Sciences, Beh Afarin St., Karim Khan Zand Ave, Tehran 15900, Iran.
| | - Fatemeh Sima Saeedian
- Department of Gastroenterology and Liver Disease, Gastrointestinal and Liver Disease Research Center (GILDRC), Tehran Firoozgar Hospital, Iran University of Medical Sciences, Beh Afarin St., Karim Khan Zand Ave, Tehran 15900, Iran.
| | - Farhad Zamani
- Department of Gastroenterology and Liver Disease, Gastrointestinal and Liver Disease Research Center (GILDRC), Tehran Firoozgar Hospital, Iran University of Medical Sciences, Beh Afarin St., Karim Khan Zand Ave, Tehran 15900, Iran.
| |
Collapse
|
115
|
Antioxidant properties of tea blunt ROS-dependent lipogenesis: beneficial effect on hepatic steatosis in a high fat-high sucrose diet NAFLD obese rat model. J Nutr Biochem 2017; 40:95-104. [DOI: 10.1016/j.jnutbio.2016.10.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/19/2016] [Accepted: 10/05/2016] [Indexed: 12/22/2022]
|
116
|
Brøns C, Grunnet LG. MECHANISMS IN ENDOCRINOLOGY: Skeletal muscle lipotoxicity in insulin resistance and type 2 diabetes: a causal mechanism or an innocent bystander? Eur J Endocrinol 2017; 176:R67-R78. [PMID: 27913612 DOI: 10.1530/eje-16-0488] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/19/2016] [Accepted: 09/14/2016] [Indexed: 12/11/2022]
Abstract
Dysfunctional adipose tissue is associated with an increased risk of developing type 2 diabetes (T2D). One characteristic of a dysfunctional adipose tissue is the reduced expandability of the subcutaneous adipose tissue leading to ectopic storage of fat in organs and/or tissues involved in the pathogenesis of T2D that can cause lipotoxicity. Accumulation of lipids in the skeletal muscle is associated with insulin resistance, but the majority of previous studies do not prove any causality. Most studies agree that it is not the intramuscular lipids per se that causes insulin resistance, but rather lipid intermediates such as diacylglycerols, fatty acyl-CoAs and ceramides and that it is the localization, composition and turnover of these intermediates that play an important role in the development of insulin resistance and T2D. Adipose tissue is a more active tissue than previously thought, and future research should thus aim at examining the exact role of lipid composition, cellular localization and the dynamics of lipid turnover on the development of insulin resistance. In addition, ectopic storage of fat has differential impact on various organs in different phenotypes at risk of developing T2D; thus, understanding how adipogenesis is regulated, the interference with metabolic outcomes and what determines the capacity of adipose tissue expandability in distinct population groups is necessary. This study is a review of the current literature on the adipose tissue expandability hypothesis and how the following ectopic lipid accumulation as a consequence of a limited adipose tissue expandability may be associated with insulin resistance in muscle and liver.
Collapse
Affiliation(s)
- Charlotte Brøns
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark
| | - Louise Groth Grunnet
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
117
|
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: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/27/2016] [Accepted: 11/21/2016] [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
Affiliation(s)
- Vanessa Souza-Mello
- Vanessa Souza-Mello, Biomedical Centre, Institute of Biology, Department of Anatomy, State University of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| |
Collapse
|
118
|
Mahamid M, Abu-Elhija O, Yassin T, Nseir W. Advanced Hepatic Fibrosis in Fatty Liver Disease Linked to Hyperplastic Colonic Polyp. Can J Gastroenterol Hepatol 2017; 2017:2054871. [PMID: 28127545 PMCID: PMC5239862 DOI: 10.1155/2017/2054871] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Aim. Our study aims to determine possible association between biopsy-proven nonalcoholic steatohepatitis (NASH) and hyperplastic polyps (HP) of the colon. Methods. A retrospective cohort observational study. All subjects underwent screening colonoscopy within two years. Data were extracted from the patient charts including demographic, anthropometric measurement, vital signs, underlying diseases, medical therapy, laboratory data, results of the liver biopsy with degree of fibrosis and necroinflammatory activity, the colonoscopy report, and the pathological report of the extracted polyp. Results. A total of 223 patients were included in our study, 123 patients with biopsy-proven NASH and 100 patients without NASH who served as the control group matched for age. 14 colonic adenomas (11% of patients) were found in the NASH group compared with 16 adenomas (16% of patients) found in the control group (P = 0.9). 28 HPs were found in the NASH group (22.7%) compared with only 8 HPs in the control group (8%) (P < 0.05). 21 from the 28 (75%) HPs diagnosed in the NASH group were observed in the high degree fibrosis patients (Fibrosis Stages 3 and 4), 6 HPs (21%) were associated with Fibrosis Stages 1 and 2, and single HP (4%) was associated with Fibrosis Stage 0. Conclusions. Our study showed an association between biopsy-proven steatohepatitis and the burden of hyperplastic polyp. The severity of hepatic fibrosis may play important role in the increased occurrence of HPs.
Collapse
Affiliation(s)
- Mahmud Mahamid
- Internal Medicine Department, Holy Family Hospital, Nazareth, Israel
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Omar Abu-Elhija
- Internal Medicine Department, Holy Family Hospital, Nazareth, Israel
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Tarik Yassin
- Internal Medicine Department, The Baruch Padeh Medical Center, Poriya, Israel
| | - William Nseir
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
- Internal Medicine Department, EMMS the Nazareth Hospital, Nazareth, Israel
| |
Collapse
|
119
|
Decaris ML, Li KW, Emson CL, Gatmaitan M, Liu S, Wang Y, Nyangau E, Colangelo M, Angel TE, Beysen C, Cui J, Hernandez C, Lazaro L, Brenner DA, Turner SM, Hellerstein MK, Loomba R. Identifying nonalcoholic fatty liver disease patients with active fibrosis by measuring extracellular matrix remodeling rates in tissue and blood. Hepatology 2017; 65:78-88. [PMID: 27706836 PMCID: PMC5516243 DOI: 10.1002/hep.28860] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 08/02/2016] [Accepted: 09/26/2016] [Indexed: 12/12/2022]
Abstract
UNLABELLED Excess collagen synthesis (fibrogenesis) in the liver plays a causal role in the progression of nonalcoholic fatty liver disease (NAFLD). Methods are needed to identify patients with more rapidly progressing disease and to demonstrate early response to treatment. We describe here a novel method to quantify hepatic fibrogenesis flux rates both directly in liver tissue and noninvasively in blood. Twenty-one patients with suspected NAFLD ingested heavy water (2 H2 O, 50-mL aliquots) two to three times daily for 3-5 weeks prior to a clinically indicated liver biopsy. Liver collagen fractional synthesis rate (FSR) and plasma lumican FSR were measured based on 2 H labeling using tandem mass spectrometry. Patients were classified by histology for fibrosis stage (F0-F4) and as having nonalcoholic fatty liver or nonalcoholic steatohepatitis (NASH). Magnetic resonance elastography measurements of liver stiffness were also performed. Hepatic collagen FSR in NAFLD increased with advancing disease stage (e.g., higher in NASH than nonalcoholic fatty liver, positive correlation with fibrosis score and liver stiffness) and correlated with hemoglobin A1C. In addition, plasma lumican FSR demonstrated a significant correlation with hepatic collagen FSR. CONCLUSION Using a well-characterized cohort of patients with biopsy-proven NAFLD, this study demonstrates that hepatic scar in NASH is actively remodeled even in advanced fibrosis, a disease that is generally regarded as static and slowly progressive. Moreover, hepatic collagen FSR correlates with established risks for fibrotic disease progression in NASH, and plasma lumican FSR correlates with hepatic collagen FSR, suggesting applications as direct or surrogate markers, respectively, of hepatic fibrogenesis in humans. (Hepatology 2017;65:78-88).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jeffrey Cui
- NAFLD Research CenterDivision of GastroenterologyDepartment of MedicineUniversity of California at San DiegoLa JollaCA
| | - Carolyn Hernandez
- NAFLD Research CenterDivision of GastroenterologyDepartment of MedicineUniversity of California at San DiegoLa JollaCA
| | - Len Lazaro
- NAFLD Research CenterDivision of GastroenterologyDepartment of MedicineUniversity of California at San DiegoLa JollaCA
| | - David A. Brenner
- NAFLD Research CenterDivision of GastroenterologyDepartment of MedicineUniversity of California at San DiegoLa JollaCA
| | | | - Marc K. Hellerstein
- KineMed, IncEmeryvilleCA,Department of Nutritional Sciences and ToxicologyUniversity of California at BerkeleyBerkeleyCA
| | - Rohit Loomba
- NAFLD Research CenterDivision of GastroenterologyDepartment of MedicineUniversity of California at San DiegoLa JollaCA
| |
Collapse
|
120
|
Webster NJG. Alternative RNA Splicing in the Pathogenesis of Liver Disease. Front Endocrinol (Lausanne) 2017; 8:133. [PMID: 28680417 PMCID: PMC5478874 DOI: 10.3389/fendo.2017.00133] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/30/2017] [Indexed: 12/27/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is becoming increasingly prevalent due to the worldwide obesity epidemic and currently affects one-third of adults or about one billion people worldwide. NAFLD is predicted to affect over 50% of the world's population by the end of the next decade. It is the most common form of liver disease and is associated with increased risk for progression to a more severe form non-alcoholic steatohepatitis, as well as insulin resistance, type 2 diabetes mellitus, cirrhosis, and eventually hepatocellular carcinoma. This review article will focus on the role of alternative splicing in normal liver physiology and dysregulation in liver disease.
Collapse
Affiliation(s)
- Nicholas J. G. Webster
- Medical Research Service, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Medicine, School of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- *Correspondence: Nicholas J. G. Webster,
| |
Collapse
|
121
|
Wang H, Wang D, Yang L, Wang Y, Jia J, Na D, Chen H, Luo Y, Liu C. Compact bone-derived mesenchymal stem cells attenuate nonalcoholic steatohepatitis in a mouse model by modulation of CD4 cells differentiation. Int Immunopharmacol 2016; 42:67-73. [PMID: 27889556 DOI: 10.1016/j.intimp.2016.11.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/03/2016] [Accepted: 11/14/2016] [Indexed: 12/29/2022]
Abstract
Increasing evidence has accrued which indicates that mesenchymal stem cells (MSCs) have a potential clinical value in the treatment of certain diseases. Globally, nonalcoholic steatohepatitis (NASH) is a widespread disorder. In the present study, MSCs were isolated successfully from compact bone and a mouse model of NASH was established as achieved with use of a methionine-choline deficient (MCD) diet. Compact bone-derived MSCs transplantation reduced MCD diet-induced weight loss, hepatic lipid peroxidation, steatosis, ballooning, lobular inflammation and fibrogenesis. It was shown that MSCs treatment hampered MCD diet-induced proliferation of CD4+ IFN-γ+ and CD4+IL-6+ T spleen cells. In addition, CD4+IL-17+ lymphocytes that associated with anti-inflammation show little change in MCD as well as in MCD+MSCs splenocytes. We conclude that MSCs may have a potential clinical value upon NASH, through their capacity to suppress activation of CD4+ IFN-γ+ and CD4+IL-6+ lymphocytes.
Collapse
Affiliation(s)
- Huafeng Wang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China.
| | - Dong Wang
- Central Blood Station of Tianjin, Tianjin, China
| | - Luhong Yang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Yanxia Wang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Junli Jia
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Dongchen Na
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Huize Chen
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Yongping Luo
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Chengfang Liu
- Department of Human anatomy, Shanxi Medical University, Taiyuan, China; Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
122
|
Mikolasevic I, Orlic L, Franjic N, Hauser G, Stimac D, Milic S. Transient elastography (FibroScan(®)) with controlled attenuation parameter in the assessment of liver steatosis and fibrosis in patients with nonalcoholic fatty liver disease - Where do we stand? World J Gastroenterol 2016; 22:7236-7251. [PMID: 27621571 PMCID: PMC4997649 DOI: 10.3748/wjg.v22.i32.7236] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/28/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide. Currently, the routinely used modalities are unable to adequately determine the levels of steatosis and fibrosis (laboratory tests and ultrasonography) or cannot be applied as a screening procedure (liver biopsy). Among the non-invasive tests, transient elastography (FibroScan(®), TE) with controlled attenuation parameter (CAP) has demonstrated good accuracy in quantifying the levels of liver steatosis and fibrosis in patients with NAFLD, the factors associated with the diagnosis and NAFLD progression. The method is fast, reliable and reproducible, with good intra- and interobserver levels of agreement, thus allowing for population-wide screening and disease follow-up. The initial inability of the procedure to accurately determine fibrosis and steatosis in obese patients has been addressed with the development of the obese-specific XL probe. TE with CAP is a viable alternative to ultrasonography, both as an initial assessment and during follow-up of patients with NAFLD. Its ability to exclude patients with advanced fibrosis may be used to identify low-risk NAFLD patients in whom liver biopsy is not needed, therefore reducing the risk of complications and the financial costs.
Collapse
|
123
|
Bensaid S, Kachenoura A, Costet N, De Ledinghen V, Vergniol J, Laine F, Turlin B, Tariel H, Senhadji L. Early diagnosis of NAFLD-NASH transition using mid infrared spectroscopy. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2016; 2016:3602-3605. [PMID: 28269075 DOI: 10.1109/embc.2016.7591507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is defined as an excessive accumulation of fat in the liver in the absence of excessive drinking of alcohol. Initially considered as benign and self-limited, NAFLD may progress to the malignant stage of non-alcoholic steatohepatitis (NASH) characterized by degenerate hepatocellular ballooning and lobular inflammation. NASH can lead to hepatic fibrosis and ultimately to cirrhosis and hepatocellular carcinoma. Unfortunately, the transition from NAFLD to NASH is difficult to detect so far. In this paper, we propose to evaluate the characterization of NASH using mid infrared fiber evanescent wave spectroscopy on blood serum. We used an heuristic variable selection method and a generalized linear model to classify NAFLD and NASH spectra. The obtained results proved that this technique is a promising non-invasive and simple diagnosis tool for NASH.
Collapse
|
124
|
Smedlund K, Dube P, Vazquez G. Early steatohepatitis in hyperlipidemic mice with endothelial-specific gain of TRPC3 function precedes changes in aortic atherosclerosis. Physiol Genomics 2016; 48:644-9. [PMID: 27449657 DOI: 10.1152/physiolgenomics.00067.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/18/2016] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its more advanced form nonalcoholic steatohepatitis (NASH) are the most common chronic liver diseases in developed countries. Moreover, NAFLD and NASH are considerable risk factors for atherosclerosis, the most frequent vascular pathology in these and other metabolic diseases. Despite this strong connection, current knowledge of the relationship between NAFLD/NASH and atherosclerosis is scarce. Recently, we studied hyperlipidemic Apoe knockout mice with endothelial-specific gain of transient receptor potential canonical 3 channel function (TgESTRPC3/ApoeKO) and found that these animals had increased burden of advanced aortic atherosclerosis (16 wk on high-fat diet) compared with nontransgenic ApoeKO littermate controls (non-Tg/ApoeKO), whereas early lesions (10 wk on high-fat diet) were not different. Here, we report that at the early stage when differences in aortic atherosclerosis are not yet manifest, the livers of TgESTRPC3/ApoeKO mice show steatosis, fibrosis, and altered hepatic enzymes compared with non-Tg/ApoeKO animals. Because differences in liver pathology were noticeable long before differences in atherosclerosis were evident, our studies suggest that TRPC3-related endothelial mechanisms that promote steatohepatitis may also contribute to atherosclerosis progression. In vitro, downregulation of TRPC3 in liver sinusoid endothelial cells reduces their susceptibility to endoplasmic reticulum stress-induced apoptosis, suggesting that a proapoptotic effect of TRPC3 may add to other fibrogenic factors in vivo. These novel findings show a positive association between augmented expression of an endothelial TRPC channel, development of early steatohepatitis, and atherosclerotic burden in a hyperlipidemic mouse model of NAFLD fed conventional Western-type diet.
Collapse
Affiliation(s)
- Kathryn Smedlund
- Department of Physiology and Pharmacology, and Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, Health Science Campus, Toledo, Ohio
| | - Prabhatachandra Dube
- Department of Physiology and Pharmacology, and Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, Health Science Campus, Toledo, Ohio
| | - Guillermo Vazquez
- Department of Physiology and Pharmacology, and Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, Health Science Campus, Toledo, Ohio
| |
Collapse
|
125
|
Jiang Tang Xiao Ke Granule, a Classic Chinese Herbal Formula, Improves the Effect of Metformin on Lipid and Glucose Metabolism in Diabetic Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:1592731. [PMID: 27418937 PMCID: PMC4932160 DOI: 10.1155/2016/1592731] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 04/30/2016] [Accepted: 05/24/2016] [Indexed: 12/19/2022]
Abstract
In the present study, the hypoglycemic, hypolipidemic, and antioxidative effects of metformin (MET) combined with Jiang Tang Xiao Ke (JTXK) granule derived from the “Di Huang Tang” were evaluated in mice with type 2 diabetes mellitus (DM) induced by high-fat diet/streptozotocin. DM mice were orally treated with MET (0.19 g/kg) either alone or combined with different doses (1.75, 3.5, or 7 g/kg) of JTXK for 4 weeks. Results showed that the serum and hepatic glucose, lipids, and oxidative stress levels were elevated in DM mice, when compared with the normal mice. MET treatment decreased FBG and serum glucagon levels of DM mice. Combination treatment with MET and JTXK 3.5 g/kg increased the hypoglycemia and insulin sensitivity at 4 weeks when compared with the DM mice treated with MET alone. However, neither MET nor MET/JTXK treatment could completely reverse the hyperglycemia in DM mice. JTXK enhanced the serum triglyceride (TG) and hepatic lipid-lowering effect of MET in a dose-dependent manner in DM mice. JTXK 1.75 and 3.5 g/kg improved the hepatoprotective effect of MET in DM mice. Synergistic effect of combination treatment with MET and JTXK on antioxidant stress was also found in DM mice compared with MET alone.
Collapse
|
126
|
Xu YY, Ge J, Zhang MH, Sun WJ, Zhang J, Yu PL, Zheng YF, Yang J, Zhu XQ. Intravenous Administration of Multiwalled Carbon Nanotubes Aggravates High-Fat Diet-Induced Nonalcoholic Steatohepatitis in Sprague Dawley Rats. Int J Toxicol 2016; 35:634-643. [PMID: 27306319 DOI: 10.1177/1091581816653363] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multiwalled carbon nanotubes (MWCNTs) have been explored in pharmaceutical applications such as tumor targeting and delivery of drugs, in which MWCNTs are given through intravenous injection. However, the biosafety of MWCNTs is of concern for such application. Therefore, in the current study, we used a fatty liver model to investigate the possible toxicity of MWCNTs to the liver, as MWCNTs were retained mainly in the liver of mice after intravenous injection. Male Sprague Dawley rats were used to generate the fatty liver model, and the effects of intravenous administration of MWCNTs on fatty liver were studied. Hematoxylin and eosin staining for hepatocellular anatomy and Masson trichrome staining for hepatic fibrosis were conducted. Histologically, MWCNTs aggravated steatohepatitis with higher nonalcoholic fatty liver disease scores. Analysis of liver injury markers indicated that MWCNTs administration resulted in chronic hepatitis, along with increased liver fat and altered liver oxidation, including the increase of P6 protein and the depletion of glutathione. In conclusion, our results suggest that MWCNTs can aggravate nonalcoholic steatohepatitis in Sprague Dawley rats, and oxidative injury may be involved in this process.
Collapse
Affiliation(s)
- Yu-Ying Xu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Juan Ge
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mo-Han Zhang
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wen-Jie Sun
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun Zhang
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Pei-Lin Yu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi-Fan Zheng
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun Yang
- Department of Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, National Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin-Qiang Zhu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
127
|
Liu W, Baker RD, Bhatia T, Zhu L, Baker SS. Pathogenesis of nonalcoholic steatohepatitis. Cell Mol Life Sci 2016; 73:1969-87. [PMID: 26894897 PMCID: PMC11108381 DOI: 10.1007/s00018-016-2161-x] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/19/2016] [Accepted: 02/09/2016] [Indexed: 02/06/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a severe form of nonalcoholic fatty liver disease and a risk factor for cirrhosis and hepatocellular carcinoma. The pathological features of NASH include steatosis, hepatocyte injury, inflammation, and various degrees of fibrosis. Steatosis reflects disordered lipid metabolism. Insulin resistance and excessive fatty acid influx to the liver are two important contributing factors. Steatosis is also likely associated with lipotoxicity and cellular stresses such as oxidative stress and endoplasmic reticulum stress, which result in hepatocyte injury. Inflammation and fibrosis are frequently triggered by various signals such as proinflammatory cytokines and chemokines, released by injuried hepatocytes and activated Kupffer cells. Although much progress has been made, the pathogenesis of NASH is not fully elucidated. The purpose of this review is to discuss the current understanding of NASH pathogenesis, mainly focusing on factors contributing to steatosis, hepatocyte injury, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Wensheng Liu
- Department of Pediatrics, Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, The State University of New York at Buffalo (SUNY Buffalo), 3435 Main Street, 422 BRB, Buffalo, NY, 14214, USA.
| | - Robert D Baker
- Department of Pediatrics, Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, The State University of New York at Buffalo (SUNY Buffalo), 3435 Main Street, 422 BRB, Buffalo, NY, 14214, USA
| | - Tavleen Bhatia
- Department of Pediatrics, Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, The State University of New York at Buffalo (SUNY Buffalo), 3435 Main Street, 422 BRB, Buffalo, NY, 14214, USA
| | - Lixin Zhu
- Department of Pediatrics, Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, The State University of New York at Buffalo (SUNY Buffalo), 3435 Main Street, 422 BRB, Buffalo, NY, 14214, USA
| | - Susan S Baker
- Department of Pediatrics, Digestive Diseases and Nutrition Center, Women and Children's Hospital of Buffalo, The State University of New York at Buffalo (SUNY Buffalo), 3435 Main Street, 422 BRB, Buffalo, NY, 14214, USA.
| |
Collapse
|
128
|
Cepero-Donates Y, Lacraz G, Ghobadi F, Rakotoarivelo V, Orkhis S, Mayhue M, Chen YG, Rola-Pleszczynski M, Menendez A, Ilangumaran S, Ramanathan S. Interleukin-15-mediated inflammation promotes non-alcoholic fatty liver disease. Cytokine 2016; 82:102-11. [PMID: 26868085 DOI: 10.1016/j.cyto.2016.01.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 01/23/2023]
Abstract
Interleukin-15 (IL-15) is essential for the homeostasis of lymphoid cells particularly memory CD8(+) T cells and NK cells. These cells are abundant in the liver, and are implicated in obesity-associated pathogenic processes. Here we characterized obesity-associated metabolic and cellular changes in the liver of mice lacking IL-15 or IL-15Rα. High fat diet-induced accumulation of lipids was diminished in the livers of mice deficient for IL-15 or IL-15Rα. Expression of enzymes involved in the transport of lipids in the liver showed modest differences. More strikingly, the liver tissues of IL15-KO and IL15Rα-KO mice showed decreased expression of chemokines CCl2, CCL5 and CXCL10 and reduced infiltration of mononuclear cells. In vitro, IL-15 stimulation induced chemokine gene expression in wildtype hepatocytes, but not in IL15Rα-deficient hepatocytes. Our results show that IL-15 is implicated in the high fat diet-induced lipid accumulation and inflammation in the liver, leading to fatty liver disease.
Collapse
Affiliation(s)
- Yuneivy Cepero-Donates
- Division of Immunology, Departments of Pediatrics, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Grégory Lacraz
- Division of Immunology, Departments of Pediatrics, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada; Hubrecht Institute, University Medical Center, Utrecht, The Netherlands
| | - Farnaz Ghobadi
- Division of Immunology, Departments of Pediatrics, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Volatiana Rakotoarivelo
- Division of Immunology, Departments of Pediatrics, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Sakina Orkhis
- Division of Immunology, Departments of Pediatrics, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Marian Mayhue
- Division of Immunology, Departments of Pediatrics, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Yi-Guang Chen
- Department of Pediatrics, Max McGee National Research Center for Juvenile Diabetes, Medical College of Wisconsin, Milwaukee, USA
| | - Marek Rola-Pleszczynski
- Division of Immunology, Departments of Pediatrics, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada; CRCHUS, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Alfredo Menendez
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada; CRCHUS, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Subburaj Ilangumaran
- Division of Immunology, Departments of Pediatrics, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada; CRCHUS, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Sheela Ramanathan
- Division of Immunology, Departments of Pediatrics, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada; CRCHUS, Sherbrooke, Québec, QC J1H 5N4, Canada.
| |
Collapse
|
129
|
Sun DQ, Liu WY, Wu SJ, Zhu GQ, Braddock M, Zhang DC, Shi KQ, Song D, Zheng MH. Increased levels of low-density lipoprotein cholesterol within the normal range as a risk factor for nonalcoholic fatty liver disease. Oncotarget 2016; 7:5728-5737. [PMID: 26735337 PMCID: PMC4868717 DOI: 10.18632/oncotarget.6799] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 12/04/2015] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Dyslipidemia exists within the setting of NAFLD and the relationship of a normal level of low-density lipoprotein cholesterol (LDL-c) with NAFLD is largely unknown. This large population-based study aimed to investigate the association between LDL-c levels within the normal range and the incidence of NAFLD. METHODS A total of 60527 subjects from 2 medical centers who had undergone liver ultrasonography were initially enrolled into this study. NAFLD was defined by ultrasonographic detection of steatosis in the absence of other liver disease. Subjects were divided into 4 groups (Q1 to Q4) by normal LDL-c quartiles : Q1: ≤ 2.00, Q2: 2.10-2.35, Q3: 2.36-2.68 and Q4: 2.69-3.12 mmol/L. The odds ratios (OR), hazard ratio (HR) and 95% confidence intervals (CIs) for NAFLD were calculated across each quartile of LDL-c, using the Q1 as reference. RESULTS The prevalence rates of NAFLD in a cross-sectional population from Q1 to Q4 were 19.34%, 25.86%, 35.65% and 42.08%, respectively. The OR for NAFLD in the cross-sectional population were 1.31 (95% CI 1.14-1.54), 1.73 (95% CI 1.46-2.04), and 1.82 (95% CI 1.49-2.23), respectively, after adjusting for known confounding variables. The HR for NAFLD in the longitudinal population were 1.23 (95% CI 1.12-1.35), 1.57 (95% CI 1.44-1.72) and 2.02 (95% CI 1.86-2.21), compared with Q1. Subjects with higher LDL-c level within the normal range had an increased cumulative incidence rate of NAFLD. CONCLUSIONS Increased levels of LDL-c within the normal range may play a significant role in the prevalence and incidence of NAFLD, independent of other confounding factors.
Collapse
Affiliation(s)
- Dan-Qin Sun
- Department of Nephrology, Affiliated Wuxi Second Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Wen-Yue Liu
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Sheng-Jie Wu
- Department of Cardiovascular Medicine, the Heart Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Gui-Qi Zhu
- Department of Infection and Liver Diseases, Liver Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Martin Braddock
- Global Medicines Development, AstraZeneca R&D, Alderley Park, United Kingdom
| | - Dong-Chu Zhang
- Wenzhou Medical Center, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Ke-Qing Shi
- Department of Infection and Liver Diseases, Liver Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou 325000, China
| | - Dan Song
- Department of Nephrology, Affiliated Wuxi Second Hospital, Nanjing Medical University, Wuxi 214002, China
| | - Ming-Hua Zheng
- Department of Infection and Liver Diseases, Liver Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
130
|
Cepero-Donates Y, Rakotoarivelo V, Mayhue M, Ma A, Chen YG, Ramanathan S. Homeostasis of IL-15 dependent lymphocyte subsets in the liver. Cytokine 2016; 82:95-101. [PMID: 26778709 DOI: 10.1016/j.cyto.2015.12.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 12/09/2015] [Accepted: 12/11/2015] [Indexed: 01/01/2023]
Abstract
IL-15 is a member of the gamma chain family of cytokines (γc - CD132). The IL-15 receptor (IL-15R) complex consists of 3 subunits: the ligand-binding IL-15Rα chain (CD215), the β chain (CD122; also used by IL-2), and the common γ chain. The biological activities of IL-15 are mostly mediated by the IL-15:IL-15Rα complex, produced by the same cell and 'trans-presented' to responder cells expressing the IL-15Rβγc. The peculiar and almost unique requirement for IL-15 to be trans-presented by IL-15Rα suggests that the biological effects of IL-15 signaling are tightly regulated even at the level of availability of IL-15. Tissue-specific deletion of IL-15Rα has shown macrophage-and dendritic cell-derived IL-15Rα mediate the homeostasis of different CD8(+) T cell subsets. Here we show that hepatocyte and macrophage- specific expression of IL-15Rα is required to maintain the homeostasis of NK and NKT cells in the liver. Thus, homeostasis of IL-15-dependent lymphocyte subsets is also regulated by trans-presentation of IL-15 by non-hematopoietic cells in the tissue environment.
Collapse
Affiliation(s)
- Yuneivy Cepero-Donates
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Volatiana Rakotoarivelo
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Marian Mayhue
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yi-Guang Chen
- Department of Pediatrics, Max McGee National Research Center for Juvenile Diabetes, Medical College of Wisconsin, Milwaukee, USA
| | - Sheela Ramanathan
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada; CRCHUS, Sherbrooke, Québec, QC J1H 5N4, Canada.
| |
Collapse
|
131
|
Wang Y, Liu Z, Zou W, Hong H, Fang H, Tong W. Molecular regulation of miRNAs and potential biomarkers in the progression of hepatic steatosis to NASH. Biomark Med 2015; 9:1189-200. [PMID: 26506944 DOI: 10.2217/bmm.15.70] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence suggests that microRNAs regulate diverse biological functions in the liver and play a very important function in metabolic-related disorders such as nonalcoholic fatty liver disease via regulating their target genes expression. In this review, we summarized the most recent progress in identification of miRNAs involving in the progression of liver steatosis and discussed the possible mechanisms by which miRNAs contribute to the diverse pathogenic liver injuries. We provide insights into the functional network of miRNAs by connecting miRNAs, their targets and biological pathways associated to hepatic steatosis and fibrosis, with important implications for our understanding of phenotypic-based disease pathogenesis. We also discuss the possible roles and challenges of miRNAs as biomarkers for drug-induced liver injury.
Collapse
Affiliation(s)
- Yuping Wang
- Division of Bioinformatics & Biostatistics, National Center for Toxicological Research, US FDA, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Zhichao Liu
- Division of Bioinformatics & Biostatistics, National Center for Toxicological Research, US FDA, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Wen Zou
- Division of Bioinformatics & Biostatistics, National Center for Toxicological Research, US FDA, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Huixiao Hong
- Division of Bioinformatics & Biostatistics, National Center for Toxicological Research, US FDA, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Hong Fang
- Office of Scientific Coordination, National Center for Toxicological Research, US FDA, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Weida Tong
- Division of Bioinformatics & Biostatistics, National Center for Toxicological Research, US FDA, 3900 NCTR Road, Jefferson, AR 72079, USA
| |
Collapse
|