51
|
Evers R, Piquette-Miller M, Polli JW, Russel FGM, Sprowl JA, Tohyama K, Ware JA, de Wildt SN, Xie W, Brouwer KLR. Disease-Associated Changes in Drug Transporters May Impact the Pharmacokinetics and/or Toxicity of Drugs: A White Paper From the International Transporter Consortium. Clin Pharmacol Ther 2018; 104:900-915. [PMID: 29756222 PMCID: PMC6424581 DOI: 10.1002/cpt.1115] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/23/2018] [Accepted: 05/07/2018] [Indexed: 12/11/2022]
Abstract
Drug transporters are critically important for the absorption, distribution, metabolism, and excretion (ADME) of many drugs and endogenous compounds. Therefore, disruption of these pathways by inhibition, induction, genetic polymorphisms, or disease can have profound effects on overall physiology, drug pharmacokinetics, drug efficacy, and toxicity. This white paper provides a review of changes in transporter function associated with acute and chronic disease states, describes regulatory pathways affecting transporter expression, and identifies opportunities to advance the field.
Collapse
Affiliation(s)
- Raymond Evers
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey, USA
| | | | - Joseph W Polli
- Mechanistic Safety and Drug Disposition, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jason A Sprowl
- Department of Pharmaceutical, Social and Administrative Sciences, School of Pharmacy, D'Youville College School, Buffalo, New York, USA
| | - Kimio Tohyama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company, Fujisawa, Kanagawa, Japan
| | - Joseph A Ware
- Department of Small Molecule Pharmaceutical Sciences, Genentech, South San Francisco, California, USA
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology and Department of Intensive Care, Radboud University Medical Center, Nijmegen, The Netherlands, and Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
52
|
Fabre T, Molina MF, Soucy G, Goulet JP, Willems B, Villeneuve JP, Bilodeau M, Shoukry NH. Type 3 cytokines IL-17A and IL-22 drive TGF-β-dependent liver fibrosis. Sci Immunol 2018; 3:eaar7754. [PMID: 30366940 DOI: 10.1126/sciimmunol.aar7754] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/04/2018] [Accepted: 09/26/2018] [Indexed: 12/17/2023]
Abstract
Inflammatory immune cells can modulate activation of hepatic stellate cells (HSCs) and progression of liver fibrosis. Type 3 inflammation characterized by production of interleukin-17A (IL-17) and IL-22 by innate and adaptive immune cells is implicated in many inflammatory conditions of the gut and can be counteracted by regulatory T cells (Tregs), but its contribution to liver fibrosis is still poorly understood. Here, we evaluated the contribution of type 3 inflammation in liver fibrosis using clinical liver biopsies, in vitro stimulation of primary HSCs, and in vivo mouse models. We report dysregulated type 3 responses in fibrotic lesions with increased IL-17+CD4+/FOXP3hiCD4+ ratio and increased IL-17 and IL-22 production in advanced liver fibrosis. Neutrophils and mast cells were the main sources of IL-17 in situ in humans. In addition, we demonstrate a new profibrotic function of IL-22 through enhancement of transforming growth factor-β signaling in HSCs in a p38 mitogen-activated protein kinase-dependent manner. In vivo, IL-22RA1 knockout mice exhibited reduced fibrosis in response to thioacetamide and carbon tetrachloride. Blocking either IL-22 or IL-17 production using aryl hydrocarbon receptor or RAR-related orphan receptor gamma-t antagonists resulted in reduced fibrosis. Together, these data have identified a pathogenic role for type 3 immune response mediated by IL-22 in driving liver fibrosis during chronic liver injury.
Collapse
Affiliation(s)
- Thomas Fabre
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Manuel Flores Molina
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Geneviève Soucy
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Département de pathologie et biologie cellulaire, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | | | - Bernard Willems
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Pierre Villeneuve
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Marc Bilodeau
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Naglaa H Shoukry
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
53
|
Aguayo-Orozco A, Bois FY, Brunak S, Taboureau O. Analysis of Time-Series Gene Expression Data to Explore Mechanisms of Chemical-Induced Hepatic Steatosis Toxicity. Front Genet 2018; 9:396. [PMID: 30279702 PMCID: PMC6153316 DOI: 10.3389/fgene.2018.00396] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a wide spectrum of disease, ranging from simple fatty liver through steatosis with inflammation and necrosis to cirrhosis. One of the most challenging problems in biomedical research and within the chemical industry is to understand the underlying mechanisms of complex disease, and complex adverse outcome pathways (AOPs). Based on a set of 28 steatotic chemicals with gene expression data measured on primary hepatocytes at three times (2, 8, and 24 h) and three doses (low, medium, and high), we identified genes and pathways, defined as molecular initiating events (MIEs) and key events (KEs) of steatosis using a combination of a time series and pathway analyses. Among the genes deregulated by these compounds, the study highlighted OSBPL9, ALDH7A1, MYADM, SLC51B, PRDX6, GPAT3, TMEM135, DLGDA5, BCO2, APO10LA, TSPAN6, NEURL1B, and DUSP1. Furthermore, pathway analysis indicated deregulation of pathways related to lipid accumulation, such as fat digestion and absorption, linoleic and linolenic acid metabolism, calcium signaling pathway, fatty acid metabolism, peroxisome, retinol metabolism, and steroid metabolic pathways in a time dependent manner. Such transcription profile analysis can help in the understanding of the steatosis evolution over time generated by chemical exposure.
Collapse
Affiliation(s)
- Alejandro Aguayo-Orozco
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frederic Yves Bois
- Institut National de l'Environnement Industriel et des Risques (INERIS), Unité Modèles pour l'Ecotoxicologie et la Toxicologie (METO), Verneuil en Halatte, France
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Olivier Taboureau
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,UMRS 973 INSERM, Université Paris Diderot, Université Sorbonne Paris Cité, Paris, France
| |
Collapse
|
54
|
Li H, Toth E, Cherrington NJ. Alcohol Metabolism in the Progression of Human Nonalcoholic Steatohepatitis. Toxicol Sci 2018; 164:428-438. [PMID: 29718361 PMCID: PMC6659028 DOI: 10.1093/toxsci/kfy106] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Alcohol metabolism is a well-characterized biological process that is dominated by the alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH) families. Nonalcoholic steatohepatitis (NASH) is the advanced inflammatory stage of nonalcoholic fatty liver disease (NAFLD) and is known to alter the metabolism and disposition of numerous drugs. The purpose of this study was to investigate the alterations in alcohol metabolism processes in response to human NASH progression. Expression and function of ADHs, ALDHs, and catalase were examined in normal, steatosis, NASH (fatty) and NASH (not fatty) human liver samples. ALDH4A1 mRNA was significantly decreased in both NASH groups, while no significant changes were observed in the mRNA levels of other alcohol-related enzymes. The protein levels of ADH1A, ADH1B, and ADH4 were each decreased in the NASH groups, which was consistent with a decreased overall ADH activity. The protein level of ALDH2 was significantly increased in both NASH groups, while ALDH1A1 and ALDH1B1 were only decreased in NASH (fatty) samples. ALDH activity represented by oxidation of acetaldehyde was decreased in the NASH (fatty) group. The protein level of catalase was decreased in both NASH groups, though activity was unchanged. Furthermore, the significant accumulation of 4-hydroxynonenal protein adduct in NASH indicated significant oxidative stress and a potential reduction in ALDH activity. Collectively, ADH and ALDH expression and function are profoundly altered in the progression of NASH, which may have a notable impact on ADH- and ALDH-associated cellular metabolism processes and lead to significant alterations in drug metabolism mediated by these enzymes.
Collapse
Affiliation(s)
- Hui Li
- Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | - Erica Toth
- Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | | |
Collapse
|
55
|
Jamwal R, de la Monte SM, Ogasawara K, Adusumalli S, Barlock BB, Akhlaghi F. Nonalcoholic Fatty Liver Disease and Diabetes Are Associated with Decreased CYP3A4 Protein Expression and Activity in Human Liver. Mol Pharm 2018; 15:2621-2632. [PMID: 29792708 DOI: 10.1021/acs.molpharmaceut.8b00159] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major cause of chronic liver disease in the Western population. We investigated the association of nonalcoholic fatty liver disease (NAFLD) and diabetes mellitus on CYP3A4 activity in human liver tissue from brain dead donors ( n = 74). Histopathologically graded livers were grouped into normal ( n = 24), nonalcoholic fatty liver (NAFL, n = 26), and nonalcoholic steatohepatitis (NASH, n = 24) categories. The rate of conversion of midazolam to its 1-hydroxy metabolite was used to assess in vitro CYP3A4 activity in human liver microsomes (HLM). A proteomics approach was utilized to quantify the protein expression of CYP3A4 and related enzymes. Moreover, a physiologically based pharmacokinetic (PBPK) model was developed to allow prediction of midazolam concentration in NAFL and NASH livers. CYP3A4 activity in NAFL and NASH was 1.9- and 3.1-fold ( p < 0.05) lower than normal donors, respectively. Intrinsic clearance (CLint) was 2.7- ( p < 0.05) and 4.1-fold ( p < 0.01) lower in donors with NAFL and NASH, respectively. CYP3A4 protein expression was significantly lower in NAFL and NASH donors ( p < 0.05) and accounted for significant midazolam hydroxylation variability in a multiple linear regression analysis (β = 0.869, r2 = 0.762, P < 0.01). Diabetes was also associated with decreased CYP3A4 activity and protein expression. Both midazolam CLint and CYP3A4 protein abundance decreased significantly with increase in hepatic fat accumulation. Age and gender did not exhibit any significant association with the observed alterations. Predicted midazolam exposure was 1.7- and 2.3-fold higher for NAFL and NASH, respectively, which may result in a longer period of sedation in these disease-states. Data suggests that NAFLD and diabetes are associated with the decreased hepatic CYP3A4 activity. Thus, further evaluation of clinical consequences of these findings on the efficacy and safety of CYP3A4 substrates is warranted.
Collapse
Affiliation(s)
- Rohitash Jamwal
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| | - Suzanne M de la Monte
- Departments of Medicine, Pathology, Neurology, and Neurosurgery , Rhode Island Hospital and the Warren Alpert Medical School of Brown University , Providence , Rhode Island 02903 , United States
| | - Ken Ogasawara
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| | - Sravani Adusumalli
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| | - Benjamin B Barlock
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| | - Fatemeh Akhlaghi
- Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| |
Collapse
|
56
|
Talal AH, Venuto CS, Younis I. Assessment of Hepatic Impairment and Implications for Pharmacokinetics of Substance Use Treatment. Clin Pharmacol Drug Dev 2018; 6:206-212. [PMID: 28263464 DOI: 10.1002/cpdd.336] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 12/30/2016] [Indexed: 12/12/2022]
Abstract
Although the liver is the primary site of metabolism and biliary excretion for many medications, data are limited on the liver's pharmacokinetic abilities in cirrhosis. Cirrhosis develops through collagen deposition, eventually culminating in end-stage liver disease that compromises hepatic drug metabolism. Consequently, the US Food and Drug Administration (FDA) recommends evaluating the pharmacokinetics of medications in subjects with hepatic impairment if hepatic metabolism constitutes more than 20% of their elimination or if they have a narrow therapeutic range. A variety of noninvasive indices and radiologic procedures can be employed to assess hepatic drug metabolism and excretion. The Child-Pugh score is the most commonly used scale for assessing hepatic impairment among drugs submitted for US FDA approval. The score, originally developed to guide operative mortality in patients undergoing hepatic resection, has not been modified since its inception 5 decades ago. Furthermore, the score was not originally intended to be a guide for potential dose modification in patients with hepatic impairment. These reasons, in combination with the availability of a variety of new imaging modalities and an enhanced understanding of hepatic biology, should foster the development of novel methods to assess the effect of hepatic impairment on liver drug metabolism.
Collapse
Affiliation(s)
- Andrew H Talal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Charles S Venuto
- Center for Human Experimental Therapeutics, University of Rochester, Rochester, NY, USA.,AIDS Clinical Trials Group Pharmacology Specialty Laboratory, New York State Center of Excellence in Bioinformatics and Life Sciences; School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Islam Younis
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
57
|
Zhu M, Wang Q, Zhou W, Liu T, Yang L, Zheng P, Zhang L, Ji G. Integrated analysis of hepatic mRNA and miRNA profiles identified molecular networks and potential biomarkers of NAFLD. Sci Rep 2018; 8:7628. [PMID: 29769539 PMCID: PMC5955949 DOI: 10.1038/s41598-018-25743-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 04/24/2018] [Indexed: 12/17/2022] Open
Abstract
To enhance our understanding of molecular mechanisms and mine novel biomarkers of non-alcoholic fatty liver disease (NAFLD), RNA sequencing was performed to gain hepatic expression profiles of mRNAs and miRNAs in NAFLD and normal rats. Using DESeq with thresholds of a two-fold change and a false discovery rate (FDR) less than 0.05, 336 mRNAs and 21 miRNAs were identified as differentially expressed. Among those, 17 miRNAs (e.g., miR-144-3p, miR-99a-3p, miR-200b-3p, miR-200b-5p, miR-200c-3p, etc.) might serve as novel biomarkers of NAFLD. MiRNA target genes (13565) were predicted by the miRWalk database. Using DAVID 6.8, the intersection (195 genes) of differentially expressed mRNAs and miRNA-predicted target genes were enriched in 47 gene ontology (GO) terms and 28 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Using Cytoscape, pathway interaction and protein-protein interaction (PPI) networks were constructed, and hub genes (e.g., Abcg8, Cyp1a1, Cyp51, Hmgcr, etc.) associated with NAFLD were obtained. Moreover, 673 miRNA-mRNA negative regulatory pairs were obtained, and networks were constructed. Finally, several representative miRNAs and mRNAs were validated by real-time qPCR. In conclusion, potential molecular mechanisms of NAFLD could be inferred from integrated analysis of mRNA and miRNA profiles, which may indicate novel biomarkers of NAFLD.
Collapse
Affiliation(s)
- Mingzhe Zhu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qianlei Wang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Tao Liu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lili Yang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Peiyong Zheng
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
58
|
Hart KM, Fabre T, Sciurba JC, Gieseck RL, Borthwick LA, Vannella KM, Acciani TH, de Queiroz Prado R, Thompson RW, White S, Soucy G, Bilodeau M, Ramalingam TR, Arron JR, Shoukry NH, Wynn TA. Type 2 immunity is protective in metabolic disease but exacerbates NAFLD collaboratively with TGF-β. Sci Transl Med 2018; 9:9/396/eaal3694. [PMID: 28659437 DOI: 10.1126/scitranslmed.aal3694] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/07/2017] [Accepted: 05/17/2017] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is now the most common progressive liver disease in developed countries and is the second leading indication for liver transplantation due to the extensive fibrosis it causes. NAFLD progression is thought to be tied to chronic low-level type 1 inflammation originating in the adipose tissue during obesity; however, the specific immunological mechanisms regulating the progression of NAFLD-associated fibrosis in the liver are unclear. To investigate the immunopathogenesis of NAFLD more completely, we investigated adipose dysfunction, nonalcoholic steatohepatitis (NASH), and fibrosis in mice that develop polarized type 1 or type 2 immune responses. Unexpectedly, obese interleukin-10 (IL-10)/IL-4-deficient mice (type 1-polarized) were highly resistant to NASH. This protection was associated with an increased hepatic interferon-γ (IFN-γ) signature. Conversely, IFN-γ-deficient mice progressed rapidly to NASH with evidence of fibrosis dependent on transforming growth factor-β (TGF-β) and IL-13 signaling. Unlike increasing type 1 inflammation and the marked loss of eosinophils seen in expanding adipose tissue, progression of NASH was associated with increasing eosinophilic type 2 liver inflammation in mice and human patient biopsies. Finally, simultaneous inhibition of TGF-β and IL-13 signaling attenuated the fibrotic machinery more completely than TGF-β alone in NAFLD-associated fibrosis. Thus, although type 2 immunity maintains healthy metabolic signaling in adipose tissues, it exacerbates the progression of NAFLD collaboratively with TGF-β in the liver.
Collapse
Affiliation(s)
- Kevin M Hart
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas Fabre
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Joshua C Sciurba
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard L Gieseck
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lee A Borthwick
- Tissue Fibrosis and Repair Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Kevin M Vannella
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas H Acciani
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rafael de Queiroz Prado
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert W Thompson
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sandra White
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Genevieve Soucy
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de pathologie et biologie cellulaire, Université de Montréal, Montréal, Québec, Canada
| | - Marc Bilodeau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Thirumalai R Ramalingam
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Naglaa H Shoukry
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Thomas A Wynn
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
59
|
Dzierlenga AL, Cherrington NJ. Misregulation of membrane trafficking processes in human nonalcoholic steatohepatitis. J Biochem Mol Toxicol 2018; 32:e22035. [PMID: 29341352 DOI: 10.1002/jbt.22035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/17/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) remodels the expression and function of genes and proteins that are critical for drug disposition. This study sought to determine whether disruption of membrane protein trafficking pathways in human NASH contributes to altered localization of multidrug resistance-associated protein 2 (MRP2). A comprehensive immunoblot analysis assessed the phosphorylation, membrane translocation, and expression of transporter membrane insertion regulators, including several protein kinases (PK), radixin, MARCKS, and Rab11. Radixin exhibited a decreased phosphorylation and total expression, whereas Rab11 had an increased membrane localization. PKCδ, PKCα, and PKA had increased membrane activation, whereas PKCε had a decreased phosphorylation and membrane expression. Radixin dephosphorylation may activate MRP2 membrane retrieval in NASH; however, the activation of Rab11/PKCδ and PKA/PKCα suggest an activation of membrane insertion pathways as well. Overall these data suggest an altered regulation of protein trafficking in human NASH, although other processes may be involved in the regulation of MRP2 localization.
Collapse
Affiliation(s)
- Anika L Dzierlenga
- Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ, USA
| | | |
Collapse
|
60
|
Effects of low bisphenol A concentration on protein expression profiles in an in vitro model of non-alcoholic fatty liver disease. Mol Cell Toxicol 2018. [DOI: 10.1007/s13273-018-0008-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
61
|
Li H, Toth E, Cherrington NJ. Asking the Right Questions With Animal Models: Methionine- and Choline-Deficient Model in Predicting Adverse Drug Reactions in Human NASH. Toxicol Sci 2018; 161:23-33. [PMID: 29145614 PMCID: PMC6454421 DOI: 10.1093/toxsci/kfx253] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In the past few decades, great conceptual and technological advances have been made in the field of toxicology, but animal model-based research still remains one of the most widely used and readily available tools for furthering our current knowledge. However, animal models are not perfect in predicting all systemic toxicity in humans. Extrapolating animal data to accurately predict human toxicities remains a challenge, and researchers are obligated to question the appropriateness of their chosen animal model. This paper provides an assessment of the utility of the methionine- and choline-deficient (MCD) diet fed animal model in reflecting human nonalcoholic steatohepatitis (NASH) and the potential risks of adverse drug reactions and toxicities that are associated with the disease. As a commonly used NASH model, the MCD model fails to exhibit most metabolic abnormalities in a similar manner to the human disease. The MCD model, on the other hand, closely resembles human NASH histology and reflects signatures of drug transporter alterations in humans. Due to the nature of the MCD model, it should be avoided in studies of NASH pathogenesis, metabolic parameter evaluation, and biomarker identification. But it can be used to accurately predict altered drug disposition due to NASH-associated transporter alterations.
Collapse
Affiliation(s)
- Hui Li
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | - Erica Toth
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | | |
Collapse
|
62
|
Systematic integrative analysis of gene expression identifies HNF4A as the central gene in pathogenesis of non-alcoholic steatohepatitis. PLoS One 2017; 12:e0189223. [PMID: 29216278 PMCID: PMC5720788 DOI: 10.1371/journal.pone.0189223] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/21/2017] [Indexed: 12/18/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the Western world, and encompasses a spectrum from simple steatosis to steatohepatitis (NASH). There is currently no approved pharmacologic therapy against NASH, partly due to an incomplete understanding of its molecular basis. The goal of this study was to determine the key differentially expressed genes (DEGs), as well as those genes and pathways central to its pathogenesis. We performed an integrative computational analysis of publicly available gene expression data in NASH from GEO (GSE17470, GSE24807, GSE37031, GSE89632). The DEGs were identified using GEOquery, and only the genes present in at least three of the studies, to a total of 190 DEGs, were considered for further analyses. The pathways, networks, molecular interactions, functional analyses were generated through the use of Ingenuity Pathway Analysis (IPA). For selected networks, we computed the centrality using igraph package in R. Among the statistically significant predicted networks (p-val < 0.05), three were of most biological interest: the first is involved in antimicrobial response, inflammatory response and immunological disease, the second in cancer, organismal injury and development and the third in metabolic diseases. We discovered that HNF4A is the central gene in the network of NASH connected to metabolic diseases and that it regulates HNF1A, an additional transcription regulator also involved in lipid metabolism. Therefore, we show, for the first time to our knowledge, that HNF4A is central to the pathogenesis of NASH. This adds to previous literature demonstrating that HNF4A regulates the transcription of genes involved in the progression of NAFLD, and that HNF4A genetic variants play a potential role in NASH progression.
Collapse
|
63
|
Cole BK, Feaver RE, Wamhoff BR, Dash A. Non-alcoholic fatty liver disease (NAFLD) models in drug discovery. Expert Opin Drug Discov 2017; 13:193-205. [PMID: 29190166 DOI: 10.1080/17460441.2018.1410135] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The progressive disease spectrum of non-alcoholic fatty liver disease (NAFLD), which includes non-alcoholic steatohepatitis (NASH), is a rapidly emerging public health crisis with no approved therapy. The diversity of various therapies under development highlights the lack of consensus around the most effective target, underscoring the need for better translatable preclinical models to study the complex progressive disease and effective therapies. Areas covered: This article reviews published literature of various mouse models of NASH used in preclinical studies, as well as complex organotypic in vitro and ex vivo liver models being developed. It discusses translational challenges associated with both kinds of models, and describes some of the studies that validate their application in NAFLD. Expert opinion: Animal models offer advantages of understanding drug distribution and effects in a whole body context, but are limited by important species differences. Human organotypic in vitro and ex vivo models with physiological relevance and translatability need to be used in a tiered manner with simpler screens. Leveraging newer technologies, like metabolomics, proteomics, and transcriptomics, and the future development of validated disease biomarkers will allow us to fully utilize the value of these models to understand disease and evaluate novel drugs in isolation or combination.
Collapse
Affiliation(s)
| | | | | | - Ajit Dash
- b Early Development Safety , Genentech Inc , South San Francisco , CA , USA
| |
Collapse
|
64
|
Li H, Canet MJ, Clarke JD, Billheimer D, Xanthakos SA, Lavine JE, Erickson RP, Cherrington NJ. Pediatric Cytochrome P450 Activity Alterations in Nonalcoholic Steatohepatitis. Drug Metab Dispos 2017; 45:1317-1325. [PMID: 28986475 PMCID: PMC5697442 DOI: 10.1124/dmd.117.077644] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/28/2017] [Indexed: 01/01/2023] Open
Abstract
Variable drug responses depend on individual variation in the activity of drug-metabolizing enzymes, including cytochrome P450 enzymes (CYP). As the most common chronic liver disease in children and adults, nonalcoholic steatohepatitis (NASH) has been identified as a source of significant interindividual variation in hepatic drug metabolism. Compared with adults, children present age-related differences in pharmacokinetics and pharmacodynamics. The purpose of this study was to determine the impact of fatty liver disease severity on the activity of a variety of CYP enzymes in children and adolescents. Healthy and nonalcoholic fatty liver disease pediatric subjects aged 12-21 years inclusive received an oral cocktail of four probe drugs: caffeine (CYP1A2, 100 mg), omeprazole (CYP2C19, 20 mg), losartan (CYP2C9, 25 mg), and midazolam (CYP3A4, 2 mg). Venous blood and urine were collected before administration and 1, 2, 4, and 6 hours after administration. Concentrations of the parent drugs and CYP-specific metabolites were quantified in plasma and urine using liquid chromatography with tandem mass spectrometry. In plasma, the decreased metabolic area under the curve (AUC) ratio, defined as the metabolite AUC to parent AUC, of omeprazole indicated significant decreases of CYP2C19 (P = 0.002) enzymatic activities in NASH adolescents, while the urine analyses did not show significant differences and were highly variable. A comparison between the present in vivo pediatric studies and a previous ex vivo study in adults indicates distinct differences in the activities of CYP1A2 and CYP2C9. These data demonstrate that pediatric NASH presents an altered pattern of CYP activity and NASH should be considered as a confounder of drug metabolism for certain CYP enzymes. These differences could lead to future investigations that may reveal unexpected variable drug responses that should be considered in pediatric dosage recommendations.
Collapse
Affiliation(s)
- Hui Li
- Department of Pharmacology and Toxicology (H.L., M.J.C., J.D.C., N.J.C.), Department of Epidemiology and Biostatistics (D.B.), and Department of Pediatrics (R.P.E.), University of Arizona, Tucson, Arizona; Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio (S.A.X.); Columbia University, New York, New York (J.E.L.)
| | - Mark J Canet
- Department of Pharmacology and Toxicology (H.L., M.J.C., J.D.C., N.J.C.), Department of Epidemiology and Biostatistics (D.B.), and Department of Pediatrics (R.P.E.), University of Arizona, Tucson, Arizona; Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio (S.A.X.); Columbia University, New York, New York (J.E.L.)
| | - John D Clarke
- Department of Pharmacology and Toxicology (H.L., M.J.C., J.D.C., N.J.C.), Department of Epidemiology and Biostatistics (D.B.), and Department of Pediatrics (R.P.E.), University of Arizona, Tucson, Arizona; Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio (S.A.X.); Columbia University, New York, New York (J.E.L.)
| | - Dean Billheimer
- Department of Pharmacology and Toxicology (H.L., M.J.C., J.D.C., N.J.C.), Department of Epidemiology and Biostatistics (D.B.), and Department of Pediatrics (R.P.E.), University of Arizona, Tucson, Arizona; Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio (S.A.X.); Columbia University, New York, New York (J.E.L.)
| | - Stavra A Xanthakos
- Department of Pharmacology and Toxicology (H.L., M.J.C., J.D.C., N.J.C.), Department of Epidemiology and Biostatistics (D.B.), and Department of Pediatrics (R.P.E.), University of Arizona, Tucson, Arizona; Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio (S.A.X.); Columbia University, New York, New York (J.E.L.)
| | - Joel E Lavine
- Department of Pharmacology and Toxicology (H.L., M.J.C., J.D.C., N.J.C.), Department of Epidemiology and Biostatistics (D.B.), and Department of Pediatrics (R.P.E.), University of Arizona, Tucson, Arizona; Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio (S.A.X.); Columbia University, New York, New York (J.E.L.)
| | - Robert P Erickson
- Department of Pharmacology and Toxicology (H.L., M.J.C., J.D.C., N.J.C.), Department of Epidemiology and Biostatistics (D.B.), and Department of Pediatrics (R.P.E.), University of Arizona, Tucson, Arizona; Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio (S.A.X.); Columbia University, New York, New York (J.E.L.)
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology (H.L., M.J.C., J.D.C., N.J.C.), Department of Epidemiology and Biostatistics (D.B.), and Department of Pediatrics (R.P.E.), University of Arizona, Tucson, Arizona; Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio (S.A.X.); Columbia University, New York, New York (J.E.L.)
| |
Collapse
|
65
|
Lyall MJ, Cartier J, Richards JA, Cobice D, Thomson JP, Meehan RR, Anderton SM, Drake AJ. Methyl donor deficient diets cause distinct alterations in lipid metabolism but are poorly representative of human NAFLD. Wellcome Open Res 2017; 2:67. [PMID: 29707653 PMCID: PMC5887079 DOI: 10.12688/wellcomeopenres.12199.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2017] [Indexed: 12/15/2022] Open
Abstract
Background: Non-alcoholic fatty liver disease (NAFLD) is a global health issue. Dietary methyl donor restriction is used to induce a NAFLD/non-alcoholic steatohepatitis (NASH) phenotype in rodents, however the extent to which this model reflects human NAFLD remains incompletely understood. To address this, we undertook hepatic transcriptional profiling of methyl donor restricted rodents and compared these to published human NAFLD datasets. Methods: Adult C57BL/6J mice were maintained on control, choline deficient (CDD) or methionine/choline deficient (MCDD) diets for four weeks; the effects on methyl donor and lipid biology were investigated by bioinformatic analysis of hepatic gene expression profiles followed by a cross-species comparison with human expression data of all stages of NAFLD. Results: Compared to controls, expression of the very low density lipoprotein (VLDL) packaging carboxylesterases (
Ces1d,
Ces1f,
Ces3b) and the NAFLD risk allele
Pnpla3 were suppressed in MCDD; with
Pnpla3 and the liver predominant
Ces isoform,
Ces3b, also suppressed in CDD. With respect to 1-carbon metabolism, down-regulation of
Chka,
Chkb,
Pcty1a,
Gnmt and
Ahcy with concurrent upregulation of
Mat2a suggests a drive to maintain S-adenosylmethionine levels. There was minimal similarity between global gene expression patterns in either dietary intervention and any stage of human NAFLD, however some common transcriptomic changes in inflammatory, fibrotic and proliferative mediators were identified in MCDD, NASH and HCC. Conclusions: This study suggests suppression of VLDL assembly machinery may contribute to hepatic lipid accumulation in these models, but that CDD and MCDD rodent diets are minimally representative of human NAFLD at the transcriptional level.
Collapse
Affiliation(s)
- Marcus J Lyall
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Jessy Cartier
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - James A Richards
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Diego Cobice
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,School of Biomedical Sciences, Biomedical Sciences Research Institute, University of Ulster, Coleraine, County Londonderry, UK
| | - John P Thomson
- MRC Human Genetics Unit, IGMM, Western General Hospital, Edinburgh, UK
| | - Richard R Meehan
- MRC Human Genetics Unit, IGMM, Western General Hospital, Edinburgh, UK
| | - Stephen M Anderton
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Centre for Immunity, Infection and Evolution, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Amanda J Drake
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
66
|
Atilano-Roque A, Roda G, Fogueri U, Kiser JJ, Joy MS. Effect of Disease Pathologies on Transporter Expression and Function. J Clin Pharmacol 2017; 56 Suppl 7:S205-21. [PMID: 27385176 DOI: 10.1002/jcph.768] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 12/12/2022]
Abstract
Transporters are important determinants of drug absorption, distribution, and excretion. The clinical relevance of drug transporters in drug disposition and toxicology depends on their localization in liver, kidney, and brain. There has been growing evidence regarding the importance of disease status on alterations in metabolizing enzymes and transporter proteins. This review focuses on uptake and efflux transporter proteins in liver, kidney, and brain and discusses mechanisms of altered transporter expression and function secondary to disease.
Collapse
Affiliation(s)
- Amandla Atilano-Roque
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Gavriel Roda
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Uma Fogueri
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Jennifer J Kiser
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Melanie S Joy
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA.,Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
67
|
Yu L, Yuan M, Wang L. The effect of omega-3 unsaturated fatty acids on non-alcoholic fatty liver disease: A systematic review and meta-analysis of RCTs. Pak J Med Sci 2017; 33:1022-1028. [PMID: 29067086 PMCID: PMC5648933 DOI: 10.12669/pjms.334.12315] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: During the treatment of diseases such as angiocardiopathy, blood lipid abnormalities and metabolic syndrome, omega-3 unsaturated fatty acids (PUFA) can reduce plasma lipids and improve cardiovascular status, thus ameliorating disease severity. We aimed to explore the effects of PUFA supplementation in patients with non-alcoholic fatty liver disease (NAFLD). Methods: A systematic literature search was performed during March 2016 for randomized controlled trials using PUFA or fish oil supplementation in patients with NAFLD or non-alcoholic steatohepatitis (NASH). All Randomized controlled trials were retrieved from MEDLINE and EMBASE database up to date (March 2016). A meta-analysis of key outcomes (serum level of liver enzymes and lipids) were identified in these studies. The mean difference (MD) and the corresponding 95% confidence intervals (CIs) were used as measures of effect size. Results: Thirteen studies were included, consisting of 266 patients in the PUFA group and 402 cases in the control group. Serum level of alanine aminotransferase (ALT) was lower in the PUFA group than that in in the controls [MD=−9.18, 95% CI (−12.41, −5.96), P <0.00001]. However, PUFA treatment did not affect aspartate aminotransferase (AST) [MD=−5.07, 95% CI (−12.65, 2.51), P= 0.19], gamma-glutamyl transferase (GGT) [MD=−1.91, 95% CI (−4.15, 0.33), P <0.009]. Conclusions: PUFA supplementation may affects serum level of ALT and improve liver function in patients with NAFLD.
Collapse
Affiliation(s)
- Le Yu
- Dr. Le Yu, Center of Infectious Diseases, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China. Division of Infectious Diseases, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Man Yuan
- Dr. Man Yuan, Center of Infectious Diseases, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China. Division of Infectious Diseases, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| | - Linchun Wang
- Linchun Wang, Center of Infectious Diseases, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China. Division of Infectious Diseases, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, China
| |
Collapse
|
68
|
Qi S, Wang C, Li C, Wang P, Liu M. Candidate genes investigation for severe nonalcoholic fatty liver disease based on bioinformatics analysis. Medicine (Baltimore) 2017; 96:e7743. [PMID: 28796060 PMCID: PMC5556226 DOI: 10.1097/md.0000000000007743] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver condition worldwide. However, its etiology and fundamental pathophysiology for the disease process are poorly understood. In this study, we thus used bioinformatics to identify candidate genes potentially causative of severe NAFLD. METHODS Gene expression profile data GSE49541 were downloaded from the Gene Expression Omnibus database. Tissues samples from 32 severe and 40 mild NAFLD patients were evaluated to identify differentially expressed genes (DEGs) between the 2 groups, followed by analyses of Gene Ontology (GO) functions and Kyoto Encyclopedia of Genes and Genomes pathways. Then, a weighted protein-protein interaction (PPI) network was constructed, and subnetworks and candidate genes were screened. Moreover, the GSE48452 data (14 normal liver tissue samples and 18 nonalcoholic steatohepatitis samples) were used to verify the results obtained from the above analyses. RESULTS A total of 100 upregulated genes and 24 downregulated ones were identified in severe NAFLD. Functional enrichment and pathway analyses showed that these DEGs were mainly associated with cell adhesion, inflammatory response, and chemokine activity. The top 5 subnetworks were selected based on the PPI network. A total of 5 hub genes, including ubiquilin 4 (UBQLN4), amyloid-beta precursor protein (APP), sex hormone-binding globulin (SHBG), cadherin-associated protein beta 1 (CTNNB1) and collagen type I alpha 1 (COL1A1), were considered to be candidate genes for NAFLD. In addition, the verification data confirmed the status of COL1A1, SHBG, and APP as candidate genes. CONCLUSION UBQLN4, APP, CTNNB1, SHBG, and COL1A1 might be involved in the development of NAFLD, and are proposed as the potential markers for predicting the development of this condition.
Collapse
Affiliation(s)
- Shan Qi
- Department of Traditional Chinese Medicine, China-Japan Union Hospital of Jilin University
| | - Changhong Wang
- Department of Traditional Chinese Medicine, China-Japan Union Hospital of Jilin University
| | - Chunfu Li
- Department of Traditional Chinese Medicine, China-Japan Union Hospital of Jilin University
| | - Pu Wang
- Clinical Medicine College, Jilin University, Changchun, Jilin Province, China
| | - Minghui Liu
- Department of Traditional Chinese Medicine, China-Japan Union Hospital of Jilin University
| |
Collapse
|
69
|
Lefebvre P, Lalloyer F, Baugé E, Pawlak M, Gheeraert C, Dehondt H, Vanhoutte J, Woitrain E, Hennuyer N, Mazuy C, Bobowski-Gérard M, Zummo FP, Derudas B, Driessen A, Hubens G, Vonghia L, Kwanten WJ, Michielsen P, Vanwolleghem T, Eeckhoute J, Verrijken A, Van Gaal L, Francque S, Staels B. Interspecies NASH disease activity whole-genome profiling identifies a fibrogenic role of PPARα-regulated dermatopontin. JCI Insight 2017; 2:92264. [PMID: 28679947 DOI: 10.1172/jci.insight.92264] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 05/19/2017] [Indexed: 12/21/2022] Open
Abstract
Nonalcoholic fatty liver disease prevalence is soaring with the obesity pandemic, but the pathogenic mechanisms leading to the progression toward active nonalcoholic steatohepatitis (NASH) and fibrosis, major causes of liver-related death, are poorly defined. To identify key components during the progression toward NASH and fibrosis, we investigated the liver transcriptome in a human cohort of NASH patients. The transition from histologically proven fatty liver to NASH and fibrosis was characterized by gene expression patterns that successively reflected altered functions in metabolism, inflammation, and epithelial-mesenchymal transition. A meta-analysis combining our and public human transcriptomic datasets with murine models of NASH and fibrosis defined a molecular signature characterizing NASH and fibrosis and evidencing abnormal inflammation and extracellular matrix (ECM) homeostasis. Dermatopontin expression was found increased in fibrosis, and reversal of fibrosis after gastric bypass correlated with decreased dermatopontin expression. Functional studies in mice identified an active role for dermatopontin in collagen deposition and fibrosis. PPARα activation lowered dermatopontin expression through a transrepressive mechanism affecting the Klf6/TGFβ1 pathway. Liver fibrotic histological damages are thus characterized by the deregulated expression of a restricted set of inflammation- and ECM-related genes. Among them, dermatopontin may be a valuable target to reverse the hepatic fibrotic process.
Collapse
Affiliation(s)
- Philippe Lefebvre
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Fanny Lalloyer
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Eric Baugé
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Michal Pawlak
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Céline Gheeraert
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Hélène Dehondt
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Jonathan Vanhoutte
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Eloise Woitrain
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Nathalie Hennuyer
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Claire Mazuy
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Marie Bobowski-Gérard
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Francesco Paolo Zummo
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bruno Derudas
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | | | | | - Luisa Vonghia
- Department of Gastroenterology and Hepatology, and.,Department of Endocrinology, Diabetology and Metabolism, University Hospital Antwerp, Edegem, Belgium
| | - Wilhelmus J Kwanten
- Department of Gastroenterology and Hepatology, and.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Peter Michielsen
- Department of Gastroenterology and Hepatology, and.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Thomas Vanwolleghem
- Department of Gastroenterology and Hepatology, and.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Jérôme Eeckhoute
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - An Verrijken
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Antwerp, Edegem, Belgium.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Luc Van Gaal
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Antwerp, Edegem, Belgium.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Sven Francque
- Department of Gastroenterology and Hepatology, and.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Bart Staels
- University Lille, Inserm, CHU-Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| |
Collapse
|
70
|
Lee SC, Arya V, Yang X, Volpe DA, Zhang L. Evaluation of transporters in drug development: Current status and contemporary issues. Adv Drug Deliv Rev 2017; 116:100-118. [PMID: 28760687 DOI: 10.1016/j.addr.2017.07.020] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/19/2017] [Accepted: 07/26/2017] [Indexed: 01/22/2023]
Abstract
Transporters govern the access of molecules to cells or their exit from cells, thereby controlling the overall distribution of drugs to their intracellular site of action. Clinically relevant drug-drug interactions mediated by transporters are of increasing interest in drug development. Drug transporters, acting alone or in concert with drug metabolizing enzymes, can play an important role in modulating drug absorption, distribution, metabolism and excretion, thus affecting the pharmacokinetics and/or pharmacodynamics of a drug. The drug interaction guidance documents from regulatory agencies include various decision criteria that may be used to predict the need for in vivo assessment of transporter-mediated drug-drug interactions. Regulatory science research continues to assess the prediction performances of various criteria as well as to examine the strength and limitations of each prediction criterion to foster discussions related to harmonized decision criteria that may be used to facilitate global drug development. This review discusses the role of transporters in drug development with a focus on methodologies in assessing transporter-mediated drug-drug interactions, challenges in both in vitro and in vivo assessments of transporters, and emerging transporter research areas including biomarkers, assessment of tissue concentrations, and effect of diseases on transporters.
Collapse
Affiliation(s)
- Sue-Chih Lee
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Vikram Arya
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Xinning Yang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Donna A Volpe
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
71
|
Clarke JD, Novak P, Lake AD, Hardwick RN, Cherrington NJ. Impaired N-linked glycosylation of uptake and efflux transporters in human non-alcoholic fatty liver disease. Liver Int 2017; 37:1074-1081. [PMID: 28097795 PMCID: PMC5479731 DOI: 10.1111/liv.13362] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/30/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS N-linked glycosylation of proteins is critical for proper protein folding and trafficking to the plasma membrane. Drug transporters are one class of proteins that have reduced function when glycosylation is impaired. N-linked glycosylation of plasma proteins has also been investigated as a biomarker for several liver diseases, including non-alcoholic fatty liver disease (NAFLD). The purpose of this study was to assess the transcriptomic expression of genes involved in protein processing and glycosylation, and to determine the glycosylation status of key drug transporters during human NAFLD progression. METHODS Human liver samples diagnosed as healthy, steatosis, and non-alcoholic steatohepatitis (NASH) were analysed for gene expression of glycosylation-related genes and for protein glycosylation using immunoblot. RESULTS Genes involved in protein processing in the ER and biosynthesis of N-glycans were significantly enriched for down-regulation in NAFLD progression. Included in the down regulated N-glycan biosynthesis category were genes involved in the oligosaccharyltransferase complex, N-glycan quality control, N-glycan precursor biosynthesis, N-glycan trimming to the core, and N-glycan extension from the core. N-glycan degradation genes were unaltered in the progression to NASH. Immunoblot analysis of the uptake transporters organic anion transporting polypeptide-1B1 (OATP1B1), OATP1B3, OATP2B1, and Sodium/Taurocholate Co-transporting Polypeptide (NTCP) and the efflux transporter multidrug resistance-associated protein 2 (MRP2) demonstrated a significant loss of glycosylation following the progression to NASH. CONCLUSIONS These data suggest that the loss of glycosylation of key uptake and efflux transporters in humans NASH may influence transporter function and contribute to altered drug disposition observed in NASH.
Collapse
Affiliation(s)
- John D Clarke
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Petr Novak
- Biology Centre ASCR, Institute of Plant Molecular Biology, Ceske Budejovice, Czech Republic
| | - April D Lake
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Rhiannon N Hardwick
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
72
|
王 鹤, 孙 鹏, 刘 克. 肝脏转运体表达和功能的变化对肝疾病的影响. Shijie Huaren Xiaohua Zazhi 2017; 25:1427-1437. [DOI: 10.11569/wcjd.v25.i16.1427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
转运体是药物吸收、分布、代谢和排泄的重要决定因素, 在肝脏表达尤为广泛. 肝脏转运体可以摄取大多数内源性物质、营养物质和外源性物质进入肝脏, 在肝脏内经过一系列的代谢转化, 最终将其外排入胆汁, 并由胆汁排到肝外. 越来越多的证据表明, 肝脏疾病状态下转运体的表达和功能会发生改变, 影响药物在体内的处置过程, 进而增加药物相互作用的可能性, 同时加大了疾病药物治疗的难度. 本文从肝脏摄取型和外排型转运体两方面出发, 针对肝脏转运体表达和功能的变化对肝疾病的影响作一综述.
Collapse
|
73
|
Han J, Dzierlenga AL, Lu Z, Billheimer DD, Torabzadehkhorasani E, Lake AD, Li H, Novak P, Shipkova P, Aranibar N, Robertson D, Reily MD, Lehman-McKeeman LD, Cherrington NJ. Metabolomic profiling distinction of human nonalcoholic fatty liver disease progression from a common rat model. Obesity (Silver Spring) 2017; 25:1069-1076. [PMID: 28452429 PMCID: PMC5513172 DOI: 10.1002/oby.21855] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 03/24/2017] [Accepted: 03/25/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Characteristic pathological changes define the progression of steatosis to nonalcoholic steatohepatitis (NASH) and are correlated to metabolic pathways. A common rodent model of NASH is the methionine and choline deficient (MCD) diet. The objective of this study was to perform full metabolomic analyses on liver samples to determine which pathways are altered most pronouncedly in this condition in humans, and to compare these changes to rodent models of nonalcoholic fatty liver disease (NAFLD). METHODS A principal component analysis for all 91 metabolites measured indicated that metabolome perturbation is greater and less varied for humans than for rodents. RESULTS Metabolome changes in human and rat NAFLD were greatest for the amino acid and bile acid metabolite families (e.g., asparagine, citrulline, gamma-aminobutyric acid, lysine); although, in many cases, the trends were reversed when compared between species (cholic acid, betaine). CONCLUSIONS Overall, these results indicate that metabolites of specific pathways may be useful biomarkers for NASH progression, although these markers may not correspond to rodent NASH models. The MCD model may be useful when studying certain end points of NASH; however, the metabolomics results indicate important differences between humans and rodents in the biochemical pathogenesis of the disease.
Collapse
Affiliation(s)
- JianHua Han
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ, USA
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Department of Clinical Laboratory, Beijing, China
| | - Anika L. Dzierlenga
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ, USA
| | - Zhengqiang Lu
- The Arizona Statistical Consulting Laboratory, University of Arizona, Tucson, AZ, USA
| | - Dean D. Billheimer
- The Arizona Statistical Consulting Laboratory, University of Arizona, Tucson, AZ, USA
| | | | - April D. Lake
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ, USA
| | - Hui Li
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ, USA
| | - Petr Novak
- Biology Centre CAS, Institute of Plant Molecular Biology, Ceske Budejovice, Czech Republic
| | - Petia Shipkova
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Princeton, NJ, USA
| | - Nelly Aranibar
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Princeton, NJ, USA
| | - Donald Robertson
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Princeton, NJ, USA
| | - Michael D. Reily
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Co., Princeton, NJ, USA
| | | | | |
Collapse
|
74
|
Thakkar N, Slizgi JR, Brouwer KLR. Effect of Liver Disease on Hepatic Transporter Expression and Function. J Pharm Sci 2017; 106:2282-2294. [PMID: 28465155 DOI: 10.1016/j.xphs.2017.04.053] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/27/2022]
Abstract
Liver disease can alter the disposition of xenobiotics and endogenous substances. Regulatory agencies such as the Food and Drug Administration and the European Medicines Evaluation Agency recommend, if possible, studying the effect of liver disease on drugs under development to guide specific dose recommendations in these patients. Although extensive research has been conducted to characterize the effect of liver disease on drug-metabolizing enzymes, emerging data have implicated that the expression and function of hepatobiliary transport proteins also are altered in liver disease. This review summarizes recent developments in the field, which may have implications for understanding altered disposition, safety, and efficacy of new and existing drugs. A brief review of liver physiology and hepatic transporter localization/function is provided. Then, the expression and function of hepatic transporters in cholestasis, hepatitis C infection, hepatocellular carcinoma, human immunodeficiency virus infection, nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, and primary biliary cirrhosis are reviewed. In the absence of clinical data, nonclinical information in animal models is presented. This review aims to advance the understanding of altered expression and function of hepatic transporters in liver disease and the implications of such changes on drug disposition.
Collapse
Affiliation(s)
- Nilay Thakkar
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Jason R Slizgi
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599.
| |
Collapse
|
75
|
Pierre V, Johnston CK, Ferslew BC, Brouwer K, Gonzalez D. Population Pharmacokinetics of Morphine in Patients With Nonalcoholic Steatohepatitis (NASH) and Healthy Adults. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:331-339. [PMID: 28417561 PMCID: PMC5445229 DOI: 10.1002/psp4.12185] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/07/2017] [Indexed: 12/11/2022]
Abstract
Altered expression and function of transporters in nonalcoholic steatohepatitis (NASH) patients may affect the pharmacokinetics (PK), efficacy, and safety of substrate drugs. A population pharmacokinetic (PopPK) analysis was performed to assess differences in morphine and morphine-3-glucuronide (M3G) disposition in NASH and healthy subjects. A total of 315 serum and 42 urine samples from 21 subjects (14 healthy; 7 NASH) were analyzed using NONMEM. Morphine and M3G PK were described by three- and one-compartment models, respectively. After accounting for the effect of total body weight on all clearance and volume of distribution parameters using an allometric scaling approach, NASH severity score (NASF; combination of fibrosis and nonalcoholic fatty liver disease activity scores) was the most significant predictor of differences in M3G exposure. The model predicted a linear decrease in the clearance of M3G with increasing NASF scores on a natural logarithmic scale. These results may provide some insight into the potential effect of NASH on the disposition of hepatic transporter substrates.
Collapse
Affiliation(s)
- V Pierre
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - C K Johnston
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Metrum Research Group LLC, Tariffville, Connecticut, USA
| | - B C Ferslew
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Klr Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - D Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
76
|
Dietrich CG, Rau M, Jahn D, Geier A. Changes in drug transport and metabolism and their clinical implications in non-alcoholic fatty liver disease. Expert Opin Drug Metab Toxicol 2017; 13:625-640. [PMID: 28359183 DOI: 10.1080/17425255.2017.1314461] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The incidence of non-alcoholic fatty liver disease (NAFLD) is rising, especially in Western countries. Drug treatment in patients with NAFLD is common since it is linked to other conditions like diabetes, obesity, and cardiovascular disease. Consequently, changes in drug metabolism may have serious clinical implications. Areas covered: A literature search for studies in animal models or patients with obesity, fatty liver, non-alcoholic steatohepatitis (NASH) or NASH cirrhosis published before November 2016 was performed. After discussing epidemiology and animal models for NAFLD, we summarized both basic as well as clinical studies investigating changes in drug transport and metabolism in NAFLD. Important drug groups were assessed separately with emphasis on clinical implications for drug treatment in patients with NAFLD. Expert opinion: Given the frequency of NAFLD even today, a high degree of drug treatment in NAFLD patients appears safe and well-tolerated despite considerable changes in hepatic uptake, distribution, metabolism and transport of drugs in these patients. NASH causes changes in biliary excretion, systemic concentrations, and renal handling of drugs leading to alterations in drug efficacy or toxicity under specific circumstances. Future clinical drug studies should focus on this special patient population in order to avoid serious adverse events in NAFLD patients.
Collapse
Affiliation(s)
- Christoph G Dietrich
- a Bethlehem Center of Health , Department of Medicine , Stolberg/Rhineland , Germany
| | - Monika Rau
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| | - Daniel Jahn
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| | - Andreas Geier
- b Division of Hepatology, Department of Medicine II , University of Würzburg , Würzburg , Germany
| |
Collapse
|
77
|
In silico search for modifier genes associated with pancreatic and liver disease in Cystic Fibrosis. PLoS One 2017; 12:e0173822. [PMID: 28339466 PMCID: PMC5365109 DOI: 10.1371/journal.pone.0173822] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/27/2017] [Indexed: 12/15/2022] Open
Abstract
Cystic Fibrosis is the most common lethal autosomal recessive disorder in the white population, affecting among other organs, the lung, the pancreas and the liver. Whereas Cystic Fibrosis is a monogenic disease, many studies reveal a very complex relationship between genotype and clinical phenotype. Indeed, the broad phenotypic spectrum observed in Cystic Fibrosis is far from being explained by obvious genotype-phenotype correlations and it is admitted that Cystic Fibrosis disease is the result of multiple factors, including effects of the environment as well as modifier genes. Our objective was to highlight new modifier genes with potential implications in the lung, pancreatic and liver outcomes of the disease. For this purpose we performed a system biology approach which combined, database mining, literature mining, gene expression study and network analysis as well as pathway enrichment analysis and protein-protein interactions. We found that IFI16, CCNE2 and IGFBP2 are potential modifiers in the altered lung function in Cystic Fibrosis. We also found that EPHX1, HLA-DQA1, HLA-DQB1, DSP and SLC33A1, GPNMB, NCF2, RASGRP1, LGALS3 and PTPN13, are potential modifiers in pancreas and liver, respectively. Associated pathways indicate that immune system is likely involved and that Ubiquitin C is probably a central node, linking Cystic Fibrosis to liver and pancreatic disease. We highlight here new modifier genes with potential implications in Cystic Fibrosis. Nevertheless, our in silico analysis requires functional analysis to give our results a physiological relevance.
Collapse
|
78
|
Li H, Clarke JD, Dzierlenga AL, Bear J, Goedken MJ, Cherrington NJ. In vivo cytochrome P450 activity alterations in diabetic nonalcoholic steatohepatitis mice. J Biochem Mol Toxicol 2017; 31:10.1002/jbt.21840. [PMID: 27712037 PMCID: PMC5426479 DOI: 10.1002/jbt.21840] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 12/17/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) has been identified as a source of significant interindividual variation in drug metabolism. A previous ex vivo study demonstrated significant changes in hepatic Cytochrome P450 (CYP) activity in human NASH. This study evaluated the in vivo activities of multiple CYP isoforms simultaneously in prominent diabetic NASH mouse models. The pharmacokinetics of CYP selective substrates: caffeine, losartan, and omeprazole changed significantly in a diabetic NASH mouse model, indicating attenuation of the activity of Cyp1a2 and Cyp2c29, respectively. Decreased mRNA expression of Cyp1a2 and Cyp2c29, as well as an overall decrease in CYP protein expression, was found in the diabetic NASH mice. Overall, these data suggest that the diabetic NASH model only partially recapitulates the human ex vivo CYP alteration pattern. Therefore, in vivo determination of the effects of NASH on CYP activity should be conducted in human, and more appropriate models are required for future drug metabolism studies in NASH.
Collapse
Affiliation(s)
- Hui Li
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - John D. Clarke
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Anika L. Dzierlenga
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - John Bear
- Statistical Consulting Lab, Univeristy of Arizona, Tucson, AZ, 85721, USA
| | - Michael J. Goedken
- Translational Sciences, Research Pathology Services, Rutgers University, New Brunswick, NJ 08854, USA
| | - Nathan J. Cherrington
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
79
|
Kostrzewski T, Cornforth T, Snow SA, Ouro-Gnao L, Rowe C, Large EM, Hughes DJ. Three-dimensional perfused human in vitro model of non-alcoholic fatty liver disease. World J Gastroenterol 2017; 23:204-215. [PMID: 28127194 PMCID: PMC5236500 DOI: 10.3748/wjg.v23.i2.204] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/25/2016] [Accepted: 11/16/2016] [Indexed: 02/07/2023] Open
Abstract
AIM To develop a human in vitro model of non-alcoholic fatty liver disease (NAFLD), utilising primary hepatocytes cultured in a three-dimensional (3D) perfused platform.
METHODS Fat and lean culture media were developed to directly investigate the effects of fat loading on primary hepatocytes cultured in a 3D perfused culture system. Oil Red O staining was used to measure fat loading in the hepatocytes and the consumption of free fatty acids (FFA) from culture medium was monitored. Hepatic functions, gene expression profiles and adipokine release were compared for cells cultured in fat and lean conditions. To determine if fat loading in the system could be modulated hepatocytes were treated with known anti-steatotic compounds.
RESULTS Hepatocytes cultured in fat medium were found to accumulate three times more fat than lean cells and fat uptake was continuous over a 14-d culture. Fat loading of hepatocytes did not cause any hepatotoxicity and significantly increased albumin production. Numerous adipokines were expressed by fatty cells and genes associated with NAFLD and liver disease were upregulated including: Insulin-like growth factor-binding protein 1, fatty acid-binding protein 3 and CYP7A1. The metabolic activity of hepatocytes cultured in fatty conditions was found to be impaired and the activities of CYP3A4 and CYP2C9 were significantly reduced, similar to observations made in NAFLD patients. The utility of the model for drug screening was demonstrated by measuring the effects of known anti-steatotic compounds. Hepatocytes, cultured under fatty conditions and treated with metformin, had a reduced cellular fat content compared to untreated controls and consumed less FFA from cell culture medium.
CONCLUSION The 3D in vitro NAFLD model recapitulates many features of clinical NAFLD and is an ideal tool for analysing the efficacy of anti-steatotic compounds.
Collapse
|
80
|
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
|
81
|
Lake AD, Chaput AL, Novak P, Cherrington NJ, Smith CL. Transcription factor binding site enrichment analysis predicts drivers of altered gene expression in nonalcoholic steatohepatitis. Biochem Pharmacol 2016; 122:62-71. [PMID: 27836672 PMCID: PMC5129657 DOI: 10.1016/j.bcp.2016.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/04/2016] [Indexed: 12/21/2022]
Abstract
The molecular mechanisms behind the transition from simple steatosis to nonalcoholic steatohepatitis (NASH) in nonalcoholic fatty liver disease (NAFLD) are not clearly understood. This hinders development of effective therapies for treatment and prevention of NASH. In this study expression profiling data from normal, steatosis, and NASH human livers were used to predict transcription factors that are misregulated as mechanistic features of NAFLD progression. Previously-published human NAFLD gene expression profiling data from normal, steatosis, and NASH livers were subjected to transcription factor binding site enrichment analysis. Selected transcription factors that bind enriched transcription factor binding sites were analyzed for changes in expression. Distinct transcription factor binding sites were enriched in genes significantly up- or down-regulated in NASH livers. Those enriched in up-regulated genes were bound by transcription factors such as FOXA, CEBP, and HNF1 family members, while those enriched in down-regulated genes were bound by nuclear receptors involved in xenobiotic sensing and lipid metabolism. Levels of mRNA and protein for selected transcription factors were significantly changed during disease progression. The study indicates that NAFLD progression involves changes in activity or expression of transcription factors that regulate genes involved in hepatic processes known to be altered in NASH. Transcription factors such as PPAR receptors, FoxA family members, and HNF4A might be targeted therapeutically to prevent NAFLD progression.
Collapse
Affiliation(s)
- April D Lake
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Alexandria L Chaput
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Petr Novak
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States; Biology Centre ASCR, Institute of Plant Molecular Biology, Branisovska 31, Ceske Budejovice CZ-37005, Czech Republic
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Catharine L Smith
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
82
|
Stroot PG. Blood oxidative stress (BLOS) is a secondary host defense system responding normally to anaerobic wound infection and inadvertently to dietary ultra-exogenous sulfide formation (USF). Med Hypotheses 2016; 98:28-34. [PMID: 28012599 DOI: 10.1016/j.mehy.2016.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/21/2016] [Accepted: 11/22/2016] [Indexed: 02/07/2023]
Abstract
Blood oxidative stress (BLOS) is the presence of white blood cells and platelets that are generating high levels of reactive oxygen species (ROS). A mathematical model links the level of BLOS or BLOS# and plasma sulfide concentration. An increase in the BLOS# reduces the plasma sulfide concentration. The reported maximum plasma sulfide concentration for defined health conditions were used to calculate the minimum BLOS#. Elevated BLOS generates high plasma concentration of ROS, which triggers multiple responses in the body that protect the host. First, insulin production by the pancreas is inhibited, which results in elevated blood glucose levels. This results in advanced glycation end products (AGE), which thicken the blood vessel wall. Elevated blood glucose levels also increases urination, which reduces the availability of substrates for infectious bacteria. Second, one or more signaling molecules are stimulated to produce vascular hypertrophy resulting in hypertension. Third, the initial stage of atherosclerosis thickens the blood vessel wall while also protecting the inner surface of the blood vessels from localized infection. The first three mechanisms provide added protection against pathogen migration through the blood vessel wall and reduce the cross-sectional area of blood vessels, which increases the retention time (RT) for improved ROS inactivation of pathogens. Fourth, genes expressed in the liver, which are associated with drug oxidation and uptake transport, are inhibited. This inhibition protects the host from any toxins produced by an anaerobic infection. Elevated BLOS also reduces plasma sulfide concentration, which inhibits wound healing and extends aerobic conditions of the wound. The normal induction of BLOS offers a short-term, cascade of several primary mechanisms for secondary defense against anaerobic infection of a wound. Normal induction of BLOS is due to ultra-exogenous sulfide formation (USF) generated by a local anaerobic infection of a wound in the natural environment. The presence of BLOS without infection is indicative of inadvertent dietary induction. Long-term dietary BLOS results in many severe inflammatory diseases and cancers that are common in an ageing population. Glands were identified as more susceptible to cancers caused by long-term dietary BLOS. Variable BLOS levels in patients of clinical trials may also be reducing effectiveness of experimental drugs and causing drug toxicity. If BLOS is confirmed as a secondary defense against infection that is inadvertently triggered by diet, then a large number of common health problems may be treated and managed by apheresis and dietary changes.
Collapse
|
83
|
Dzierlenga AL, Clarke JD, Cherrington NJ. Nonalcoholic Steatohepatitis Modulates Membrane Protein Retrieval and Insertion Processes. Drug Metab Dispos 2016; 44:1799-1807. [PMID: 27604106 PMCID: PMC5074469 DOI: 10.1124/dmd.116.071415] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/25/2016] [Indexed: 01/29/2023] Open
Abstract
Interindividual variability in drug response in nonalcoholic steatohepatitis (NASH) can be mediated by altered regulation of drug metabolizing enzymes and transporters. Among these is the mislocalization of multidrug resistance-associated protein (MRP2)/Mrp2 away from the canalicular membrane, which results in decreased transport of MRP2/Mrp2 substrates. The exact mechanism of this mislocalization is unknown, although increased activation of membrane retrieval processes may be one possibility. The current study measures the activation status of various mediators implicated in the active membrane retrieval or insertion of membrane proteins to identify which processes may be important in rodent methionine and choline deficient diet-induced NASH. The mediators currently known to be associated with transporter mislocalization are stimulated by oxidative stressors and choleretic stimuli, which play a role in the pathogenesis of NASH. The activation of protein kinases PKA, PKCα, PKCδ, and PKCε and substrates radixin, myristoylated alanine-rich C-kinase substrate, and Rab11 were measured by comparing the expression, phosphorylation, and membrane translocation between control and NASH. Many of the mediators exhibited altered activation in NASH rats. Consistent with membrane retrieval of Mrp2, NASH rats exhibited a decreased phosphorylation of radixin and increased membrane localization of PKCδ and PKCε, thought to be mediators of radixin dephosphorylation. Altered activation of PKCδ, PKA, and PKCα may impair the Rab11-mediated active insertion of Mrp2. Overall, these data suggest alterations in membrane retrieval and insertion processes that may contribute to altered localization of membrane proteins in NASH.
Collapse
Affiliation(s)
- A L Dzierlenga
- Department of Pharmacology & Toxicology, University of Arizona, Tucson, Arizona
| | - J D Clarke
- Department of Pharmacology & Toxicology, University of Arizona, Tucson, Arizona
| | - N J Cherrington
- Department of Pharmacology & Toxicology, University of Arizona, Tucson, Arizona
| |
Collapse
|
84
|
Abstract
INTRODUCTION Drug induced steatohepatitis (DISH), a form of drug induced liver injury (DILI) is characterized by intracellular accumulation of lipids in hepatocytes and subsequent inflammatory events, in some ways similar to the pathology seen with other metabolic, viral and genetic causes of non alcoholic fatty liver disease and steatohepatitis (NAFLD and NASH). Areas covered: This paper provides a comprehensive review of the main underlying mechanisms by which various drugs cause DISH, and outlines existing preclinical tools to predict it and study underlying pathways involved. The translational hurdles of these models are discussed, with the example of an organotypic liver system designed to address them. Finally, we describe the clinical assessment and management of DISH. Expert Opinion: The complexity of the interconnected mechanistic pathways underlying DISH makes it important that preclinical evaluation of drugs is done in a physiologically and metabolically relevant context. Advanced organotypic tissue models, coupled with translational functional biomarkers and next-generational pan-omic measurements, may offer the best shot at gathering mechanistic knowledge and potential of a drug causing steatohepatitis. Ultimately this information could also help predict, detect or guide the development of specific treatments for DISH, which is an unmet need as of today.
Collapse
Affiliation(s)
- Ajit Dash
- a HemoShear Therapeutics LLC , Charlottesville , VA , USA
| | | | - Arun J Sanyal
- b Department of Internal Medicine, School of Medicine , Virginia Commonwealth University , Richmond , VA , USA
| | | |
Collapse
|
85
|
Nachshon A, Abu-Toamih Atamni HJ, Steuerman Y, Sheikh-Hamed R, Dorman A, Mott R, Dohm JC, Lehrach H, Sultan M, Shamir R, Sauer S, Himmelbauer H, Iraqi FA, Gat-Viks I. Dissecting the Effect of Genetic Variation on the Hepatic Expression of Drug Disposition Genes across the Collaborative Cross Mouse Strains. Front Genet 2016; 7:172. [PMID: 27761138 PMCID: PMC5050206 DOI: 10.3389/fgene.2016.00172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/09/2016] [Indexed: 12/26/2022] Open
Abstract
A central challenge in pharmaceutical research is to investigate genetic variation in response to drugs. The Collaborative Cross (CC) mouse reference population is a promising model for pharmacogenomic studies because of its large amount of genetic variation, genetic reproducibility, and dense recombination sites. While the CC lines are phenotypically diverse, their genetic diversity in drug disposition processes, such as detoxification reactions, is still largely uncharacterized. Here we systematically measured RNA-sequencing expression profiles from livers of 29 CC lines under baseline conditions. We then leveraged a reference collection of metabolic biotransformation pathways to map potential relations between drugs and their underlying expression quantitative trait loci (eQTLs). By applying this approach on proximal eQTLs, including eQTLs acting on the overall expression of genes and on the expression of particular transcript isoforms, we were able to construct the organization of hepatic eQTL-drug connectivity across the CC population. The analysis revealed a substantial impact of genetic variation acting on drug biotransformation, allowed mapping of potential joint genetic effects in the context of individual drugs, and demonstrated crosstalk between drug metabolism and lipid metabolism. Our findings provide a resource for investigating drug disposition in the CC strains, and offer a new paradigm for integrating biotransformation reactions to corresponding variations in DNA sequences.
Collapse
Affiliation(s)
- Aharon Nachshon
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel-Aviv University Tel-Aviv, Israel
| | - Hanifa J Abu-Toamih Atamni
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel- Aviv University Tel-Aviv, Israel
| | - Yael Steuerman
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel-Aviv University Tel-Aviv, Israel
| | - Roa'a Sheikh-Hamed
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel- Aviv University Tel-Aviv, Israel
| | - Alexandra Dorman
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel- Aviv University Tel-Aviv, Israel
| | - Richard Mott
- Genetics Institute, University College of London London, UK
| | - Juliane C Dohm
- Genomics Unit, Center for Genomic RegulationBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna (BOKU)Vienna, Austria
| | - Hans Lehrach
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics Berlin, Germany
| | - Marc Sultan
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics Berlin, Germany
| | - Ron Shamir
- The Blavatnik School of Computer Science, Tel Aviv University Tel Aviv, Israel
| | - Sascha Sauer
- Department of Vertebrate Genomics, Max Planck Institute for Molecular GeneticsBerlin, Germany; CU Systems Medicine, University of WürzburgWürzburg, Germany
| | - Heinz Himmelbauer
- Genomics Unit, Center for Genomic RegulationBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna (BOKU)Vienna, Austria
| | - Fuad A Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel- Aviv University Tel-Aviv, Israel
| | - Irit Gat-Viks
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel-Aviv University Tel-Aviv, Israel
| |
Collapse
|
86
|
Teufel A, Itzel T, Erhart W, Brosch M, Wang XY, Kim YO, von Schönfels W, Herrmann A, Brückner S, Stickel F, Dufour JF, Chavakis T, Hellerbrand C, Spang R, Maass T, Becker T, Schreiber S, Schafmayer C, Schuppan D, Hampe J. Comparison of Gene Expression Patterns Between Mouse Models of Nonalcoholic Fatty Liver Disease and Liver Tissues From Patients. Gastroenterology 2016; 151:513-525.e0. [PMID: 27318147 DOI: 10.1053/j.gastro.2016.05.051] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/23/2016] [Accepted: 05/27/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder in industrialized countries. Mouse models of NAFLD have been used in studies of pathogenesis and treatment, and have certain features of the human disease. We performed a systematic transcriptome-wide analysis of liver tissues from patients at different stages of NAFLD progression (ranging from healthy obese individuals to those with steatosis), as well as rodent models of NAFLD, to identify those that most closely resemble human disease progression in terms of gene expression patterns. METHODS We performed a systematic evaluation of genome-wide messenger RNA expression using liver tissues collected from mice fed a standard chow diet (controls) and 9 mouse models of NAFLD: mice on a high-fat diet (with or without fructose), mice on a Western-type diet, mice on a methionine- and choline-deficient diet, mice on a high-fat diet given streptozotocin, and mice with disruption of Pten in hepatocytes. We compared gene expression patterns with those of liver tissues from 25 patients with nonalcoholic steatohepatitis (NASH), 27 patients with NAFLD, 15 healthy obese individuals, and 39 healthy nonobese individuals (controls). Liver samples were obtained from patients undergoing liver biopsy for suspected NAFLD or NASH, or during liver or bariatric surgeries. Data sets were analyzed using the limma R-package. Overlap of functional profiles was analyzed by gene set enrichment analysis profiles. RESULTS We found differences between human and mouse transcriptomes to be significantly larger than differences between disease stages or models. Of the 65 genes with significantly altered expression in patients with NASH and 177 genes with significantly altered expression in patients with NAFLD, compared with controls, only 1-18 of these genes also differed significantly in expression between mouse models of NAFLD and control mice. However, expression of genes that regulate pathways associated with the development of NAFLD were altered in some mouse models (such as pathways associated with lipid metabolism). On a pathway level, gene expression patterns in livers of mice on the high-fat diet were associated more closely with human fatty liver disease than other models. CONCLUSIONS In comparing gene expression profiles between liver tissues from different mouse models of NAFLD and patients with different stages of NAFLD, we found very little overlap. Our data set is available for studies of pathways that contribute to the development of NASH and NAFLD and selection of the most applicable mouse models (http://www.nash-profiler.com).
Collapse
Affiliation(s)
- Andreas Teufel
- Department of Medicine I, University Hospital, Regensburg, Germany.
| | - Timo Itzel
- Department of Medicine I, University Hospital, Regensburg, Germany
| | - Wiebke Erhart
- Department of Internal Medicine I, University Hospital, Kiel, Germany
| | - Mario Brosch
- Medical Department 1, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Xiao Yu Wang
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Mainz, Germany
| | - Yong Ook Kim
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Mainz, Germany
| | | | | | - Stefan Brückner
- Medical Department 1, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| | - Felix Stickel
- Department of Clinical Research, Division of Hepatology, University of Berne, Berne, Switzerland
| | - Jean-François Dufour
- Department of Clinical Research, Division of Hepatology, University of Berne, Berne, Switzerland
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Technical University Dresden, Dresden, Germany
| | | | - Rainer Spang
- Statistical Bioinfomatics, Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Thorsten Maass
- Department of Medicine I, University Hospital, Regensburg, Germany
| | - Thomas Becker
- Department of Visceral and Thoracic Surgery, University Hospital, Kiel, Germany
| | - Stefan Schreiber
- Department of Internal Medicine I, University Hospital, Kiel, Germany
| | - Clemens Schafmayer
- Department of Visceral and Thoracic Surgery, University Hospital, Kiel, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jochen Hampe
- Medical Department 1, University Hospital Dresden, Technical University Dresden, Dresden, Germany
| |
Collapse
|
87
|
Dzierlenga AL, Clarke JD, Klein DM, Anumol T, Snyder SA, Li H, Cherrington NJ. Biliary Elimination of Pemetrexed Is Dependent on Mrp2 in Rats: Potential Mechanism of Variable Response in Nonalcoholic Steatohepatitis. J Pharmacol Exp Ther 2016; 358:246-53. [PMID: 27233293 PMCID: PMC4959105 DOI: 10.1124/jpet.116.234310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 05/26/2016] [Indexed: 12/22/2022] Open
Abstract
Hepatic multidrug resistance-associated protein 2 (MRP2) provides the biliary elimination pathway for many xenobiotics. Disruption of this pathway contributes to retention of these compounds and may ultimately lead to adverse drug reactions. MRP2 mislocalization from the canalicular membrane has been observed in nonalcoholic steatohepatitis (NASH), the late stage of nonalcoholic fatty liver disease, which is characterized by fat accumulation, oxidative stress, inflammation, and fibrosis. MRP2/Mrp2 mislocalization is observed in both human NASH and the rodent methionine and choline-deficient (MCD) diet model, but the extent to which it impacts overall transport capacity of MRP2 is unknown. Pemetrexed is an antifolate chemotherapeutic indicated for non-small cell lung cancer, yet its hepatobiliary elimination pathway has yet to be determined. The purpose of this study was to quantify the loss of Mrp2 function in NASH using an obligate Mrp2 transport substrate. To determine whether pemetrexed is an obligate Mrp2 substrate, its cumulative biliary elimination was compared between wild-type and Mrp2(-/-) rats. No pemetrexed was detected in the bile of Mrp2(-/-) rats, indicating pemetrexed is completely reliant on Mrp2 function for biliary elimination. Comparing the biliary elimination of pemetrexed between MCD and control animals identified a transporter-dependent decrease in biliary excretion of 60% in NASH. This study identifies Mrp2 as the exclusive biliary elimination mechanism for pemetrexed, making it a useful in vivo probe substrate for Mrp2 function, and quantifying the loss of function in NASH. This mechanistic feature may provide useful insight into the impact of NASH on interindividual variability in response to pemetrexed.
Collapse
Affiliation(s)
- Anika L Dzierlenga
- Department of Pharmacology and Toxicology (A.L.D., J.D.C., D.M.K., H.Y.L., N.J.C.); Department of Chemical and Environmental Engineering (T.A., S.A.S.), University of Arizona, Tucson, Arizona
| | - John D Clarke
- Department of Pharmacology and Toxicology (A.L.D., J.D.C., D.M.K., H.Y.L., N.J.C.); Department of Chemical and Environmental Engineering (T.A., S.A.S.), University of Arizona, Tucson, Arizona
| | - David M Klein
- Department of Pharmacology and Toxicology (A.L.D., J.D.C., D.M.K., H.Y.L., N.J.C.); Department of Chemical and Environmental Engineering (T.A., S.A.S.), University of Arizona, Tucson, Arizona
| | - Tarun Anumol
- Department of Pharmacology and Toxicology (A.L.D., J.D.C., D.M.K., H.Y.L., N.J.C.); Department of Chemical and Environmental Engineering (T.A., S.A.S.), University of Arizona, Tucson, Arizona
| | - Shane A Snyder
- Department of Pharmacology and Toxicology (A.L.D., J.D.C., D.M.K., H.Y.L., N.J.C.); Department of Chemical and Environmental Engineering (T.A., S.A.S.), University of Arizona, Tucson, Arizona
| | - HongYu Li
- Department of Pharmacology and Toxicology (A.L.D., J.D.C., D.M.K., H.Y.L., N.J.C.); Department of Chemical and Environmental Engineering (T.A., S.A.S.), University of Arizona, Tucson, Arizona
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology (A.L.D., J.D.C., D.M.K., H.Y.L., N.J.C.); Department of Chemical and Environmental Engineering (T.A., S.A.S.), University of Arizona, Tucson, Arizona
| |
Collapse
|
88
|
Aragonès G, Auguet T, Berlanga A, Guiu-Jurado E, Martinez S, Armengol S, Sabench F, Ras R, Hernandez M, Aguilar C, Colom J, Sirvent JJ, Del Castillo D, Richart C. Increased Circulating Levels of Alpha-Ketoglutarate in Morbidly Obese Women with Non-Alcoholic Fatty Liver Disease. PLoS One 2016; 11:e0154601. [PMID: 27123846 PMCID: PMC4849715 DOI: 10.1371/journal.pone.0154601] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/16/2016] [Indexed: 01/06/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) causes a wide spectrum of liver damage, ranging from simple steatosis to cirrhosis. However, simple steatosis (SS) and steatohepatitis (NASH) cannot yet be distinguished by clinical or laboratory features. The aim of this study was to assess the relationship between alpha-ketoglutarate and the degrees of NAFLD in morbidly obese patients. Materials and Methods We used a gas chromatography-quadruple time-of-flight-mass spectrometry analysis to quantify alpha-ketoglutarate in serum from normal-weight subjects (n = 30) and morbidly obese women (n = 97) with or without NAFLD. Results We found that serum levels of alpha-ketoglutarate were significantly higher in morbidly obese women than in normal-weight women. We showed that circulating levels of alpha-ketoglutarate were lower in lean controls and morbidly obese patients without NAFLD. We also found that alpha-ketoglutarate serum levels were higher in both SS and NASH than in normal liver of morbidly obese patients. However, there was no difference between SS and NASH. Moreover, we observed that circulating levels of alpha-ketoglutarate were associated with glucose metabolism parameters, lipid profile, hepatic enzymes and steatosis degree. In addition, diagnostic performance of alpha-ketoglutarate has been analyzed in NAFLD patients. The AUROC curves from patients with liver steatosis exhibited an acceptable clinical utility. Finally, we showed that the combination of biomarkers (AST, ALT and alpha-ketoglutarate) had the highest accuracy in diagnosing liver steatosis. Conclusion These findings suggest that alpha-ketoglutarate can determine the presence of non-alcoholic fatty liver in morbidly obese patients but it is not valid a biomarker for NASH.
Collapse
Affiliation(s)
- Gemma Aragonès
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Teresa Auguet
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
- Servei Medicina Interna, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
| | - Alba Berlanga
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Esther Guiu-Jurado
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Salomé Martinez
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
| | - Sandra Armengol
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Fàtima Sabench
- Servei de Cirurgia. Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Rosa Ras
- Group of Research on Omic Methodologies (GROM). Centre for Omic Sciences (COS), Reus, Spain
| | - Mercè Hernandez
- Servei de Cirurgia. Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Carmen Aguilar
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
| | - Josep Colom
- Servei Medicina Interna, Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Joan Josep Sirvent
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
| | - Daniel Del Castillo
- Servei de Cirurgia. Hospital Sant Joan de Reus. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204, Reus, Spain
| | - Cristóbal Richart
- Grup de Recerca GEMMAIR (AGAUR)- Medicina Aplicada. Departament de Medicina i Cirurgia. Universitat Rovira i Virgili (URV), Institut d’Investigació Sanitària Pere Virgili (IISPV), 43007, Tarragona, Spain
- Servei Medicina Interna, Hospital Universitari Joan XXIII Tarragona. Mallafré Guasch, 4, 43007, Tarragona, Spain
- * E-mail:
| |
Collapse
|
89
|
Ditzel EJ, Li H, Foy CE, Perrera AB, Parker P, Renquist BJ, Cherrington NJ, Camenisch TD. Altered Hepatic Transport by Fetal Arsenite Exposure in Diet-Induced Fatty Liver Disease. J Biochem Mol Toxicol 2016; 30:321-30. [PMID: 26890134 DOI: 10.1002/jbt.21796] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/09/2016] [Accepted: 01/15/2016] [Indexed: 12/21/2022]
Abstract
Non-alcoholic fatty liver disease can result in changes to drug metabolism and disposition potentiating adverse drug reactions. Furthermore, arsenite exposure during development compounds the severity of diet-induced fatty liver disease. This study examines the effects of arsenite potentiated diet-induced fatty liver disease on hepatic transport in male mice. Changes were detected for Mrp2/3/4 hepatic transporter gene expression as well as for Oatp1a4/2b1/1b2. Plasma concentrations of Mrp and Oatp substrates were increased in arsenic exposure groups compared with diet-only controls. In addition, murine embryonic hepatocytes and adult primary hepatocytes show significantly altered transporter expression after exposure to arsenite alone: a previously unreported phenomenon. These data indicate that developmental exposure to arsenite leads to changes in hepatic transport which could increase the risk for ADRs during fatty liver disease.
Collapse
Affiliation(s)
- Eric J Ditzel
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.
| | - Hui Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Caroline E Foy
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Alec B Perrera
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Patricia Parker
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Benjamin J Renquist
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.,Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA
| | - Todd D Camenisch
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.,Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA.,Steele Children's Research Center, Arizona Health Sciences Center, The University of Arizona, Tucson, AZ, 85724, USA.,Sarver Heart Center, The University of Arizona, Tucson, AZ, 85724, USA.,Bio5 Institute, The University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
90
|
Michaut A, Le Guillou D, Moreau C, Bucher S, McGill MR, Martinais S, Gicquel T, Morel I, Robin MA, Jaeschke H, Fromenty B. A cellular model to study drug-induced liver injury in nonalcoholic fatty liver disease: Application to acetaminophen. Toxicol Appl Pharmacol 2015; 292:40-55. [PMID: 26739624 DOI: 10.1016/j.taap.2015.12.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/18/2015] [Accepted: 12/22/2015] [Indexed: 12/22/2022]
Abstract
Obesity and nonalcoholic fatty liver disease (NAFLD) can increase susceptibility to hepatotoxicity induced by some xenobiotics including drugs, but the involved mechanisms are poorly understood. For acetaminophen (APAP), a role of hepatic cytochrome P450 2E1 (CYP2E1) is suspected since the activity of this enzyme is consistently enhanced during NAFLD. The first aim of our study was to set up a cellular model of NAFLD characterized not only by triglyceride accumulation but also by higher CYP2E1 activity. To this end, human HepaRG cells were incubated for one week with stearic acid or oleic acid, in the presence of different concentrations of insulin. Although cellular triglycerides and the expression of lipid-responsive genes were similar with both fatty acids, CYP2E1 activity was significantly increased only by stearic acid. CYP2E1 activity was reduced by insulin and this effect was reproduced in cultured primary human hepatocytes. Next, APAP cytotoxicity was assessed in HepaRG cells with or without lipid accretion and CYP2E1 induction. Experiments with a large range of APAP concentrations showed that the loss of ATP and glutathione was almost always greater in the presence of stearic acid. In cells pretreated with the CYP2E1 inhibitor chlormethiazole, recovery of ATP was significantly higher in the presence of stearate with low (2.5mM) or high (20mM) concentrations of APAP. Levels of APAP-glucuronide were significantly enhanced by insulin. Hence, HepaRG cells can be used as a valuable model of NAFLD to unveil important metabolic and hormonal factors which can increase susceptibility to drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Anaïs Michaut
- INSERM, U991, Université de Rennes 1, Rennes, France
| | | | - Caroline Moreau
- INSERM, U991, Université de Rennes 1, Rennes, France; Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | - Simon Bucher
- INSERM, U991, Université de Rennes 1, Rennes, France
| | - Mitchell R McGill
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Thomas Gicquel
- INSERM, U991, Université de Rennes 1, Rennes, France; Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | - Isabelle Morel
- INSERM, U991, Université de Rennes 1, Rennes, France; Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | | | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | | |
Collapse
|
91
|
Rondini EA, Pant A, Kocarek TA. Transcriptional Regulation of Cytosolic Sulfotransferase 1C2 by Intermediates of the Cholesterol Biosynthetic Pathway in Primary Cultured Rat Hepatocytes. J Pharmacol Exp Ther 2015; 355:429-41. [PMID: 26427720 PMCID: PMC4658488 DOI: 10.1124/jpet.115.226365] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/30/2015] [Indexed: 01/19/2023] Open
Abstract
Cytosolic sulfotransferase 1C2 (SULT1C2) is expressed in the kidney, stomach, and liver of rats; however, the mechanisms regulating expression of this enzyme are not known. We evaluated transcriptional regulation of SULT1C2 by mevalonate (MVA)-derived intermediates in primary cultured rat hepatocytes using several cholesterol synthesis inhibitors. Blocking production of mevalonate with the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor pravastatin (30 μM), reduced SULT1C2 mRNA content by ∼40% whereas the squalene synthase inhibitor squalestatin (SQ1, 0.1 μM), which causes accumulation of nonsterol isoprenoids, increased mRNA content by 4-fold. Treatment with MVA (10 mM) strongly induced SULT1C2 mRNA by 12-fold, and this effect was blocked by inhibiting squalene epoxidase but not by more distal cholesterol inhibitors, indicating the effects of MVA are mediated by postsqualene metabolites. Using rapid amplification of cDNA ends (RACE), we characterized the 5' end of SULT1C2 mRNA and used this information to generate constructs for promoter analysis. SQ1 and MVA increased reporter activity by ∼1.6- and 3-fold, respectively, from a construct beginning 49 base pairs (bp) upstream from the longest 5'-RACE product (-3140:-49). Sequence deletions from this construct revealed a hepatocyte nuclear factor 1 (HNF1) element (-2558), and mutation of this element reduced basal (75%) and MVA-induced (30%) reporter activity and attenuated promoter activation following overexpression of HNF1α or 1β. However, the effects of SQ1 were localized to a more proximal promoter region (-281:-49). Collectively, our findings demonstrate that cholesterol biosynthetic intermediates influence SULT1C2 expression in rat primary hepatocytes. Further, HNF1 appears to play an important role in mediating basal and MVA-induced SULT1C2 transcription.
Collapse
Affiliation(s)
- Elizabeth A Rondini
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan
| | - Asmita Pant
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan
| | - Thomas A Kocarek
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan
| |
Collapse
|
92
|
Genome-scale metabolic modelling of hepatocytes reveals serine deficiency in patients with non-alcoholic fatty liver disease. Nat Commun 2015; 5:3083. [PMID: 24419221 DOI: 10.1038/ncomms4083] [Citation(s) in RCA: 408] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 12/10/2013] [Indexed: 02/07/2023] Open
Abstract
Several liver disorders result from perturbations in the metabolism of hepatocytes, and their underlying mechanisms can be outlined through the use of genome-scale metabolic models (GEMs). Here we reconstruct a consensus GEM for hepatocytes, which we call iHepatocytes2322, that extends previous models by including an extensive description of lipid metabolism. We build iHepatocytes2322 using Human Metabolic Reaction 2.0 database and proteomics data in Human Protein Atlas, which experimentally validates the incorporated reactions. The reconstruction process enables improved annotation of the proteomics data using the network centric view of iHepatocytes2322. We then use iHepatocytes2322 to analyse transcriptomics data obtained from patients with non-alcoholic fatty liver disease. We show that blood concentrations of chondroitin and heparan sulphates are suitable for diagnosing non-alcoholic steatohepatitis and for the staging of non-alcoholic fatty liver disease. Furthermore, we observe serine deficiency in patients with NASH and identify PSPH, SHMT1 and BCAT1 as potential therapeutic targets for the treatment of non-alcoholic steatohepatitis.
Collapse
|
93
|
Brouwer KLR, Aleksunes LM, Brandys B, Giacoia GP, Knipp G, Lukacova V, Meibohm B, Nigam SK, Rieder M, de Wildt SN. Human Ontogeny of Drug Transporters: Review and Recommendations of the Pediatric Transporter Working Group. Clin Pharmacol Ther 2015; 98:266-87. [PMID: 26088472 DOI: 10.1002/cpt.176] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 12/19/2022]
Abstract
The critical importance of membrane-bound transporters in pharmacotherapy is widely recognized, but little is known about drug transporter activity in children. In this white paper, the Pediatric Transporter Working Group presents a systematic review of the ontogeny of clinically relevant membrane transporters (e.g., SLC, ABC superfamilies) in intestine, liver, and kidney. Different developmental patterns for individual transporters emerge, but much remains unknown. Recommendations to increase our understanding of membrane transporters in pediatric pharmacotherapy are presented.
Collapse
Affiliation(s)
- K L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - L M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers, the State University of New Jersey, Ernest Mario School of Pharmacy, Piscataway, New Jersey, USA
| | - B Brandys
- NIH Library, National Institutes of Health, Bethesda, Maryland, USA
| | - G P Giacoia
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Rockville, Maryland, USA
| | - G Knipp
- College of Pharmacy, Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana, USA
| | - V Lukacova
- Simulations Plus, lnc., Lancaster, California, USA
| | - B Meibohm
- University of Tennessee Health Science Center, College of Pharmacy, Memphis, Tennessee, USA
| | - S K Nigam
- University of California San Diego, La Jolla, California, USA
| | - M Rieder
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada
| | - S N de Wildt
- Erasmus MC Sophia Children's Hospital, Intensive Care and Department of Pediatric Surgery, Rotterdam, the Netherlands
| | | |
Collapse
|
94
|
Kimoto E, Li R, Scialis RJ, Lai Y, Varma MVS. Hepatic Disposition of Gemfibrozil and Its Major Metabolite Gemfibrozil 1-O-β-Glucuronide. Mol Pharm 2015; 12:3943-52. [DOI: 10.1021/acs.molpharmaceut.5b00411] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Emi Kimoto
- Department
of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Rui Li
- Systems
Modeling and Simulation, Department of Pharmacokinetics, Dynamics,
and Metabolism, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, United States
| | - Renato J. Scialis
- Department
of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Yurong Lai
- Departments
of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, , Princeton, New Jersey 08540, United States
| | - Manthena V. S. Varma
- Department
of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| |
Collapse
|
95
|
Clarke JD, Dzierlenga AL, Nelson NR, Li H, Werts S, Goedken MJ, Cherrington NJ. Mechanism of Altered Metformin Distribution in Nonalcoholic Steatohepatitis. Diabetes 2015; 64:3305-13. [PMID: 26016715 PMCID: PMC4542448 DOI: 10.2337/db14-1947] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 05/19/2015] [Indexed: 12/13/2022]
Abstract
Metformin is an antihyperglycemic drug that is widely prescribed for type 2 diabetes mellitus and is currently being investigated for the treatment of nonalcoholic steatohepatitis (NASH). NASH is known to alter hepatic membrane transporter expression and drug disposition similarly in humans and rodent models of NASH. Metformin is almost exclusively eliminated through the kidney primarily through active secretion mediated by Oct1, Oct2, and Mate1. The purpose of this study was to determine how NASH affects kidney transporter expression and metformin pharmacokinetics. A single oral dose of [(14)C]metformin was administered to C57BL/6J (wild type [WT]) and diabetic ob/ob mice fed either a control diet or a methionine- and choline-deficient (MCD) diet. Metformin plasma concentrations were slightly increased in the WT/MCD and ob/control groups, whereas plasma concentrations were 4.8-fold higher in ob/MCD mice compared with WT/control. The MCD diet significantly increased plasma half-life and mean residence time and correspondingly decreased oral clearance in both genotypes. These changes in disposition were caused by ob/ob- and MCD diet-specific decreases in the kidney mRNA expression of Oct2 and Mate1, whereas Oct1 mRNA expression was only decreased in ob/MCD mice. These results indicate that the diabetic ob/ob genotype and the MCD disease model alter kidney transporter expression and alter the pharmacokinetics of metformin, potentially increasing the risk of drug toxicity.
Collapse
Affiliation(s)
- John D Clarke
- Department of Pharmacology and Toxicology, The University of Arizona, Tucson, AZ
| | - Anika L Dzierlenga
- Department of Pharmacology and Toxicology, The University of Arizona, Tucson, AZ
| | - Nicholas R Nelson
- Department of Pharmacology and Toxicology, The University of Arizona, Tucson, AZ
| | - Hui Li
- Department of Pharmacology and Toxicology, The University of Arizona, Tucson, AZ
| | - Samantha Werts
- Department of Pharmacology and Toxicology, The University of Arizona, Tucson, AZ
| | - Michael J Goedken
- Translational Sciences, Research Pathology Services, Rutgers University, New Brunswick, NJ
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, The University of Arizona, Tucson, AZ
| |
Collapse
|
96
|
Drug Induced Steatohepatitis: An Uncommon Culprit of a Common Disease. BIOMED RESEARCH INTERNATIONAL 2015; 2015:168905. [PMID: 26273591 PMCID: PMC4529891 DOI: 10.1155/2015/168905] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/27/2015] [Accepted: 02/04/2015] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a leading cause of liver disease in developed countries. Its frequency is increasing in the general population mostly due to the widespread occurrence of obesity and the metabolic syndrome. Although drugs and dietary supplements are viewed as a major cause of acute liver injury, drug induced steatosis and steatohepatitis are considered a rare form of drug induced liver injury (DILI). The complex mechanism leading to hepatic steatosis caused by commonly used drugs such as amiodarone, methotrexate, tamoxifen, valproic acid, glucocorticoids, and others is not fully understood. It relates not only to induction of the metabolic syndrome by some drugs but also to their impact on important molecular pathways including increased hepatocytes lipogenesis, decreased secretion of fatty acids, and interruption of mitochondrial β-oxidation as well as altered expression of genes responsible for drug metabolism. Better familiarity with this type of liver injury is important for early recognition of drug hepatotoxicity and crucial for preventing severe forms of liver injury and cirrhosis. Moreover, understanding the mechanisms leading to drug induced hepatic steatosis may provide much needed clues to the mechanism and potential prevention of the more common form of metabolic steatohepatitis.
Collapse
|
97
|
Canet MJ, Merrell MD, Hardwick RN, Bataille AM, Campion SN, Ferreira DW, Xanthakos SA, Manautou JE, A-Kader HH, Erickson RP, Cherrington NJ. Altered regulation of hepatic efflux transporters disrupts acetaminophen disposition in pediatric nonalcoholic steatohepatitis. Drug Metab Dispos 2015; 43:829-35. [PMID: 25788542 PMCID: PMC4429682 DOI: 10.1124/dmd.114.062703] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 03/12/2015] [Indexed: 12/31/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, representing a spectrum of liver pathologies that include simple hepatic steatosis and the more advanced nonalcoholic steatohepatitis (NASH). The current study was conducted to determine whether pediatric NASH also results in altered disposition of acetaminophen (APAP) and its two primary metabolites, APAP-sulfate and APAP-glucuronide. Pediatric patients with hepatic steatosis (n = 9) or NASH (n = 3) and healthy patients (n = 12) were recruited in a small pilot study design. All patients received a single 1000-mg dose of APAP. Blood and urine samples were collected at 1, 2, and 4 hours postdose, and APAP and APAP metabolites were determined by high-performance liquid chromatography. Moreover, human liver tissues from patients diagnosed with various stages of NAFLD were acquired from the Liver Tissue Cell Distribution System to investigate the regulation of the membrane transporters, multidrug resistance-associated protein 2 and 3 (MRP2 and MRP3, respectively). Patients with the more severe disease (i.e., NASH) had increased serum and urinary levels of APAP-glucuronide along with decreased serum levels of APAP-sulfate. Moreover, an induction of hepatic MRP3 and altered canalicular localization of the biliary efflux transporter, MRP2, describes the likely mechanism for the observed increase in plasma retention of APAP-glucuronide, whereas altered regulation of sulfur activation genes may explain decreased sulfonation activity in NASH. APAP-glucuronide and APAP-sulfate disposition is altered in NASH and is likely due to hepatic membrane transporter dysregulation as well as altered intracellular sulfur activation.
Collapse
Affiliation(s)
- Mark J Canet
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Matthew D Merrell
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Rhiannon N Hardwick
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Amy M Bataille
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Sarah N Campion
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Daniel W Ferreira
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Stavra A Xanthakos
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Jose E Manautou
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - H Hesham A-Kader
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Robert P Erickson
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| | - Nathan J Cherrington
- Departments of Pharmacology and Toxicology (M.J.C., M.D.M., R.N.H., N.J.C.) and Pediatrics (H.A.K., R.P.E.), University of Arizona, Tucson, Arizona; School of Pharmacy, University of Connecticut, Storrs, Connecticut (A.M.B., D.W.F., J.E.M.); Drug Safety Research and Development, Pfizer, Inc., New York, New York (S.N.C.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio (S.A.X.)
| |
Collapse
|
98
|
Arendt BM, Comelli EM, Ma DWL, Lou W, Teterina A, Kim T, Fung SK, Wong DKH, McGilvray I, Fischer SE, Allard JP. Altered hepatic gene expression in nonalcoholic fatty liver disease is associated with lower hepatic n-3 and n-6 polyunsaturated fatty acids. Hepatology 2015; 61:1565-78. [PMID: 25581263 DOI: 10.1002/hep.27695] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 12/31/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED In nonalcoholic fatty liver disease, hepatic gene expression and fatty acid (FA) composition have been reported independently, but a comprehensive gene expression profiling in relation to FA composition is lacking. The aim was to assess this relationship. In a cross-sectional study, hepatic gene expression (Illumina Microarray) was first compared among 20 patients with simple steatosis (SS), 19 with nonalcoholic steatohepatitis (NASH), and 24 healthy controls. The FA composition in hepatic total lipids was compared between SS and NASH, and associations between gene expression and FAs were examined. Gene expression differed mainly between healthy controls and patients (SS and NASH), including genes related to unsaturated FA metabolism. Twenty-two genes were differentially expressed between NASH and SS; most of them correlated with disease severity and related more to cancer progression than to lipid metabolism. Biologically active long-chain polyunsaturated FAs (PUFAs; eicosapentaenoic acid + docosahexaenoic acid, arachidonic acid) in hepatic total lipids were lower in NASH than in SS. This may be related to overexpression of FADS1, FADS2, and PNPLA3. The degree and direction of correlations between PUFAs and gene expression were different among SS and NASH, which may suggest that low PUFA content in NASH modulates gene expression in a different way compared with SS or, alternatively, that gene expression influences PUFA content differently depending on disease severity (SS versus NASH). CONCLUSION Well-defined subjects with either healthy liver, SS, or NASH showed distinct hepatic gene expression profiles including genes involved in unsaturated FA metabolism. In patients with NASH, hepatic PUFAs were lower and associations with gene expression were different compared to SS.
Collapse
Affiliation(s)
- Bianca M Arendt
- Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Stoldt AK, Derno M, Nürnberg G, Weitzel JM, Otten W, Starke A, Wolffram S, Metges CC. Effects of a 6-wk intraduodenal supplementation with quercetin on energy metabolism and indicators of liver damage in periparturient dairy cows. J Dairy Sci 2015; 98:4509-20. [PMID: 25935242 DOI: 10.3168/jds.2014-9053] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 03/14/2015] [Indexed: 11/19/2022]
Abstract
Periparturient dairy cows experience metabolic challenges that result in a negative energy balance (EB) and a range of postpartum health problems. To compensate for the negative EB, cows mobilize fatty acids from adipose tissues, which can lead to fatty liver disease, a periparturient metabolic disorder. Flavonoids, such as quercetin (Q), are polyphenolic substances found in all higher plants and have hepatoprotective potential and the ability to prevent or reduce lipid accumulation in the liver. In ruminants, few studies on the metabolic effects of Q are available, and thus this study was conducted to determine whether Q has beneficial effects on EB, lipid metabolism, and hepatoprotective effects in periparturient dairy cows. Quercetin was supplemented intraduodenally to circumvent Q degradation in the rumen. Cows (n=10) with duodenal fistulas were monitored for 7wk. Beginning 3wk before expected calving, 5 cows were treated with 100mg of quercetin dihydrate per kilogram of body weight daily in a 0.9% sodium chloride solution for a total period of 6wk, whereas the control cows received only the sodium chloride solution. The plasma flavonoid levels were higher in the Q-treated cows than in the control cows. A tendency for higher postpartum (pp) than antepartum (ap) plasma flavonoid levels was observed in the Q-treated cows than in the controls, which was potentially caused by a reduced capacity to metabolize Q. However, the metabolic status of the Q-treated cows did not differ from that of the control cows. The pp increases in plasma aspartate aminotransferase and glutamate dehydrogenase activities were less in the Q-treated cows than in the control cows. The Q had no effect on energy expenditures, but from ap to pp the cows had a slight decline in respiratory quotients. Irrespective of the treatment group, the oxidation of fat peaked after calving, suggesting that the increase occurred because of an increased supply of fatty acids from lipomobilization. In conclusion, supplementation with Q resulted in lower pp plasma aminotransferase and glutamate dehydrogenase, which indicated reduced liver damage. However, the direct effects of Q on the liver and the implications for animal performance remain to be investigated.
Collapse
Affiliation(s)
- Ann-Kathrin Stoldt
- Institute of Nutritional Physiology "Oskar Kellner,", Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Michael Derno
- Institute of Nutritional Physiology "Oskar Kellner,", Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Gerd Nürnberg
- Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Joachim M Weitzel
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Winfried Otten
- Institute of Behavioural Physiology, Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Alexander Starke
- Clinic for Ruminants and Swine, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Siegfried Wolffram
- Institute of Animal Nutrition and Physiology, Christian-Albrechts University of Kiel, 24118 Kiel, Germany
| | - Cornelia C Metges
- Institute of Nutritional Physiology "Oskar Kellner,", Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany.
| |
Collapse
|
100
|
Ye H, Liu W. Transcriptional networks implicated in human nonalcoholic fatty liver disease. Mol Genet Genomics 2015; 290:1793-804. [PMID: 25851235 DOI: 10.1007/s00438-015-1037-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/27/2015] [Indexed: 02/06/2023]
Abstract
The transcriptome of nonalcoholic fatty liver disease (NAFLD) was investigated in several studies. However, the implications of transcriptional networks in progressive NAFLD are not clear and mechanisms inducing transition from nonalcoholic simple fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) are still elusive. The aims of this study were to (1) construct networks for progressive NAFLD, (2) identify hub genes and functional modules in these networks and (3) infer potential linkages among hub genes, transcription factors and microRNAs (miRNA) for NAFLD progression. A systems biology approach by combining differential expression analysis and weighted gene co-expression network analysis (WGCNA) was utilized to dissect transcriptional profiles in 19 normal, 10 NAFL and 16 NASH patients. Based on this framework, 3 modules related to chromosome organization, proteasomal ubiquitin-dependent protein degradation and immune response were identified in NASH network. Furthermore, 9 modules of co-expressed genes associated with NAFL/NASH transition were found. Further characterization of these modules defined 13 highly connected hub genes in NAFLD progression network. Interestingly, 11 significantly changed miRNAs were predicted to target 10 of the 13 hub genes. Characterization of modules and hub genes that may be regulated by miRNAs could facilitate the identification of candidate genes and pathways responsible for NAFL/NASH transition and lead to a better understanding of NAFLD pathogenesis. The identified modules and hub genes may point to potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Hua Ye
- Department of Gastroenterology, Ningbo Medical Treatment Center Lihuili Hospital, Ningbo, 315040, China
| | - Wei Liu
- School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
- Department of Pathology, Human Centrifuge Medical Training Center, Institute of Aviation Medicine of Chinese PLA Air Force, Beijing, 100089, China.
| |
Collapse
|