151
|
Yu Y, Cai J, She Z, Li H. Insights into the Epidemiology, Pathogenesis, and Therapeutics of Nonalcoholic Fatty Liver Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801585. [PMID: 30828530 PMCID: PMC6382298 DOI: 10.1002/advs.201801585] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/14/2018] [Indexed: 05/05/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease which affects ≈25% of the adult population worldwide, placing a tremendous burden on human health. The disease spectrum ranges from simple steatosis to steatohepatitis, fibrosis, and ultimately, cirrhosis and carcinoma, which are becoming leading reasons for liver transplantation. NAFLD is a complex multifactorial disease involving myriad genetic, metabolic, and environmental factors; it is closely associated with insulin resistance, metabolic syndrome, obesity, diabetes, and many other diseases. Over the past few decades, countless studies focusing on the investigation of noninvasive diagnosis, pathogenesis, and therapeutics have revealed different aspects of the mechanism and progression of NAFLD. However, effective pharmaceuticals are still in development. Here, the current epidemiology, diagnosis, animal models, pathogenesis, and treatment strategies for NAFLD are comprehensively reviewed, emphasizing the outstanding breakthroughs in the above fields and promising medications in and beyond phase II.
Collapse
Affiliation(s)
- Yao Yu
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| | - Jingjing Cai
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| | - Zhigang She
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| | - Hongliang Li
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| |
Collapse
|
152
|
Chen B, Ma Y, Xue X, Wei J, Hu G, Lin Y. Tetramethylpyrazine reduces inflammation in the livers of mice fed a high fat diet. Mol Med Rep 2019; 19:2561-2568. [PMID: 30720104 PMCID: PMC6423567 DOI: 10.3892/mmr.2019.9928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to assess the protective effects of tetramethylpyrazine (TMP) on the livers of mice fed a high fat diet. The mice were divided into five groups: Regular diet; high fat diet; simvastatin‑treated; and low and high dose TMP‑treated groups. The results demonstrated that, compared with the control group, serum glucose, total cholesterol (TC) and low‑density lipoprotein cholesterol levels were increased in the model group. Additionally, compared with the model group, simvastatin lowered the TC level, whereas TMP did not. Compared with the control group, the level of malondialdehyde (MDA) in the liver tissue was increased and the level of glutathione peroxidase (GSH‑pX) in the liver tissue was decreased in the model group. Furthermore, compared with the model group, TMP decreased the level of MDA and increased the level of GSH‑Px; however, simvastatin did not have these effects. Immunohistochemistry and western blotting were performed; the results showed that, compared with the control group, the levels of inflammatory factors (tumor necrosis factor‑α and interleukin‑6) in the liver tissue were increased, and the ratio of phosphorylated (p)‑nuclear factor κB (NF‑κB)/NF‑κB was also increased in the model group. The addition of TMP and simvastatin demonstrated that, compared with the model group, the inflammatory factor levels and the ratio of p‑NF‑κB/NF‑κB were decreased. In addition, liver lipid deposition was examined in the model group using hematoxylin and eosin staining and Oil Red O staining, and the results showed that TMP and simvastatin reduced liver lipid deposition. Furthermore, compared with the control group, the reactive oxygen species (ROS) level in the liver tissue was increased. Compared with that in the model group, TMP and simvastatin decreased the ROS level. In conclusion, TMP, similar to simvastatin, exerted a notable hepatoprotective effect on mice fed a high fat diet with non‑alcoholic fatty liver disease, by inhibiting inflammatory factors and the p‑NF‑κB/ROS signaling pathway.
Collapse
Affiliation(s)
- Bing Chen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Yaluan Ma
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Xin Xue
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Jie Wei
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| | - Gang Hu
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| | - Yajun Lin
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| |
Collapse
|
153
|
Nakashima A, Sugimoto R, Suzuki K, Shirakata Y, Hashiguchi T, Yoshida C, Nakano Y. Anti-fibrotic activity of Euglena gracilis and paramylon in a mouse model of non-alcoholic steatohepatitis. Food Sci Nutr 2019; 7:139-147. [PMID: 30680167 PMCID: PMC6341149 DOI: 10.1002/fsn3.828] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 12/18/2022] Open
Abstract
Progression to non-alcoholic steatohepatitis (NASH) manifests as hepatitis, fibrosis, and sometimes carcinoma, resulting in liver failure. Various clinical trials have indicated that several pharmacological agents, including angiotensin II receptor blockers (ARBs) or farnesoid X receptor (FXR) agonists, are effective in NASH treatment. In addition, functional foods are expected to be important alternatives for treating or preventing NASH. Recently, focus has been directed toward microalgae as dietary supplements, mainly for lifestyle-related diseases, because they contain various nutrients and functional ingredients. Specifically, a unicellular microalga Euglena gracilis stores a unique β-1,3-glucan particle called paramylon that stimulates the immune system. In this study, we evaluated the effects of Euglena and paramylon on NASH in Stelic Animal Model (STAM) mice using Sirius red staining and confirmed that oral administration of Euglena or paramylon inhibits the process of liver fibrosis. Moreover, compared with controls, paramylon decreased non-alcoholic fatty liver disease (NAFLD) activity scores related to inflammation. These results indicate that the oral administration of Euglena and paramylon inhibits fibrosis and ameliorates NASH.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yoshihisa Nakano
- Center for Research and Development of BioresourcesOsaka Prefecture UniversityOsakaJapan
| |
Collapse
|
154
|
Nonalcoholic Fatty Liver Disease: Basic Pathogenetic Mechanisms in the Progression From NAFLD to NASH. Transplantation 2019; 103:e1-e13. [DOI: 10.1097/tp.0000000000002480] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
155
|
Tang A, Destrempes F, Kazemirad S, Garcia-Duitama J, Nguyen BN, Cloutier G. Quantitative ultrasound and machine learning for assessment of steatohepatitis in a rat model. Eur Radiol 2018; 29:2175-2184. [DOI: 10.1007/s00330-018-5915-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/29/2018] [Accepted: 11/23/2018] [Indexed: 12/13/2022]
|
156
|
Hou C, Feng W, Wei S, Wang Y, Xu X, Wei J, Ma Z, Du Y, Guo J, He Y, Kong F, Tang R, Zheng K. Bioinformatics Analysis of Key Differentially Expressed Genes in Nonalcoholic Fatty Liver Disease Mice Models. Gene Expr 2018; 19:25-35. [PMID: 30135001 PMCID: PMC6290321 DOI: 10.3727/105221618x15341831737687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global health problem characterized by excessive accumulation of fat in the liver without effect of other pathological factors including hepatitis infection and alcohol abuse. Current studies indicate that gene factors play important roles in the development of NAFLD. However, the molecular characteristics of differentially expressed genes (DEGs) and associated mechanisms with NAFLD have not been well elucidated. Using two microarray data associated with the gene expression profiling in liver tissues of NAFLD mice models, we identified and selected several common key DEGs that contributed to NAFLD. Based on bioinformatics analysis, we discovered that the DEGs were associated with a variety of biological processes, cellular components, and molecular functions and were also related to several significant pathways. Via pathway crosstalk analysis based on overlapping DEGs, we observed that the identified pathways could form large and complex crosstalk networks. Besides, large and complex protein interaction networks of DEGs were further constructed. In addition, many hub host factors with a high degree of connectivity were identified based on interaction networks. Furthermore, significant modules in interaction networks were found, and the DEGs in the identified modules were found to be enriched with distinct pathways. Taken together, these results suggest that the key DEGs, associated pathways, and modules contribute to the development of NAFLD and might be used as novel molecular targets for the treatment of NAFLD.
Collapse
Affiliation(s)
- Chao Hou
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Wenwen Feng
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Shan Wei
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Yulin Wang
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Xiaoyi Xu
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Jin Wei
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Ziliang Ma
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Yongsheng Du
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Jialin Guo
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Yu He
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- †Department of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Fanyun Kong
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- ‡National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Renxian Tang
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- ‡National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Kuiyang Zheng
- *Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
- ‡National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| |
Collapse
|
157
|
Zhu C, Kim K, Wang X, Bartolome A, Salomao M, Dongiovanni P, Meroni M, Graham MJ, Yates KP, Diehl AM, Schwabe RF, Tabas I, Valenti L, Lavine JE, Pajvani UB. Hepatocyte Notch activation induces liver fibrosis in nonalcoholic steatohepatitis. Sci Transl Med 2018; 10:10/468/eaat0344. [PMID: 30463916 PMCID: PMC6822168 DOI: 10.1126/scitranslmed.aat0344] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/03/2018] [Accepted: 10/31/2018] [Indexed: 01/07/2023]
Abstract
Fibrosis is the major determinant of morbidity and mortality in patients with nonalcoholic steatohepatitis (NASH) but has no approved pharmacotherapy in part because of incomplete understanding of its pathogenic mechanisms. Here, we report that hepatocyte Notch activity tracks with disease severity and treatment response in patients with NASH and is similarly increased in a mouse model of diet-induced NASH and liver fibrosis. Hepatocyte-specific Notch loss-of-function mouse models showed attenuated NASH-associated liver fibrosis, demonstrating causality to obesity-induced liver pathology. Conversely, forced activation of hepatocyte Notch induced fibrosis in both chow- and NASH diet-fed mice by increasing Sox9-dependent Osteopontin (Opn) expression and secretion from hepatocytes, which activate resident hepatic stellate cells. In a cross-sectional study, we found that OPN explains the positive correlation between liver Notch activity and fibrosis stage in patients. Further, we developed a Notch inhibitor [Nicastrin antisense oligonucleotide (Ncst ASO)] that reduced fibrosis in NASH diet-fed mice. In summary, these studies demonstrate the pathological role and therapeutic accessibility of the maladaptive hepatocyte Notch response in NASH-associated liver fibrosis.
Collapse
Affiliation(s)
- Changyu Zhu
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - KyeongJin Kim
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Alberto Bartolome
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Marcela Salomao
- Department of Pathology, Mayo Clinic, Phoenix, AZ 85054, USA
| | - Paola Dongiovanni
- Department of Pathophysiology and Transplantation, Università degli Studi Milano, and Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan 20122, Italy
| | - Marica Meroni
- Department of Pathophysiology and Transplantation, Università degli Studi Milano, and Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan 20122, Italy
| | | | - Katherine P. Yates
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Robert F. Schwabe
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Ira Tabas
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi Milano, and Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan 20122, Italy
| | - Joel E. Lavine
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Utpal B. Pajvani
- Department of Medicine, Columbia University, New York, NY 10032, USA.,Corresponding author.
| |
Collapse
|
158
|
Maricic I, Marrero I, Eguchi A, Nakamura R, Johnson CD, Dasgupta S, Hernandez CD, Nguyen PS, Swafford AD, Knight R, Feldstein AE, Loomba R, Kumar V. Differential Activation of Hepatic Invariant NKT Cell Subsets Plays a Key Role in Progression of Nonalcoholic Steatohepatitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:3017-3035. [PMID: 30322964 PMCID: PMC6219905 DOI: 10.4049/jimmunol.1800614] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/07/2018] [Indexed: 02/07/2023]
Abstract
Innate immune mechanisms play an important role in inflammatory chronic liver diseases. In this study, we investigated the role of type I or invariant NKT (iNKT) cell subsets in the progression of nonalcoholic steatohepatitis (NASH). We used α-galactosylceramide/CD1d tetramers and clonotypic mAb together with intracytoplasmic cytokine staining to analyze iNKT cells in choline-deficient l-amino acid-defined (CDAA)-induced murine NASH model and in human PBMCs, respectively. Cytokine secretion of hepatic iNKT cells in CDAA-fed C57BL/6 mice altered from predominantly IL-17+ to IFN-γ+ and IL-4+ during NASH progression along with the downmodulation of TCR and NK1.1 expression. Importantly, steatosis, steatohepatitis, and fibrosis were dependent upon the presence of iNKT cells. Hepatic stellate cell activation and infiltration of neutrophils, Kupffer cells, and CD8+ T cells as well as expression of key proinflammatory and fibrogenic genes were significantly blunted in Jα18-/- mice and in C57BL/6 mice treated with an iNKT-inhibitory RAR-γ agonist. Gut microbial diversity was significantly impacted in Jα18-/- and in CDAA diet-fed mice. An increased frequency of CXCR3+IFN-γ+T-bet+ and IL-17A+ iNKT cells was found in PBMC from NASH patients in comparison with nonalcoholic fatty liver patients or healthy controls. Consistent with their in vivo activation, iNKT cells from NASH patients remained hyporesponsive to ex-vivo stimulation with α-galactosylceramide. Accumulation of plasmacytoid dendritic cells in both mice and NASH patients suggest their role in activation of iNKT cells. In summary, our findings indicate that the differential activation of iNKT cells play a key role in mediating diet-induced hepatic steatosis and fibrosis in mice and its potential involvement in NASH progression in humans.
Collapse
Affiliation(s)
- Igor Maricic
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Idania Marrero
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Akiko Eguchi
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
| | - Ryota Nakamura
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
| | - Casey D Johnson
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
| | - Suryasarathi Dasgupta
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Carolyn D Hernandez
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Phirum Sam Nguyen
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Austin D Swafford
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA 92093; and
| | - Ariel E Feldstein
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
- Nonalcoholic Fatty Liver Disease Research Center, University of California San Diego, La Jolla, CA 92093
| | - Rohit Loomba
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093
- Nonalcoholic Fatty Liver Disease Research Center, University of California San Diego, La Jolla, CA 92093
| | - Vipin Kumar
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093;
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093
- Nonalcoholic Fatty Liver Disease Research Center, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
159
|
Adams LC, Lübbe F, Bressem K, Wagner M, Hamm B, Makowski MR. Non-alcoholic fatty liver disease in underweight patients with inflammatory bowel disease: A case-control study. PLoS One 2018; 13:e0206450. [PMID: 30427909 PMCID: PMC6241122 DOI: 10.1371/journal.pone.0206450] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/13/2018] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) was shown to also occur in lean and underweight patients. So far, the prevalence of NAFLD in underweight individuals with and without inflammatory bowel disease (IBD) is insufficiently enlightened. In this cross-sectional age, gender and disease-matched case-control study, underweight patients (BMI<18.5 kg/m2) with inflammatory bowel disease (IBD), who underwent abdominal MRI at 1.5 T/3 T with fat-saturated fast-spin-echo imaging from 10/2005-07/2018 were analysed (control-to-case-ratio 1:1, n = 130). All patients were additionally investigated for duration, history of surgery, medical treatment, laboratory values, liver and spleen diameters. On MRI, liver fat was quantified by two observers based on the relative signal loss on T2-weighted fast spin-echo MR images with fat saturation compared to images without fat saturation. The prevalence of NAFLD/liver steatosis, defined as a measured intrahepatic fat content of at least 5%, was significantly higher in underweight IBD patients than in normal weight patients (87.6% versus 21.5%, p<0.001). Compared to the cases, the liver fat content of the controls was reduced by -0.19 units on average (-19%; 95%Cl: -0.20; -0.14). Similar results were obtained for the subgroup of non-IBD individuals (n = 12; -0.25 units on average (-25%); 95%Cl: -0.35; -0.14). Patients with extremely low body weight (BMI <17.5 kg/m2) showed the highest liver fat content (+0.15 units on average (+15%) compared to underweight patients with a BMI of 17.5-18.5 kg/m2 (p<0.05)). Furthermore, underweight patients showed slightly increased liver enzymes and liver diameters. There were no indications of significant differences in disease duration, type of medications or surgery between cases and controls and also, there were no significant differences between observers or field strengths (p>0.05). The prevalence of liver steatosis was higher among underweight IBD and non-IBD patients compared to normal weight controls. Also, underweight patients showed slightly increased liver enzymes and liver diameters, hinting at initial metabolic disturbances.
Collapse
Grants
- Deutsche Forschungsgemeinschaft
- BIH/Charité – Universitätsmedizin Berlin (DE)
- BH has received research grants for the Department of Radiology, Charité – Universitätsmedizin Berlin from the following companies: 1. Abbott, 2. Actelion Pharmaceuticals, 3. Bayer Schering Pharma, 4. Bayer Vital, 5. BRACCO Group, 6. Bristol-Myers Squibb, 7. Charite research organisation GmbH, 8. Deutsche Krebshilfe, 9. Dt. Stiftung für Herzforschung, 10. Essex Pharma, 11. EU Programmes, 12. Fibrex Medical Inc., 13. Focused Ultrasound Surgery Foundation, 14. Fraunhofer Gesellschaft, 15. Guerbet, 16. INC Research, 17. lnSightec Ud., 18. IPSEN Pharma, 19. Kendlel MorphoSys AG, 20. Lilly GmbH, 21. Lundbeck GmbH, 22. MeVis Medical Solutions AG, 23. Nexus Oncology, 24. Novartis, 25. Parexel Clinical Research Organisation Service, 26. Perceptive, 27. Pfizer GmbH, 28. Philipps, 29. Sanofis-Aventis S.A, 30. Siemens, 31. Spectranetics GmbH, 32. Terumo Medical Corporation, 33. TNS Healthcare GMbH, 34. Toshiba, 35. UCB Pharma, 36. Wyeth Pharma, 37. Zukunftsfond Berlin (TSB), 38. Amgen, 39. AO Foundation, 40. BARD, 41. BBraun, 42. Boehring Ingelheimer, 43. Brainsgate, 44. PPD (Clinical Research Organisation), 45. CELLACT Pharma, 46. Celgene, 47. CeloNova BioSciences, 48. Covance, 49. DC Deviees, Ine. USA, 50. Ganymed, 51. Gilead Sciences, 52. Glaxo Smith Kline, 53. ICON (Clinical Research Organisation), 54. Jansen, 55. LUX Bioseienees, 56. MedPass, 57. Merek, 58. Mologen, 59. Nuvisan, 60. Pluristem, 61. Quintiles, 62. Roehe, 63. Sehumaeher GmbH (Sponsoring eines Workshops), 64. Seattle Geneties, 65. Symphogen, 66. TauRx Therapeuties Ud., 67. Accovion, 68. AIO: Arbeitsgemeinschaft Internistische Onkologie, 69. ASR Advanced sleep research, 70. Astellas, 71. Theradex, 72. Galena Biopharma, 73. Chiltern, 74. PRAint, 75. lnspiremd, 76. Medronic, 77. Respicardia, 78. Silena Therapeutics, 79. Spectrum Pharmaceuticals, 80. St. Jude., 81. TEVA, 82. Theorem, 83. Abbvie, 84. Aesculap, 85. Biotronik, 86. Inventivhealth, 87. ISA Therapeutics, 88. LYSARC, 89. MSD, 90. novocure, 91. Ockham oncology, 92. Premier-research, 93. Psi-cro, 94. Tetec-ag, 94. Tetec-ag, 95. Winicker-norimed, 96. Achaogen Inc, 97. ADIR, 98. AstraZenaca AB, 99. Demira Inc, 100.Euroscreen S.A., 101. Galmed Research and Development Ltd., 102. GETNE, 103. Guidant Europe NV, 104. Holaira Inc., 105. Immunomedics Inc., 106. Innate Pharma, 107. Isis Pharmaceuticals Inc, 108. Kantar Health GmbH, 109. MedImmune Inc, 110. Medpace Germany GmbH (CRO), 111. Merrimack Pharmaceuticals Inc, 112. Millenium Pharmaceuticals Inc, 113. Orion Corporation Orion Pharma, 114. Pharmacyclics Inc, 115. PIQUR Therapeutics Ltd, 116. Pulmonx International Sárl, 117. Servier (CRO), 118. SGS Life Science Services (CRO), 119. Treshold Pharmaceuticals Inc.
Collapse
Affiliation(s)
- Lisa C. Adams
- Department of Radiology, Charité, Berlin, Germany
- * E-mail:
| | - Falk Lübbe
- Department of Radiology, Charité, Berlin, Germany
| | - Keno Bressem
- Department of Radiology, Charité, Berlin, Germany
| | | | - Bernd Hamm
- Department of Radiology, Charité, Berlin, Germany
| | | |
Collapse
|
160
|
Inoue-Yamauchi A, Itagaki H, Oda H. Eicosapentaenoic acid attenuates obesity-related hepatocellular carcinogenesis. Carcinogenesis 2018; 39:28-35. [PMID: 29040439 PMCID: PMC5862334 DOI: 10.1093/carcin/bgx112] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 10/10/2017] [Indexed: 12/17/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), the hepatic manifestation of obesity, is an emerging risk factor for hepatocellular carcinoma (HCC). Accumulating evidence has shown that chronic inflammation represents a plausible link between obesity and HCC and that the pro-inflammatory cytokine interleukin (IL)-6 contributes to the development of obesity-related HCC. In the present study, we aimed to examine the therapeutic potential of the omega-3 polyunsaturated fatty acid, eicosapentaenoic acid (EPA), which exerts anti-inflammatory effects. The results showed that the development of carcinogen-induced HCC was significantly less in mice fed a high-fat diet (HFD) supplemented with EPA than in those fed HFD only, suggesting that EPA attenuates the development of obesity-related HCC. Although EPA did not appear to affect obesity-linked inflammation, it suppressed the activation of the pro-tumorigenic IL-6 effector STAT3, contributing to the inhibition of tumor growth. These findings suggest a clinical implication of EPA as a treatment for obesity-related HCC.
Collapse
Affiliation(s)
| | - Hiroko Itagaki
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan.,Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hideaki Oda
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
161
|
Van der Graaff D, Kwanten WJ, Couturier FJ, Govaerts JS, Verlinden W, Brosius I, D'Hondt M, Driessen A, De Winter BY, De Man JG, Michielsen PP, Francque SM. Severe steatosis induces portal hypertension by systemic arterial hyporeactivity and hepatic vasoconstrictor hyperreactivity in rats. J Transl Med 2018; 98:1263-1275. [PMID: 29326427 DOI: 10.1038/s41374-017-0018-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/30/2017] [Accepted: 12/13/2017] [Indexed: 12/27/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease. The presence of portal hypertension has been demonstrated in NAFLD prior to development of inflammation or fibrosis, and is a result of extrahepatic and intrahepatic factors, principally driven by vascular dysfunction. An increased intrahepatic vascular resistance potentially contributes to progression of NAFLD via intralobular hypoxia. However, the exact mechanisms underlying vascular dysfunction in NAFLD remain unknown. This study investigates systemic hemodynamics and both aortic and intrahepatic vascular reactivity in a rat model of severe steatosis. Wistar rats were fed a methionine-choline-deficient diet, inducing steatosis, or control diet for 4 weeks. In vivo hemodynamic measurements, aortic contractility studies, and in situ liver perfusion experiments were performed. The mean arterial blood pressure was lower and portal blood pressure was higher in steatosis compared to controls. The maximal contraction force in aortic rings from steatotic rats was markedly reduced compared to controls. While blockade of nitric oxide (NO) production did not reveal any differences, cyclooxygenase (COX) blockade reduced aortic reactivity in both controls and steatosis, whereas effects were more pronounced in controls. Effects could be attributed to COX-2 iso-enzyme activity. In in situ liver perfusion experiments, exogenous NO donation or endogenous NO stimulation reduced the transhepatic pressure gradient (THPG), whereas NO synthase blockade increased the THPG only in steatosis, but not in controls. Alpha-1-adrenergic stimulation and endothelin-1 induced a significantly more pronounced increase in THPG in steatosis compared to controls. Our results demonstrate that severe steatosis, without inflammation or fibrosis, induces portal hypertension and signs of a hyperdynamic circulation, accompanied by extrahepatic arterial hyporeactivity and intrahepatic vascular hyperreactivity. The arterial hyporeactivity seems to be NO-independent, but appears to be mediated by specific COX-2-related mechanisms. Besides, the increased intrahepatic vascular resistance in steatosis appears not to be NO-related but rather to vasoconstrictor hyperreactivity.
Collapse
Affiliation(s)
- Denise Van der Graaff
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Wilhelmus J Kwanten
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Filip J Couturier
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Jesse S Govaerts
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Verlinden
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Isabel Brosius
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Michiel D'Hondt
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Ann Driessen
- Department of Pathology, Antwerp University Hospital, Laboratory of Pathology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Joris G De Man
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter P Michielsen
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Sven M Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium. .,Laboratory of Experimental Medicine and Pediatrics (LEMP), Division of Gastroenterology-Hepatology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
162
|
Cruces-Sande M, Vila-Bedmar R, Arcones AC, González-Rodríguez Á, Rada P, Gutiérrez-de-Juan V, Vargas-Castrillón J, Iruzubieta P, Sánchez-González C, Formentini L, Crespo J, García-Monzón C, Martínez-Chantar ML, Valverde ÁM, Mayor F, Murga C. Involvement of G protein-coupled receptor kinase 2 (GRK2) in the development of non-alcoholic steatosis and steatohepatitis in mice and humans. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3655-3667. [PMID: 30261289 DOI: 10.1016/j.bbadis.2018.09.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/17/2018] [Accepted: 09/19/2018] [Indexed: 01/04/2023]
Abstract
Insulin resistance (IR) and obesity are important risk factors for non-alcoholic fatty liver disease (NAFLD). G protein-coupled receptor kinase 2 (GRK2) is involved in the development of IR and obesity in vivo. However, its possible contribution to NAFLD and/or non-alcoholic steatohepatitis (NASH) independently of its role on IR or fat mass accretion has not been explored. Here, we used wild-type (WT) or GRK2 hemizygous (GRK2±) mice fed a high-fat diet (HFD) or a methionine and choline-deficient diet (MCD) as a model of NASH independent of adiposity and IR. GRK2± mice were protected from HFD-induced NAFLD. Moreover, MCD feeding caused an increased in triglyceride content and liver-to-body weight ratio in WT mice, features that were attenuated in GRK2± mice. According to their NAFLD activity score, MCD-fed GRK2± mice were diagnosed with simple steatosis and not overt NASH. They also showed reduced expression of lipogenic and lipid-uptake markers and less signs of inflammation in the liver. GRK2± mice preserved hepatic protective mechanisms as enhanced autophagy and mitochondrial fusion and biogenesis, together with reduced endoplasmic reticulum stress. GRK2 protein was increased in MCD-fed WT but not in GRK2± mice, and enhanced GRK2 expression potentiated palmitic acid-triggered lipid accumulation in human hepatocytes directly relating GRK2 levels to steatosis. GRK2 protein and mRNA levels were increased in human liver biopsies from simple steatosis or NASH patients in two different human cohorts. Our results describe a functional relationship between GRK2 levels and hepatic lipid accumulation and implicate GRK2 in the establishment and/or development of NASH.
Collapse
Affiliation(s)
- Marta Cruces-Sande
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), Madrid, Spain
| | - Rocío Vila-Bedmar
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), Madrid, Spain
| | - Alba C Arcones
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), Madrid, Spain
| | - Águeda González-Rodríguez
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; Unidad de Investigación, Hospital Universitario Santa Cristina, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, Spain
| | - Patricia Rada
- Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC/UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Virginia Gutiérrez-de-Juan
- Center for Cooperative Research in Bioscience (CIC bioGUNE), Liver Disease Lab, Derio, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, Spain
| | - Javier Vargas-Castrillón
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; Unidad de Investigación, Hospital Universitario Santa Cristina, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, Spain
| | - Paula Iruzubieta
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Infection, Immunity and Digestive Pathology Group, Research Institute Marqués de Valdecilla (IDIVAL), Santander, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, Spain
| | - Cristina Sánchez-González
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), ISCIII, Spain
| | - Laura Formentini
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), ISCIII, Spain
| | - Javier Crespo
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Infection, Immunity and Digestive Pathology Group, Research Institute Marqués de Valdecilla (IDIVAL), Santander, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, Spain
| | - Carmelo García-Monzón
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; Unidad de Investigación, Hospital Universitario Santa Cristina, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, Spain
| | - María L Martínez-Chantar
- Center for Cooperative Research in Bioscience (CIC bioGUNE), Liver Disease Lab, Derio, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, Spain
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC/UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Federico Mayor
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), Madrid, Spain.
| | - Cristina Murga
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Madrid, Spain; CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), Madrid, Spain.
| |
Collapse
|
163
|
Sid V, Siow YL, Shang Y, Woo CW, O K. High-fat diet consumption reduces hepatic folate transporter expression via nuclear respiratory factor-1. J Mol Med (Berl) 2018; 96:1203-1213. [PMID: 30178194 DOI: 10.1007/s00109-018-1688-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 08/03/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Folate is an essential micronutrient for biological function. The liver, a primary organ for folate metabolism and storage, plays an important role in folate homeostasis. Proton-coupled folate transporter (PCFT) and reduced folate carrier (RFC) are the major folate transporters responsible for folate uptake at basolateral membrane of hepatocytes. Low serum folate levels are frequently associated with obesity. We investigated the mechanism that regulated folate status in a mouse model with diet-induced obesity. Mice (C57BL/6J) were fed a high-fat diet (60% kcal fat) for 8 weeks. Mice displayed increased hepatic lipid accumulation and decreased folate levels in the liver and serum compared to mice fed a normal chow diet (10% kcal fat). High-fat diet-fed mice had low expression of PCFT and RFC and decreased nuclear respiratory factor-1 (NRF-1)/DNA-binding activity. Treatment with NRF-1 siRNA or palmitic acid reduced folate transporter expression in hepatocytes. Inhibition of NRF-1 mediated folate transporter expression significantly reduced intracellular folate levels. These results suggest that chronic consumption of high-fat diets impairs folate transporter expression via NRF-1-dependent mechanism, leading to reduced hepatic folate storage. Understanding the regulation of folate homeostasis in obesity may have an important implication in current guideline of folate intake. KEY MESSAGES: Serum and liver folate levels are decreased in diet-induced obese mice. Chronic high-fat diet consumption impairs expression of hepatic PCFT and RFC. NRF-1 regulates hepatic folate transporters expression and folate levels.
Collapse
Affiliation(s)
- Victoria Sid
- St. Boniface Hospital Research Centre, Winnipeg, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Yaw L Siow
- St. Boniface Hospital Research Centre, Winnipeg, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
- Agriculture and Agri-Food Canada, Winnipeg, Canada
| | - Yue Shang
- St. Boniface Hospital Research Centre, Winnipeg, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, Canada
| | - Connie W Woo
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, SAR, China.
| | - Karmin O
- St. Boniface Hospital Research Centre, Winnipeg, Canada.
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.
- Department of Animal Science, University of Manitoba, Winnipeg, Canada.
- Laboratory of Integrative Biology, CCARM, St. Boniface Hospital Research Centre, 351 Tache Avenue, Winnipeg, Manitoba, R2H 2A6, Canada.
| |
Collapse
|
164
|
Liu XJ, Duan NN, Liu C, Niu C, Liu XP, Wu J. Characterization of a murine nonalcoholic steatohepatitis model induced by high fat high calorie diet plus fructose and glucose in drinking water. J Transl Med 2018; 98:1184-1199. [PMID: 29959418 DOI: 10.1038/s41374-018-0074-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/20/2018] [Accepted: 04/20/2018] [Indexed: 02/07/2023] Open
Abstract
There are varieties of murine models of nonalcoholic steatohepatitis (NASH) with different pathophysiologic characteristics. For preclinical assessment, a standardized model would allow comparisons of various pharmacotherapeutic candidates in efficacy, pharmacokinetics, pharmaco-metabolism, and adverse effects under a same system. The present study aims to characterize murine NASH models by comparing end-points of major abnormalities. NASH was induced by feeding high fructose/glucose in drinking water (HF/G), high-fat/calorie diet (HFCD), and in combination (HFCD-HF/G) in mice for 8 or 16 weeks. HF/G feeding caused a minimal fat accumulation and increase in free fatty acids (FFA). In contrast, HFCD-HF/G feeding resulted in a remarkable increase in body weight, subcutaneous and visceral adipose tissue, macrosteatosis with a nearly seven-fold increase in triglyceride and FFA content, accompanied with marked hepatocellular injury, inflammatory responses, fibrosis, and insulin resistance, and represented as typical NASH in histopathology, metabolic, and adipokine profiles in a progressive manner. Meanwhile, mice fed HFCD displayed significant steatosis, necroptosis, fibrosis, insulin resistance, metabolic, and adipokine profiles, and the extent is less than those fed HFCD-HF/G. Significant MCP-1, CCR-2, and NLRP-1/3 activation were found in mice fed HFCD and HFCD-HF/G for 16 weeks, whereas gene expression of CPT-1 and ACOX-1 was down-regulated in these two groups in comparison to the controls. Nuclear receptors, such as SREBP-1c, FXR, LXR-α, PPAR-α, and PPAR-γ, were strikingly elevated in the HFCD-HF/G group. In conclusion, feeding HFCD-HF/G resulted in a reliable NASH model in mice with remarkable necroptosis, steatosis, fibrosis, and insulin resistance as well as a disordered profile of lipid metabolism and adipokine, and HFCD caused significant NASH features in histopathology and metabolic profiles only at a late stage. Whereas HF/G feeding barely led to minimal fat accumulation, some changes at molecular levels and metabolic disturbance in mice.
Collapse
Affiliation(s)
- Xue-Jing Liu
- Department of Medical Microbiology, Key Laboratory of Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Na-Na Duan
- Department of Medical Microbiology, Key Laboratory of Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.,Stomatological Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, 215005, China
| | - Chang Liu
- Department of Medical Microbiology, Key Laboratory of Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chen Niu
- Department of Medical Microbiology, Key Laboratory of Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Xiu-Ping Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Jian Wu
- Department of Medical Microbiology, Key Laboratory of Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China. .,Shanghai Institute of Liver Diseases, Fudan University Shanghai Medical College, Shanghai, 200032, China.
| |
Collapse
|
165
|
Brown ZJ, Heinrich B, Greten TF. Mouse models of hepatocellular carcinoma: an overview and highlights for immunotherapy research. Nat Rev Gastroenterol Hepatol 2018; 15:536-554. [PMID: 29904153 DOI: 10.1038/s41575-018-0033-6] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mouse models are the basis of preclinical and translational research in hepatocellular carcinoma (HCC). Multiple methods exist to induce tumour formation in mice, including genetically engineered mouse models, chemotoxic agents, intrahepatic or intrasplenic injection of tumour cells and xenograft approaches. Additionally, as HCC generally develops in the context of diseased liver, methods exist to induce liver disease in mice to mimic viral hepatitis, fatty liver disease, fibrosis, alcohol-induced liver disease and cholestasis. Similar to HCC in humans, response to therapy in mouse models is monitored with imaging modalities such as CT or MRI, as well as additional techniques involving bioluminescence. As immunotherapy is increasingly applied to HCC, mouse models for these approaches are required for preclinical data. In studying cancer immunotherapy, it is important to consider aspects of antitumour immune responses and to produce a model that mimics the complexity of the immune system. This Review provides an overview of the different mouse models of HCC, presenting techniques to prepare an HCC mouse model and discussing different approaches to help researchers choose an appropriate model for a specific hypothesis. Specific aspects of immunotherapy research in HCC and the applied mouse models in this field are also highlighted.
Collapse
Affiliation(s)
- Zachary J Brown
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bernd Heinrich
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
166
|
Na AY, Jo JJ, Kwon OK, Shrestha R, Cho PJ, Kim KM, Ki SH, Lee TH, Jeon TW, Jeong TC, Lee S. Investigation of nonalcoholic fatty liver disease-induced drug metabolism by comparative global toxicoproteomics. Toxicol Appl Pharmacol 2018; 352:28-37. [DOI: 10.1016/j.taap.2018.05.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/18/2018] [Accepted: 05/19/2018] [Indexed: 02/06/2023]
|
167
|
Liss KH, McCommis KS, Chambers KT, Pietka TA, Schweitzer GG, Park SL, Nalbantoglu ILK, Weinheimer CJ, Hall AM, Finck BN. The impact of diet-induced hepatic steatosis in a murine model of hepatic ischemia/reperfusion injury. Liver Transpl 2018; 24:908-921. [PMID: 29729104 PMCID: PMC6097916 DOI: 10.1002/lt.25189] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/22/2017] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
The prevalence of obesity-associated nonalcoholic fatty liver disease has significantly increased over the past decade, and end-stage liver disease secondary to nonalcoholic steatohepatitis has become 1 of the most common indications for liver transplantation. This both increases the demand for organs and decreases the availability of donor livers deemed suitable for transplantation. Although in the past many steatotic livers were discarded due to concerns over enhanced susceptibility to ischemia/reperfusion injury (IRI) and organ failure, the discrepancy between supply and demand has resulted in increasing use of expanded criteria donor organs including steatotic livers. However, it remains controversial whether steatotic livers can be safely used for transplantation and how best to improve the performance of steatotic grafts. We aimed to evaluate the impact of diet-induced hepatic steatosis in a murine model of IRI. Using a diet of high trans-fat, fructose, and cholesterol (HTF-C) and a diet high in saturated fats, sucrose, and cholesterol (Western diet), we were able to establish models of mixed macrovesicular and microvesicular steatosis (HTF-C) and microvesicular steatosis (Western). We found that the presence of hepatic steatosis, whether it is predominantly macrovesicular or microvesicular, significantly worsens IRI as measured by plasma alanine aminotransferase levels and inflammatory cytokine concentration, and histological evaluation for necrosis. Additionally, we report on a novel finding in which hepatic IRI in the setting of steatosis results in the induction of the necroptosis factors, receptor interacting protein kinase (RIPK) 3, RIPK1, and mixed-lineage kinase domain-like. These data lay the groundwork for additional experimentation to test potential therapeutic approaches to limit IRI in steatotic livers by using a genetically tractable system. Liver Transplantation 24 908-921 2018 AASLD.
Collapse
Affiliation(s)
- Kim H.H. Liss
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Kyle S. McCommis
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Kari T. Chambers
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Terri A. Pietka
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Sara L. Park
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - ILKe Nalbantoglu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Carla J. Weinheimer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Angela M. Hall
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Brian N. Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
168
|
Fang J, Zhou Z, Chang NF, Wan YL, Tsui PH. Ultrasound parametric imaging of hepatic steatosis using the homodyned-K distribution: An animal study. ULTRASONICS 2018; 87:91-102. [PMID: 29476945 DOI: 10.1016/j.ultras.2018.02.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/12/2018] [Accepted: 02/14/2018] [Indexed: 06/08/2023]
Abstract
Hepatic steatosis is an abnormal state where excess lipid mass is accumulated in hepatocyte vesicles. Backscattered ultrasound signals received from the liver contain useful information regarding the degree of steatosis in the liver. The homodyned-K (HK) distribution has been demonstrated as a general model for ultrasound backscattering. The estimator based on the first three integer moments (denoted as "FTM") of the intensity has potential for practical applications because of its simplicity and low computational complexity. This study explored the diagnostic performance of HK parametric imaging based on the FTM method in the assessment of hepatic steatosis. Phantom experiments were initially conducted using the sliding window technique to determine an appropriate window size length (WSL) for HK parametric imaging. Subsequently, hepatic steatosis was induced in male Wistar rats fed a methionine- and choline-deficient (MCD) diet for 0 (i.e., normal control), 1, 2, 4, 6, and 8 weeks (n = 36; six rats in each group). After completing the scheduled MCD diet, ultrasound B-mode and HK imaging of the rat livers were performed in vivo and histopathological examinations were conducted to score the degree of hepatic steatosis. HK parameters μ (related to scatterer number density) and k (related to scatterer periodicity) were expressed as functions of the steatosis stage in terms of the median and interquartile range (IQR). Receiver operating characteristic (ROC) curve analysis was conducted to assess the diagnostic performance levels of the μ and k parameters. The results showed that an appropriate WSL for HK parametric imaging is seven times the pulse length of the transducer. The median value of the μ parameter increased monotonically from 0.194 (IQR: 0.18-0.23) to 0.893 (IQR: 0.64-1.04) as the steatosis stage increased. Concurrently, the median value of the k parameter increased from 0.279 (IQR: 0.26-0.31) to 0.5 (IQR: 0.41-0.54) in the early stages (normal to mild) and decreased to 0.39 (IQR: 0.29-0.45) in the advanced stages (moderate to severe). The areas under the ROC curves obtained using (μ, k) were (0.947, 0.804), (0.914, 0.575), and (0.813, 0.604) for the steatosis stages of ≥mild, ≥moderate, and ≥severe, respectively. The current findings suggest that ultrasound HK parametric imaging based on FTM estimation has great potential for future clinical diagnoses of hepatic steatosis.
Collapse
Affiliation(s)
- Jui Fang
- Ph. D. Program in Biomedical Engineering, College of Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Zhuhuang Zhou
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China; Faculty of Information Technology, Beijing University of Technology, Beijing, China
| | - Ning-Fang Chang
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yung-Liang Wan
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University and Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Po-Hsiang Tsui
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University and Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| |
Collapse
|
169
|
Wang W, Zhang D, Yang R, Xia W, Qian K, Shi Z, Brown R, Zhou H, Xi Y, Shi L, Chen L, Xu F, Sun X, Zhu D, Gong DW. Hepatic and cardiac beneficial effects of a long-acting Fc-apelin fusion protein in diet-induced obese mice. Diabetes Metab Res Rev 2018; 34:e2997. [PMID: 29577579 DOI: 10.1002/dmrr.2997] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 02/10/2018] [Accepted: 02/11/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Apelin is a peptide ligand of the G-protein-coupled receptor APJ and exhibits anti-diabetes and anti-heart failure activities. However, short serum half-life of the apelin peptide limits its potential clinical applications. This study aimed to develop a long-acting apelin analog. METHODS To extend apelin's in vivo half-life, we made a recombinant protein by fusing the IgG Fc fragment to apelin-13 (Fc-apelin-13), conducted pharmacokinetics studies in mice, and determined in vitro biological activities in suppressing cyclic adenosine monophosphate and activating extracellular signal-regulated kinase signalling by reporter assays. We investigated the effects of Fc-apelin-13 on food intake, body weight, fasting blood glucose and insulin levels, glucose tolerance test, hepatic steatosis, and cardiac function and fibrosis by subcutaneous administration of Fc-apelin-13 in diet-induced obese mice for 4 weeks. RESULTS The estimated half-life of Fc-apelin-13 in blood was approximately 33 hours. Reporter assays showed that Fc-apelin-13 was active in suppressing cyclic adenosine monophosphate response element and activating serum response element activities. Four weeks of Fc-apelin-13 treatment in obese mice did not affect food intake and body weight, but resulted in a significant improvement of glucose tolerance, and a decrease in hepatic steatosis and fibrosis, as well as in serum alanine transaminase levels. Moreover, cardiac stroke volume and output were increased and cardiac fibrosis was decreased in the treated mice. CONCLUSIONS Fc-apelin-13 fusion protein has an extended in vivo half-life and exerts multiple benefits on obese mice with respect to the improvement of glucose disposal, amelioration of liver steatosis and heart fibrosis, and increase of cardiac output. Hence, Fc-apelin-13 is potentially a therapeutic for obesity-associated disease conditions.
Collapse
Affiliation(s)
- Weimin Wang
- Department of Endocrinology, Drum Tower Hospital of Nanjing Medical University, Nanjing, China
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dongming Zhang
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rongze Yang
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wei Xia
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kun Qian
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zhengrong Shi
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert Brown
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Huifen Zhou
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yue Xi
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lin Shi
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ling Chen
- Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Feng Xu
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Xiaojian Sun
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital of Nanjing Medical University, Nanjing, China
| | - Da-Wei Gong
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
170
|
Yan T, Wang H, Cao L, Wang Q, Takahashi S, Yagai T, Li G, Krausz KW, Wang G, Gonzalez FJ, Hao H. Glycyrrhizin Alleviates Nonalcoholic Steatohepatitis via Modulating Bile Acids and Meta-Inflammation. Drug Metab Dispos 2018; 46:1310-1319. [PMID: 29959134 DOI: 10.1124/dmd.118.082008] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/27/2018] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the progressive stage of nonalcoholic fatty liver disease that may ultimately lead to cirrhosis and liver cancer, and there are few therapeutic options for its treatment. Glycyrrhizin (GL), extracted from the traditional Chinese medicine liquorice, has potent hepatoprotective effects in both preclinical animal models and in humans. However, little is currently known about its effects and mechanisms in treating NASH. To explore the effects of GL on NASH, GL or its active metabolite glycyrrhetinic acid (GA) was administered to mice treated with a methionine- and choline-deficient (MCD) diet-induced NASH model, and histologic and biochemical analyses were used to measure the degree of lipid disruption, liver inflammation, and fibrosis. GL significantly improved MCD diet-induced hepatic steatosis, inflammation, and fibrosis and inhibited activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome. GL significantly attenuated serum bile acid accumulation in MCD diet-fed mice partially by restoring inflammation-mediated hepatic farnesoid X receptor inhibition. In Raw 264.7 macrophage cells, both GL and GA inhibited deoxycholic acid-induced NLRP3 inflammasome-associated inflammation. Notably, both intraperitoneal injection of GL's active metabolite GA and oral administration of GL prevented NASH in mice, indicating that GL may attenuate NASH via its active metabolite GA. These results reveal that GL, via restoration of bile acid homeostasis and inhibition of inflammatory injury, can be a therapeutic option for treatment of NASH.
Collapse
Affiliation(s)
- Tingting Yan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Hong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Lijuan Cao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Qiong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Shogo Takahashi
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Tomoki Yagai
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Guolin Li
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Kristopher W Krausz
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Frank J Gonzalez
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China (Ti. Y., H.W., L.C., G.W., H.H.); and Laboratory of Metabolism, Center for Cancer Research, National Institutes of Health National Cancer Institute, Bethesda, Maryland (Ti. Y., Q.W., S.T., To.Y., G.L., K.W.K., F.J.G.)
| |
Collapse
|
171
|
Wang X, Zhang X, Ma L, Li S. Simultaneous quantification of hepatic MRI-PDFF and R2* in a rabbit model with nonalcoholic fatty liver disease. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1107-1114. [PMID: 29934919 DOI: 10.1007/s11427-017-9279-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022]
Abstract
Quantification of hepatic fat and iron content is important for early detection and monitoring of nonalcoholic fatty liver disease (NAFLD) patients. This study evaluated quantification efficiency of hepatic proton density fat fraction (PDFF) by MRI using NAFLD rabbits. R2* was also measured to investigate whether it correlates with fat levels in NAFLD. NAFLD rabbit model was successfully established by high fat and cholesterol diet. Rabbits underwent MRI examination for fat and iron analyses, compared with liver histological findings. MR examinations were performed on a 3.0T MR system using multi-echo 3D gradient recalled echo (GRE) sequence. MRI-PDFF showed significant differences between different steatosis grades with medians of 3.72% (normal), 5.43% (mild), 9.11% (moderate) and 11.17% (severe), whereas this was not observed in R2*. Close correlation between MRI-PDFF and histological steatosis was observed (r=0.78, P=0.000). Hepatic iron deposit was not found in any rabbits. There was no correlation between R2* and either liver MRI-PDFF or histological steatosis. MR measuring MRI-PDFF and R2* simultaneously provides promising quantification of steatosis and iron. Rabbit NAFLD model confirmed accuracy of MRI-PDFF for liver fat quantification. R2* measurement and relationship between fat and iron of NAFLD liver need further experimental investigation.
Collapse
Affiliation(s)
- Xiaomin Wang
- Department of Radiology, Chinese PLA General Hospital, Beijing, 100853, China.,School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Xiaojing Zhang
- Department of Radiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lin Ma
- Department of Radiology, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Shengli Li
- Laboratory Animal Center, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
172
|
Mohammadghasemi F, Abbasi M, Rudkhaneei K, Aghajany-Nasab M. Beneficial effect of apple vinegar on reproductive parameters in male rat model of nonalcoholic fatty liver disease. Andrologia 2018; 50:e13065. [DOI: 10.1111/and.13065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 04/18/2018] [Accepted: 05/06/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Fahimeh Mohammadghasemi
- Cellular & Molecular Research Center; School Of Medicine; Guilan University of Medical Sciences; Rasht Iran
| | - Masumeh Abbasi
- Department Of Anatomy; School Of Medicine; Guilan University of Medical Sciences; Rasht Iran
| | - Kamran Rudkhaneei
- Department Of Anatomy; School Of Medicine; Guilan University of Medical Sciences; Rasht Iran
| | - Monireh Aghajany-Nasab
- Cellular & Molecular Research Center; School Of Medicine; Guilan University of Medical Sciences; Rasht Iran
- Department Of Biochemistry & Biophysic; School Of Medicine; Guilan University of Medical Sciences; Rasht Iran
| |
Collapse
|
173
|
Treatment with Lobeglitazone Attenuates Hepatic Steatosis in Diet-Induced Obese Mice. PPAR Res 2018; 2018:4292509. [PMID: 30008738 PMCID: PMC6020545 DOI: 10.1155/2018/4292509] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/22/2018] [Accepted: 05/20/2018] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is strongly associated with insulin resistance. The peroxisome proliferator-activated receptor (PPAR) activators, thiazolidinediones, (TZDs), are insulin sensitizers used as a treatment for NAFLD. However, TZDs are a controversial treatment for NAFLD because of conflicting results regarding hepatic steatosis and fibrosis. To evaluate a possible effective drug for treatment of NAFLD, we investigated the effects of a newly developed TZD, lobeglitazone, with an emphasis on hepatic lipid metabolism. Lobeglitazone treatment for 4 weeks in high fat diet- (HFD-) induced obese mice (HL group) improved insulin resistance and glucose intolerance compared to HFD-induced obese mice (HU group). The gene levels related to hepatic gluconeogenesis also decreased after treatment by lobeglitazone. The livers of mice in the HL group showed histologically reduced lipid accumulation, with lowered total plasma cholesterol and triglyceride levels. In addition, the HL group significantly decreased the hepatic expression of genes associated with lipid synthesis, cholesterol biosynthesis, and lipid droplet development and increased the hepatic expression of genes associated with fatty acid β-oxidation, thus suggesting that lobeglitazone decreased hepatic steatosis and reversed hepatic lipid dysregulation. Livers with steatohepatitis contained increased levels of PPARγ and phosphorylated PPARγ at serine 273, leading to downregulation of expression of genes associated with insulin sensitivity. Notably, the treatment of lobeglitazone increased the protein levels of PPARα and diminished levels of PPARγ phosphorylated at serine 273, which were increased by a HFD, suggesting that induction of PPARα and posttranslational modification of PPARγ in livers by lobeglitazone might be an underlying mechanism of the improvement seen in NAFLD. Taken together, our data showed that lobeglitazone might be an effective treatment for NAFLD.
Collapse
|
174
|
Brown ZJ, Heinrich B, Greten TF. Establishment of Orthotopic Liver Tumors by Surgical Intrahepatic Tumor Injection in Mice with Underlying Non-Alcoholic Fatty Liver Disease. Methods Protoc 2018; 1:mps1020021. [PMID: 31164564 PMCID: PMC6526445 DOI: 10.3390/mps1020021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 12/18/2022] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) and its advanced form, nonalcoholic steatohepatitis (NASH), is increasing, and as such its contribution to the development of hepatocellular carcinoma is also rising. NAFLD has been shown to influence the immune tumor microenvironment. Therefore, development of pre-clinical mouse models in the context of NAFLD are increasingly important. Here, we describe a mouse model designed to recapitulate the findings of NAFLD followed by rapid induction of orthotopic liver tumors with intrahepatic tumor injection. Additionally, we utilized bioluminescent imaging to monitor tumor growth and response to therapy. The development of one dominant tumor nodule allows precise separation of tumor and liver tissue. This is useful for immunotherapy studies as mononuclear cells from the tumor and the surrounding liver tissue can be analyzed separately.
Collapse
Affiliation(s)
- Zachary J Brown
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Bernd Heinrich
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
175
|
Wilson CH, Kumar S. Caspases in metabolic disease and their therapeutic potential. Cell Death Differ 2018; 25:1010-1024. [PMID: 29743560 PMCID: PMC5988802 DOI: 10.1038/s41418-018-0111-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/26/2018] [Accepted: 03/22/2018] [Indexed: 12/13/2022] Open
Abstract
Caspases, a family of cysteine-dependent aspartate-specific proteases, are central to the maintenance of cellular and organismal homoeostasis by functioning as key mediators of the inflammatory response and/or apoptosis. Both metabolic inflammation and apoptosis play a central role in the pathogenesis of metabolic disease such as obesity and the progression of nonalcoholic steatohepatisis (NASH) to more severe liver disease. Obesity and nonalcoholic fatty liver disease (NAFLD) are the leading global health challenges associated with the development of numerous comorbidities including insulin resistance, type-2 diabetes and early mortality. Despite the high prevalence, current treatment strategies including lifestyle, dietary, pharmaceutical and surgical interventions, are often limited in their efficacy to manage or treat obesity, and there are currently no clinical therapies for NAFLD/NASH. As mediators of inflammation and cell death, caspases are attractive therapeutic targets for the treatment of these metabolic diseases. As such, pan-caspase inhibitors that act by blocking apoptosis have reached phase I/II clinical trials in severe liver disease. However, there is still a lack of knowledge of the specific and differential functions of individual caspases. In addition, cross-talk between alternate cell death pathways is a growing concern for long-term caspase inhibition. Evidence is emerging of the important cell-death-independent, non-apoptotic functions of caspases in metabolic homoeostasis that may be of therapeutic value. Here, we review the current evidence for roles of caspases in metabolic disease and discuss their potential targeting as a therapeutic strategy.
Collapse
Affiliation(s)
- Claire H Wilson
- Centre for Cancer Biology, University of South Australia & SA Pathology, Adelaide, SA, 5001, Australia.
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia & SA Pathology, Adelaide, SA, 5001, Australia.
| |
Collapse
|
176
|
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]
|
177
|
Britton L, Bridle K, Reiling J, Santrampurwala N, Wockner L, Ching H, Stuart K, Subramaniam VN, Jeffrey G, St Pierre T, House M, Gummer J, Trengove R, Olynyk J, Crawford D, Adams L. Hepatic iron concentration correlates with insulin sensitivity in nonalcoholic fatty liver disease. Hepatol Commun 2018; 2:644-653. [PMID: 29881816 PMCID: PMC5983226 DOI: 10.1002/hep4.1190] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/23/2018] [Accepted: 04/01/2018] [Indexed: 01/01/2023] Open
Abstract
Rodent and cell‐culture models support a role for iron‐related adipokine dysregulation and insulin resistance in the pathogenesis of nonalcoholic fatty liver disease (NAFLD); however, substantial human data are lacking. We examined the relationship between measures of iron status, adipokines, and insulin resistance in patients with NAFLD in the presence and absence of venesection. This study forms part of the Impact of Iron on Insulin Resistance and Liver Histology in Nonalcoholic Steatohepatitis (IIRON2) study, a prospective randomized controlled trial of venesection for adults with NAFLD. Paired serum samples at baseline and 6 months (end of treatment) in controls (n = 28) and patients who had venesection (n = 23) were assayed for adiponectin, leptin, resistin, retinol binding protein‐4, tumor necrosis factor α, and interleukin‐6, using a Quantibody, customized, multiplexed enzyme‐linked immunosorbent assay array. Hepatic iron concentration (HIC) was determined using MR FerriScan. Unexpectedly, analysis revealed a significant positive correlation between baseline serum adiponectin concentration and HIC, which strengthened after correction for age, sex, and body mass index (rho = 0.36; P = 0.007). In addition, there were significant inverse correlations between HIC and measures of insulin resistance (adipose tissue insulin resistance (Adipo‐IR), serum insulin, serum glucose, homeostasis model assessment of insulin resistance, hemoglobin A1c, and hepatic steatosis), whereas a positive correlation was noted with the insulin sensitivity index. Changes in serum adipokines over 6 months did not differ between the control and venesection groups. Conclusion: HIC positively correlates with serum adiponectin and insulin sensitivity in patients with NAFLD. Further study is required to establish causality and mechanistic explanations for these associations and their relevance in the pathogenesis of insulin resistance and NAFLD. (Hepatology Communications 2018;2:644‐653)
Collapse
Affiliation(s)
- Laurence Britton
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia.,Department of Gastroenterology Princess Alexandra Hospital Woolloongabba Australia.,QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Kim Bridle
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia
| | - Janske Reiling
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia.,Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Maastricht the Netherlands
| | - Nishreen Santrampurwala
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia.,QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Leesa Wockner
- QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Helena Ching
- Medical School, Faculty of Health Sciences University of Western Australia Crawley Australia
| | - Katherine Stuart
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,Department of Gastroenterology Princess Alexandra Hospital Woolloongabba Australia
| | - V Nathan Subramaniam
- QIMR Berghofer Medical Research Institute Brisbane Australia.,Institute of Health and Biomedical Innovation and School of Biomedical Sciences Queensland University of Technology Kelvin Grove Australia
| | - Gary Jeffrey
- Medical School, Faculty of Health Sciences University of Western Australia Crawley Australia.,Department of Hepatology Sir Charles Gairdner Hospital Perth Australia
| | - Tim St Pierre
- School of Physics University of Western Australia Crawley Australia
| | - Michael House
- School of Physics University of Western Australia Crawley Australia
| | - Joel Gummer
- Separation Science and Metabolomics Laboratory (Metabolomics Australia, Western Australia node) Murdoch University Murdoch Australia
| | - Robert Trengove
- Separation Science and Metabolomics Laboratory (Metabolomics Australia, Western Australia node) Murdoch University Murdoch Australia
| | - John Olynyk
- Department of Gastroenterology Fiona Stanley and Fremantle Hospital Group Murdoch Australia.,School of Health and Medical Sciences Edith Cowan University Joondalup Australia
| | - Darrell Crawford
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia
| | - Leon Adams
- Medical School, Faculty of Health Sciences University of Western Australia Crawley Australia.,Department of Hepatology Sir Charles Gairdner Hospital Perth Australia
| |
Collapse
|
178
|
Vasseur P, Dion S, Filliol A, Genet V, Lucas-Clerc C, Jean-Philippe G, Silvain C, Lecron JC, Piquet-Pellorce C, Samson M. Endogenous IL-33 has no effect on the progression of fibrosis during experimental steatohepatitis. Oncotarget 2018; 8:48563-48574. [PMID: 28611297 PMCID: PMC5564708 DOI: 10.18632/oncotarget.18335] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/15/2017] [Indexed: 01/01/2023] Open
Abstract
Interleukin (IL)-33 has been recently reported to be strongly pro-fibrogenic in various models of liver disease. Our aim was to study the role of endogenous IL-33 in a diet-induced model of steatohepatitis. IL-33 deficient mice and wild type (WT) littermates received a high-fat diet (HFD), or a standard diet for 12 weeks. The HFD-induced steatohepatitis was associated with an upregulation of IL-33 transcripts and protein. An insulin tolerance test revealed lower systemic insulin sensitivity in IL-33-/—HFD mice than in WT-HFD mice. Nevertheless, IL-33 deficiency did not affect the severity of liver inflammation by histological and transcriptomic analyses, nor the quantity of liver fibrosis. Livers from HFD mice had more myeloid populations, markedly fewer NKT cells and higher proportion of ST2+ Treg cells and ST2+ type 2 innate lymphoid cells (ILC2), all unaffected by IL-33 deficiency. In conclusion, deficiency of endogenous IL-33 does not affect the evolution of experimental diet-induced steatohepatitis towards liver fibrosis.
Collapse
Affiliation(s)
- Philippe Vasseur
- Service d'Hépato-Gastroentérologie, Centre Hospitalier Nord Deux-Sèvres, Thouars, France.,Laboratoire Inflammation Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France
| | - Sarah Dion
- Institut National de la Santé et de la Recherche Médicale, Institut de Recherche Santé Environnement & Travail, Université de Rennes, Rennes, France
| | - Aveline Filliol
- Institut National de la Santé et de la Recherche Médicale, Institut de Recherche Santé Environnement & Travail, Université de Rennes, Rennes, France
| | - Valentine Genet
- Institut National de la Santé et de la Recherche Médicale, Institut de Recherche Santé Environnement & Travail, Université de Rennes, Rennes, France
| | - Catherine Lucas-Clerc
- Service de Biochimie, Centre Hospitalier Universitaire, Rennes, Université de Rennes, Rennes, France
| | - Girard Jean-Philippe
- Institut de Pharmacologie et de Biologie Structurale, Centre National de la Recherche Scientifique, Université de Toulouse, Toulouse, France
| | - Christine Silvain
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France.,Service d'Hépato-Gastroentérologie, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Jean-Claude Lecron
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France.,Service d'Immunologie et Inflammation, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Claire Piquet-Pellorce
- Institut National de la Santé et de la Recherche Médicale, Institut de Recherche Santé Environnement & Travail, Université de Rennes, Rennes, France
| | - Michel Samson
- Institut National de la Santé et de la Recherche Médicale, Institut de Recherche Santé Environnement & Travail, Université de Rennes, Rennes, France
| |
Collapse
|
179
|
Zou A, Magee N, Deng F, Lehn S, Zhong C, Zhang Y. Hepatocyte nuclear receptor SHP suppresses inflammation and fibrosis in a mouse model of nonalcoholic steatohepatitis. J Biol Chem 2018; 293:8656-8671. [PMID: 29666185 DOI: 10.1074/jbc.ra117.001653] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 03/17/2018] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a burgeoning health problem worldwide, ranging from nonalcoholic fatty liver (NAFL, steatosis without hepatocellular injury) to the more aggressive nonalcoholic steatohepatitis (NASH, steatosis with ballooning, inflammation, or fibrosis). Although many studies have greatly contributed to the elucidation of NAFLD pathogenesis, the disease progression from NAFL to NASH remains incompletely understood. Nuclear receptor small heterodimer partner (Nr0b2, SHP) is a transcriptional regulator critical for the regulation of bile acid, glucose, and lipid metabolism. Here, we show that SHP levels are decreased in the livers of patients with NASH and in diet-induced mouse NASH. Exposing primary mouse hepatocytes to palmitic acid and lipopolysaccharide in vitro, we demonstrated that the suppression of Shp expression in hepatocytes is due to c-Jun N-terminal kinase (JNK) activation, which stimulates c-Jun-mediated transcriptional repression of Shp Interestingly, in vivo induction of hepatocyte-specific SHP in steatotic mouse liver ameliorated NASH progression by attenuating liver inflammation and fibrosis, but not steatosis. Moreover, a key mechanism linking the anti-inflammatory role of hepatocyte-specific SHP expression to inflammation involved SHP-induced suppression of NF-κB p65-mediated induction of chemokine (C-C motif) ligand 2 (CCL2), which activates macrophage proinflammatory polarization and migration. In summary, our results indicate that a JNK/SHP/NF-κB/CCL2 regulatory network controls communications between hepatocytes and macrophages and contributes to the disease progression from NAFL to NASH. Our findings may benefit the development of new management or prevention strategies for NASH.
Collapse
Affiliation(s)
- An Zou
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Nancy Magee
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Fengyan Deng
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Sarah Lehn
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Cuncong Zhong
- the Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, Kansas 66045, and
| | - Yuxia Zhang
- From the Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160, .,the Liver Center, University of Kansas Medical Center, Kansas City, Kansas 66160
| |
Collapse
|
180
|
Britton L, Bridle K, Jaskowski LA, He J, Ng C, Ruelcke JE, Mohamed A, Reiling J, Santrampurwala N, Hill MM, Whitehead JP, Subramaniam VN, Crawford DH. Iron Inhibits the Secretion of Apolipoprotein E in Cultured Human Adipocytes. Cell Mol Gastroenterol Hepatol 2018; 6:215-217.e8. [PMID: 30105281 PMCID: PMC6085534 DOI: 10.1016/j.jcmgh.2018.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/02/2018] [Indexed: 12/11/2022]
Affiliation(s)
- L.J. Britton
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
- Department of Gastroenterology, Princess Alexandra Hospital, Queensland, Australia
- Mater Research, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Kim Bridle
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Lesley-Anne Jaskowski
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Jingjing He
- Mater Research, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Choaping Ng
- Mater Research, Translational Research Institute, Woolloongabba, Queensland, Australia
- School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Jayde E. Ruelcke
- The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Queensland, Australia
| | - Ahmed Mohamed
- The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Janske Reiling
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Nishreen Santrampurwala
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Michelle M. Hill
- The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jonathan P. Whitehead
- Mater Research, Translational Research Institute, Woolloongabba, Queensland, Australia
- School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - V. Nathan Subramaniam
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Darrell H.G. Crawford
- Gallipoli Medical Research Institute, Greenslopes Private Hospital, Greenslopes, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
181
|
Xu B, Jiang M, Chu Y, Wang W, Chen D, Li X, Zhang Z, Zhang D, Fan D, Nie Y, Shao F, Wu K, Liang J. Gasdermin D plays a key role as a pyroptosis executor of non-alcoholic steatohepatitis in humans and mice. J Hepatol 2018; 68:773-782. [PMID: 29273476 DOI: 10.1016/j.jhep.2017.11.040] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Gasdermin D (GSDMD)-executed programmed necrosis is involved in inflammation and controls interleukin (IL)-1β release. However, the role of GSDMD in non-alcoholic steatohepatitis (NASH) remains unclear. We investigated the role of GSDMD in the pathogenesis of steatohepatitis. METHODS Human liver tissues from patients with non-alcoholic fatty liver disease (NAFLD) and control individuals were obtained to evaluate GSDMD expression. Gsdmd knockout (Gsdmd-/-) mice, obese db/db mice and their wild-type (WT) littermates were fed with methionine-choline deficient (MCD) or control diet to induce steatohepatitis. The Gsdmd-/- and WT mice were also used in a high-fat diet (HFD)-induced NAFLD model. In addition, Alb-Cre mice were administered an adeno-associated virus (AAV) vector that expressed the gasdermin-N domain (AAV9-FLEX-GSDMD-N) and were fed with either MCD or control diet for 10 days. RESULTS GSDMD and its pyroptosis-inducing fragment GSDMD-N were upregulated in liver tissues of human NAFLD/NASH. Importantly, hepatic GSDMD-N protein levels were significantly higher in human NASH and correlated with the NAFLD activity score and fibrosis. GSDMD-N remained a potential biomarker for the diagnosis of NASH. MCD-fed Gsdmd-/- mice exhibit decreased severity of steatosis and inflammation compared with WT littermates. GSDMD was associated with the secretion of pro-inflammatory cytokines (IL-1β, TNF-α, and MCP-1 [CCL2]) and persistent activation of the NF-ĸB signaling pathway. Gsdmd-/- mice showed lower steatosis, mainly because of reduced expression of the lipogenic gene Srebp1c (Srebf1) and upregulated expression of lipolytic genes, including Pparα, Aco [Klk15], Lcad [Acadl], Cyp4a10 and Cyp4a14. Alb-Cre mice administered with AAV9-FLEX-GSDMD-N showed significantly aggravated steatohepatitis when fed with MCD diet. CONCLUSION As an executor of pyroptosis, GSDMD plays a key role in the pathogenesis of steatohepatitis, by controlling cytokine secretion, NF-ĸB activation, and lipogenesis. LAY SUMMARY Non-alcoholic fatty liver disease has become one of the most feared chronic liver diseases, because it is the most rapidly growing indication for adult liver transplantation and a major cause of hepatocellular carcinoma. However, the mechanisms involved in the transformation of simple steatosis to steatohepatitis remain unclear. Herein, we show that gasdermin D driven pyroptosis is prominent in patients with non-alcoholic steatohepatitis (NASH), and gasdermin-N domain remains a potential biomarker for the diagnosis of NASH. Gasdermin D plays a key role in the pathogenesis of NASH by regulating lipogenesis, the inflammatory response, and the NF-ĸB signaling pathway, revealing potential treatment targets for NASH in humans.
Collapse
Affiliation(s)
- Bing Xu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Mingzuo Jiang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Yi Chu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Weijie Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Di Chen
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Xiaowei Li
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Zhao Zhang
- Lintong Aerial Medical Evaluation and Training Center of Air Force, PLA, Xi'an, Shaanxi, MI 710032, China
| | - Di Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China
| | - Feng Shao
- National Institute of Biological Sciences, Number 7 Science Park Road, Zhongguancun Life Science Park, Beijing, MI 102206, China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China.
| | - Jie Liang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, MI 710032, China.
| |
Collapse
|
182
|
Fischer IP, Irmler M, Meyer CW, Sachs SJ, Neff F, Hrabě de Angelis M, Beckers J, Tschöp MH, Hofmann SM, Ussar S. A history of obesity leaves an inflammatory fingerprint in liver and adipose tissue. Int J Obes (Lond) 2018; 42:507-517. [PMID: 28901330 PMCID: PMC5880583 DOI: 10.1038/ijo.2017.224] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/28/2017] [Accepted: 09/04/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND/OBJECTIVES Dieting is a popular yet often ineffective way to lower body weight, as the majority of people regain most of their pre-dieting weights in a relatively short time. The underlying molecular mechanisms driving weight regain and the increased risk for metabolic disease are still incompletely understood. Here we investigate the molecular alterations inherited from a history of obesity. METHODS In our model, male high-fat diet (HFD)-fed obese C57BL/6J mice were switched to a low caloric chow diet, resulting in a decline of body weight to that of lean mice. We measured body composition, as well as metrics of glucose, insulin and lipid homeostasis. This was accompanied by histological and gene expression analysis of adipose tissue and liver to assess adipose tissue inflammation and hepatosteatosis. Moreover, acute hypothalamic response to (re-) exposure to HFD was assessed by qPCR. RESULTS & CONCLUSIONS Within 7 weeks after diet switch, most obesity-associated phenotypes, such as body mass, glucose intolerance and blood metabolite levels were reversed. However, hepatic inflammation, hepatic steatosis as well as hypertrophy and inflammation of perigonadal, but not subcutaneous, adipocytes persisted in formerly obese mice. Transcriptional profiling of liver and perigonadal fat revealed an upregulation of pathways associated with immune function and cellularity. Thus, we show that weight reduction leaves signs of inflammation in liver and perigonadal fat, indicating that persisting proinflammatory signals in liver and adipose tissue could contribute to an increased risk of formerly obese subjects to develop the metabolic syndrome upon recurring weight gain.
Collapse
Affiliation(s)
- I P Fischer
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, Garching, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - M Irmler
- Institute for Experimental Genetics, Helmholtz Zentrum München, München-Neuherberg, Germany
| | - C W Meyer
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - S J Sachs
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV der LMU, Munich, Germany
- Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Zentrum München, Garching, Germany
| | - F Neff
- Institute for Pathology, Helmholtz Zentrum München, München-Neuherberg, Germany
| | - M Hrabě de Angelis
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Experimental Genetics, Helmholtz Zentrum München, München-Neuherberg, Germany
- Technische Universität München, Lehrstuhl für Experimentelle Genetik, Freising, Germany
| | - J Beckers
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Experimental Genetics, Helmholtz Zentrum München, München-Neuherberg, Germany
- Technische Universität München, Lehrstuhl für Experimentelle Genetik, Freising, Germany
| | - M H Tschöp
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Garching, Germany
| | - S M Hofmann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV der LMU, Munich, Germany
- Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Zentrum München, Garching, Germany
| | - S Ussar
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, Garching, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
183
|
|
184
|
Chu H, Williams B, Schnabl B. Gut microbiota, fatty liver disease, and hepatocellular carcinoma. LIVER RESEARCH 2018; 2:43-51. [PMID: 30416839 PMCID: PMC6223644 DOI: 10.1016/j.livres.2017.11.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intestinal bacteria contribute to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Recently developed microbial profiling techniques are beginning to shed light on the nature of the changes in the gut microbiota that accompany NAFLD and non-alcoholic steatohepatitis (NASH). In this review, we summarize the role of gut microbiota in the development of NAFLD, NASH, and hepatocellular carcinoma (HCC). We highlight the mechanisms by which gut microbiota contribute to NAFLD/NASH, including through alterations in gut epithelial permeability, choline metabolism, endogenous alcohol production, release of inflammatory cytokines, regulation of hepatic Toll-like receptor (TLR), and bile acid metabolism. In addition, we analyze possible mechanisms for enhanced hepatic carcinogenesis, including alterations in bile acid metabolism, release of inflammatory cytokines, and expression of TLR-4. Finally, we describe therapeutic approaches for NAFLD/NASH and preventive strategies for HCC involving modulation of the intestinal microbiota or affected host pathways. Although recent studies have provided useful information, large-scale prospective studies are required to better characterize the intestinal microbiota and metabolome, in order to demonstrate a causative role for changes in the gut microbiota in the etiology of NAFLD/NASH, to identify new therapeutic strategies for NAFLD/NASH, and to develop more effective methods of preventing HCC.
Collapse
Affiliation(s)
- Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Brandon Williams
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA,epartment of Medicine, VA San Diego Healthcare System, San Diego, CA, USA,Corresponding author. Department of Medicine, University of California San Diego, Biomedical Research Facility 2 (BRF2), La Jolla, CA, USA. (B. Schnabl)
| |
Collapse
|
185
|
Kim E, Shin JH, Seok PR, Kim MS, Yoo SH, Kim Y. Phyllodulcin, a natural functional sweetener, improves diabetic metabolic changes by regulating hepatic lipogenesis, inflammation, oxidative stress, fibrosis, and gluconeogenesis in db/db mice. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
186
|
Morrison MC, Kleemann R, van Koppen A, Hanemaaijer R, Verschuren L. Key Inflammatory Processes in Human NASH Are Reflected in Ldlr -/-.Leiden Mice: A Translational Gene Profiling Study. Front Physiol 2018. [PMID: 29527177 PMCID: PMC5829089 DOI: 10.3389/fphys.2018.00132] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Introduction: It is generally accepted that metabolic inflammation in the liver is an important driver of disease progression in NASH and associated matrix remodeling/fibrosis. However, the exact molecular inflammatory mechanisms are poorly defined in human studies. Investigation of key pathogenic mechanisms requires the use of pre-clinical models, for instance for time-resolved studies. Such models must reflect molecular disease processes of importance in patients. Herein we characterized inflammation in NASH patients on the molecular level by transcriptomics and investigated whether key human disease pathways can be recapitulated experimentally in Ldlr−/−.Leiden mice, an established pre-clinical model of NASH. Methods: Human molecular inflammatory processes were defined using a publicly available NASH gene expression profiling dataset (GSE48452) allowing the comparison of biopsy-confirmed NASH patients with normal controls. Gene profiling data from high-fat diet (HFD)-fed Ldlr−/−.Leiden mice (GSE109345) were used for assessment of the translational value of these mice. Results: In human NASH livers, we observed regulation of 65 canonical pathways of which the majority was involved in inflammation (32%), lipid metabolism (16%), and extracellular matrix/remodeling (12%). A similar distribution of pathways across these categories, inflammation (36%), lipid metabolism (24%) and extracellular matrix/remodeling (8%) was observed in HFD-fed Ldlr−/−.Leiden mice. Detailed evaluation of these pathways revealed that a substantial proportion (11 out of 13) of human NASH inflammatory pathways was recapitulated in Ldlr−/−.Leiden mice. Furthermore, the activation state of identified master regulators of inflammation (i.e., specific transcription factors, cytokines, and growth factors) in human NASH was largely reflected in Ldlr−/−.Leiden mice, further substantiating its translational value. Conclusion: Human NASH is characterized by upregulation of specific inflammatory processes (e.g., “Fcγ Receptor-mediated Phagocytosis in Macrophages and Monocytes,” “PI3K signaling in B Lymphocytes”) and master regulators (e.g., TNF, CSF2, TGFB1). The majority of these processes and regulators are modulated in the same direction in Ldlr−/−.Leiden mice fed HFD with a human-like macronutrient composition, thus demonstrating that specific experimental conditions recapitulate human disease on the molecular level of disease pathways and upstream/master regulators.
Collapse
Affiliation(s)
- Martine C Morrison
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands.,Department of Vascular Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Arianne van Koppen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Roeland Hanemaaijer
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| |
Collapse
|
187
|
Li X, Liu L, Li R, Wu A, Lu J, Wu Q, Jia J, Zhao M, Song H. Hepatic loss of Lissencephaly 1 ( Lis1) induces fatty liver and accelerates liver tumorigenesis in mice. J Biol Chem 2018; 293:5160-5171. [PMID: 29475944 PMCID: PMC5892582 DOI: 10.1074/jbc.ra117.001474] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/20/2018] [Indexed: 12/22/2022] Open
Abstract
The liver is a major organ in lipid metabolism, and its malfunction leads to various diseases. Nonalcoholic fatty liver disease, the most common chronic liver disorder in developed countries, is characterized by the abnormal retention of excess lipid within hepatocytes and predisposes individuals to liver cancer. We previously reported that the levels of Lissencephaly 1 (LIS1, also known as PAFAH1B1) are down-regulated in human hepatocellular carcinoma. Following up on this observation, we found that genetic deletion of Lis1 in the mouse liver increases lipid accumulation and inflammation in this organ. Further analysis revealed that loss of Lis1 triggers endoplasmic reticulum (ER) stress and reduces triglyceride secretion. Attenuation of ER stress by addition of tauroursodeoxycholic acid (TUDCA) diminished lipid accumulation in the Lis1-deficient hepatocytes. Moreover, the Golgi stacks were disorganized in Lis1-deficient liver cells. Of note, the Lis1 liver-knockout mice exhibited increased hepatocyte ploidy and accelerated development of liver cancer after exposure to the liver carcinogen diethylnitrosamine (DEN). Taken together, these findings suggest that reduced Lis1 levels can spur the development of liver diseases from steatosis to liver cancer and provide a useful model for delineating the molecular pathways that lead to these diseases.
Collapse
Affiliation(s)
- Xiaoling Li
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China and
| | - Liansheng Liu
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China and
| | - Ran Li
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China and
| | - Ailing Wu
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China and
| | - Jinqiu Lu
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China and
| | - Qingzhe Wu
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China and
| | - Junling Jia
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China and
| | - Mujun Zhao
- the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hai Song
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China and
| |
Collapse
|
188
|
Performance Evaluations on Using Entropy of Ultrasound Log-Compressed Envelope Images for Hepatic Steatosis Assessment: An In Vivo Animal Study. ENTROPY 2018; 20:e20020120. [PMID: 33265211 PMCID: PMC7512613 DOI: 10.3390/e20020120] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/05/2018] [Accepted: 02/09/2018] [Indexed: 12/11/2022]
Abstract
Ultrasound B-mode imaging based on log-compressed envelope data has been widely applied to examine hepatic steatosis. Modeling raw backscattered signals returned from the liver parenchyma by using statistical distributions can provide additional information to assist in hepatic steatosis diagnosis. Since raw data are not always available in modern ultrasound systems, information entropy, which is a widely known nonmodel-based approach, may allow ultrasound backscattering analysis using B-scan for assessing hepatic steatosis. In this study, we explored the feasibility of using ultrasound entropy imaging constructed using log-compressed backscattered envelopes for assessing hepatic steatosis. Different stages of hepatic steatosis were induced in male Wistar rats fed with a methionine- and choline-deficient diet for 0 (i.e., normal control) and 1, 1.5, and 2 weeks (n = 48; 12 rats in each group). In vivo scanning of rat livers was performed using a commercial ultrasound machine (Model 3000, Terason, Burlington, MA, USA) equipped with a 7-MHz linear array transducer (Model 10L5, Terason) for ultrasound B-mode and entropy imaging based on uncompressed (HE image) and log-compressed envelopes (HB image), which were subsequently compared with histopathological examinations. Receiver operating characteristic (ROC) curve analysis and areas under the ROC curves (AUC) were used to assess diagnostic performance levels. The results showed that ultrasound entropy imaging can be used to assess hepatic steatosis. The AUCs obtained from HE imaging for diagnosing different steatosis stages were 0.93 (≥mild), 0.89 (≥moderate), and 0.89 (≥severe), respectively. HB imaging produced AUCs ranging from 0.74 (≥mild) to 0.84 (≥severe) as long as a higher number of bins was used to reconstruct the signal histogram for estimating entropy. The results indicated that entropy use enables ultrasound parametric imaging based on log-compressed envelope signals with great potential for diagnosing hepatic steatosis.
Collapse
|
189
|
Wu J. Utilization of animal models to investigate nonalcoholic steatohepatitis-associated hepatocellular carcinoma. Oncotarget 2018; 7:42762-42776. [PMID: 27072576 PMCID: PMC5173170 DOI: 10.18632/oncotarget.8641] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) comprises a spectrum of liver disorders with fat accumulation from simple fatty liver, nonalcoholic steatohepatitis (NASH), fibrosis/cirrhosis and NAFLD/NASH-associated hepatocellular carcinoma (HCC). NASH is a progressive form of NAFLD and requires medical attention. One of 5-10 NASH patients may progress to end-state liver disease (ESLD or cirrhosis) in 5-10 years; meanwhile, life-threatening complications of ESLD and HCC account for major mortality. An increasing burden of NAFLD in clinics, elucidation of its pathogenesis and progression, and assessment of the efficacy of potential therapeutics demand reliable animal models. Most NASH-associated HCC occurs in cirrhotic subjects; however, HCC does appear in NASH patients without cirrhosis. Lipotoxicity, oxidant stress, insulin resistance, endoplasmic reticulum stress, altered adipokine and lymphokine profiles and gut microbiome changes affect NAFLD progression and constitute key pathobiologic interplays. How these factors promote malignant transformation in a microenvironment of steatotic inflammation and fibrosis/cirrhosis, and lead to development of neoplasms is one of critical questions faced in the hepatology field. The present review summarizes the characteristics of emerging rodent NASH-HCC models, and discusses the challenges in utilizing these models to unveil the mysteries of NASH-associated HCC development.
Collapse
Affiliation(s)
- Jian Wu
- Key Laboratory of Molecular Virology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Institute of Liver Diseases, Fudan University, Shanghai, China
| |
Collapse
|
190
|
Ferroportin Expression in Adipocytes Does Not Contribute to Iron Homeostasis or Metabolic Responses to a High Calorie Diet. Cell Mol Gastroenterol Hepatol 2018; 5:319-331. [PMID: 29552621 PMCID: PMC5852331 DOI: 10.1016/j.jcmgh.2018.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/03/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND & AIMS Iron has an increasingly recognized role in the regulation of adipose tissue function, including the expression of adipokines involved in the pathogenesis of nonalcoholic fatty liver disease. The cellular iron exporter, ferroportin, has been proposed as being a key determinant of adipocyte iron homeostasis. METHODS We studied an adipocyte-specific ferroportin (Fpn1) knockout mouse model, using an Adipoq-Cre recombinase driven Fpn1 deletion and fed mice according to the fast food diet model of nonalcoholic steatohepatitis. RESULTS We showed successful selective deletion of Fpn1 in adipocytes, but found that this did not lead to increased adipocyte iron stores as measured by atomic absorption spectroscopy or histologically quantified iron granules after staining with 3,3'-diaminobenzidine-enhanced Perls' stain. Mice with adipocyte-specific Fpn1 deletion did not show dysregulation of adiponectin, leptin, resistin, or retinol-binding protein-4 expression. Similarly, adipocyte-specific Fpn1 deletion did not affect insulin sensitivity during hyperinsulinemic-euglycemic clamp studies or lead to histologic evidence of increased liver injury. We have shown, however, that the fast food diet model of nonalcoholic steatohepatitis generates an increase in adipose tissue macrophage infiltration with crown-like structures, as seen in human beings, further validating the utility of this model. CONCLUSIONS Ferroportin may not be a key determinant of adipocyte iron homeostasis in this knockout model. Further studies are needed to determine the mechanisms of iron metabolism in adipocytes and adipose tissue.
Collapse
Key Words
- AAS, atomic absorption spectroscopy
- ANOVA, analysis of variance
- AUC, area under the curve
- Adipoq, adiponectin
- Adipose Tissue
- EFP, epididymal fat pad
- FKO, ferroportin knockout
- Ferroportin
- Ferroportin Flox, Fpn1fl/fl
- Fpn1, ferroportin
- HIC, hepatic iron concentration
- Hamp1, hepcidin
- Iron
- NAFLD, nonalcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- Nonalcoholic Fatty Liver Disease
- PCR, polymerase chain reaction
- RBP-4, retinol binding protein-4
- Tfr1, transferrin receptor-1
- bp, base pair
- cDNA, complementary DNA
- mRNA, messenger RNA
Collapse
|
191
|
Teranishi H, Hayashi M, Higa R, Mori K, Miyazawa T, Hino J, Amano Y, Tozawa R, Ida T, Hanada T, Miyazato M, Hanada R, Kangawa K, Nakao K. Role of neuromedin U in accelerating of non-alcoholic steatohepatitis in mice. Peptides 2018; 99:134-141. [PMID: 29017855 DOI: 10.1016/j.peptides.2017.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 12/16/2022]
Abstract
Neuromedin U (NMU), a neuropeptide originally isolated from porcine spinal cord, has multiple physiological functions and is involved in obesity and inflammation. Excessive fat accumulation in the liver leads to non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), which is closely associated with obesity. NAFLD and NASH develop and progress via complex pathophysiological processes, and it remains unclear to what extend the NMU system contributes to the risk of obesity-related disorders such as NAFLD and NASH. Here, we demonstrate that the NMU system plays a role in NAFLD/NASH pathogenesis. In the normal mouse liver, NMU mRNA was not detectable, and expression of the mRNA encoding neuromedin U receptor 1 (NMUR1), the peripheral receptor of NMU, was low. However, the expression of both was significantly increased in the livers of NASH mice. Furthermore, overproduction of NMU induced the mouse liver by hydrodynamic injection, exacerbated NASH pathogenesis. These data indicate a novel role for the peripheral NMU system, providing new insights into the pathogenesis of NAFLD/NASH.
Collapse
Affiliation(s)
- Hitoshi Teranishi
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masafumi Hayashi
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Ryoko Higa
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Cell Biology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kenji Mori
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Takashi Miyazawa
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Yuichiro Amano
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Ryuichi Tozawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takanori Ida
- Interdisciplinary Research Organization, University of Miyazaki, Miyazaki, Japan
| | - Toshikatsu Hanada
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Cell Biology, Faculty of Medicine, Oita University, Oita, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Reiko Hanada
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan.
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Kazuwa Nakao
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
192
|
Jensen VS, Hvid H, Damgaard J, Nygaard H, Ingvorsen C, Wulff EM, Lykkesfeldt J, Fledelius C. Dietary fat stimulates development of NAFLD more potently than dietary fructose in Sprague-Dawley rats. Diabetol Metab Syndr 2018; 10:4. [PMID: 29410708 PMCID: PMC5781341 DOI: 10.1186/s13098-018-0307-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/16/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In humans and animal models, excessive intake of dietary fat, fructose and cholesterol has been linked to the development of non-alcoholic fatty liver disease (NAFLD). However, the individual roles of the dietary components remain unclear. To investigate this further, we compared the effects of a high-fat diet, a high-fructose diet and a combination diet with added cholesterol on the development of NAFLD in rats. METHODS Forty male Sprague-Dawley rats were randomized into four groups receiving either a control-diet (Control: 10% fat); a high-fat diet (HFD: 60% fat, 20% carbohydrate), a high-fructose diet [HFr: 10% fat, 70% carbohydrate (mainly fructose)] or a high-fat/high-fructose/high-cholesterol-diet (NASH: 40% fat, 40% carbohydrate (mainly fructose), 2% cholesterol) for 16 weeks. RESULTS After 16 weeks, liver histology revealed extensive steatosis and inflammation in both NASH- and HFD-fed rats, while hepatic changes in HFr-rats were much more subtle. These findings were corroborated by significantly elevated hepatic triglyceride content in both NASH- (p < 0.01) and HFD-fed rats (p < 0.0001), elevated hepatic cholesterol levels in NASH-fed rats (p < 0.0001), but no changes in HFr-fed rats, compared to Control. On the contrary, only HFr-fed rats developed dyslipidemia as characterized by higher levels of plasma triglycerides compared to all other groups (p < 0.0001). Hepatic dysfunction and inflammation was confirmed in HFD-fed rats by elevated levels of hepatic MCP-1 (p < 0.0001), TNF-alpha (p < 0.001) and plasma β-hydroxybutyrate (p < 0.0001), and in NASH-fed rats by elevated levels of hepatic MCP-1 (p < 0.01), increased hepatic macrophage infiltration (p < 0.001), and higher plasma levels of alanine aminotransferase (p < 0.0001) aspartate aminotransferase (p < 0.05), haptoglobin (p < 0.001) and TIMP-1 (p < 0.01) compared to Control. CONCLUSION These findings show that dietary fat and cholesterol are the primary drivers of NAFLD development and progression in rats, while fructose mostly exerts its effect on the circulating lipid pool.
Collapse
Affiliation(s)
- Victoria Svop Jensen
- Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark
- Insulin Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Henning Hvid
- Insulin Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Jesper Damgaard
- Insulin Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Helle Nygaard
- Insulin Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Camilla Ingvorsen
- Histology and Imaging, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Erik Max Wulff
- Obesity and Diabetes Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Jens Lykkesfeldt
- Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark
| | - Christian Fledelius
- Insulin Pharmacology, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| |
Collapse
|
193
|
In vivo redox metabolic imaging of mitochondria assesses disease progression in non-alcoholic steatohepatitis. Sci Rep 2017; 7:17170. [PMID: 29215054 PMCID: PMC5719423 DOI: 10.1038/s41598-017-17447-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 11/27/2017] [Indexed: 12/19/2022] Open
Abstract
Given the rising incidence of non-alcoholic fatty liver disease (NAFLD) in both adults and children, the development of a non-invasive diagnostic method for assessing disease progression to non-alcoholic steatohepatitis (NASH) has become an important research goal. Currently available non-invasive imaging technologies are only able to assess fat accumulation in the liver. Therefore, these methods are not suitable for a precise diagnosis of NASH. The standard diagnostic technique for NASH, liver biopsy, has several drawbacks, including the higher risk of complications that accompanies invasive procedures. Here, we demonstrated that in vivo mitochondrial redox metabolism was dramatically altered at an early stage, before histopathological changes, and NASH could be accurately diagnosed by in vivo dynamic nuclear polarization-magnetic resonance imaging, with carbamoyl-PROXYL as a molecular imaging probe. In addition, this technique was feasible for the diagnosis of NASH compared with histopathological findings from biopsies. Our data reveal a novel method for monitoring the dynamics of redox metabolic changes in NAFLD/NASH.
Collapse
|
194
|
He W, Xu Y, Zhang C, Lu J, Li J, Xiang D, Yang J, Chang M, Liu D. Hepatoprotective effect of calculus bovis sativus on nonalcoholic fatty liver disease in mice by inhibiting oxidative stress and apoptosis of hepatocytes. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3449-3460. [PMID: 29255346 PMCID: PMC5723121 DOI: 10.2147/dddt.s150187] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Calculus bovis (CB, niu-huang) is a high-class therapeutic drug that is often used in traditional Chinese medicine. CB helps to eliminate heat and toxic components, and prevents the accumulation of phlegm and blood stasis in the liver. In Asian countries, CB Sativus (CBS), an ideal substitute for natural CB, is presently extensively used for long-term treatment of chronic liver diseases. The present study aimed to evaluate the effects and potential mechanism(s) of action of CBS on mice with fructose-induced nonalcoholic fatty liver disease (NAFLD). The NAFLD model was established in C57BL/6 mice by exclusively feeding fluids containing 30% fructose for 8 consecutive weeks. After these 8 weeks, mice were given CBS (50 mg/kg/day or 100 mg/kg/day) for 2 consecutive weeks. Treatment with CBS reversed the fructose-induced impaired glucose tolerance. Compared with the model group, in which mice received 8 weeks of high-fructose diet and 2 weeks of 0.5% sodium carboxymethyl cellulose, CBS treatment significantly decreased the levels of fasting serum glucose, fasting insulin, triglyceride, and total cholesterol, and increased levels of high-density lipoprotein-cholesterol. CBS treatment also significantly decreased the levels of triglyceride, total cholesterol, and free fatty acid in the liver. The activity of superoxide dismutase in the liver was increased after treatment with CBS, however, levels of malondialdehyde and reactive oxygen species decreased. Histopathological examination showed that liver steatosis and injury were significantly reduced in CBS-treated mice. The expression of fatty acid synthase, nuclear factor kappa-light-chain-enhancer of activated B cells, Cysteinyl aspartate-specific proteinase-3, and synonyms B-cell leukemia/lymphoma-2 gene-associated X protein were downregulated after treatment with CBS, whereas the expression of nuclear factor erythroid-2-related factor 2 was upregulated. In conclusion, CBS treatment exerted therapeutic effects in the liver of mice with NAFLD, which may be associated with amelioration of metabolic disorders, enhanced antioxidant effects, and alleviation of apoptosis.
Collapse
Affiliation(s)
- Wenxi He
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanjiao Xu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chengliang Zhang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingli Lu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Juan Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dong Xiang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinyu Yang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mujun Chang
- Center for Translational Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dong Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
195
|
Ejima C, Kuroda H, Ishizaki S. A novel diet-induced murine model of steatohepatitis with fibrosis for screening and evaluation of drug candidates for nonalcoholic steatohepatitis. Physiol Rep 2017; 4:4/21/e13016. [PMID: 27821715 PMCID: PMC5112494 DOI: 10.14814/phy2.13016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/23/2016] [Accepted: 10/03/2016] [Indexed: 12/14/2022] Open
Abstract
Many animal models of nonalcoholic steatohepatitis have been reported. While these models exhibit mild onset of hepatitis and fibrosis, induction is often slow. For faster screening of drug candidates, there is a compelling need for convenient animal models of steatohepatitis and nonalcoholic steatohepatitis in which fatty liver and hepatitis are stably induced within a short period. Here, we analyzed the hepatic lipid composition in nonalcoholic steatohepatitis, and used this information to successfully establish a murine model where steatohepatitis is induced within only 1 week using a novel diet (steatohepatitis‐inducing high‐fat diet, STHD‐01) high in saturated fatty acids and cholesterol. After receiving STHD‐01 for 1 week, normal mice (C57BL/6J) showed elevated markers of fatty liver and hepatitis, including hepatic triglycerides and plasma alanine aminotransferase; the administration of angiotensin receptor blockers reduced these symptoms. Furthermore, we confirmed that STHD‐01 administration for 36 weeks induced not only sustained elevation of hepatic triglyceride and plasma alanine aminotransferase levels, but also fibrosis and tumor formation. Pretreatment with the carcinogen diethylnitrosamine accelerated tumor formation, and hepatic lesions were observed within 30 weeks of STHD‐01 feeding following diethylnitrosamine pretreatment. Finally, branched‐chain amino acids, known to reduce the risk for hepatocellular carcinoma in preclinical models, were effective in reducing the progression of liver fibrosis induced by STHD‐01 feeding after diethylnitrosamine pretreatment. We concluded that STHD‐01 administration successfully induces steatohepatitis within a short period of time. The proposed murine model is suitable for studying the long‐term effects of pharmaceutical agents targeting steatohepatitis, fibrosis, and tumor formation.
Collapse
Affiliation(s)
- Chieko Ejima
- Research Institute, EA Pharma Co. Ltd., Kanagawa, Japan
| | - Haruna Kuroda
- Research Institute, EA Pharma Co. Ltd., Kanagawa, Japan
| | | |
Collapse
|
196
|
van Koppen A, Verschuren L, van den Hoek AM, Verheij J, Morrison MC, Li K, Nagabukuro H, Costessi A, Caspers MP, van den Broek TJ, Sagartz J, Kluft C, Beysen C, Emson C, van Gool AJ, Goldschmeding R, Stoop R, Bobeldijk-Pastorova I, Turner SM, Hanauer G, Hanemaaijer R. Uncovering a Predictive Molecular Signature for the Onset of NASH-Related Fibrosis in a Translational NASH Mouse Model. Cell Mol Gastroenterol Hepatol 2017; 5:83-98.e10. [PMID: 29276754 PMCID: PMC5738456 DOI: 10.1016/j.jcmgh.2017.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/06/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS The incidence of nonalcoholic steatohepatitis (NASH) is increasing. The pathophysiological mechanisms of NASH and the sequence of events leading to hepatic fibrosis are incompletely understood. The aim of this study was to gain insight into the dynamics of key molecular processes involved in NASH and to rank early markers for hepatic fibrosis. METHODS A time-course study in low-density lipoprotein-receptor knockout. Leiden mice on a high-fat diet was performed to identify the temporal dynamics of key processes contributing to NASH and fibrosis. An integrative systems biology approach was used to elucidate candidate markers linked to the active fibrosis process by combining transcriptomics, dynamic proteomics, and histopathology. The translational value of these findings were confirmed using human NASH data sets. RESULTS High-fat-diet feeding resulted in obesity, hyperlipidemia, insulin resistance, and NASH with fibrosis in a time-dependent manner. Temporal dynamics of key molecular processes involved in the development of NASH were identified, including lipid metabolism, inflammation, oxidative stress, and fibrosis. A data-integrative approach enabled identification of the active fibrotic process preceding histopathologic detection using a novel molecular fibrosis signature. Human studies were used to identify overlap of genes and processes and to perform a network biology-based prioritization to rank top candidate markers representing the early manifestation of fibrosis. CONCLUSIONS An early predictive molecular signature was identified that marked the active profibrotic process before histopathologic fibrosis becomes manifest. Early detection of the onset of NASH and fibrosis enables identification of novel blood-based biomarkers to stratify patients at risk, development of new therapeutics, and help shorten (pre)clinical experimental time frames.
Collapse
Key Words
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- DEG, differentially expressed genes
- Diagnosis
- ECM, extracellular matrix
- HFD, high-fat diet
- IPA, Ingenuity Pathway Analysis
- LDLr-/-, low-density lipoprotein receptor knock out
- Liver Disease
- Metabolic Syndrome
- NAFLD, nonalcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- Systems Biology
- THBS1, thrombospontin-1
Collapse
Affiliation(s)
- Arianne van Koppen
- Department of Metabolic Health Research, TNO, Leiden, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, TNO, Zeist, The Netherlands
| | | | - Joanne Verheij
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Kelvin Li
- Kinemed, Inc, Emeryville, California
| | | | | | | | | | | | | | | | | | - Alain J. van Gool
- Department of Microbiology and Systems Biology, TNO, Zeist, The Netherlands
- Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Reinout Stoop
- Department of Metabolic Health Research, TNO, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
197
|
Fuchs CD, Claudel T, Scharnagl H, Stojakovic T, Trauner M. FXR controls CHOP expression in steatohepatitis. FEBS Lett 2017; 591:3360-3368. [PMID: 28895119 PMCID: PMC5698708 DOI: 10.1002/1873-3468.12845] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/24/2017] [Accepted: 09/05/2017] [Indexed: 01/22/2023]
Abstract
The farnesoid X receptor (FXR) and C/EBP homologous protein (CHOP) have critical functions in hepatic lipid metabolism. Here, we aimed to explore a potential relationship between FXR and CHOP. We fed wild‐type (WT) and FXR KO mice a MCD diet (model of steatohepatitis) and found that Chop mRNA expression is upregulated in WT but not FXR KO mice. The absence of FXR aggravates hepatic inflammation after MCD feeding. In HepG2 cells, we found that Chop expression is regulated in a FXR/Retinoid X receptor (RXR)‐dependent manner. We identified a FXR/RXR‐binding site in the human CHOP promoter, demonstrating a highly conserved regulatory pathway. Our study shows that FXR/RXR regulates Chop expression in a mouse model of steatohepatitis, providing novel insights into pathogenesis of this disorder.
Collapse
Affiliation(s)
- Claudia D Fuchs
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| |
Collapse
|
198
|
Kumar R, Mohan S. Non-alcoholic Fatty Liver Disease in Lean Subjects: Characteristics and Implications. J Clin Transl Hepatol 2017; 5:216-223. [PMID: 28936403 PMCID: PMC5606968 DOI: 10.14218/jcth.2016.00068] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/07/2017] [Accepted: 05/15/2017] [Indexed: 12/21/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is commonly diagnosed in obese subjects; however, it is not rare among lean individuals. Given the absence of traditional risk factors, it tends to remain under-recognised. The metabolic profiles of lean NAFLD patients are frequently comparable to those of obese NAFLD patients. Though results from several studies have been mixed, it has been generally revealed that lean subjects with NAFLD have minor insulin resistance compared to that in obese NAFLD. Several genetic variants are associated with NAFLD without insulin resistance. Some data suggest that the prevalence of steatohepatitis and advanced fibrosis do not differ significantly between lean and obese NAFLD; however, the former tend to have less severe disease at presentation. The underlying pathophysiology of lean NAFLD may be quite different. Genetic predispositions, fructose- and cholesterol-rich diet, visceral adiposity and dyslipidaemia have potential roles in the pathogenic underpinnings. Lean NAFLD may pose a risk for metabolic disturbances, cardiovascular morbidity or overall mortality. Secondary causes of hepatic steatosis are also needed to be ruled out in lean subjects with NAFLD. The effectiveness of various treatment modalities, such as exercise and pharmacotherapy, on lean NAFLD is not known. Weight loss is expected to help lean NAFLD patients who have visceral obesity. Further investigation is needed for many aspects of lean NAFLD, including mechanistic pathogenesis, risk assessment, natural history and therapeutic approach.
Collapse
Affiliation(s)
- Ramesh Kumar
- *Correspondence to: Dr Ramesh Kumar, Department of Gastroenterology, Institute of Gastrosciences, Medical Divisions, Paras HMR Institute, Patna 800014, India. Tel: +612-7107700-7852, E-mail:
| | | |
Collapse
|
199
|
Lee SM, Dorotea D, Jung I, Nakabayashi T, Miyata T, Ha H. TM5441, a plasminogen activator inhibitor-1 inhibitor, protects against high fat diet-induced non-alcoholic fatty liver disease. Oncotarget 2017; 8:89746-89760. [PMID: 29163785 PMCID: PMC5685706 DOI: 10.18632/oncotarget.21120] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/03/2017] [Indexed: 12/16/2022] Open
Abstract
Recent evidences showed that elevation of plasminogen activator inhibitor 1 (PAI-1) was responsible in mediating obesity-induced non-alcoholic fatty liver disease (NAFLD) and metabolic disorders. Here, we investigated the effect of TM5441, an oral PAI-1 inhibitor that lacks of bleeding risk, on high-fat diet (HFD)-induced NAFLD. HFD-fed C57BL/6J mice was daily treated with 20 mg/kg TM5441. To examine the preventive effect, 10-week-treatment was started along with initiation of HFD; alternatively, 4-week-treatment was started in mice with glucose intolerance in the interventional strategy. In vivo study showed that early and delayed treatment decreased hepatic steatosis. Particularly, early treatment prevented the progression of hepatic inflammation and fibrosis in HFD mice. Interestingly, both strategies abrogated hepatic insulin resistance and mitochondrial dysfunction, presented by enhanced p-Akt and p-GSK3β, reduced p-JNK signaling, along with p-AMPK and PGC-1α activation. Consistently, TM5441 treatment in the presence of either PAI-1 exposure or TNF-α stimulated-PAI-1 activity showed a restoration of mitochondrial biogenesis related genes expression on HepG2 cells. Thus, improvement of insulin sensitivity and mitochondrial function was imperative to partially explain the therapeutic effects of TM5441, a novel agent targeting HFD-induced NAFLD.
Collapse
Affiliation(s)
- Seon Myeong Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Debra Dorotea
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Inji Jung
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Tetsuo Nakabayashi
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
200
|
Abstract
Diet has become one of the top risk factors for poor health. The incidence of cardiometabolic disease in the United Sates, in Mexico, and in most countries is driven fundamentally by changes in diet quality. Weight gain has been typically framed as a problem of excess caloric intake, but, as reviewed in this paper, subtle changes in the quality of diet are associated with long-term weight gain. In order to successfully address obesity and diabetes, researchers and policy makers have to better understand how weight gain in the long term is modulated and to change the focus of research and public policy from one based on counting calories to one based on diet quality and its determinants at various levels.
Collapse
Affiliation(s)
- Dariush Mozaffarian
- D. Mozaffarian is with the Friedman School of Nutrition Science & Policy, Tufts University, Boston, Massachusetts, USA.
| |
Collapse
|