1
|
Schönke M, Rensen PC. Mouse Models for the Study of Liver Fibrosis Regression In Vivo and Ex Vivo. J Clin Transl Hepatol 2024; 12:930-938. [PMID: 39544245 PMCID: PMC11557367 DOI: 10.14218/jcth.2024.00212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/10/2024] [Accepted: 09/29/2024] [Indexed: 11/17/2024] Open
Abstract
This review discussed experimental mouse models used in the pre-clinical study of liver fibrosis regression, a pivotal process in preventing the progression of metabolic dysfunction-associated steatohepatitis to irreversible liver cirrhosis. These models provide a valuable resource for understanding the cellular and molecular processes underlying fibrosis regression in different contexts. The primary focus of this review is on the most commonly used models with diet- or hepatotoxin-induced fibrosis, but it also touches upon genetic models and mouse models with biliary atresia or parasite-induced fibrosis. In addition to emphasizing in vivo models, we briefly summarized current in vitro approaches designed for studying fibrosis regression and provided an outlook on evolving methodologies that aim to refine and reduce the number of experimental animals needed for these studies. Together, these models contribute significantly to unraveling the underlying mechanisms of liver fibrosis regression and offer insights into potential therapeutic interventions. By presenting a comprehensive overview of these models and highlighting their respective advantages and limitations, this review serves as a roadmap for future research.
Collapse
Affiliation(s)
- Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C.N. Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Lad A, Hunyadi J, Connolly J, Breidenbach JD, Khalaf FK, Dube P, Zhang S, Kleinhenz AL, Baliu-Rodriguez D, Isailovic D, Hinds TD, Gatto-Weis C, Stanoszek LM, Blomquist TM, Malhotra D, Haller ST, Kennedy DJ. Antioxidant Therapy Significantly Attenuates Hepatotoxicity following Low Dose Exposure to Microcystin-LR in a Murine Model of Diet-Induced Non-Alcoholic Fatty Liver Disease. Antioxidants (Basel) 2022; 11:1625. [PMID: 36009344 PMCID: PMC9404967 DOI: 10.3390/antiox11081625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 12/15/2022] Open
Abstract
We have previously shown in a murine model of Non-alcoholic Fatty Liver Disease (NAFLD) that chronic, low-dose exposure to the Harmful Algal Bloom cyanotoxin microcystin-LR (MC-LR), resulted in significant hepatotoxicity including micro-vesicular lipid accumulation, impaired toxin metabolism as well as dysregulation of the key signaling pathways involved in inflammation, immune response and oxidative stress. On this background we hypothesized that augmentation of hepatic drug metabolism pathways with targeted antioxidant therapies would improve MC-LR metabolism and reduce hepatic injury in NAFLD mice exposed to MC-LR. We chose N-acetylcysteine (NAC, 40 mM), a known antioxidant that augments the glutathione detoxification pathway and a novel peptide (pNaKtide, 25 mg/kg) which is targeted to interrupting a specific Src-kinase mediated pro-oxidant amplification mechanism. Histological analysis showed significant increase in hepatic inflammation in NAFLD mice exposed to MC-LR which was attenuated on treatment with both NAC and pNaKtide (both p ≤ 0.05). Oxidative stress, as measured by 8-OHDG levels in urine and protein carbonylation in liver sections, was also significantly downregulated upon treatment with both antioxidants after MC-LR exposure. Genetic analysis of key drug transporters including Abcb1a, Phase I enzyme-Cyp3a11 and Phase II metabolic enzymes-Pkm (Pyruvate kinase, muscle), Pklr (Pyruvate kinase, liver, and red blood cell) and Gad1 (Glutamic acid decarboxylase) was significantly altered by MC-LR exposure as compared to the non-exposed control group (all p ≤ 0.05). These changes were significantly attenuated with both pNaKtide and NAC treatment. These results suggest that MC-LR metabolism and detoxification is significantly impaired in the setting of NAFLD, and that these pathways can potentially be reversed with targeted antioxidant treatment.
Collapse
Affiliation(s)
- Apurva Lad
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Jonathan Hunyadi
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Jacob Connolly
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | | | - Fatimah K. Khalaf
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
- Department of Clinical Pharmacy, University of Alkafeel, Najaf 54001, Iraq
| | - Prabhatchandra Dube
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Shungang Zhang
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Andrew L. Kleinhenz
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - David Baliu-Rodriguez
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Dragan Isailovic
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Cara Gatto-Weis
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Lauren M. Stanoszek
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Thomas M. Blomquist
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Deepak Malhotra
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Steven T. Haller
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - David J. Kennedy
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
3
|
Genetics of Polygenic Metabolic Liver Disease. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11596-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
4
|
Liver-specific knockout of B cell lymphoma 6 suppresses progression of non-alcoholic steatohepatitis in mice. Sci Rep 2020; 10:9704. [PMID: 32546802 PMCID: PMC7297717 DOI: 10.1038/s41598-020-66539-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/21/2020] [Indexed: 12/19/2022] Open
Abstract
The prevalence of non-alcoholic steatohepatitis (NASH) rapidly increases with metabolic disorders such as dyslipidaemia, high blood pressure, and hyperglycaemia. B cell lymphoma 6 (Bcl6), a transcriptional repressor, is essential for the formation of germinal centre B cells. In this study, we analysed the role of Bcl6 in NASH progression-associated pathological changes, such as hepatic lipid accumulation, liver fibrosis, and hepatocarcinogenesis. The roles of Bcl6 in NASH were analysed using liver-specific Bcl6 knockout (Bcl6-LKO) and control wild-type (WT) mice. The murine NASH model was established by feeding the mice with choline-deficient, L-amino-acid-defined, high-fat diet (CDAHFD). Feeding the WT mice with CDAHFD for 7 weeks induced the formation of histopathological features resembling human NASH, such as hepatic lipid accumulation, hepatocellular injury, and fibrosis. These histopathological changes were significantly attenuated in Bcl6-LKO mice. Additionally, feeding the male WT mice with CDAHFD for 38 weeks induced the formation of liver tumours, which was suppressed in Bcl6-LKO mice. These findings indicate that Bcl6 is involved in the progression of NASH and NASH-derived tumours.
Collapse
|
5
|
Roh YS, Kim JW, Park S, Shon C, Kim S, Eo SK, Kwon JK, Lim CW, Kim B. Toll-Like Receptor-7 Signaling Promotes Nonalcoholic Steatohepatitis by Inhibiting Regulatory T Cells in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2574-2588. [PMID: 30125542 DOI: 10.1016/j.ajpath.2018.07.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 06/09/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022]
Abstract
Toll-like receptor 7 (TLR7) signaling regulates the production of type 1 interferons (IFNs) and proinflammatory cytokines, such as tumor necrosis factor (TNF)-α, implicated in the control of regulatory T (Treg) cell activity. However, the mechanistic interplay between TLR7 signaling and Treg cells in nonalcoholic steatohepatitis (NASH) has not been elucidated. Our aim was to clarify the role of TLR7 signaling in the pathogenesis of NASH. Steatohepatitis was induced in wild-type (WT), TLR7-deficient, IFN-α/β receptor 1-deficient, and Treg cell-depleted mice. TLR7-deficient and IFN-α/β receptor 1-deficient mice were more protective to steatohepatitis than WT mice. Of interest, both TNF-α and type 1 IFN promoted apoptosis of Treg cells involved in the prevention of NASH. Indeed, Treg cell-depleted mice had aggravated steatohepatitis compared with WT mice. Finally, treatment with immunoregulatory sequence 661, an antagonist of TLR7, efficiently ameliorated NASH in vivo. These results demonstrate that TLR7 signaling can induce TNF-α production in Kupffer cells and type I IFN production in dendritic cells. These cytokines subsequently induce hepatocyte death and inhibit Treg cells activities, leading to the progression of NASH. Thus, manipulating the TLR7-Treg cell axis might be used as a novel therapeutic strategy to treat NASH.
Collapse
|
6
|
Königshofer P, Brusilovskaya K, Schwabl P, Reiberger T. Animal models of portal hypertension. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1019-1030. [PMID: 30055295 DOI: 10.1016/j.bbadis.2018.07.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 07/14/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
Abstract
Chronic liver diseases ultimately lead to cirrhosis and portal hypertension (PHT). Indeed, PHT is a major cause of severe complications, while medical treatment is limited to non-selective beta blockers. Sophisticated animal models are needed to investigate novel treatment options for different etiologies of liver disease, effective anti-fibrotic agents as well as vasoactive drugs against PHT. In this review, we present some of the most common animal models of liver disease and PHT - including pre-hepatic, intra-hepatic and post-hepatic PHT in rodents. Methodology for induction, considerations for disease etiology, advantages and limitations and practical issues of these animal models are discussed. The appropriate and sensible use of animal models in preclinical research supporting the 3R concept of replacement, reduction and refinement is highlighted.
Collapse
Affiliation(s)
- P Königshofer
- Div. of Gastroenterology and Hepatology, Dept. of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna, Austria
| | - K Brusilovskaya
- Div. of Gastroenterology and Hepatology, Dept. of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna, Austria
| | - P Schwabl
- Div. of Gastroenterology and Hepatology, Dept. of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna, Austria
| | - T Reiberger
- Div. of Gastroenterology and Hepatology, Dept. of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Hemodynamic Lab, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Bertola A. WITHDRAWN: Rodent models of fatty liver diseases. LIVER RESEARCH 2018. [DOI: 10.1016/j.livres.2018.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
8
|
Stephenson K, Kennedy L, Hargrove L, Demieville J, Thomson J, Alpini G, Francis H. Updates on Dietary Models of Nonalcoholic Fatty Liver Disease: Current Studies and Insights. Gene Expr 2018; 18:5-17. [PMID: 29096730 PMCID: PMC5860971 DOI: 10.3727/105221617x15093707969658] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a disease of increasing interest, as its prevalence is on the rise. NAFLD has been linked to metabolic syndrome, which is becoming more common due to the Western diet. Because NAFLD can lead to cirrhosis and related complications including hepatocellular carcinoma, the increasing prevalence is concerning, and medical therapy aimed at treating NAFLD is of great interest. Researchers studying the effects of medical therapy on NAFLD use dietary mouse models. The two main types of mouse model diets are the methionine- and choline-deficient (MCD) diet and the Western-like diet (WD). Although both induce NAFLD, the mechanisms are very different. We reviewed several studies conducted within the last 5 years that used MCD diet or WD mouse models in order to mimic this disease in a way most similar to humans. The MCD diet inconsistently induces NAFLD and fibrosis and does not completely induce metabolic syndrome. Thus, the clinical significance of the MCD diet is questionable. In contrast, WD mouse models consisting of high fat, cholesterol, and a combination of high-fructose corn syrup, sucrose, fructose, or glucose not only lead to metabolic syndrome but also induce NAFLD with fibrosis, making these choices most suitable for research.
Collapse
Affiliation(s)
- Kristen Stephenson
- *Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX, USA
| | - Lindsey Kennedy
- †Research, Central Texas Veterans Health Care System, Temple, TX, USA
- ‡Department of Medicine, Texas A&M Health Science Center, Temple, Texas, USA
| | - Laura Hargrove
- ‡Department of Medicine, Texas A&M Health Science Center, Temple, Texas, USA
| | | | - Joanne Thomson
- †Research, Central Texas Veterans Health Care System, Temple, TX, USA
| | - Gianfranco Alpini
- *Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX, USA
- †Research, Central Texas Veterans Health Care System, Temple, TX, USA
- ‡Department of Medicine, Texas A&M Health Science Center, Temple, Texas, USA
| | - Heather Francis
- *Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX, USA
- †Research, Central Texas Veterans Health Care System, Temple, TX, USA
- ‡Department of Medicine, Texas A&M Health Science Center, Temple, Texas, USA
| |
Collapse
|
9
|
|
10
|
Duwaerts CC, Amin AM, Siao K, Her C, Fitch M, Beysen C, Turner SM, Goodsell A, Baron JL, Grenert JP, Cho SJ, Maher JJ. Specific Macronutrients Exert Unique Influences on the Adipose-Liver Axis to Promote Hepatic Steatosis in Mice. Cell Mol Gastroenterol Hepatol 2017; 4. [PMID: 28649594 PMCID: PMC5472193 DOI: 10.1016/j.jcmgh.2017.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The factors that distinguish metabolically healthy obesity from metabolically unhealthy obesity are not well understood. Diet has been implicated as a determinant of the unhealthy obesity phenotype, but which aspects of the diet induce dysmetabolism are unknown. The goal of this study was to investigate whether specific macronutrients or macronutrient combinations provoke dysmetabolism in the context of isocaloric, high-energy diets. METHODS Mice were fed 4 high-energy diets identical in calorie and nutrient content but different in nutrient composition for 3 weeks to 6 months. The test diets contained 42% carbohydrate (sucrose or starch) and 42% fat (oleate or palmitate). Weight and glucose tolerance were monitored; blood and tissues were collected for histology, gene expression, and immunophenotyping. RESULTS Mice gained weight on all 4 test diets but differed significantly in other metabolic outcomes. Animals fed the starch-oleate diet developed more severe hepatic steatosis than those on other formulas. Stable isotope incorporation showed that the excess hepatic steatosis in starch-oleate-fed mice derived from exaggerated adipose tissue lipolysis. In these mice, adipose tissue lipolysis coincided with adipocyte necrosis and inflammation. Notably, the liver and adipose tissue abnormalities provoked by starch-oleate feeding were reproduced when mice were fed a mixed-nutrient Western diet with 42% carbohydrate and 42% fat. CONCLUSIONS The macronutrient composition of the diet exerts a significant influence on metabolic outcome, independent of calories and nutrient proportions. Starch-oleate appears to cause hepatic steatosis by inducing progressive adipose tissue injury. Starch-oleate phenocopies the effect of a Western diet; consequently, it may provide clues to the mechanism whereby specific nutrients cause metabolically unhealthy obesity.
Collapse
Affiliation(s)
- Caroline C. Duwaerts
- Department of Medicine, University of California, San Francisco, California,The Liver Center, University of California, San Francisco, California
| | - Amin M. Amin
- Department of Medicine, University of California, San Francisco, California,The Liver Center, University of California, San Francisco, California
| | - Kevin Siao
- Department of Medicine, University of California, San Francisco, California,The Liver Center, University of California, San Francisco, California
| | - Chris Her
- Department of Medicine, University of California, San Francisco, California,The Liver Center, University of California, San Francisco, California
| | - Mark Fitch
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California
| | | | | | - Amanda Goodsell
- Department of Medicine, University of California, San Francisco, California,The Liver Center, University of California, San Francisco, California
| | - Jody L. Baron
- Department of Medicine, University of California, San Francisco, California,The Liver Center, University of California, San Francisco, California
| | - James P. Grenert
- The Liver Center, University of California, San Francisco, California,Department of Pathology, University of California, San Francisco, California
| | - Soo-Jin Cho
- Department of Pathology, University of California, San Francisco, California
| | - Jacquelyn J. Maher
- Department of Medicine, University of California, San Francisco, California,The Liver Center, University of California, San Francisco, California,Correspondence Address correspondence to: Jacquelyn J. Maher, MD, Liver Center Laboratory, 1001 Potrero Avenue, Building 40, Room 4102, San Francisco, California 94110. fax: (415) 641-0517.Liver Center Laboratory1001 Potrero Avenue, Building 40, Room 4102San FranciscoCalifornia 94110
| |
Collapse
|
11
|
Bloomer SA, Olivier AK, Bergmann OM, Mathahs MM, Broadhurst KA, Hicsasmaz H, Brown KE. Strain- and time-dependent alterations in hepatic iron metabolism in a murine model of nonalcoholic steatohepatitis. Cell Biochem Funct 2017; 34:628-639. [PMID: 27935134 DOI: 10.1002/cbf.3238] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 01/29/2023]
Abstract
Nonalcoholic steatohepatitis is a common liver disease that is often accompanied by dysregulated iron metabolism. The aim of the study was to test the hypothesis that aberrant iron metabolism in nonalcoholic steatohepatitis is modulated by genetic susceptibility to inflammation and oxidative stress. Hepatic histology and iron content were assessed in 3 inbred strains of mice (C57BL/6, BALB/c, and C3H/HeJ) fed an atherogenic diet (AD). Hepatic expression of genes relevant to iron metabolism, inflammation, and oxidative stress were quantitated by real-time reverse transcription-polymerase chain reaction. At 6 weeks on the AD, histologic injury and induction of inflammatory and oxidative stress-associated gene expression were most pronounced in C57BL/6. At 18 weeks on the AD, these parameters were similar in C57BL/6 and BALB/c. Atherogenic diet-fed C3H/HeJ showed milder responses at both time points. The AD was associated with decreased hepatic iron concentrations in all strains at 6 and 18 weeks. The decrease in hepatic iron concentrations did not correlate with changes in hepcidin expression and was not associated with altered expression of iron transporters. These findings are similar to those observed in models of obesity-induced steatosis and indicate that hepatic steatosis can be associated with depletion of iron stores that is not explained by upregulation of hepcidin expression by inflammation. SIGNIFICANCE OF THE STUDY Nonalcoholic steatohepatitis (NASH) is a common liver disease that often accompanies the metabolic syndrome. The latter condition has been linked to iron deficiency and diminished intestinal iron absorption, likely the result of hepcidin upregulation by chronic inflammation. Paradoxically, some NASH patients accumulate excess hepatic iron, which may increase fibrosis and cancer risk. Iron accumulation has been attributed to suppression of hepcidin by oxidative stress. The objective of this study was to investigate the contributions of inflammation and oxidative stress to altered hepatic iron metabolism in a murine model of NASH using inbred strains of mice with differing susceptibilities to injury.
Collapse
Affiliation(s)
- Steven A Bloomer
- Division of Science and Engineering, Penn State Abington, Abington, PA, USA
| | - Alicia K Olivier
- Division of Comparative Pathology, Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Pathobiology and Population Medicine, Mississippi State University, Starkville, MS, USA
| | - Ottar M Bergmann
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, Section of Gastroenterology and Hepatology, The National University Hospital of Iceland, Reykjavik, Iceland
| | - M Meleah Mathahs
- Iowa City Veterans Administration Medical Center, Iowa City, IA, USA
| | | | | | - Kyle E Brown
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Iowa City Veterans Administration Medical Center, Iowa City, IA, USA.,Program in Free Radical and Radiation Biology, Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
12
|
Page A, Paoli P, Salvador EM, White S, French J, Mann J. Hepatic stellate cell transdifferentiation involves genome-wide remodeling of the DNA methylation landscape. J Hepatol 2016; 64:661-73. [PMID: 26632634 PMCID: PMC4904781 DOI: 10.1016/j.jhep.2015.11.024] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/13/2015] [Accepted: 11/09/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS DNA methylation (5-mC) is an epigenetic mark that is an established regulator of transcriptional repression with an important role in liver fibrosis. Currently, there is very little knowledge available as to how DNA methylation controls the phenotype of hepatic stellate cell (HSC), the key cell type responsible for onset and progression of liver fibrosis. Moreover, recently discovered DNA hydroxymethylation (5-hmC) is involved in transcriptional activation and its patterns are often altered in human diseases. The aim of this study is to investigate the role of DNA methylation/hydroxymethylation in liver fibrosis. METHODS Levels of 5-mC and 5-hmC were assessed by slot blot in a range of animal liver fibrosis models and human liver diseases. Expression levels of TET and DNMT enzymes were measured by qRT-PCR and Western blotting. Reduced representation bisulfite sequencing (RRBS) method was used to examine 5-mC and 5-hmC patterns in quiescent and in vivo activated rat HSC. RESULTS We demonstrate global alteration in 5-mC and 5-hmC and their regulatory enzymes that accompany liver fibrosis and HSC transdifferentiation. Using RRBS, we show exact genomic positions of changed methylation patterns in quiescent and in vivo activated rat HSC. In addition, we demonstrate that reduction in DNMT3a expression leads to attenuation of pro-fibrogenic phenotype in activated HSC. CONCLUSIONS Our data suggest that DNA 5-mC/5-hmC is a crucial step in HSC activation and therefore fibrogenesis. Changes in DNA methylation during HSC activation may bring new insights into the molecular events underpinning fibrogenesis and may provide biomarkers for disease progression as well as potential new drug targets.
Collapse
Affiliation(s)
- Agata Page
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4 Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Pier Paoli
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4 Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Eva Moran Salvador
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4 Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Steve White
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4 Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Jeremy French
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4 Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Jelena Mann
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4 Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
13
|
Hui ST, Parks BW, Org E, Norheim F, Che N, Pan C, Castellani LW, Charugundla S, Dirks DL, Psychogios N, Neuhaus I, Gerszten RE, Kirchgessner T, Gargalovic PS, Lusis AJ. The genetic architecture of NAFLD among inbred strains of mice. eLife 2015; 4:e05607. [PMID: 26067236 PMCID: PMC4493743 DOI: 10.7554/elife.05607] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 06/11/2015] [Indexed: 02/06/2023] Open
Abstract
To identify genetic and environmental factors contributing to the pathogenesis of non-alcoholic fatty liver disease, we examined liver steatosis and related clinical and molecular traits in more than 100 unique inbred mouse strains, which were fed a diet rich in fat and carbohydrates. A >30-fold variation in hepatic TG accumulation was observed among the strains. Genome-wide association studies revealed three loci associated with hepatic TG accumulation. Utilizing transcriptomic data from the liver and adipose tissue, we identified several high-confidence candidate genes for hepatic steatosis, including Gde1, a glycerophosphodiester phosphodiesterase not previously implicated in triglyceride metabolism. We confirmed the role of Gde1 by in vivo hepatic over-expression and shRNA knockdown studies. We hypothesize that Gde1 expression increases TG production by contributing to the production of glycerol-3-phosphate. Our multi-level data, including transcript levels, metabolite levels, and gut microbiota composition, provide a framework for understanding genetic and environmental interactions underlying hepatic steatosis.
Collapse
Affiliation(s)
- Simon T Hui
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Brian W Parks
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Elin Org
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nam Che
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Lawrence W Castellani
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Sarada Charugundla
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Darwin L Dirks
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Nikolaos Psychogios
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Isaac Neuhaus
- Department of Computational Genomics, Bristol-Myers Squibb, Princeton, United States
| | - Robert E Gerszten
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Todd Kirchgessner
- Department of Cardiovascular Drug Discovery, Bristol-Myers Squibb, Princeton, United States
| | - Peter S Gargalovic
- Department of Computational Genomics, Bristol-Myers Squibb, Princeton, United States
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
14
|
Identification of eQTLs for hepatic Xbp1s and Socs3 gene expression in mice fed a high-fat, high-caloric diet. G3-GENES GENOMES GENETICS 2015; 5:487-96. [PMID: 25617409 PMCID: PMC4390565 DOI: 10.1534/g3.115.016626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a highly prevalent form of human hepatic disease and feeding mice a high-fat, high-caloric (HFHC) diet is a standard model of NAFLD. To better understand the genetic basis of NAFLD, we conducted an expression quantitative trait locus (eQTL) analysis of mice fed a HFHC diet. Two-hundred sixty-five (A/J × C57BL/6J) F2 male mice were fed a HFHC diet for 8 wk. eQTL analysis was utilized to identify genomic regions that regulate hepatic gene expression of Xbp1s and Socs3. We identified two overlapping loci for Xbp1s and Socs3 on Chr 1 (164.0–185.4 Mb and 174.4–190.5 Mb, respectively) and Chr 11 (41.1–73.1 Mb and 44.0–68.6 Mb, respectively), and an additional locus for Socs3 on Chr 12 (109.9–117.4 Mb). C57BL/6J-Chr 11A/J/ NaJ mice fed a HFHC diet manifested the A/J phenotype of increased Xbp1s and Socs3 gene expression (P < 0.05), whereas C57BL/6J-Chr 1A/J/ NaJ mice retained the C57BL/6J phenotype. In addition, we replicated the eQTLs on Chr 1 and Chr 12 (LOD scores ≥3.5) using mice from the BXD murine reference panel challenged with CCl4 to induce chronic liver injury and fibrosis. We have identified overlapping eQTLs for Xbp1 and Socs3 on Chr 1 and Chr 11, and consomic mice confirmed that replacing the C57BL/6J Chr 11 with the A/J Chr 11 resulted in an A/J phenotype for Xbp1 and Socs3 gene expression. Identification of the genes for these eQTLs will lead to a better understanding of the genetic factors responsible for NAFLD and potentially other hepatic diseases.
Collapse
|
15
|
Asai A, Chou PM, Bu HF, Wang X, Rao MS, Jiang A, DiDonato CJ, Tan XD. Dissociation of hepatic insulin resistance from susceptibility of nonalcoholic fatty liver disease induced by a high-fat and high-carbohydrate diet in mice. Am J Physiol Gastrointest Liver Physiol 2014; 306:G496-504. [PMID: 24436353 PMCID: PMC3949024 DOI: 10.1152/ajpgi.00291.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liver steatosis in nonalcoholic fatty liver disease is affected by genetics and diet. It is associated with insulin resistance (IR) in hepatic and peripheral tissues. Here, we aimed to characterize the severity of diet-induced steatosis, obesity, and IR in two phylogenetically distant mouse strains, C57BL/6J and DBA/2J. To this end, mice (male, 8 wk old) were fed a high-fat and high-carbohydrate (HFHC) or control diet for 16 wk followed by the application of a combination of classic physiological, biochemical, and pathological studies to determine obesity and hepatic steatosis. Peripheral IR was characterized by measuring blood glucose level, serum insulin level, homeostasis model assessment of IR, glucose intolerance, insulin intolerance, and AKT phosphorylation in adipose tissues, whereas the level of hepatic IR was determined by measuring insulin-triggered hepatic AKT phosphorylation. We discovered that both C57BL/6J and DBA/2J mice developed obesity to a similar degree without the feature of liver inflammation after being fed an HFHC diet for 16 wk. C57BL/6J mice in the HFHC diet group exhibited severe pan-lobular steatosis, a marked increase in hepatic triglyceride levels, and profound peripheral IR. In contrast, DBA/2J mice in the HFHC diet group developed only a mild degree of pericentrilobular hepatic steatosis that was associated with moderate changes in peripheral IR. Interestingly, both C57BL/6J and DBA/2J developed severe hepatic IR after HFHC diet treatment. Collectively, these data suggest that the severity of diet-induced hepatic steatosis is correlated to the level of peripheral IR, not with the severity of obesity and hepatic IR. Peripheral rather than hepatic IR is a dominant factor of pathophysiology in nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Akihiro Asai
- 1Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children's Hospital of Chicago Research Center and ,2Departments of Pediatrics and
| | - Pauline M. Chou
- 3Pathology, Feinberg School of Medicine, Northwestern University, Chicago; and
| | - Heng-Fu Bu
- 1Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children's Hospital of Chicago Research Center and ,2Departments of Pediatrics and
| | - Xiao Wang
- 1Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children's Hospital of Chicago Research Center and ,2Departments of Pediatrics and
| | - M. Sambasiva Rao
- 3Pathology, Feinberg School of Medicine, Northwestern University, Chicago; and
| | - Anthony Jiang
- 1Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children's Hospital of Chicago Research Center and
| | | | - Xiao-Di Tan
- 1Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children's Hospital of Chicago Research Center and ,2Departments of Pediatrics and ,3Pathology, Feinberg School of Medicine, Northwestern University, Chicago; and ,4Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
16
|
Sutti S, Jindal A, Locatelli I, Vacchiano M, Gigliotti L, Bozzola C, Albano E. Adaptive immune responses triggered by oxidative stress contribute to hepatic inflammation in NASH. Hepatology 2014; 59:886-897. [PMID: 24115128 DOI: 10.1002/hep.26749] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 09/11/2013] [Indexed: 12/11/2022]
Abstract
UNLABELLED Previous studies have shown that human nonalcoholic steatohepatitis (NASH) is often associated with the presence of circulating antibodies against protein adducted by lipid peroxidation products. Here we used the methionine-choline deficient (MCD) model of NASH to characterize the possible involvement of adaptive immunity in NASH. In mice fed up to 8 weeks with the MCD diet the extension of liver injury and lobular inflammation paralleled the development of immunoglobulin G (IgG) against malonyldialdehyde (MDA) and 4-hydroxynonenal (4-HNE)-derived antigens as well as with the hepatic recruitment of CD4(+) and CD8(+) T-lymphocytes responsive to the same antigens. Moreover, in these animals the individual IgG reactivity against MDA-adducts positively correlated with transaminase release and hepatic tumor necrosis factor alpha (TNF-α) expression. To substantiate the role of immune responses triggered by oxidative stress in the progression of NASH, mice were immunized with MDA-adducted bovine serum albumin (MDA-BSA) before feeding the MCD diet. MDA-BSA immunization did not affect control mice livers, but further stimulated transaminase release, lobular inflammation, and the hepatic expression of proinflammatory cytokine in MCD-fed mice. The increased severity of NASH in immunized MCD-fed mice involved liver recruitment and the T helper (Th)-1 activation of CD4(+) T cells that, in turn, further stimulated macrophage M1 responses. Moreover, hepatic fibrosis was also evident in these animals in relation with an IL-15-mediated increase of natural killer T-cells (NKT) and the up-regulation in liver production of osteopontin by NKT cells and hepatic macrophages. CONCLUSION These results indicate that oxidative stress can contribute to the progression of NASH by stimulating both humoral and cellular immune responses, pointing to the possible role of adaptive immunity in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University "Amedeo Avogadro" of East Piedmont, Novara, Italy
| | | | | | | | | | | | | |
Collapse
|
17
|
Liedtke C, Luedde T, Sauerbruch T, Scholten D, Streetz K, Tacke F, Tolba R, Trautwein C, Trebicka J, Weiskirchen R. Experimental liver fibrosis research: update on animal models, legal issues and translational aspects. FIBROGENESIS & TISSUE REPAIR 2013; 6:19. [PMID: 24274743 PMCID: PMC3850878 DOI: 10.1186/1755-1536-6-19] [Citation(s) in RCA: 251] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 09/11/2013] [Indexed: 12/13/2022]
Abstract
Liver fibrosis is defined as excessive extracellular matrix deposition and is based on complex interactions between matrix-producing hepatic stellate cells and an abundance of liver-resident and infiltrating cells. Investigation of these processes requires in vitro and in vivo experimental work in animals. However, the use of animals in translational research will be increasingly challenged, at least in countries of the European Union, because of the adoption of new animal welfare rules in 2013. These rules will create an urgent need for optimized standard operating procedures regarding animal experimentation and improved international communication in the liver fibrosis community. This review gives an update on current animal models, techniques and underlying pathomechanisms with the aim of fostering a critical discussion of the limitations and potential of up-to-date animal experimentation. We discuss potential complications in experimental liver fibrosis and provide examples of how the findings of studies in which these models are used can be translated to human disease and therapy. In this review, we want to motivate the international community to design more standardized animal models which might help to address the legally requested replacement, refinement and reduction of animals in fibrosis research.
Collapse
Affiliation(s)
- Christian Liedtke
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Tom Luedde
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Tilman Sauerbruch
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - David Scholten
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Konrad Streetz
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - René Tolba
- Institute of Laboratory Animal Science, RWTH University Hospital Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Ralf Weiskirchen
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH University Hospital Aachen, Aachen D-52074, Germany
| |
Collapse
|
18
|
Walkin L, Herrick SE, Summers A, Brenchley PE, Hoff CM, Korstanje R, Margetts PJ. The role of mouse strain differences in the susceptibility to fibrosis: a systematic review. FIBROGENESIS & TISSUE REPAIR 2013; 6:18. [PMID: 24294831 PMCID: PMC3849643 DOI: 10.1186/1755-1536-6-18] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 09/09/2013] [Indexed: 12/21/2022]
Abstract
In humans, a number of genetic factors have been linked to the development of fibrosis in a variety of different organs. Seeking a wider understanding of this observation in man is ethically important. There is mounting evidence suggesting that inbred mouse strains with different genetic backgrounds demonstrate variable susceptibility to a fibrotic injury. We performed a systematic review of the literature describing strain and organ specific response to injury in order to determine whether genetic susceptibility plays a role in fibrogenesis. Data were collected from studies that were deemed eligible for analysis based on set inclusion criteria, and findings were assessed in relation to strain of mouse, type of injury and organ of investigation. A total of 44 studies were included covering 21 mouse strains and focusing on fibrosis in the lung, liver, kidney, intestine and heart. There is evidence that mouse strain differences influence susceptibility to fibrosis and this appears to be organ specific. For instance, C57BL/6J mice are resistant to hepatic, renal and cardiac fibrosis but susceptible to pulmonary and intestinal fibrosis. However, BALB/c mice are resistant to pulmonary fibrosis but susceptible to hepatic fibrosis. Few studies have assessed the effect of the same injury stimulus in different organ systems using the same strains of mouse. Such mouse strain studies may prove useful in elucidating the genetic as well as epigenetic factors in humans that could help determine why some people are more susceptible to the development of certain organ specific fibrosis than others.
Collapse
Affiliation(s)
- Louise Walkin
- School of Medicine, Faculty of Medical and Human Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- 3.107 Blond McIndoe Laboratory, Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | - Sarah E Herrick
- School of Medicine, Faculty of Medical and Human Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Angela Summers
- Manchester Institute of Nephrology and Transplantation, Manchester Royal Infirmary, Grafton St, Manchester M13 9WL, UK
| | - Paul E Brenchley
- Manchester Institute of Nephrology and Transplantation, Manchester Royal Infirmary, Grafton St, Manchester M13 9WL, UK
| | - Catherine M Hoff
- Baxter Healthcare, Renal Division Scientific Affairs, Baxter Healthcare Corporation, McGaw Park, Chicago, Illinois, 60015-4625, USA
| | - Ron Korstanje
- The Jackson Laboratory, 600 Main St, Bar Harbor, Maine 04609, USA
| | - Peter J Margetts
- Department of Pathology and Molecular Medicine and Division of Nephrology, McMaster University, 1200 Main St West, Hamilton, Ontario, L8S4L8, Canada
| |
Collapse
|
19
|
In vitro and in vivo models of non-alcoholic fatty liver disease (NAFLD). Int J Mol Sci 2013; 14:11963-80. [PMID: 23739675 PMCID: PMC3709766 DOI: 10.3390/ijms140611963] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/17/2013] [Accepted: 05/22/2013] [Indexed: 12/28/2022] Open
Abstract
By now, non-alcoholic fatty liver disease (NAFLD) is considered to be among the most common liver diseases world-wide. NAFLD encompasses a broad spectrum of pathological conditions ranging from simple steatosis to steatohepatitis, fibrosis and finally even cirrhosis; however, only a minority of patients progress to end-stages of the disease, and the course of the disease progression to the later stages seems to be slow, developing progressively over several years. Key risk factors including overweight, insulin resistance, a sedentary life-style and an altered dietary pattern, as well as genetic factors and disturbances of the intestinal barrier function have been identified in recent years. Despite intense research efforts that lead to the identification of these risk factors, knowledge about disease initiation and molecular mechanisms involved in progression is still limited. This review summarizes diet-induced and genetic animal models, as well as cell culture models commonly used in recent years to add to the understanding of the mechanisms involved in NAFLD, also referring to their advantages and disadvantages.
Collapse
|
20
|
Locatelli I, Sutti S, Vacchiano M, Bozzola C, Albano E. NF-κB1 deficiency stimulates the progression of non-alcoholic steatohepatitis (NASH) in mice by promoting NKT-cell-mediated responses. Clin Sci (Lond) 2013; 124:279-287. [PMID: 22970906 DOI: 10.1042/cs20120289] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Growing evidence indicates that NF-κB (nuclear factor κB) activation contributes to the pathogenesis of NASH (non-alcoholic steatohepatisis). Among the NF-κB subunits, p50/NF-κB1 has regulatory activities down-modulating NF-κB-mediated responses. In the present study, we investigated the effects of NF-κB1 deficiency on the progression of NASH induced by feeding mice on an MCD (methionine/choline-deficient) diet. Following 4 weeks on the MCD diet, steatosis, ALT (alanine aminotransferase) release, hepatocyte apoptosis, lobular inflammation and TNFα (tumour necrosis factor α) production were higher in NF-κB1(-/-) (NF-κB1-knockout) mice than in WT (wild-type) mice. NF-κB1(-/-) mice also showed appreciable centrilobular collagen deposition, an increased number of activated hepatic stellate cells and higher type-I procollagen-α and TIMP-1 (tissue inhibitor of metalloproteases-1) mRNA expression. Although NF-κB p50 homodimers regulate macrophage activation, the number of hepatic macrophages and liver mRNAs for iNOS (inducible NO synthase), IL (interleukin)-12p40, CCL2 (CC chemokine ligand 2) and CXCL10 (CXC chemokine ligand 10) were comparable in the two strains. NASH was associated with an increase in liver infiltrating T-cells that was more evident in MCD-fed NF-κB1(-/-) than in similarly treated WT mice. Flow cytorimetry showed that T-cell recruitment involved effector CD8+ T-cells without changes in the helper CD4+ T-cell fraction. Furthermore, although NASH lowered hepatic NKT cells [NK (natural killer) T-cells] in WT mice, the NKT cell pool was selectively increased in the livers of MCD-fed NF-κB1(-/-) mice. Such NKT cell recruitment was associated with an early overexpression of IL-15, a cytokine controlling NKT cell survival and maturation. In the livers of MCD-fed NF-κB1(-/-) mice, but not in those of WT littermates, we also observed an up-regulation in the production of NKT-related cytokines IFN (interferon)-γ and osteopontin. Taken together, these results indicate that NF-κB1 down-modulation enhanced NASH progression to fibrosis by favouring NKT cell recruitment, stressing the contribution of NKT cells in the pathogenesis of NASH.
Collapse
Affiliation(s)
- Irene Locatelli
- Department of Health Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University 'Amedeo Avogadro' of East Piedmont, Novara, Italy
| | | | | | | | | |
Collapse
|
21
|
Matsumoto M, Hada N, Sakamaki Y, Uno A, Shiga T, Tanaka C, Ito T, Katsume A, Sudoh M. An improved mouse model that rapidly develops fibrosis in non-alcoholic steatohepatitis. Int J Exp Pathol 2013; 94:93-103. [PMID: 23305254 PMCID: PMC3607137 DOI: 10.1111/iep.12008] [Citation(s) in RCA: 366] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Accepted: 10/29/2012] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a progressive fibrotic disease, the pathogenesis of which has not been fully elucidated. One of the most common models used in NASH research is a nutritional model where NASH is induced by feeding a diet deficient in both methionine and choline. However, the dietary methionine-/choline-deficient model in mice can cause severe weight loss and liver atrophy, which are not characteristics of NASH seen in human patients. Exclusive, long-term feeding with a high-fat diet (HFD) produced fatty liver and obesity in mice, but the HFD for several months did not affect fibrosis. We aimed to establish a mouse model of NASH with fibrosis by optimizing the methionine content in the HFD. Male mice were fed a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) consisting of 60 kcal% fat and 0.1% methionine by weight. After 1–14 weeks of being fed CDAHFD, the mice were killed. C57BL/6J mice maintained or gained weight when fed CDAHFD, while A/J mice showed a steady decline in body weight (of up to 20% of initial weight). In both strains of mice, plasma levels of alanine aminotransferase increased from week 1, when hepatic steatosis was also observed. By week 6, C57BL/6J mice had developed enlarged fatty liver with fibrosis as assessed by Masson's trichrome staining and by hydroxyproline assay. Therefore, this improved CDAHFD model may be a mouse model of rapidly progressive liver fibrosis and be potentially useful for better understanding human NASH disease and in the development of efficient therapies for this condition.
Collapse
Affiliation(s)
- Masahiko Matsumoto
- Fuji-Gotemba Research Laboratories, Chugai Research Institute for Medical Science Inc, Gotemba, Shizuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Quantitative trait loci affecting liver fat content in mice. G3-GENES GENOMES GENETICS 2012; 2:1019-25. [PMID: 22973538 PMCID: PMC3429915 DOI: 10.1534/g3.112.003343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 06/20/2012] [Indexed: 01/13/2023]
Abstract
Nonalcoholic fatty liver disease, a condition in which excess fat accumulates in the liver, is strongly associated with the metabolic syndrome, including obesity and other related conditions. This disease has the potential to progress from steatosis to steatohepatitis, fibrosis, and cirrhosis. The recent increase in the prevalence of the metabolic syndrome is largely driven by changes in diet and activity levels. Individual variation in the response to this obesogenic environment, however, is attributable in part to genetic variation between individuals, but very few mammalian genetic loci have been identified with effects on fat accumulation in the liver. To study the genetic basis for variation in liver fat content in response to dietary fat, liver fat proportion was determined using quantitative magnetic resonance imaging in 478 mice from 16 LG/J X SM/J recombinant inbred strains fed either a high-fat (42% kcal from fat) or low-fat (15% kcal from fat) diet. An analysis of variance confirmed that there is a genetic basis for variation in liver fat content within the population with significant effects of sex and diet. Three quantitative trail loci that contribute to liver fat content also were mapped.
Collapse
|
23
|
Maina V, Sutti S, Locatelli I, Vidali M, Mombello C, Bozzola C, Albano E. Bias in macrophage activation pattern influences non-alcoholic steatohepatitis (NASH) in mice. Clin Sci (Lond) 2012; 122:545-553. [PMID: 22142284 DOI: 10.1042/cs20110366] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In humans, there is large inter-individual variability in the evolution of NAFLD (non-alcoholic fatty liver disease) to NASH (non-alcoholic steatohepatitis). To investigate this issue, NASH was induced with an MCD (methionine-choline-deficient) diet in C57BL/6 and Balb/c mice that are characterized by different biases in Th1/Th2 and macrophage (M1/M2) responses. Following 4 weeks on the MCD diet, steatosis and lobular inflammation were prevalent in C57BL/6 (Th1, M1 oriented) than in Balb/c (Th2, M2 oriented) mice. Consistently, hepatic TNFα (tumour necrosis factor α) mRNA expression and circulating TNFα levels were higher in MCD-fed C57BL/6 than in MCD-fed Balb/c mice. The Th1/Th2 bias did not account for the increased NASH severity, as in both strains MCD feeding did not significantly modify the liver mRNA expression of the Th1 markers IFNγ (interferon γ) and T-bet or that of the Th2 markers IL-4 (interleukin 4) and GATA-3. Conversely, MCD-fed C57BL/6 mice displayed higher liver mRNAs for the macrophage M1 activation markers iNOS (inducible NO synthase), IL-12p40 and CXCL10 (CXC chemokine ligand 10) than similarly treated Balb/c mice, without effects on the M2 polarization markers IL-10 and MGL-1 (macrophage galactose-type C-type lectin-1). Circulating IL-12 was also higher in MCD-fed C57BL/6 than in MCD-fed Balb/c mice. The analysis of macrophages isolated from the livers of MCD-fed animals confirmed an enhanced expression of M1 markers in C57BL/6 mice. Among all of the MCD-treated mice, liver iNOS, IL-12p40 and CXCL10 mRNA levels positively correlated with the frequency of hepatic necro-inflammatory foci. We concluded that the macrophage M1 bias in C57BL/6 mice may account for the increased severity of NASH in this strain, suggesting macrophage responses as important contributors to NAFLD progression.
Collapse
Affiliation(s)
- Virginia Maina
- Department of Medical Sciences and Interdisciplinary Research Centre for Autoimmune Diseases, University Amedeo Avogadro of East Piedmont, Via Solaroli 17, 28100 Novara, Italy
| | | | | | | | | | | | | |
Collapse
|
24
|
Haplotype Association Mapping Identifies a Candidate Gene Region in Mice Infected With Staphylococcus aureus. G3-GENES GENOMES GENETICS 2012; 2:693-700. [PMID: 22690378 PMCID: PMC3362298 DOI: 10.1534/g3.112.002501] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 04/11/2012] [Indexed: 12/15/2022]
Abstract
Exposure to Staphylococcus aureus has a variety of outcomes, from asymptomatic colonization to fatal infection. Strong evidence suggests that host genetics play an important role in susceptibility, but the specific host genetic factors involved are not known. The availability of genome-wide single nucleotide polymorphism (SNP) data for inbred Mus musculus strains means that haplotype association mapping can be used to identify candidate susceptibility genes. We applied haplotype association mapping to Perlegen SNP data and kidney bacterial counts from Staphylococcus aureus-infected mice from 13 inbred strains and detected an associated block on chromosome 7. Strong experimental evidence supports the result: a separate study demonstrated the presence of a susceptibility locus on chromosome 7 using consomic mice. The associated block contains no genes, but lies within the gene cluster of the 26-member extended kallikrein gene family, whose members have well-recognized roles in the generation of antimicrobial peptides and the regulation of inflammation. Efficient mixed-model association (EMMA) testing of all SNPs with two alleles and located within the gene cluster boundaries finds two significant associations: one of the three polymorphisms defining the associated block and one in the gene closest to the block, Klk1b11. In addition, we find that 7 of the 26 kallikrein genes are differentially expressed between susceptible and resistant mice, including the Klk1b11 gene. These genes represent a promising set of candidate genes influencing susceptibility to Staphylococcus aureus.
Collapse
|
25
|
Charlton M, Krishnan A, Viker K, Sanderson S, Cazanave S, McConico A, Masuoko H, Gores G. Fast food diet mouse: novel small animal model of NASH with ballooning, progressive fibrosis, and high physiological fidelity to the human condition. Am J Physiol Gastrointest Liver Physiol 2011; 301:G825-34. [PMID: 21836057 PMCID: PMC3220319 DOI: 10.1152/ajpgi.00145.2011] [Citation(s) in RCA: 346] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Although there are small animal platforms that recapitulate some of the histological features of nonalcoholic fatty liver disease, there are no small animal models of nonalcoholic steatohepatitis (NASH) with consistent hepatocellular ballooning and progressive fibrosis that also exhibit fidelity to the human condition physiologically. We examined the metabolic and histological effects of a diet on the basis of the composition of "fast food" (high saturated fats, cholesterol, and fructose). Mice (n = 8 in each group) were assigned to diets as follows: 1) standard chow (SC), i.e., 13% energy as fat [1% saturated fatty acids (SFA)], 2) high fat (HF), i.e., 60% energy as fat (1% SFA), and 3) fast food (FF), i.e., 40% energy as fat (12% SFA, 2% cholesterol). All three diets were supplemented with high fructose. All diets produced obesity. The HF and FF diets produced insulin resistance. Liver histology was normal in animals fed the SC diet. Steatohepatitis with pronounced ballooning and progressive fibrosis (stage 2) was observed in mice fed the FF diet. Although the HF diet produced obesity, insulin resistance, and some steatosis; inflammation was minimal, and there was no increase in fibrosis. The FF diet produced a gene expression signature of increased fibrosis, inflammation, and endoplasmic reticulum stress and lipoapoptosis. A diet based on high cholesterol, high saturated fat, and high fructose recapitulates features of the metabolic syndrome and NASH with progressive fibrosis. This represents a novel small animal model of fibrosing NASH with high fidelity to the human condition. These results highlight the contribution of dietary composition to the development of nonalcoholic fatty liver disease and NASH.
Collapse
Affiliation(s)
- Michael Charlton
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Rodent models of fatty liver disease are essential research tools that provide a window into disease pathogenesis and a testing ground for prevention and treatment. Models come in many varieties involving dietary and genetic manipulations, and sometimes both. High-energy diets that induce obesity do not uniformly cause fatty liver disease; this has prompted close scrutiny of specific macronutrients and nutrient combinations to determine which have the greatest potential for hepatotoxicity. At the same time, diets that do not cause obesity or the metabolic syndrome but do cause severe steatohepatitis have been exploited to study factors important to progressive liver injury, including cell death, oxidative stress, and immune activation. Rodents with a genetic predisposition to overeating offer yet another model in which to explore the evolution of fatty liver disease. In some animals that overeat, steatohepatitis can develop even without resorting to a high-energy diet. Importantly, these models and others have been used to document that aerobic exercise can prevent or reduce fatty liver disease. This review focuses primarily on lessons learned about steatohepatitis from manipulations of diet and eating behavior. Numerous additional insights about hepatic lipid metabolism, which have been gained from genetically engineered mice, are also mentioned.
Collapse
Affiliation(s)
- Jacquelyn J Maher
- Liver Center and Department of Medicine, University of California, San Francisco San Francisco, California, USA.
| |
Collapse
|
27
|
Berres ML, Koenen RR, Rueland A, Zaldivar MM, Heinrichs D, Sahin H, Schmitz P, Streetz KL, Berg T, Gassler N, Weiskirchen R, Proudfoot A, Weber C, Trautwein C, Wasmuth HE. Antagonism of the chemokine Ccl5 ameliorates experimental liver fibrosis in mice. J Clin Invest 2010; 120:4129-4140. [PMID: 20978355 PMCID: PMC2964968 DOI: 10.1172/jci41732] [Citation(s) in RCA: 216] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 09/01/2010] [Indexed: 12/13/2022] Open
Abstract
Activation of hepatic stellate cells in response to chronic inflammation represents a crucial step in the development of liver fibrosis. However, the molecules involved in the interaction between immune cells and stellate cells remain obscure. Herein, we identify the chemokine CCL5 (also known as RANTES), which is induced in murine and human liver after injury, as a central mediator of this interaction. First, we showed in patients with liver fibrosis that CCL5 haplotypes and intrahepatic CCL5 mRNA expression were associated with severe liver fibrosis. Consistent with this, we detected Ccl5 mRNA and CCL5 protein in 2 mouse models of liver fibrosis, induced by either injection of carbon tetrachloride (CCl4) or feeding on a methionine and choline-deficient (MCD) diet. In these models, Ccl5-/- mice exhibited decreased hepatic fibrosis, with reduced stellate cell activation and immune cell infiltration. Transplantation of Ccl5-deficient bone marrow into WT recipients attenuated liver fibrosis, identifying infiltrating hematopoietic cells as the main source of Ccl5. We then showed that treatment with the CCL5 receptor antagonist Met-CCL5 inhibited cultured stellate cell migration, proliferation, and chemokine and collagen secretion. Importantly, in vivo administration of Met-CCL5 greatly ameliorated liver fibrosis in mice and was able to accelerate fibrosis regression. Our results define a successful therapeutic approach to reduce experimental liver fibrosis by antagonizing Ccl5 receptors.
Collapse
Affiliation(s)
- Marie-Luise Berres
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Rory R. Koenen
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Anna Rueland
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Mirko Moreno Zaldivar
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Daniel Heinrichs
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Hacer Sahin
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Petra Schmitz
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Konrad L. Streetz
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Thomas Berg
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Nikolaus Gassler
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Ralf Weiskirchen
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Amanda Proudfoot
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Christian Weber
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Christian Trautwein
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Hermann E. Wasmuth
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| |
Collapse
|
28
|
Soon RK, Yan JS, Grenert JP, Maher JJ. Stress signaling in the methionine-choline-deficient model of murine fatty liver disease. Gastroenterology 2010; 139:1730-9, 1739.e1. [PMID: 20682321 PMCID: PMC2967598 DOI: 10.1053/j.gastro.2010.07.046] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 06/25/2010] [Accepted: 07/22/2010] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Stress signaling, both within and outside the endoplasmic reticulum, has been linked to metabolic dysregulation and hepatic steatosis. Methionine-choline-deficient (MCD) diets cause severe fatty liver disease and have the potential to cause many types of cellular stress. The purpose of this study was to characterize hepatic stress in MCD-fed mice and explore the relationship between MCD-mediated stress and liver injury. METHODS Stress signaling was examined in mice fed MCD formulas for 4-21 days. Signaling also was evaluated in mice fed MCD formulas supplemented with clofibrate, which inhibits hepatic triglyceride accumulation. The role of the pro-apoptotic stress protein C/EBP homologous protein (CHOP) in MCD-mediated liver injury was assessed by comparing the responses of wild-type and CHOP-deficient mice to an MCD diet. RESULTS MCD feeding caused steatohepatitis coincident with the activation of cJun N-terminal kinase and caspase-12. In contrast, MCD feeding did not activate inositol-requiring protein-1 and actually suppressed the expression of X-box protein-1s. MCD feeding caused weak stimulation of double-stranded RNA-activated protein kinase-like endoplasmic reticulum-resident kinase, but robust activation of general control nonderepressible-2, followed by the phosphorylation of eukaryotic initiating factor-2α and induction of CHOP. Clofibrate eliminated MCD-mediated hepatic steatosis but did not inhibit diet-induced stress. CHOP deficiency did not alleviate, and in fact worsened, MCD-mediated liver disease. CONCLUSIONS MCD feeding causes an integrated stress response in the liver rather than a classic unfolded protein response. This stress response does not by itself lead to liver injury. CHOP, despite its identity as a mediator of stress-related cell death, does not play a central role in the pathogenesis of MCD-mediated liver disease.
Collapse
Affiliation(s)
- Russell K. Soon
- Department of Medicine, University of California, San Francisco, 94110, The Liver Center, University of California, San Francisco, 94110
| | - Jim S. Yan
- Department of Medicine, University of California, San Francisco, 94110, The Liver Center, University of California, San Francisco, 94110
| | - James P. Grenert
- Department of Pathology, University of California, San Francisco, 94110, The Liver Center, University of California, San Francisco, 94110
| | - Jacquelyn J. Maher
- Department of Medicine, University of California, San Francisco, 94110, The Liver Center, University of California, San Francisco, 94110
| |
Collapse
|
29
|
Burgess-Herbert SL, Tsaih SW, Stylianou IM, Walsh K, Cox AJ, Paigen B. An experimental assessment of in silico haplotype association mapping in laboratory mice. BMC Genet 2009; 10:81. [PMID: 20003225 PMCID: PMC2797012 DOI: 10.1186/1471-2156-10-81] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Accepted: 12/09/2009] [Indexed: 11/10/2022] Open
Abstract
Background To assess the utility of haplotype association mapping (HAM) as a quantitative trait locus (QTL) discovery tool, we conducted HAM analyses for red blood cell count (RBC) and high density lipoprotein cholesterol (HDL) in mice. We then experimentally tested each HAM QTL using published crosses or new F2 intercrosses guided by the haplotype at the HAM peaks. Results The HAM for RBC, using 33 classic inbred lines, revealed 8 QTLs; 2 of these were true positives as shown by published crosses. A HAM-guided (C57BL/6J × CBA/J)F2 intercross we carried out verified 2 more as true positives and 4 as false positives. The HAM for HDL, using 81 strains including recombinant inbred lines and chromosome substitution strains, detected 46 QTLs. Of these, 36 were true positives as shown by published crosses. A HAM-guided (C57BL/6J × A/J)F2 intercross that we carried out verified 2 more as true positives and 8 as false positives. By testing each HAM QTL for RBC and HDL, we demonstrated that 78% of the 54 HAM peaks were true positives and 22% were false positives. Interestingly, all false positives were in significant allelic association with one or more real QTL. Conclusion Because type I errors (false positives) can be detected experimentally, we conclude that HAM is useful for QTL detection and narrowing. We advocate the powerful and economical combined approach demonstrated here: the use of HAM for QTL discovery, followed by mitigation of the false positive problem by testing the HAM-predicted QTLs with small HAM-guided experimental crosses.
Collapse
|
30
|
Yamazaki Y, Kakizaki S, Takizawa D, Ichikawa T, Sato K, Takagi H, Mori M. Interstrain differences in susceptibility to non-alcoholic steatohepatitis. J Gastroenterol Hepatol 2008; 23:276-282. [PMID: 17868334 DOI: 10.1111/j.1440-1746.2007.05150.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM The pathophysiological mechanisms leading to the development of non-alcoholic steatohepatitis (NASH) remain unclear. There are differences in the susceptibility to NASH between the different species and sexes. The investigation of the precise mechanism of interstrain differences may provide new means by which the pathophysiological mechanisms of NASH may be understood. METHODS C57BL/6N and C3H/HeN mice were administered a methionine- and choline-deficient (MCD) diet to establish a dietary model of NASH. RESULTS An elevation of the serum alanine aminotransferase and increased infiltration of inflammatory cells were predominant in C57BL/6N mice at 8 weeks. The increase in the steatosis and lipid contents in the liver was greater in C57BL/6N mice than in C3H/HeN mice. The indices of lipid peroxidation demonstrated by F2-isoprostanes or 8-hydroxy-2'-deoxyguanosine also increased in the livers of C57BL/6N mice. Furthermore, Sirius red staining revealed an increase in the degree of fibrosis in C57BL/6N mice given the MCD diet. As a result, the C57BL/6N strain had a higher susceptibility to NASH than the C3H/HeN mice. The carnitine palmitoyltransferase 1A (in beta-oxidation) mRNA and mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (in ketogenesis) mRNA were downregulated in the C57BL/6N mice in comparison with C3H/HeN mice. There were no differences in the expression of microsomal triglyceride transfer protein or sterol regulatory element binding protein 1 between the C57BL/6N and C3H/HeN mice. CONCLUSION There were interstrain differences in susceptibility to NASH observed in a rodent dietary model. Further evaluations of the precise molecular mechanism of this interstrain difference may provide some indications of the pathophysiological mechanisms of NASH in humans.
Collapse
Affiliation(s)
- Yuichi Yamazaki
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | | | | | | | | | | | | |
Collapse
|
31
|
|