1
|
Ma S, Li X, Wu C, Wulayin K, Li M, Zhou L, Lin S, Hu Z, Tuerxun M, Lin B, Chen L. The potential value of fatty acid binding protein 1 in Chronic HBV-related liver disease progression assessment. BMC Infect Dis 2024; 24:1214. [PMID: 39468469 PMCID: PMC11520826 DOI: 10.1186/s12879-024-10114-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/23/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Fatty acid binding protein 1 (FABP1), a low molecular weight intracellular protein, has been proposed as a potential useful serum biomarker for liver injury. However, limited investigations have been conducted in chronic hepatitis B virus (HBV)-related liver disease. OBJECTIVE To investigate the diagnostic potential of FABP1 in disease progression among patients with chronic HBV-related liver disease. METHODS A prospective study was conducted on 293 patients with chronic HBV-related liver diseases, including chronic asymptomatic carrier (ASC), chronic hepatitis B (CHB). FABP1 was measured in serum samples collected at admission and some selected liver biopsies. RESULTS Immunohistochemical analysis revealed abundant cytoplasmic expression of FABP1 in hepatocytes. A significant negative correlation was observed between FABP1 expression and inflammation grades in liver tissue (Spearman's r = -0.355, P = 0.017). However, no statistically significant correlation was found with fibrosis (P > 0.05). Serum FABP1 levels in the case group were significantly higher than in the healthy control (HC) group [median: 804.2 (687.8, 939.2) vs. 709.1 (626.2, 807.8) ng/ml, Z = -5.505, P < 0.001] and showed correlations with alanine aminotransferase (ALT), aspartate aminotransferase (AST); total bilirubin (TBIL); direct bilirubin (DBIL); albumin (ALB), etc. Its levels progressively increased with the advancement from ASC to CHB, with significant differences compared to the HC group (P < 0.001), especially in ASC patients with high HBV DNA (exceeding 106 IU/ml, P = 0.019), HBeAg positive (P = 0.013) and ALT higher than 0.5 times upper limit of normal (ULN)(P = 0.035). Meanwhile, serum FABP1 in CHB patients with higher TBIL(P = 0.005) or the severe CHB were higher (P = 0.002). CONCLUSION Our study demonstrated a significant inverse correlation between FABP1 levels and the severity of inflammation grades in patients with HBV-related liver diseases. Furthermore, elevated serum FABP1 levels were observed in these patients, suggesting its potential as a biomarker for assessing HBV-related liver damage to initiate antiviral therapy. Additionally, further evaluation is required to determine its potential as a biomarker for assessing disease severity.
Collapse
Affiliation(s)
- Shasha Ma
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
- Division of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China
| | - Xiaoyan Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Chao Wu
- Department of Infectious Diseases, The First People's Hospital of Kashi (Kashgar) Prefecture, Xinjiang, 844000, People's Republic of China
- National Regional Medical Center for Infectious Diseases, Kashi Hospital Affiliated to Sun Yat-Sen University, Xinjiang, 844000, People's Republic of China
| | - Kuerbannisa Wulayin
- Department of Infectious Diseases, The First People's Hospital of Kashi (Kashgar) Prefecture, Xinjiang, 844000, People's Republic of China
- National Regional Medical Center for Infectious Diseases, Kashi Hospital Affiliated to Sun Yat-Sen University, Xinjiang, 844000, People's Republic of China
| | - Mingna Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Lian Zhou
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Shutao Lin
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Zhaoxia Hu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Maimaitiaili Tuerxun
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China.
- National Regional Medical Center for Infectious Diseases, Kashi Hospital Affiliated to Sun Yat-Sen University, Xinjiang, 844000, People's Republic of China.
| | - Bingliang Lin
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China.
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, Guangdong, 510080, People's Republic of China.
| | - Lubiao Chen
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China.
| |
Collapse
|
2
|
Chen L, Wang B, Li H, Mao J, Liang Z, Chen Y, Yu M, Liu Y, Liao Z, Yang Y, Wu X, Wang H, Yang Y, Xiang R, Zhang L, Li Z. Design, synthesis, and biological evaluation of novel highly selective non-carboxylic acid FABP1 inhibitors. Eur J Med Chem 2024; 276:116705. [PMID: 39067439 DOI: 10.1016/j.ejmech.2024.116705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Histologic spectrum studies in patients revealed fatty acid binding proteins 1 (FABP1) as a potential new target for the treatment of metabolic associated fatty liver disease. However, there is no FABP1 inhibitor has been reported except the first-in-class FABP1 inhibitor bearing acid moiety reported by our laboratory. Herein, we firstly report the structure-activity relationship of novel non-carboxylic acid FABP1 inhibitors, which resulted in the identification of the potent and selective FABP1 inhibitor 30. The IC50 value of compound 30 for subtype FABP4 in the same family was greater than 80 μM. Moreover, compound 30 significantly alleviated the hepatic steatosis in DIO mice, which is equivalent to that of clinical drug obeticholic acid. This study might be provided a promising probe for the development of FABP1 inhibitors and thus can help to further elucidate the pharmacology of FABP1.
Collapse
Affiliation(s)
- Lianru Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Bin Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Hongming Li
- Fujian Provincial Key Laboratory of Hepatic Drug Research, Ningde, 355300, PR China
| | - Jianming Mao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Zhiling Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Ya Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Mingyang Yu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yuxia Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Zibin Liao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yuanqian Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiaojing Wu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Huazheng Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yonghong Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Ruojing Xiang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Luyong Zhang
- Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, PR China.
| |
Collapse
|
3
|
Chen Y, Yu M, Chen L, Mao J, Wang W, Yang Z, Cao Z, Liu Y, Wei M, Zhang L, Li Z. Design, synthesis, and biological evaluation of first-in-class FABP1 inhibitors for the treatment of NASH. Eur J Med Chem 2024; 270:116358. [PMID: 38574638 DOI: 10.1016/j.ejmech.2024.116358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024]
Abstract
The fatty acid-binding protein 1 (FABP1) is a fatty acid transporter protein that is considered as an emerging target for metabolic diseases. Despite forceful evidence that the inhibition of FABP1 is essential for ameliorating NASH, pharmacological control and validation of FABP1 are hindered by a lack of relevant inhibitors as pharmacological tool. Therefore, the development of effective FABP1 inhibitors is a current focus of research. Herein, we firstly reported the comprehensive structure-activity relationship (SAR) study of novel FABP1 inhibitors derived from high throughput screening of our in-house library, which resulting in the identification of the optimal compound 44 (IC50 = 4.46 ± 0.54 μM). Molecular docking studies revealed that 44 forms stable hydrogen bonds with amino acids around the active pocket of FABP1. Moreover, 44 alleviated the typical histological features of fatty liver in NASH mice, including steatosis, lobular inflammation, ballooning and fibrosis. Additionally, 44 has been demonstrated to have lipid metabolism regulating, anti-oxidative stress and hepatoprotective properties. This study might be provided a promising insight into the field of NASH and inspiration for the development of FABP1 inhibitors.
Collapse
Affiliation(s)
- Ya Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Mingyang Yu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Lianru Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jianming Mao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Wenxin Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Zhongcheng Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Zhijun Cao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yuxia Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Min Wei
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Luyong Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, PR China.
| |
Collapse
|
4
|
Yin X, Dong L, Wang X, Qin Z, Ma Y, Ke X, Li Y, Wang Q, Mi Y, Lyu Q, Xu X, Zheng P, Tang Y. Perilipin 5 regulates hepatic stellate cell activation and high-fat diet-induced non-alcoholic fatty liver disease. Animal Model Exp Med 2024; 7:166-178. [PMID: 37202925 PMCID: PMC11079159 DOI: 10.1002/ame2.12327] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/21/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases globally. Hepatic stellate cells (HSCs) are the major effector cells of liver fibrosis. HSCs contain abundant lipid droplets (LDs) in their cytoplasm during quiescence. Perilipin 5 (PLIN 5) is a LD surface-associated protein that plays a crucial role in lipid homeostasis. However, little is known about the role of PLIN 5 in HSC activation. METHODS PLIN 5 was overexpressed in HSCs of Sprague-Dawley rats by lentivirus transfection. At the same time, PLIN 5 gene knockout mice were constructed and fed with a high-fat diet (HFD) for 20 weeks to study the role of PLIN 5 in NAFLD. The corresponding reagent kits were used to measure TG, GSH, Caspase 3 activity, ATP level, and mitochondrial DNA copy number. Metabolomic analysis of mice liver tissue metabolism was performed based on UPLC-MS/MS. AMPK, mitochondrial function, cell proliferation, and apoptosis-related genes and proteins were detected by western blotting and qPCR. RESULTS Overexpression of PLIN 5 in activated HSCs led to a decrease in ATP levels in mitochondria, inhibition of cell proliferation, and a significant increase in cell apoptosis through AMPK activation. In addition, compared with the HFD-fed C57BL/6J mice, PLIN 5 knockout mice fed with HFD showed reduced liver fat deposition, decreased LD abundance and size, and reduced liver fibrosis. CONCLUSION These findings highlight the unique regulatory role of PLIN 5 in HSCs and the role of PLIN 5 in the fibrosis process of NAFLD.
Collapse
Affiliation(s)
- Xuecui Yin
- Department of Internal Medicinethe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Lin Dong
- Department of Pediatricsthe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xiaohan Wang
- Department of Pediatricsthe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenzhen Qin
- Department of Internal Medicinethe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuying Ma
- Department of Internal Medicinethe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xiaofei Ke
- Department of Pediatricsthe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ya Li
- Department of Internal Medicinethe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Qingde Wang
- Department of Internal Medicinethe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yang Mi
- Department of Internal Medicinethe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Quanjun Lyu
- Department of Clinical Nutritionthe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xia Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Co‐innovation Center of Henan Province for New drug R & D and Preclinical Safety, School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Pengyuan Zheng
- Department of Internal Medicinethe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Youcai Tang
- Department of Internal Medicinethe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Department of Pediatrics, Gastroenterology, Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury and Henan Provincial Outstanding Overseas Scientists Chronic Liver Injury Studiothe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
5
|
Kaesler N, Cheng M, Nagai J, O’Sullivan J, Peisker F, Bindels EM, Babler A, Moellmann J, Droste P, Franciosa G, Dugourd A, Saez-Rodriguez J, Neuss S, Lehrke M, Boor P, Goettsch C, Olsen JV, Speer T, Lu TS, Lim K, Floege J, Denby L, Costa I, Kramann R. Mapping cardiac remodeling in chronic kidney disease. SCIENCE ADVANCES 2023; 9:eadj4846. [PMID: 38000021 PMCID: PMC10672229 DOI: 10.1126/sciadv.adj4846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023]
Abstract
Patients with advanced chronic kidney disease (CKD) mostly die from sudden cardiac death and recurrent heart failure. The mechanisms of cardiac remodeling are largely unclear. To dissect molecular and cellular mechanisms of cardiac remodeling in CKD in an unbiased fashion, we performed left ventricular single-nuclear RNA sequencing in two mouse models of CKD. Our data showed a hypertrophic response trajectory of cardiomyocytes with stress signaling and metabolic changes driven by soluble uremia-related factors. We mapped fibroblast to myofibroblast differentiation in this process and identified notable changes in the cardiac vasculature, suggesting inflammation and dysfunction. An integrated analysis of cardiac cellular responses to uremic toxins pointed toward endothelin-1 and methylglyoxal being involved in capillary dysfunction and TNFα driving cardiomyocyte hypertrophy in CKD, which was validated in vitro and in vivo. TNFα inhibition in vivo ameliorated the cardiac phenotype in CKD. Thus, interventional approaches directed against uremic toxins, such as TNFα, hold promise to ameliorate cardiac remodeling in CKD.
Collapse
Affiliation(s)
- Nadine Kaesler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Mingbo Cheng
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James Nagai
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James O’Sullivan
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Fabian Peisker
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Eric M. J. Bindels
- Department of Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne Babler
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Patrick Droste
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Giulia Franciosa
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Aurelien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Sabine Neuss
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Thimoteus Speer
- Department of Medicine (Nephrology), Goethe University Frankfurt, Frankfurt, Germany
| | - Tzong-Shi Lu
- Brigham and Women’s Hospital, Renal Division, Boston, MA, USA
| | - Kenneth Lim
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jürgen Floege
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Laura Denby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ivan Costa
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
6
|
Fu Z, Yin H, Liu J, He Y, Song S, Peng X, Huang X, Lai Y, Li S, Luo Q, Su J, Yang P. Therapeutic effects of fatty acid binding protein 1 in mice with pulmonary fibrosis by regulating alveolar epithelial regeneration. BMJ Open Respir Res 2023; 10:e001568. [PMID: 37940355 PMCID: PMC10632910 DOI: 10.1136/bmjresp-2022-001568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 10/20/2023] [Indexed: 11/10/2023] Open
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis is a progressive fibrotic lung disease with limited therapeutic options and high lethality, related to alveolar type II epithelial (ATII) cell dysregulation, the abnormal repair of alveolar epithelial cells and activation of fibroblasts promote the development of pulmonary fibrosis. Fatty acid binding protein 1 (FABP1) was significantly downregulated in the fibrotic state by proteomics screening in our previous date, and the ATII cell dysregulation can be mediated by FABP1 via regulating fatty acid metabolism and intracellular transport. The aim of this study was to evaluate the role and potential mechanism of FABP1 in the development of pulmonary fibrosis. METHODS Proteomics screening was used to detect changes of the protein profiles in two different types (induced by bleomycin and silica, respectively) of pulmonary fibrosis models. The localisation of FABP1 in mouse lung was detected by Immunofluorescence and immunohistochemistry. Experimental methods such as lung pathology, micro-CT, western blotting, small animal imaging in vivo, EdU, etc were used to verify the role of FABP1 in pulmonary fibrosis. RESULTS The expression of FABP1 in the mouse lung was significantly reduced in the model of pulmonary fibrosis from our proteomic analysis and immunological methods, the double immunofluorescence staining showed that FABP1 was mainly localised in type II alveolar epithelial cells. Additionally, the expression of FABP1 was negatively correlated with the progression of pulmonary fibrosis. Further in vivo and in vitro experiments showed that overexpression of FABP1 alleviated pulmonary fibrosis by protecting alveolar epithelium from injury and promoting cell survival. CONCLUSION Our findings provide a proof-of-principle that FABP1 may represent an effective treatment for pulmonary fibrosis by regulating alveolar epithelial regeneration, which may be associated with the fatty acid metabolism in ATII cells.
Collapse
Affiliation(s)
- Zhenli Fu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hang Yin
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiani Liu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying He
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shengren Song
- Department of Respiratory Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiaomin Peng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xihui Huang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yunxin Lai
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuang Li
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qun Luo
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Su
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Penghui Yang
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
You H, Wen X, Wang X, Zhu C, Chen H, Bu L, Zhang J, Qu S. Derlin-1 ameliorates nonalcoholic hepatic steatosis by promoting ubiquitylation and degradation of FABP1. Free Radic Biol Med 2023; 207:260-271. [PMID: 37499886 DOI: 10.1016/j.freeradbiomed.2023.07.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND AND AIMS The functions of liver fatty acid binding protein 1 (FABP1) in the regulation of nonalcoholic fatty liver disease (NAFLD) have been previously established. However, how FABP1 expression is dynamically regulated in metabolic disorders is unclear. Previous studies have reported that ubiquitin proteasome-mediated degradation of FABP1 is involved, but the mechanism remains unknown. METHODS Dysregulated expression of hepatic FABP1 and Derlin-1 was observed in NAFLD patients. We performed mice hepatic tissue coimmunoprecipitation based mass spectrum assays. Interaction between Derlin-1 and FABP1, and its impact on FABP1 ubiquitination status was evaluated by coimmunoprecipitation. The role of Derlin-1 in lipid deposition was tested using adenovirus-mediated overexpression in C57BL/6 mice, as well as by Derlin-1 overexpression or knockdown in HepG2 cells. RESULTS As a subunit of the endoplasmic reticulum-associated degradation complex, Derlin-1 was negatively associated with NAFLD patients, interacted with and ubiquitinated FABP1. Derlin-1 suppressed FABP1 levels and inhibited lipid deposition through a FABP1-dependent pathway. Additionally, Trim25, an E3 ubiquitin ligase present in the endoplasmic reticulum, was recruited to promote Derlin-1-related polyubiquitylation of FABP1, thereby creating a ubiquitin-associated network for FABP1 regulation. Derlin-1 overexpression ameliorated hepatic steatosis in both C57BL/6 mice and HepG2 cells, and contributed to attenuated weight gain, lower liver weight, and visceral fat mass. CONCLUSIONS FABP1 was degraded by Derlin-1 through ubiquitin modification. Negative regulation of FABP1 by Derlin-1 overexpression, suppressed lipid metabolism and alleviated lipid deposition in vivo and in vitro. Hence, Derlin-1 activation in hepatocytes may represent a potential therapeutic strategy for NAFLD.
Collapse
Affiliation(s)
- Hui You
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China; Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Xin Wen
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Xingchun Wang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China; Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Cuiling Zhu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Haibing Chen
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Le Bu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China.
| | - Jun Zhang
- Research Center for Translational Medicine at East Hospital, Tongji University School of Medicine, Tongji University, Shanghai, 200092, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200072, PR China.
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China; Shanghai Center of Thyroid Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China.
| |
Collapse
|
8
|
Yin KL, Li M, Song PP, Duan YX, Ye WT, Tang W, Kokudo N, Gao Q, Liao R. Unraveling the Emerging Niche Role of Hepatic Stellate Cell-derived Exosomes in Liver Diseases. J Clin Transl Hepatol 2023; 11:441-451. [PMID: 36643031 PMCID: PMC9817040 DOI: 10.14218/jcth.2022.00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 01/18/2023] Open
Abstract
Hepatic stellate cells (HSCs) play an essential role in various liver diseases, and exosomes are critical mediators of intercellular communication in local and distant microenvironments. Cellular crosstalk between HSCs and surrounding multiple tissue-resident cells promotes or inhibits the activation of HSCs. Substantial evidence has revealed that HSC-derived exosomes are involved in the occurrence and development of liver diseases through the regulation of retinoid metabolism, lipid metabolism, glucose metabolism, protein metabolism, and mitochondrial metabolism. HSC-derived exosomes are underpinned by vehicle molecules, such as mRNAs and microRNAs, that function in, and significantly affect, the processes of various liver diseases, such as acute liver injury, alcoholic liver disease, nonalcoholic fatty liver disease, viral hepatitis, fibrosis, and cancer. As such, numerous exosomes derived from HSCs or HSC-associated exosomes have attracted attention because of their biological roles and translational applications as potential targets for therapeutic targets. Herein, we review the pathophysiological and metabolic processes associated with HSC-derived exosomes, their roles in various liver diseases and their potential clinical application.
Collapse
Affiliation(s)
- Kun-Li Yin
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Li
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Pei-Pei Song
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Yu-Xin Duan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen-Tao Ye
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Tang
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kokudo
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
- Correspondence to: Qiang Gao, Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, 180 Fenglin Road, Shanghai 200032, China. ORCID: https://orcid.org/0000-0002-6695-9906. ; Rui Liao, Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China. ORCID: https://orcid.org/0000-0002-0057-2792. E-mail:
| | - Rui Liao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Correspondence to: Qiang Gao, Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, 180 Fenglin Road, Shanghai 200032, China. ORCID: https://orcid.org/0000-0002-6695-9906. ; Rui Liao, Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China. ORCID: https://orcid.org/0000-0002-0057-2792. E-mail:
| |
Collapse
|
9
|
Inci MK, Park SH, Helsley RN, Attia SL, Softic S. Fructose impairs fat oxidation: Implications for the mechanism of western diet-induced NAFLD. J Nutr Biochem 2023; 114:109224. [PMID: 36403701 PMCID: PMC11042502 DOI: 10.1016/j.jnutbio.2022.109224] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/29/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022]
Abstract
Increased fructose intake from sugar-sweetened beverages and highly processed sweets is a well-recognized risk factor for the development of obesity and its complications. Fructose strongly supports lipogenesis on a normal chow diet by providing both, a substrate for lipid synthesis and activation of lipogenic transcription factors. However, the negative health consequences of dietary sugar are best observed with the concomitant intake of a HFD. Indeed, the most commonly used obesogenic research diets, such as "Western diet", contain both fructose and a high amount of fat. In spite of its common use, how the combined intake of fructose and fat synergistically supports development of metabolic complications is not fully elucidated. Here we present the preponderance of evidence that fructose consumption decreases oxidation of dietary fat in human and animal studies. We provide a detailed review of the mitochondrial β-oxidation pathway. Fructose affects hepatic activation of fatty acyl-CoAs, decreases acylcarnitine production and impairs the carnitine shuttle. Mechanistically, fructose suppresses transcriptional activity of PPARα and its target CPT1α, the rate limiting enzyme of acylcarnitine production. These effects of fructose may be, in part, mediated by protein acetylation. Acetylation of PGC1α, a co-activator of PPARα and acetylation of CPT1α, in part, account for fructose-impaired acylcarnitine production. Interestingly, metabolic effects of fructose in the liver can be largely overcome by carnitine supplementation. In summary, fructose decreases oxidation of dietary fat in the liver, in part, by impairing acylcarnitine production, offering one explanation for the synergistic effects of these nutrients on the development of metabolic complications, such as NAFLD.
Collapse
Affiliation(s)
| | - Se-Hyung Park
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Robert N Helsley
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA; Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Suzanna L Attia
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Samir Softic
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA; Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Nie K, Gao Y, Chen S, Wang Z, Wang H, Tang Y, Su H, Lu F, Dong H, Fang K. Diosgenin attenuates non-alcoholic fatty liver disease in type 2 diabetes through regulating SIRT6-related fatty acid uptake. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154661. [PMID: 36682299 DOI: 10.1016/j.phymed.2023.154661] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/01/2023] [Accepted: 01/09/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND More than 70% of patients with type 2 diabetes (T2DM) concomitantly suffer from Non-alcoholic fatty liver disease (NAFLD), and the coexistence and interaction of them increases the intractability of NAFLD. With the protective effect against hepatic steatosis and liver fibrosis, SIRT6 is becoming a notable target of NAFLD. Diosgenin, an active monomer from Chinese herbs, has been reported to protect against NAFLD. PURPOSE This study aims to figure out the mechanism how diosgenin alleviate NAFLD in T2DM and the relationship with SIRT6. METHODS In vivo studies used spontaneous diabetic db/db mice and divided them into two parts. The first part included four groups consisting of control (Con) group, model (Mod) group, low dose of diosgenin (DL) group and high dose of diosgenin (DH) group. The second part included four groups consisting of Con group, Mod group, DH+OSS (OSS_128167, inhibitor of SIRT6) group, MDL (MDL800, agonist of SIRT6) group. HepG2 cell line was selected in study in vitro, which was mainly composed of six groups including Con group, palmitic acid (PA) group, PA+DL group, PA+DH group, PA+DH+OSS group, PA+MDL group. OGTT, Biochemical biomarker (including TG, TC, AST, ALT), inflammatory biomarker (including IL-6 and TNF-α) were measured. HE, Oil Red O, and DHE staining were conducted. Immunohistochemistry, immunofluorescence, mRNA-seq, and qPCR were used to explore the mechanism. RESULTS Results in the first part of study in vivo indicated that diosgenin protected against lipid accumulation, oxidative stress, cell injury, and light inflammatory of liver in db/db mice and regulated the expression of SIRT6 and fatty acid transporter including CD36, FATP2, FABP1. The effect of diosgenin could be reversed in DH+OSS group and the same effect was observed in MDL group in the second part of study in vivo. The same results were also noted in followed study in vitro. Diosgenin inhibited the fatty acids uptake and regulated the expression of SIRT6 and fatty acid transporter including CD36, FATP2, and FABP1 in PA-induced hepG2 cells, and which was reversed in DH+OSS group and resembled in MDL group. CONCLUSIONS Diosgenin could attenuate non-alcoholic fatty liver disease in type 2 diabetes through regulating SIRT6-related fatty acid uptake.
Collapse
Affiliation(s)
- Kexin Nie
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yang Gao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shen Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhi Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hongzhan Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yueheng Tang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hao Su
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Fuer Lu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Ke Fang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
11
|
Liu H, Hallauer Hastings M, Kitchen R, Xiao C, Baldovino Guerra JR, Kuznetsov A, Rosenzweig A. Beneficial Effects of Moderate Hepatic Activin A Expression on Metabolic Pathways, Inflammation, and Atherosclerosis. Arterioscler Thromb Vasc Biol 2023; 43:330-349. [PMID: 36453275 DOI: 10.1161/atvbaha.122.318138] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Atherosclerosis is an inflammatory vascular disease marked by hyperlipidemia and hematopoietic stem cell expansion. Activin A, a member of the Activin/GDF/TGFβ/BMP (growth/differentiation factor/transforming growth factor beta/bone morphogenetic protein) family is broadly expressed and increases in human atherosclerosis, but its functional effects in vivo in this context remain unclear. METHODS We studied LDLR-/- mice on a Western diet for 12 weeks and used adeno-associated viral vectors with a liver-specific TBG (thyroxine-binding globulin) promoter to express Activin A or GFP (control). Atherosclerotic lesions were analyzed by oil red staining. Blood lipid profiling was performed by high-performance liquid chromatography, and immune cells were evaluated by flow cytometry. Liver RNA-sequencing was performed to explore the underlying mechanisms. RESULTS Activin A expression decreased in both livers and aortae from LDLR-/- mice fed a Western diet compared with standard laboratory diet. Adenoassociated virus-TBG-Activin A increased Activin A hepatic expression ≈10-fold at 12 weeks; P<0.001) and circulating Activin A levels ≈2000 pg/ml versus ≈50 pg/ml; P<0.001, compared with controls). Hepatic Activin A expression decreased plasma total and LDL (low-density lipoprotein) cholesterol ≈60% and ≈40%, respectively), reduced inflammatory cells in aortae and proliferating hematopoietic stem cells in bone marrow, and reduced atherosclerotic lesion and necrotic core area in aortae. Activin A also attenuated liver steatosis and expression of the lipogenesis genes, Srebp1 and Srebp2. RNA sequencing revealed Activin A not only blocked expression of genes involved in hepatic de novo lipogenesis but also fatty acid uptake and liver inflammation. In addition, Activin A expressed in the liver also reduced white fat tissue accumulation, decreased adipocyte size, and improved glucose tolerance. CONCLUSIONS Our studies reveal hepatic Activin A expression reduces inflammation, hematopoietic stem cell expansion, liver steatosis, circulating cholesterol, and fat accumulation, which likely all contribute to the observed protection against atherosclerosis. The reduced Activin A observed in LDLR-/- mice on a Western diet seems maladaptive and deleterious for atherogenesis.
Collapse
Affiliation(s)
- Huan Liu
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | | | - Robert Kitchen
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | - Chunyang Xiao
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | | | - Alexandra Kuznetsov
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| | - Anthony Rosenzweig
- Cardiovascular Research Center, Massachusetts General Hospital, and Harvard Medical School, Boston
| |
Collapse
|
12
|
Diethyldithiocarbamate inhibits the activation of hepatic stellate cells via PPARα/FABP1 in mice with non-alcoholic steatohepatitis. Biochem Biophys Res Commun 2023; 641:192-199. [PMID: 36535078 DOI: 10.1016/j.bbrc.2022.12.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Activation of hepatic stellate cells (HSCs) is the main course of liver fibrosis which is positively correlated with adverse clinical outcomes in non-alcoholic steatohepatitis (NASH). Diethyldithiocarbamate (DDC) attenuates NASH related liver fibrosis in mice, but its underlying mechanisms remains unclear. In this study, the data showed that DDC inhibited the activation of HSCs in high fat choline-deficient, L-amino acid-defined (CDAA) diet induced NASH. Double Immunofluorescence analysis showed that the baseline expression of peroxisome proliferator-activated receptor α (PPARα) is high in HSCs in normal mouse liver and notably decreases in the NASH liver, indicating that PPARα might be associated with the activation of HSCs. While, DDC upregulated PPARα in HSCs in the NASH liver. Mixture of free fatty acid was used to induce steatosis of hepatocytes. Human HSCs (LX-2 cells) were activated after co-cultured with steatotic hepatocytes, and DDC inhibited the activation of LX-2 cells. Meanwhile, DDC upregulated PPARα and FABP1, and promoted the accumulation of LDs in LX-2 cells. PPARα small interfering RNA blocked these effect of DDC. These findings suggest that PPARα is associated with the activation of HSCs in the context of NASH. DDC improves NASH related fibrosis through inhibiting the activation of HSCs via PPARα/FABP1.
Collapse
|
13
|
Yan T, Luo Y, Yan N, Hamada K, Zhao N, Xia Y, Wang P, Zhao C, Qi D, Yang S, Sun L, Cai J, Wang Q, Jiang C, Gavrilova O, Krausz KW, Patel DP, Yu X, Wu X, Hao H, Liu W, Qu A, Gonzalez FJ. Intestinal peroxisome proliferator-activated receptor α-fatty acid-binding protein 1 axis modulates nonalcoholic steatohepatitis. Hepatology 2023; 77:239-255. [PMID: 35460276 PMCID: PMC9970020 DOI: 10.1002/hep.32538] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/08/2022] [Accepted: 04/20/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND AIMS Peroxisome proliferator-activated receptor α (PPARα) regulates fatty acid transport and catabolism in liver. However, the role of intestinal PPARα in lipid homeostasis is largely unknown. Here, intestinal PPARα was examined for its modulation of obesity and NASH. APPROACH AND RESULTS Intestinal PPARα was activated and fatty acid-binding protein 1 (FABP1) up-regulated in humans with obesity and high-fat diet (HFD)-fed mice as revealed by using human intestine specimens or HFD/high-fat, high-cholesterol, and high-fructose diet (HFCFD)-fed C57BL/6N mice and PPARA -humanized, peroxisome proliferator response element-luciferase mice. Intestine-specific Ppara or Fabp1 disruption in mice fed a HFD or HFCFD decreased obesity-associated metabolic disorders and NASH. Molecular analyses by luciferase reporter assays and chromatin immunoprecipitation assays in combination with fatty acid uptake assays in primary intestinal organoids revealed that intestinal PPARα induced the expression of FABP1 that in turn mediated the effects of intestinal PPARα in modulating fatty acid uptake. The PPARα antagonist GW6471 improved obesity and NASH, dependent on intestinal PPARα or FABP1. Double-knockout ( Ppara/Fabp1ΔIE ) mice demonstrated that intestinal Ppara disruption failed to further decrease obesity and NASH in the absence of intestinal FABP1. Translationally, GW6471 reduced human PPARA-driven intestinal fatty acid uptake and improved obesity-related metabolic dysfunctions in PPARA -humanized, but not Ppara -null, mice. CONCLUSIONS Intestinal PPARα signaling promotes NASH progression through regulating dietary fatty acid uptake through modulation of FABP1, which provides a compelling therapeutic target for NASH treatment.
Collapse
Affiliation(s)
- Tingting Yan
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yuhong Luo
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nana Yan
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, P.R. China
| | - Keisuke Hamada
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nan Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, Beijing, P.R. China
| | - Yangliu Xia
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ping Wang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Changdong Zhao
- Department of Gastroenterology, Second People’s Hospital of Lianyungang City, Lianyungang, P.R. China
| | - Dan Qi
- Department of Pathology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Shoumei Yang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Lulu Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jie Cai
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Qiong Wang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P.R. China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P.R. China
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kristopher W. Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daxesh P. Patel
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiaoting Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, Beijing, P.R. China
| | - Xuan Wu
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, P.R. China
- Department of Laboratory Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai, P.R. China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, P.R. China
| | - Weiwei Liu
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, P.R. China
- Department of Laboratory Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai, P.R. China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Key Laboratory of Remodeling‐Related Cardiovascular Diseases, Ministry of Education, Beijing, P.R. China
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
14
|
Wang M, Su Y, Hou C, Ren K, Liu X, Zhao S, Wang Y, Liu X. Targeted Lipidomics Analysis of Lysine 179 Acetylation of ACSF2 in Rat Hepatic Stellate Cells. Prostaglandins Other Lipid Mediat 2022; 163:106671. [PMID: 36028068 DOI: 10.1016/j.prostaglandins.2022.106671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022]
Abstract
Activation of hepatic stellate cells (HSCs) is generally recognized as a central driver of liver fibrosis. Metabolism of fatty acids (FA) play a critical role in the activation of HSCs. Proteomics analysis on lysine acetylation of proteins in activated HSCs in our previous study indicated that acetylation of the lysine residues on ACSF2 is one of the most significantly upregulated sites in activated-HSCs and K179 is its important acetylation site. However, the role of acetylation at K179 of ACSF2 on activation of HSCs and free fatty acids (FFA) metabolism remains largely unknown. The reported study demonstrates that acetylation at K179 of ACSF2 promoted HSCs activation. The targeted lipidomic analysis indicated K179 acetylation of ACSF2 mainly affected long chain fatty acids (LCFA) metabolism, especially oleic acid, elaidic acid and palmitoleic acid. And the liquid chromatography mass spectrometry (LC-MS) analysis further demonstrated the formation of many long-chain acyl-CoAs were catalyzed by acetylation at K179 of ACSF2 including oleic acid, elaidic acid and palmitoleic acid. In conclusion, this study indicated that ACSF2 may be a potential therapeutic targets by regulating the metabolism of LCFA for liver fibrosis.
Collapse
Affiliation(s)
- Meili Wang
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Ying Su
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenjian Hou
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Kehan Ren
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiaoli Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Shanyu Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yuxiang Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Xiuping Liu
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China.
| |
Collapse
|
15
|
Betaine Reduces Lipid Anabolism and Promotes Lipid Transport in Mice Fed a High-Fat Diet by Influencing Intestinal Protein Expression. Foods 2022; 11:foods11162421. [PMID: 36010422 PMCID: PMC9407371 DOI: 10.3390/foods11162421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/30/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Betaine is more efficient than choline and methionine methyl donors, as it can increase nitrogen storage, promote fat mobilisation and fatty acid oxidation and change body fat content and distribution. Lipid is absorbed primarily in the small intestine after consumption, which is also the basis of lipid metabolism. This study was conducted to establish a mouse model of obesity in Kunming mice of the same age and similar body weight, and to assess the effect of betaine on the intestinal protein expression profile of mice using a proteomic approach. Analysis showed that betaine supplementation reversed the reduction in expression of proteins related to lipid metabolism and transport in the intestine of mice induced by a high-fat diet (HFD). For example, the addition of betaine resulted in a significant upregulation of microsomal triglyceride transfer protein (Mttp), apolipoprotein A-IV (Apoa4), fatty-acid-binding protein 1 (Fabp1) and fatty-acid-binding protein 2 (Fabp2) expression compared to the HFD group (p < 0.05), which exhibited accelerated lipid absorption and then translocation from the intestine into the body’s circulation, in addition to a significant increase in Acetyl-CoA acyltransferase (Acaa1a) protein expression, hastening lipid metabolism in the intestine (p < 0.05). Simultaneously, a significant reduction in protein expression of alpha-enolase 1 (Eno1) as the key enzyme for gluconeogenesis in mice in the betaine-supplemented group resulted in a reduction in lipid synthesis in the intestine (p < 0.05). These findings provide useful information for understanding the changes in the protein profile of the small intestine in response to betaine supplementation and the potential physiological regulation of diets’ nutrient absorption.
Collapse
|
16
|
Simvastatin Improves Microcirculatory Function in Nonalcoholic Fatty Liver Disease and Downregulates Oxidative and ALE-RAGE Stress. Nutrients 2022; 14:nu14030716. [PMID: 35277075 PMCID: PMC8838100 DOI: 10.3390/nu14030716] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/12/2022] Open
Abstract
Increased reactive oxidative stress, lipid peroxidation, inflammation, and fibrosis, which contribute to tissue damage and development and progression of nonalcoholic liver disease (NAFLD), play important roles in microcirculatory disorders. We investigated the effect of the modulatory properties of simvastatin (SV) on the liver and adipose tissue microcirculation as well as metabolic and oxidative stress parameters, including the advanced lipoxidation end product–receptors of advanced glycation end products (ALE-RAGE) pathway. SV was administered to an NAFLD model constructed using a high-fat–high-carbohydrate diet (HFHC). HFHC caused metabolic changes indicative of nonalcoholic steatohepatitis; treatment with SV protected the mice from developing NAFLD. SV prevented microcirculatory dysfunction in HFHC-fed mice, as evidenced by decreased leukocyte recruitment to hepatic and fat microcirculation, decreased hepatic stellate cell activation, and improved hepatic capillary network architecture and density. SV restored basal microvascular blood flow in the liver and adipose tissue and restored the endothelium-dependent vasodilatory response of adipose tissue to acetylcholine. SV treatment restored antioxidant enzyme activity and decreased lipid peroxidation, ALE-RAGE pathway activation, steatosis, fibrosis, and inflammatory parameters. Thus, SV may improve microcirculatory function in NAFLD by downregulating oxidative and ALE-RAGE stress and improving steatosis, fibrosis, and inflammatory parameters.
Collapse
|
17
|
Kostallari E, Wei B, Sicard D, Li J, Cooper SA, Gao J, Dehankar M, Li Y, Cao S, Yin M, Tschumperlin DJ, Shah VH. Stiffness is associated with hepatic stellate cell heterogeneity during liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2022; 322:G234-G246. [PMID: 34941452 PMCID: PMC8793867 DOI: 10.1152/ajpgi.00254.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The fibrogenic wound-healing response in liver increases stiffness. Stiffness mechanotransduction, in turn, amplifies fibrogenesis. Here, we aimed to understand the distribution of stiffness in fibrotic liver, how it impacts hepatic stellate cell (HSC) heterogeneity, and identify mechanisms by which stiffness amplifies fibrogenic responses. Magnetic resonance elastography and atomic force microscopy demonstrated a heterogeneous distribution of liver stiffness at macroscopic and microscopic levels, respectively, in a carbon tetrachloride (CCl4) mouse model of liver fibrosis as compared with controls. High stiffness was mainly attributed to extracellular matrix dense areas. To identify a stiffness-sensitive HSC subpopulation, we performed single-cell RNA sequencing (scRNA-seq) on primary HSCs derived from healthy versus CCl4-treated mice. A subcluster of HSCs was matrix-associated with the most upregulated pathway in this subpopulation being focal adhesion signaling, including a specific protein termed four and a half LIM domains protein 2 (FHL2). In vitro, FHL2 expression was increased in primary human HSCs cultured on stiff matrix as compared with HSCs on soft matrix. Moreover, FHL2 knockdown inhibited fibronectin and collagen 1 expression, whereas its overexpression promoted matrix production. In summary, we demonstrate stiffness heterogeneity at the whole organ, lobular, and cellular level, which drives an amplification loop of fibrogenesis through specific focal adhesion molecular pathways.NEW & NOTEWORTHY The fibrogenic wound-healing response in liver increases stiffness. Here, macro and microheterogeneity of liver stiffness correlate with HSC heterogeneity in a hepatic fibrosis mouse model. Fibrogenic HSCs localized in stiff collagen-high areas upregulate the expression of focal adhesion molecule FHL2, which, in turn, promotes extracellular matrix protein expression. These results demonstrate that stiffness heterogeneity at the whole organ, lobular, and cellular level drives an amplification loop of fibrogenesis through specific focal adhesion molecular pathways.
Collapse
Affiliation(s)
- Enis Kostallari
- 1Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Bo Wei
- 1Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota,2Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Delphine Sicard
- 3Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Jiahui Li
- 4Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Shawna A. Cooper
- 5Department of Biochemistry and Molecular Biology, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota
| | - Jinhang Gao
- 1Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota,2Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Mrunal Dehankar
- 6Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Ying Li
- 6Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Sheng Cao
- 1Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Meng Yin
- 4Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | - Vijay H. Shah
- 1Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
18
|
Mandala A, Chen WJ, Armstrong A, Malhotra MR, Chavalmane S, McCommis KS, Chen A, Carpenter D, Biswas P, Gnana-Prakasam JP. PPAR α agonist fenofibrate attenuates iron-induced liver injury in mice by modulating the Sirt3 and β-catenin signaling. Am J Physiol Gastrointest Liver Physiol 2021; 321:G262-G269. [PMID: 34287090 PMCID: PMC8461793 DOI: 10.1152/ajpgi.00129.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/22/2021] [Accepted: 07/07/2021] [Indexed: 01/31/2023]
Abstract
Iron accumulation is frequently associated with chronic liver diseases. However, our knowledge on how iron contributes to the liver injury is limited. Aberrant Wnt/β-catenin signaling is a hallmark of several hepatic pathologies. We recently reported that peroxisome proliferator-activated receptor α (PPARα) agonist, fenofibrate, prevents iron-induced oxidative stress and β-catenin signaling by chelating the iron. Sirtuin3 (Sirt3), a type of NAD+-dependent deacetylase, that plays a critical role in metabolic regulation was found to prevent ischemia reperfusion injury (IRI) by normalizing the Wnt/β-catenin pathway. In the present study, we explored if fenofibrate prevents iron-induced liver injury by regulating the Sirt3 and β-catenin signaling. In vitro and in vivo iron treatment resulted in the downregulation of PPARα, Sirt3, active β-catenin, and its downstream target gene c-Myc in the mouse liver. Pharmacological activation of Sirt3, both in vitro and in vivo, by Honokiol (HK), a known activator of Sirt3, abrogated the inhibitory effect of iron overload on active β-catenin expression and prevented the iron-induced upregulation of α smooth muscle actin (αSMA) and TGFβ expression. Intrinsically, PPARα knockout mice showed significant downregulation of hepatic Sirt3 levels. In addition, treatment of iron overload mice with PPARα agonist fenofibrate reduced hepatic iron accumulation and prevented iron-induced downregulation of liver Sirt3 and active β-catenin, mitigating the progression of fibrosis. Thus, our results establish a novel link between hepatic iron and PPARα, Sirt3, and β-catenin signaling. Further exploration on the mechanisms by which fenofibrate ameliorates iron-induced liver injury likely has significant therapeutic impact on iron-associated chronic liver diseases.NEW & NOTEWORTHY Hepatic intracellular iron accumulation has been implicated in the pathophysiology of chronic liver diseases. In this study, we identified a novel mechanism involved in the progression of fibrosis. Excess iron accumulation in liver caused downregulation of PPARα-Sirt3-Wnt signaling leading to fibrosis. This work has significant translational potential as PPARα agonist fenofibrate could be an attractive therapeutic drug for the treatment of liver disorders associated with iron overload.
Collapse
Affiliation(s)
- Ashok Mandala
- Department of Ophthalmology, Saint Louis University, St. Louis, Missouri
| | - William J Chen
- Department of Ophthalmology, Saint Louis University, St. Louis, Missouri
| | - Austin Armstrong
- Department of Ophthalmology, Saint Louis University, St. Louis, Missouri
| | - Milan R Malhotra
- Department of Ophthalmology, Saint Louis University, St. Louis, Missouri
| | - Sanmathi Chavalmane
- Department of Energy, Environmental and Chemical Engineering, Washington University, St. Louis, Missouri
| | - Kyle S McCommis
- Department of Biochemistry & Molecular Biology, Saint Louis University, St. Louis, Missouri
| | - Anping Chen
- Department of Pathology, Saint Louis University, St. Louis, Missouri
| | | | - Pratim Biswas
- Department of Engineering, University of Miami, Coral Gables, Florida
| | - Jaya P Gnana-Prakasam
- Department of Ophthalmology, Saint Louis University, St. Louis, Missouri
- Department of Biochemistry & Molecular Biology, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
19
|
Newberry EP, Hall Z, Xie Y, Molitor EA, Bayguinov PO, Strout GW, Fitzpatrick JA, Brunt EM, Griffin JL, Davidson NO. Liver-Specific Deletion of Mouse Tm6sf2 Promotes Steatosis, Fibrosis, and Hepatocellular Cancer. Hepatology 2021; 74:1203-1219. [PMID: 33638902 PMCID: PMC8390580 DOI: 10.1002/hep.31771] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/17/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Human transmembrane 6 superfamily 2 (TM6SF2) variant rs58542926 is associated with NAFLD and HCC. However, conflicting reports in germline Tm6sf2 knockout mice suggest no change or decreased very low density lipoprotein (VLDL) secretion and either unchanged or increased hepatic steatosis, with no increased fibrosis. We generated liver-specific Tm6Sf2 knockout mice (Tm6 LKO) to study VLDL secretion and the impact on development and progression of NAFLD. APPROACH AND RESULTS Two independent lines of Tm6 LKO mice exhibited spontaneous hepatic steatosis. Targeted lipidomic analyses showed increased triglyceride species whose distribution and abundance phenocopied findings in mice with liver-specific deletion of microsomal triglyceride transfer protein. The VLDL triglyceride secretion was reduced with small, underlipidated particles and unchanged or increased apolipoprotein B. Liver-specific adeno-associated viral, serotype 8 (AAV8) rescue using either wild-type or mutant E167K-Tm6 reduced hepatic steatosis and improved VLDL secretion. The Tm6 LKO mice fed a high milk-fat diet for 3 weeks exhibited increased steatosis and fibrosis, and those phenotypes were further exacerbated when mice were fed fibrogenic, high fat/fructose diets for 20 weeks. In two models of HCC, either neonatal mice injected with streptozotocin (NASH/STAM) and high-fat fed or with diethylnitrosamine injection plus fibrogenic diet feeding, Tm6 LKO mice exhibited increased steatosis, greater tumor burden, and increased tumor area versus Tm6 flox controls. Additionally, diethylnitrosamine-injected and fibrogenic diet-fed Tm6 LKO mice administered wild-type Tm6 or E167K-mutant Tm6 AAV8 revealed significant tumor attenuation, with tumor burden inversely correlated with Tm6 protein levels. CONCLUSIONS Liver-specific Tm6sf2 deletion impairs VLDL secretion, promoting hepatic steatosis, fibrosis, and accelerated development of HCC, which was mitigated with AAV8- mediated rescue.
Collapse
Affiliation(s)
- Elizabeth P. Newberry
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Zoe Hall
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom,Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Yan Xie
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Elizabeth A. Molitor
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Peter O. Bayguinov
- Washington University Center for Cellular Imaging, Washington University in Saint Louis, St. Louis, MO 63130
| | - Gregory W. Strout
- Washington University Center for Cellular Imaging, Washington University in Saint Louis, St. Louis, MO 63130
| | - James A.J. Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University in Saint Louis, St. Louis, MO 63130;,Departments of Cell Biology & Physiology and Neuroscience, Washington University School of Medicine, Louis, St. Louis, MO 63130;,Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63130
| | - Elizabeth M. Brunt
- Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Julian L. Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom,Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Nicholas O. Davidson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110;,To whom correspondence should be addressed: Nicholas O. Davidson, MD, DSc, Gastroenterology Division, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110.
| |
Collapse
|
20
|
α-Lipoic Acid Alleviates Hepatic Lipid Deposition by Inhibiting FASN Expression via miR-3548 in Rats. Nutrients 2021; 13:nu13072331. [PMID: 34371841 PMCID: PMC8308747 DOI: 10.3390/nu13072331] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 11/16/2022] Open
Abstract
Excessive liver lipid deposition is a vital risk factor for the development of many diseases. Here, we fed Sprague-Dawley rats with a control or α-lipoic acid-supplemented diet (0.2%) for 5 weeks to elucidate the effects of α-lipoic acid on preventive ability, hepatic lipid metabolism-related gene expression, and the involved regulatory mechanisms. In the current study, α-lipoic acid supplementation lowered plasma triglyceride level and hepatic triglyceride content. Reduced hepatic lipid deposition was closely associated with inhibiting fatty acid-binding protein 1 and fatty acid synthase expression, as well as increasing phosphorylated hormone-sensitive lipase expression at the protein level in α-lipoic acid-exposed rats. Hepatic miRNA sequencing revealed increased expression of miR-3548 targeting the 3'untranslated region of Fasn mRNA, and the direct regulatory link between miRNA-3548 and FASN was verified by dual-luciferase reporter assay. Taken together, α-lipoic acid lowered hepatic lipid accumulation, which involved changes in miRNA-mediated lipogenic genes.
Collapse
|
21
|
Wu X, Shu L, Zhang Z, Li J, Zong J, Cheong LY, Ye D, Lam KSL, Song E, Wang C, Xu A, Hoo RLC. Adipocyte Fatty Acid Binding Protein Promotes the Onset and Progression of Liver Fibrosis via Mediating the Crosstalk between Liver Sinusoidal Endothelial Cells and Hepatic Stellate Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2003721. [PMID: 34105268 PMCID: PMC8188197 DOI: 10.1002/advs.202003721] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/10/2021] [Indexed: 05/16/2023]
Abstract
Development of liver fibrosis results in drastic changes in the liver microenvironment, which in turn accelerates disease progression. Although the pathological function of various hepatic cells in fibrogenesis is identified, the crosstalk between them remains obscure. The present study demonstrates that hepatic expression of adipocyte fatty acid binding protein (A-FABP) is induced especially in the liver sinusoidal endothelial cells (LSECs) in mice after bile duct ligation (BDL). Genetic ablation and pharmacological inhibition of A-FABP attenuate BDL- or carbon tetrachloride-induced liver fibrosis in mice associating with reduced collagen accumulation, LSEC capillarization, and hepatic stellate cell (HSC) activation. Mechanistically, elevated A-FABP promotes LSEC capillarization by activating Hedgehog signaling, thus impairs the gatekeeper function of LSEC on HSC activation. LSEC-derived A-FABP also acts on HSCs in paracrine manner to potentiate the transactivation of transforming growth factor β1 (TGFβ1) by activating c-Jun N-terminal kinase (JNK)/c-Jun signaling. Elevated TGFβ1 subsequently exaggerates liver fibrosis. These findings uncover a novel pathological mechanism of liver fibrosis in which LSEC-derived A-FABP is a key regulator modulating the onset and progression of the disease. Targeting A-FABP may represent a potential approach against liver fibrosis.
Collapse
Affiliation(s)
- Xiaoping Wu
- State Key Laboratory of Pharmaceutical BiotechnologyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
| | - Lingling Shu
- State Key Laboratory of Pharmaceutical BiotechnologyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- Department of MedicineLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Zixuan Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
| | - Jingjing Li
- Department of Pharmacology and PharmacyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
| | - Jiuyu Zong
- State Key Laboratory of Pharmaceutical BiotechnologyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical BiotechnologyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- Department of MedicineLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
| | - Dewei Ye
- Joint Laboratory of Guangdong and Hong Kong on Metabolic DiseasesGuangdong Pharmaceutical UniversityGuangzhou510000China
| | - Karen S. L. Lam
- State Key Laboratory of Pharmaceutical BiotechnologyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- Department of MedicineLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
| | - Erfei Song
- Department of Metabolic and Bariatric SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdong510630China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdong510630China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical BiotechnologyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- Department of MedicineLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
| | - Ruby L. C. Hoo
- State Key Laboratory of Pharmaceutical BiotechnologyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLKS Faculty of Medicinethe University of Hong KongHong Kong999077China
- HKU‐Shenzhen Institute of Research and Innovation (HKU‐SIRI)Shenzhen518057China
| |
Collapse
|
22
|
Wang Y, Jia M, Liang C, Sheng N, Wang X, Wang F, Luo Y, Jiang J, Cai L, Niu H, Zhu D, Nesa EU, Young CY, Yuan H. Anterior gradient 2 increases long-chain fatty acid uptake via stabilizing FABP1 and facilitates lipid accumulation. Int J Biol Sci 2021; 17:834-847. [PMID: 33767592 PMCID: PMC7975708 DOI: 10.7150/ijbs.57099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 01/23/2021] [Indexed: 12/19/2022] Open
Abstract
Anterior gradient 2 (AGR2), a protein disulfide isomerase (PDI), is a well-established oncogene. Here, we found that Agr2-/- mice had a decreased fat mass and hepatic and serum lipid levels compared with their wild-type littermates after fasting, and exhibited reduced high-fat diet (HFD)-induced fat accumulation. Transgenic mice overexpressing AGR2 (Agr2/Tg) readily gained fat weight on a HFD but not a normal diet. Proteomic analysis of hepatic samples from Agr2-/- mice revealed that depletion of AGR2 impaired long-chain fatty acid uptake and activation but did not affect de novo hepatic lipogenesis. Further investigations led to the identification of several effector substrates, particularly fatty acid binding protein-1 (FABP1) as essential for the AGR2-mediated effects. AGR2 was coexpressed with FABP1, and knockdown of AGR2 resulted in a reduction in FABP1 stability. Physical interactions of AGR2 and FABP1 depended on the PDI motif in AGR2 and the formation of a disulfide bond between these two proteins. Overexpression of AGR2 but not a mutant AGR2 protein lacking PDI activity suppressed lipid accumulation in cells lacking FABP1. Moreover, AGR2 deficiency significantly reduced fatty acid absorption in the intestine, which might be resulted from decreased fatty acid transporter CD36 in mice. These findings demonstrated a novel role of AGR2 in fatty-acid uptake and activation in both the liver and intestine, which contributed to the AGR2-mediated lipid accumulation, suggesting that AGR2 is an important regulator of whole-body lipid metabolism and down-regulation of AGR2 may antagonize the development of obesity.
Collapse
Affiliation(s)
- Yunqiu Wang
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
| | - Mengqi Jia
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Chuanjie Liang
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
| | - Nan Sheng
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Xiaodan Wang
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Fang Wang
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Yanhai Luo
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
| | - Jin Jiang
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Liangyu Cai
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Huanmin Niu
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Deyu Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
| | - Effat Un Nesa
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Charles Yf Young
- Department of Urology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester 55905, MN, USA
| | - Huiqing Yuan
- Key Laboratory of Experimental Teratology of Ministry of Education, Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250014, China
| |
Collapse
|
23
|
Eguchi A, Iwasa M. The Role of Elevated Liver-Type Fatty Acid-Binding Proteins in Liver Diseases. Pharm Res 2021; 38:89-95. [PMID: 33534129 DOI: 10.1007/s11095-021-02998-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/04/2020] [Indexed: 01/27/2023]
Abstract
Liver-type fatty acid-binding protein (L-FABP) is mainly expressed in the liver as well as the proximal tubular epithelial cells in the kidney. In general, the proteins and enzymes existing within the hepatocytes have the potential to become biomarkers, for instance alanine aminotransferase, which reflects hepatocellular damage. However, due to reduced hepatocellular function in late stage of chronic liver diseases (e.g. cirrhosis), proteins and enzymes relating to hepatocellular damage are not always accurate measures of disease progression. Recently, several publications have demonstrated elevated serum L-FABP levels during the progression of human liver diseases, including hepatocellular carcinoma (HCC), and were a prognostic factor for survival in acute and chronic liver disease patients. However, the study regarding serum L-FABP levels and hepatic L-FABP expression in liver diseases is not sufficient to understand the molecular mechanism of L-FABP during the progression of these disease states. In this review, we focus on the use of serum and/or hepatic L-FABP expression as a biomarker in human liver diseases, including mechanistic potential in HCC.
Collapse
Affiliation(s)
- Akiko Eguchi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
- JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
| | - Motoh Iwasa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| |
Collapse
|
24
|
Abstract
Hepatic stellate cells (HSCs) are resident non-parenchymal liver pericytes whose plasticity enables them to regulate a remarkable range of physiologic and pathologic responses. To support their functions in health and disease, HSCs engage pathways regulating carbohydrate, mitochondrial, lipid, and retinoid homeostasis. In chronic liver injury, HSCs drive hepatic fibrosis and are implicated in inflammation and cancer. To do so, the cells activate, or transdifferentiate, from a quiescent state into proliferative, motile myofibroblasts that secrete extracellular matrix, which demands rapid adaptation to meet a heightened energy need. Adaptations include reprogramming of central carbon metabolism, enhanced mitochondrial number and activity, endoplasmic reticulum stress, and liberation of free fatty acids through autophagy-dependent hydrolysis of retinyl esters that are stored in cytoplasmic droplets. As an archetype for pericytes in other tissues, recognition of the HSC's metabolic drivers and vulnerabilities offer the potential to target these pathways therapeutically to enhance parenchymal growth and modulate repair.
Collapse
Affiliation(s)
- Parth Trivedi
- Division of Liver Diseases, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shuang Wang
- Division of Liver Diseases, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Scott L Friedman
- Division of Liver Diseases, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
25
|
Kulkarni AV, Sharma M, Kumar P, Simhadri V, Sowmya TR, Mitnala S, Nageshwar Reddy D, Nagaraja Rao P. Adipocyte Fatty Acid-Binding Protein as a Predictor of Outcome in Alcohol-induced Acute-On-Chronic Liver Failure. J Clin Exp Hepatol 2021; 11:201-208. [PMID: 33746445 PMCID: PMC7953014 DOI: 10.1016/j.jceh.2020.07.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/18/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Alcohol is the leading cause of acute-on-chronic liver failure (ACLF). Several severity scores predict the outcome of ACLF. However, there is a lack of simple biomarkers in predicting the outcome of these sick patients. Fatty acid-binding proteins (FABPs) are small cytosolic proteins that play a major role in lipid metabolism, energy homeostasis, and inflammation, but, have not been investigated in alcohol-induced ACLF (A-ACLF). OBJECTIVES The primary objective was to assess the correlation between serum adipocyte-FABP (A-FABP) and liver-FABP (L-FABP) levels on mortality at day 90. Secondary objectives were to compare the levels between controls and A-ACLF, correlate L-FABP, and A-FABP levels on the development of organ failure/sepsis at day 90. METHODS In this prospective observational pilot study, we included patients with A-ACLF and age-matched healthy controls. FABP's were analyzed by enzyme-linked immunosorbent assay method. The patients were followed up for 90 days. RESULTS Twenty-five patients with A-ACLF (mean age: 40years; mean model for end-stage liver disease NA: 29.8; median Modified Maddrey's discriminant function [mDF]: 95) and 12 controls (mean age: 36.83yrs) were included in the study. A-FABP and L-FABP levels were significantly high in patients with A-ACLF than controls. Forty-four percent of patients with A-ACLF developed sepsis, 48% developed organ failure, and 44% expired by day 90. On multivariate Cox regression analysis, A-FABP (hazard ratio [HR]: 1.27 [1.08-1.5]; P = 0.003), Asian Pacific Association for the Study of Liver ACLF research consortium score (HR: 3.3[1.15-9.54]; P = 0.02), L-FABP (HR: 0.69 [0.52-0.91]; P = 0.009), and serum protein levels (HR: 0.03 [0.003-0.36]; P = 0.005) predicted mortality. A-FABP (1.17 [1.07-1.29]; P = 0.001), and serum bilirubin (1.05 [0.99-1.12]; P = 0.06) predicted development of organ failure, and only mDF (HR: 1.04 [1.01-1.07]; P = 0.009) predicted the development of sepsis on multivariate analysis. Fifteen patients received steroid therapy, of which 13.34% were nonresponders. CONCLUSIONS In a selected group of patients with A-ACLF, A-FABP is highly sensitive at predicting mortality and outcome. If validated in a large, diverse sample, A-FABP can be used as a simple biomarker for prognostication in A-ACLF.
Collapse
Affiliation(s)
- Anand V. Kulkarni
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India,Address for correspondence: Department of Hepatology & Liver Transplantation, Asian Institute of Gastroenterology, Hyderabad, India.
| | - Mithun Sharma
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | - Pramod Kumar
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | - Venu Simhadri
- Department of Research, Asian Healthcare Foundation, Hyderabad, India
| | | | - Sasikala Mitnala
- Department of Research, Asian Healthcare Foundation, Hyderabad, India
| | | | - Padaki Nagaraja Rao
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| |
Collapse
|
26
|
Chen X, Acquaah-Mensah GK, Denning KL, Peterson JM, Wang K, Denvir J, Hong F, Cederbaum AI, Lu Y. High-fat diet induces fibrosis in mice lacking CYP2A5 and PPARα: a new model for steatohepatitis-associated fibrosis. Am J Physiol Gastrointest Liver Physiol 2020; 319:G626-G635. [PMID: 32877213 PMCID: PMC8087345 DOI: 10.1152/ajpgi.00213.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obesity is linked to nonalcoholic steatohepatitis. Peroxisome proliferator-activated receptor-α (PPARα) regulates lipid metabolism. Cytochrome P-450 2A5 (CYP2A5) is a potential antioxidant and CYP2A5 induction by ethanol is CYP2E1 dependent. High-fat diet (HFD)-induced obesity and steatosis are more severe in CYP2A5 knockout (cyp2a5-/-) mice than in wild-type mice although PPARα is elevated in cyp2a5-/- mice. To examine why the upregulated PPARα failed to prevent the enhanced steatosis in cyp2a5-/- mice, we abrogate the upregulated PPARα in cyp2a5-/- mice by cross-breeding cyp2a5-/- mice with PPARα knockout (pparα-/-) mice to create pparα-/-/cyp2a5-/- mice. The pparα-/-/cyp2a5-/- mice, pparα-/- mice, and cyp2a5-/- mice were fed HFD to induce steatosis. After HFD feeding, more severe steatosis was developed in pparα-/-/cyp2a5-/- mice than in pparα-/- mice and cyp2a5-/- mice. The pparα-/-/cyp2a5-/- mice and pparα-/- mice exhibited comparable and impaired lipid metabolism. Elevated serum alanine transaminase and liver interleukin-1β, liver inflammatory cell infiltration, and foci of hepatocellular ballooning were observed in pparα-/-/cyp2a5-/- mice but not in pparα-/- mice and cyp2a5-/- mice. In pparα-/-/cyp2a5-/- mice, although redox-sensitive transcription factor nuclear factor erythroid 2-related factor 2 and its target antioxidant genes were upregulated as a compensation, thioredoxin was suppressed, and phosphorylation of JNK and formation of nitrotyrosine adduct were increased. Liver glutathione was decreased, and lipid peroxidation was increased. Interestingly, inflammation and fibrosis were all observed within the clusters of lipid droplets, and these lipid droplet clusters were all located inside the area with CYP2E1-positive staining. These results suggest that HFD-induced fibrosis in pparα-/-/cyp2a5-/- mice is associated with steatosis, and CYP2A5 interacts with PPARα to participate in regulating steatohepatitis-associated fibrosis.
Collapse
Affiliation(s)
- Xue Chen
- 1Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - George K. Acquaah-Mensah
- 2Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Worcester, Massachusetts
| | - Krista L. Denning
- 3Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Jonathan M. Peterson
- 4Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee
| | - Kesheng Wang
- 5Department of Family and Community Health, School of Nursing, Health Sciences Center, West Virginia University, Morgantown, West Virginia
| | - James Denvir
- 1Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Feng Hong
- 6Institute of Liver Diseases, Affiliated Hospital of Jining Medical University, Jining, China
| | - Arthur I. Cederbaum
- 7Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yongke Lu
- 1Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia,8Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| |
Collapse
|
27
|
The biology of lipid droplet-bound mitochondria. Semin Cell Dev Biol 2020; 108:55-64. [PMID: 32446655 DOI: 10.1016/j.semcdb.2020.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 12/12/2022]
Abstract
Proper regulation of cellular lipid storage and oxidation is indispensable for the maintenance of cellular energy homeostasis and health. Mitochondrial function has been shown to be a main determinant of functional lipid storage and oxidation, which is of particular interest for the adipose tissue, as it is the main site of triacylglyceride storage in lipid droplets (LDs). Recent studies have identified a subpopulation of mitochondria attached to LDs, peridroplet mitochondria (PDM) that can be separated from cytoplasmic mitochondria (CM) by centrifugation. PDM have distinct bioenergetics, proteome, cristae organization and dynamics that support LD build-up, however their role in adipose tissue biology remains largely unexplored. Therefore, understanding the molecular basis of LD homeostasis and their relationship to mitochondrial function and attachment in adipocytes is of major importance.
Collapse
|
28
|
SUMOylation inhibitors synergize with FXR agonists in combating liver fibrosis. Nat Commun 2020; 11:240. [PMID: 31932588 PMCID: PMC6957516 DOI: 10.1038/s41467-019-14138-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Farnesoid X receptor (FXR) is a promising target for nonalcoholic steatohepatitis (NASH) and fibrosis. Although various FXR agonists have shown anti-fibrotic effects in diverse preclinical animal models, the response rate and efficacies in clinical trials were not optimum. Here we report that prophylactic but not therapeutic administration of obeticholic acid (OCA) prevents hepatic stellate cell (HSC) activation and fibrogenesis. Activated HSCs show limited response to OCA and other FXR agonists due to enhanced FXR SUMOylation. SUMOylation inhibitors rescue FXR signaling and thereby increasing the efficacy of OCA against HSC activation and fibrosis. FXR upregulates Perilipin-1, a direct target gene of FXR, to stabilize lipid droplets and thereby prevent HSC activation. Therapeutic coadministration of OCA and SUMOylation inhibitors drastically impedes liver fibrosis induced by CCl4, bile duct ligation, and more importantly NASH. In conclusion, we propose a promising therapeutic approach by combining SUMOylation inhibitors and FXR agonists for liver fibrosis. FXR agonists have been investigated for the treatment of non-alcoholic steatohepatitis and liver fibrosis but the clinical efficacy is not optimal. Here the authors show that enhanced FXR SUMOylation in activated hepatic stellate cells reduces FXR signaling and that this can be rescued by SUMOylation inhibitors.
Collapse
|
29
|
Lu YC, Chang CC, Wang CP, Hung WC, Tsai IT, Tang WH, Wu CC, Wei CT, Chung FM, Lee YJ, Hsu CC. Circulating fatty acid-binding protein 1 (FABP1) and nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus. Int J Med Sci 2020; 17:182-190. [PMID: 32038102 PMCID: PMC6990891 DOI: 10.7150/ijms.40417] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/19/2019] [Indexed: 12/30/2022] Open
Abstract
Background: Fatty acid-binding protein 1 (FABP1) (also known as liver-type fatty acid-binding protein or LFABP) is a protein that is mainly expressed in the liver, and is associated with hepatocyte injury in acute transplant rejection. Reduced levels of FABP1 in mice livers have been shown to be effective against nonalcoholic fatty liver disease (NAFLD). In this study, we investigated the association between plasma FABP1 levels and NAFLD in patients with type 2 diabetes mellitus (T2DM). Methods: We enrolled 267 T2DM patients. Clinical and biochemical parameters were measured. The severity of NAFLD was assessed by ultrasound. FABP1 levels were determined using by enzyme-linked immunosorbent assays. Results: FABP1 levels were higher in patients with overt NAFLD, defined as more than a moderate degree of fatty liver compared to those without NAFLD. Age- and sex-adjusted analysis of FABP1 showed positive associations with body mass index (BMI), waist circumference, homeostasis model assessment estimate of β-cell function, creatinine, and fatty liver index, but showed negative associations with albumin and estimated glomerular filtration rate (eGFR). The odds ratio (OR) for the risk of overt NAFLD with increasing levels of sex-specific FABP1 was significantly increased (OR 2.63 [95% CI 1.30-5.73] vs. 4.94 [2.25-11.48]). The OR in the second and third tertiles of FABP1 remained significant after adjustments for BMI, triglycerides, high-density lipoprotein cholesterol, HbA1C, homeostasis model assessment estimate of insulin resistance, white blood cell count, hepatic enzymes, and eGFR. Conclusion: Our results indicate that FABP1 may play a role in the pathogenesis of NAFLD in patients with T2DM.
Collapse
Affiliation(s)
- Yung-Chuan Lu
- Division of Endocrinology and Metabolism, E-Da Hospital, Kaohsiung, 82445 Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan
| | - Chi-Chang Chang
- Department of Obstetrics & Gynecology, E-Da Hospital, Kaohsiung, 82445 Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan.,Department of Obstetrics & Gynecology, E-Da Dachang Hospital, Kaohsiung 80794 Taiwan
| | - Chao-Ping Wang
- Division of Cardiology, E-Da Hospital, Kaohsiung, 82445 Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan
| | - Wei-Chin Hung
- Division of Cardiology, E-Da Hospital, Kaohsiung, 82445 Taiwan.,The School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan
| | - I-Ting Tsai
- Departmen of Emergency, E-Da Hospital, Kaohsiung, 82445 Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan
| | - Wei-Hua Tang
- Lee's Endocrinology Clinic, Pingtung, 90000 Taiwan
| | - Cheng-Ching Wu
- Division of Cardiology, E-Da Hospital, Kaohsiung, 82445 Taiwan.,The School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan.,Division of Cardiology, Department of Internal Medicine, E-Da Cancer Hospital, Kaohsiung 82445 Taiwan
| | - Ching-Ting Wei
- Division of General Surgery, Department of Surgery, E-Da Hospital, Kaohsiung, 82445 Taiwan
| | - Fu-Mei Chung
- Division of Cardiology, E-Da Hospital, Kaohsiung, 82445 Taiwan
| | | | - Chia-Chang Hsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, 82445 Taiwan.,The School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, 82445 Taiwan.,Health Examination Center, E-Da Dachang Hospital, Kaohsiung, Taiwan
| |
Collapse
|
30
|
Khomich O, Ivanov AV, Bartosch B. Metabolic Hallmarks of Hepatic Stellate Cells in Liver Fibrosis. Cells 2019; 9:E24. [PMID: 31861818 PMCID: PMC7016711 DOI: 10.3390/cells9010024] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/09/2019] [Accepted: 12/18/2019] [Indexed: 12/17/2022] Open
Abstract
Liver fibrosis is a regenerative process that occurs after injury. It is characterized by the deposition of connective tissue by specialized fibroblasts and concomitant proliferative responses. Chronic damage that stimulates fibrogenic processes in the long-term may result in the deposition of excess matrix tissue and impairment of liver functions. End-stage fibrosis is referred to as cirrhosis and predisposes strongly to the loss of liver functions (decompensation) and hepatocellular carcinoma. Liver fibrosis is a pathology common to a number of different chronic liver diseases, including alcoholic liver disease, non-alcoholic fatty liver disease, and viral hepatitis. The predominant cell type responsible for fibrogenesis is hepatic stellate cells (HSCs). In response to inflammatory stimuli or hepatocyte death, HSCs undergo trans-differentiation to myofibroblast-like cells. Recent evidence shows that metabolic alterations in HSCs are important for the trans-differentiation process and thus offer new possibilities for therapeutic interventions. The aim of this review is to summarize current knowledge of the metabolic changes that occur during HSC activation with a particular focus on the retinol and lipid metabolism, the central carbon metabolism, and associated redox or stress-related signaling pathways.
Collapse
Affiliation(s)
- Olga Khomich
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR_5286, Centre Léon Bérard, CEDEX 03, 69424 Lyon, France;
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexander V. Ivanov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Birke Bartosch
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR_5286, Centre Léon Bérard, CEDEX 03, 69424 Lyon, France;
| |
Collapse
|
31
|
Zhao J, Han M, Zhou L, Liang P, Wang Y, Feng S, Lu H, Yuan X, Han K, Chen X, Liu S, Cheng J. TAF and TDF attenuate liver fibrosis through NS5ATP9, TGFβ1/Smad3, and NF-κB/NLRP3 inflammasome signaling pathways. Hepatol Int 2019; 14:145-160. [PMID: 31758498 DOI: 10.1007/s12072-019-09997-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/23/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND This study aimed to investigate the roles and mechanisms of tenofovir alafenamide fumarate (TAF)/tenofovir disoproxil fumarate (TDF) in treating liver fibrosis. METHODS The effects of TAF/TDF on carbon tetrachloride (CCl4)-induced liver fibrosis in C57BL/6 wild-type or nonstructural protein 5A transactivated protein 9 (NS5ATP9) knockout mice were studied. The differentiation, activation, and proliferation of LX-2 cells after TAF/TDF treatment were tested in vitro. The expression of NS5ATP9 and activities of transforming growth factor-β1 (TGFβ1)/Sekelsky mothers against decapentaplegic homolog 3 (Smad3) and NF-κB/NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome signaling pathways were detected in TAF/TDF-treated mice and LX-2 cells. The genes related to extracellular matrix accumulation were detected in vivo and in vitro after NS5ATP9 silencing or knockout. RESULTS TAF/TDF significantly inhibited CCl4-induced liver fibrosis in mice, and regulated the differentiation, activation, and proliferation of hepatic stellate cells (HSCs). Furthermore, TAF/TDF suppressed the activities of TGFβ1/Smad3 and NF-κB/NLRP3 inflammasome signaling pathways in vivo and in vitro. NS5ATP9 inhibited liver fibrosis through TGFβ1/Smad3 and NF-κB signaling pathways. TAF/TDF upregulated the expression of NS5ATP9 in vivo and in vitro. Finally, TAF/TDF could only show marginal therapeutic effects when NS5ATP9 was silenced and knocked out in vivo and in vitro. CONCLUSIONS TAF/TDF prevented progression and promoted reversion of liver fibrosis through assembling TGFβ1/Smad3 and NF-κB/NLRP3 inflammasome signaling pathways via upregulating the expression of NS5ATP9. TAF/TDF also regulated the differentiation, activation, and proliferation of HSCs. The findings provided strong evidence for the role of TAF/TDF as a new promising therapeutic strategy in liver fibrosis.
Collapse
Affiliation(s)
- Jing Zhao
- Peking University Ditan Teaching Hospital, Beijing, 100015, China
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Ming Han
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Li Zhou
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
- Department of Infectious Disease, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Pu Liang
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Yun Wang
- Peking University Ditan Teaching Hospital, Beijing, 100015, China
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Shenghu Feng
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
- Department of Infectious Diseases and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hongping Lu
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Xiaoxue Yuan
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Kai Han
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Xiaofan Chen
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
- Department of Infectious Diseases, Center for Liver Diseases, Peking University First Hospital, Beijing, 100034, China
| | - Shunai Liu
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Jun Cheng
- Peking University Ditan Teaching Hospital, Beijing, 100015, China.
- Institiute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing, 100191, China.
| |
Collapse
|
32
|
Han XQ, Xu SQ, Lin JG. Curcumin Recovers Intracellular Lipid Droplet Formation Through Increasing Perilipin 5 Gene Expression in Activated Hepatic Stellate Cells In Vitro. Curr Med Sci 2019; 39:766-777. [DOI: 10.1007/s11596-019-2104-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/02/2019] [Indexed: 02/06/2023]
|
33
|
Pi H, Liu M, Xi Y, Chen M, Tian L, Xie J, Chen M, Wang Z, Yang M, Yu Z, Zhou Z, Gao F. Long-term exercise prevents hepatic steatosis: a novel role of FABP1 in regulation of autophagy-lysosomal machinery. FASEB J 2019; 33:11870-11883. [PMID: 31366243 DOI: 10.1096/fj.201900812r] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most prevalent diseases worldwide. Exercise is a first-line therapy and an important preventive measure for patients with NAFLD, but the underlying mechanisms are not clear. C57BL/6 mice were fed a high-fat diet (HFD) and subjected to 12 wk swimming exercise. Exercise protected against hepatic lipid accumulation and alleviated hepatocyte damage in HFD mice. Tandem mass tag-based quantitative proteomic analyses and ingenuity pathway analysis revealed that exercise down-regulated fatty acid-binding protein (FABP)1 signaling pathway, which was most closely associated with lipid metabolism. Moreover, exercise significantly decreased FABP1 expression, and liver-specific overexpression of FABP1 abolished the protective effect of exercise in NAFLD mice. Specifically, exercise significantly increased autophagic flux via restoring lysosomal function, including lysosomal proteolysis and lysosomal acidification maintenance, contributing to enhancement in autophagic clearance and subsequently alleviation of hepatic steatosis. Conversely, Fabp1 overexpression in the mouse liver blocked the protective effect of exercise via inhibiting autophagy flux. The present study identified FABP1 inhibition-mediated replenishment of the autophagy-lysosomal machinery as a novel endogenous mechanism whereby long-term exercise improves lipid homeostasis and ameliorates hepatic steatosis in NAFLD.-Pi, H., Liu, M., Xi, Y., Chen, M., Tian, L., Xie, J., Chen, M., Wang, Z., Yang, M., Yu, Z., Zhou, Z., Gao, F. Long-term exercise prevents hepatic steatosis: a novel role of FABP1 in regulation of autophagy-lysosomal machinery.
Collapse
Affiliation(s)
- Huifeng Pi
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China.,Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Mengyu Liu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Yu Xi
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Department of Emergency Medicine-First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengyan Chen
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Li Tian
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Jia Xie
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Mingliang Chen
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology, Ministry of Education, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhen Wang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Min Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Department of Emergency Medicine-First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Gao
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
34
|
Eguchi A, Hasegawa H, Iwasa M, Tamai Y, Ohata K, Oikawa T, Sugaya T, Takei Y. Serum Liver-Type Fatty Acid-Binding Protein Is a Possible Prognostic Factor in Human Chronic Liver Diseases From Chronic Hepatitis to Liver Cirrhosis and Hepatocellular Carcinoma. Hepatol Commun 2019; 3:825-837. [PMID: 31168516 PMCID: PMC6545868 DOI: 10.1002/hep4.1350] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 03/15/2019] [Indexed: 01/27/2023] Open
Abstract
Liver‐type fatty acid–binding protein (L‐FABP) is a key regulator of fatty acid metabolism, but serum L‐FABP levels are not well investigated in chronic liver diseases. We aimed to elucidate the prognostic ability of serum L‐FABP in human chronic liver diseases and compare it with the albumin‐bilirubin (ALBI) score. In 242 chronic liver disease patients, including chronic hepatitis (CH, n = 100), liver cirrhosis (LC, n = 142), and presence of hepatocellular carcinoma (HCC, n = 144), serum L‐FABP levels were correlated with liver function (P < 0.0001), increased in LC compared with CH (P < 0.01), and correlated to ALBI score (P < 0.0001). Serum L‐FABP levels were increased in the presence of HCC (P < 0.0001), correlating to des‐gamma‐carboxy prothrombin (P < 0.0001), alpha‐fetoprotein (P = 0.009), and Barcelona‐Clinic Liver Cancer stage. In the average follow‐up period of 1,054 days, serum L‐FABP levels were elevated (P < 0.0001) in patients who eventually died. The area under the curve (AUC) of serum L‐FABP (0.764) was higher than that of ALB (0.709), and the patients with serum L‐FABP ≤ 6.8 ng/mL had significantly longer rates of survival (P < 0.0001). Serum L‐FABP (hazard ratio [HR] 4.0; P < 0.001), HCC (HR 3.7; P = 0.001), ALBI score (HR 2.7; P < 0.001), and age (HR 1.0; P = 0.049) were independent predictors of survival. In the subgroup who maintained liver function, the AUC of serum L‐FABP (0.751) was higher than that of ALB (0.643). In this subgroup, serum L‐FABP (HR 4.4; P = 0.002) and HCC (HR 13.9; P < 0.001) were independent predictors of survival. Conclusion: Serum L‐FABP is a possible predictor of survival in chronic liver diseases from CH to LC and HCC, including any subgroup that maintains liver function.
Collapse
Affiliation(s)
- Akiko Eguchi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine Mie University Mie Japan.,Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology Saitama Japan
| | - Hiroshi Hasegawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine Mie University Mie Japan
| | - Motoh Iwasa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine Mie University Mie Japan
| | - Yasuyuki Tamai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine Mie University Mie Japan
| | | | | | | | - Yoshiyuki Takei
- Department of Gastroenterology and Hepatology, Graduate School of Medicine Mie University Mie Japan
| |
Collapse
|
35
|
Cao H, Zhou Z, Wang L, Liu G, Sun Y, Wang Y, Wang T, Liang Y. Screening of Potential PFOS Alternatives To Decrease Liver Bioaccumulation: Experimental and Computational Approaches. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:2811-2819. [PMID: 30735364 DOI: 10.1021/acs.est.8b05564] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Perfluorooctanesulfonate (PFOS) is a persistent organic pollutant with significant bioaccumulation potential in liver tissues. Exposure to PFOS could cause increase of liver weight, induce adenomas of the liver, and cause hepatomegaly. Alternatives of PFOS might be designed and synthesized that have significantly lower liver bioaccumulation. In this study, we conducted animal exposure experiments to investigate tissue accumulations of 14 per- and polyfluoroalkyl substances. Correlation analysis demonstrated that accumulation of the compounds in rat liver had strong correlations with their binding affinities of liver fatty acid binding protein (LFABP). Thus, we combined a quantitative structure-activity relationship model with molecular dynamics (MD) simulations to develop computational models to predict the LFABP binding affinities of two newly synthesized alternatives, perfluorodecalin-2-sulfonic acid and N-diperfluorobutanoic acid. The binding characteristics of the PFOS alternatives for LFABP were elaborated to explore how the different structural modifications of molecules influenced the underlying binding mechanisms. Subsequent animal experiments demonstrated that the binding free energy calculations based on the MD simulations provided a good indicator to reflect the relative degree of liver accumulation of the PFOS alternatives in the same exposure doses and durations. Our findings from the combination of experimental exposure and computational model can provide helpful information to design potential alternatives of PFOS with weak LFABP binding capability and low liver accumulation.
Collapse
Affiliation(s)
- Huiming Cao
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances , Jianghan University , Wuhan 430056 , P. R. China
- Institute of Environment and Health , Jianghan University , Wuhan 430056 , P. R. China
| | - Zhen Zhou
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances , Jianghan University , Wuhan 430056 , P. R. China
- Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, School of Chemical and Environmental Engineering , Jianghan University , Wuhan 430056 , P. R. China
| | - Ling Wang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances , Jianghan University , Wuhan 430056 , P. R. China
- Institute of Environment and Health , Jianghan University , Wuhan 430056 , P. R. China
| | - Guangliang Liu
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances , Jianghan University , Wuhan 430056 , P. R. China
- Institute of Environment and Health , Jianghan University , Wuhan 430056 , P. R. China
| | - Yuzhen Sun
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances , Jianghan University , Wuhan 430056 , P. R. China
- Institute of Environment and Health , Jianghan University , Wuhan 430056 , P. R. China
| | - Yawei Wang
- Institute of Environment and Health , Jianghan University , Wuhan 430056 , P. R. China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , P.O. Box 2871, Beijing 100085 , P. R. China
| | - Thanh Wang
- MTM Research Centre, School of Science and Technology , Örebro University , Örebro 70182 , Sweden
| | - Yong Liang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances , Jianghan University , Wuhan 430056 , P. R. China
- Institute of Environment and Health , Jianghan University , Wuhan 430056 , P. R. China
| |
Collapse
|
36
|
Newberry EP, Xie Y, Lodeiro C, Solis R, Moritz W, Kennedy S, Barron L, Onufer E, Alpini G, Zhou T, Blaner WS, Chen A, Davidson NO. Hepatocyte and stellate cell deletion of liver fatty acid binding protein reveals distinct roles in fibrogenic injury. FASEB J 2019; 33:4610-4625. [PMID: 30576225 PMCID: PMC6404585 DOI: 10.1096/fj.201801976r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/26/2018] [Indexed: 12/13/2022]
Abstract
Liver fatty acid binding protein (L-Fabp) modulates lipid trafficking in enterocytes, hepatocytes, and hepatic stellate cells (HSCs). We examined hepatocyte vs. HSC L-Fabp deletion in hepatic metabolic adaptation and fibrotic injury. Floxed L-Fabp mice were bred to different transgenic Cre mice or injected with adeno-associated virus type 8 (AAV8) Cre and fed diets to promote steatosis and fibrosis or were subjected to either bile duct ligation or CCl4 injury. Albumin-Cre-mediated L-Fabp deletion revealed recombination in hepatocytes and HSCs; these findings were confirmed with 2 other floxed alleles. Glial fibrillary acid protein-Cre and platelet-derived growth factor receptor β-Cre-mediated L-Fabp deletion demonstrated recombination only in HSCs. Mice with albumin promoter-driven Cre recombinase (Alb-Cre)-mediated or AAV8-mediated L-Fabp deletion were protected against food withdrawal-induced steatosis. Mice with Alb-Cre-mediated L-Fabp deletion were protected against high saturated fat-induced steatosis and fibrosis, phenocopying germline L-Fabp-/- mice. Mice with HSC-specific L-Fabp deletion exhibited retinyl ester depletion yet demonstrated no alterations in fibrosis. On the other hand, fibrogenic resolution after CCl4 administration was impaired in mice with Alb-Cre-mediated L-Fabp deletion. These findings suggest cell type-specific roles for L-Fabp in mitigating hepatic steatosis and in modulating fibrogenic injury and reversal.-Newberry, E. P., Xie, Y., Lodeiro, C., Solis, R., Moritz, W., Kennedy, S., Barron, L., Onufer, E., Alpini, G., Zhou, T., Blaner, W. S., Chen, A., Davidson, N. O. Hepatocyte and stellate cell deletion of liver fatty acid binding protein reveal distinct roles in fibrogenic injury.
Collapse
Affiliation(s)
- Elizabeth P. Newberry
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yan Xie
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carlos Lodeiro
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Roberto Solis
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - William Moritz
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Susan Kennedy
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lauren Barron
- Pediatric Surgery Division, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Emily Onufer
- Pediatric Surgery Division, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gianfranco Alpini
- Department of Medical Physiology and Internal Medicine, Texas A&M University, Temple, Texas, USA
- Department of Internal Medicine, Texas A&M University, Temple, Texas, USA
| | - Tianhao Zhou
- Department of Medical Physiology and Internal Medicine, Texas A&M University, Temple, Texas, USA
- Department of Internal Medicine, Texas A&M University, Temple, Texas, USA
| | - William S. Blaner
- Department of Medicine, Columbia University, New York, New York, USA; and
| | - Anping Chen
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Nicholas O. Davidson
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
37
|
Clugston RD, Gao MA, Blaner WS. The Hepatic Lipidome: A Gateway to Understanding the Pathogenes is of Alcohol-Induced Fatty Liver. Curr Mol Pharmacol 2019; 10:195-206. [PMID: 26278391 DOI: 10.2174/1874467208666150817111419] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 12/30/2022]
Abstract
Chronic alcohol consumption can lead to the development of alcoholic fatty liver disease. The underlying pathogenic mechanisms however, have not been fully elucidated. Here, we review the current state of the art regarding the application of lipidomics to study alcohol's effect on hepatic lipids. It is clear that alcohol has a profound effect on the hepatic lipidome, with documented changes in the major lipid categories (i.e. fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, sterol lipids and prenol lipids). Alcohol's most striking effect is the marked change in the hepatic fatty acyl pool. This effect includes increased levels of 18-carbon fatty acyl chains incorporated into multiple lipid species, as well as a general shift toward increased unsaturation of fatty acyl moieties. In addition to our literature review, we also make several recommendations to consider when designing lipidomic studies into alcohol's effects. These recommendations include integration of lipidomic data with other measures of lipid metabolism, inclusion of multiple experimental time points, and presentation of quantitative data. We believe rigorous analysis of the hepatic lipidome can yield new insight into the pathogenesis of alcohol-induced fatty liver. While the existing literature has been largely descriptive, the field is poised to apply lipidomics to yield a new level of understanding on alcohol's effects on hepatic lipid metabolism.
Collapse
Affiliation(s)
- Robin D Clugston
- Department of Physiology, University of Alberta, Edmonton, AB, T6G 2H7. Canada
| | - Madeleine A Gao
- Department of Medicine, Columbia University, New York, NY, 10032. United States
| | - William S Blaner
- Department of Medicine, Columbia University, New York, NY, 10032. United States
| |
Collapse
|
38
|
Dornas W, Glaise D, Bodin A, Sharanek A, Burban A, Le Guillou D, Robert S, Dutertre S, Aninat C, Corlu A, Lagente V. Endotoxin regulates matrix genes increasing reactive oxygen species generation by intercellular communication between palmitate-treated hepatocyte and stellate cell. J Cell Physiol 2018; 234:122-133. [PMID: 30191979 DOI: 10.1002/jcp.27175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/16/2018] [Indexed: 12/19/2022]
Abstract
Previous studies have shown that gut-derived bacterial endotoxins contribute in the progression of simple steatosis to steatohepatitis, although the mechanism(s) remains inaccurate to date. As hepatic stellate cells (HSC) play a pivotal role in the accumulation of excessive extracellular matrix (ECM), leading to collagen deposition, fibrosis, and perpetuation of inflammatory response, an in vitro model was developed to investigate the crosstalk between HSC and hepatocytes (human hepatoma cell) pretreated with palmitate. Bacterial lipopolysaccharide (LPS) stimulated HSC with phosphorylation of the p38 mitogen-activated protein kinase/NF-κB pathway, while several important pro-inflammatory cytokines were upregulated in the presence of hepatocyte-HSC. Concurrently, fibrosis-related genes were regulated by palmitate and the inflammatory effect of endotoxin where cells were more exposed or sensitive to reactive oxygen species (ROS). This interaction was accompanied by increased expression of the mitochondrial master regulator, proliferator-activated receptor gamma coactivator alpha, and a cytoprotective effect of the agent N-acetylcysteine suppressing ROS production, transforming growth factor-β1, and tissue inhibitor of metalloproteinase-1. In summary, our results demonstrate that pro-inflammatory mediators LPS-induced promote ECM rearrangement in hepatic cells transcriptionally committed to the regulation of genes encoding enzymes for fatty acid metabolism in light of differences that might require an alternative therapeutic approach targeting ROS regulation.
Collapse
Affiliation(s)
- Waleska Dornas
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Denise Glaise
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Aude Bodin
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Ahmad Sharanek
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Audrey Burban
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Dounia Le Guillou
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Sacha Robert
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Stephanie Dutertre
- Microscopy Rennes Imaging Center UMS CNRS 3480/US INSERM 018, Biosit, Université de Rennes 1, Rennes, France
| | - Caroline Aninat
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Anne Corlu
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| | - Vincent Lagente
- Nutrition Metabolisms and Cancer Institute, Institut National de la Santé et de la Recherche Médicale U1241, INRA, Université de Rennes 1, Université Bretagne Loire, Rennes, France
| |
Collapse
|
39
|
Animal models of NAFLD from the pathologist's point of view. Biochim Biophys Acta Mol Basis Dis 2018; 1865:929-942. [PMID: 29746920 DOI: 10.1016/j.bbadis.2018.04.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/25/2018] [Accepted: 04/30/2018] [Indexed: 01/18/2023]
Abstract
Fatty liver disease is a multifactorial world-wide health problem resulting from a complex interplay between liver, adipose tissue and intestine and initiated by alcohol abuse, overeating, various types of intoxication, adverse drug reactions and genetic or acquired metabolic defects. Depending on etiology fatty liver disease is commonly categorized as alcoholic or non-alcoholic. Both types may progress from simple steatosis to the necro-inflammatory lesion of alcoholic (ASH) and non-alcoholic steatohepatitis (NASH), respectively, and finally to cirrhosis and hepatocellular carcinoma. Animal models are helpful to clarify aspects of pathogenesis and progression. Generally, they are classified as nutritional (dietary), toxin-induced and genetic, respectively, or represent a combination of these factors. Numerous reviews are dealing with NASH animal models designed to imitate as closely as possible the metabolic situation associated with human disease. This review focuses on currently used mouse models of NASH with particular emphasis on liver morphology. Despite metabolic similarities most models (except those with chemically or genetically induced porphyria or keratin 18-deficiency) fail to develop the morphologic key features of NASH, namely hepatocyte ballooning and formation of histologically and immunohistochemically well-defined Mallory-Denk-Bodies (MDBs). Although MDBs are not universally detectable in ballooned hepatocytes in NASH their experimental reproduction and analysis may, however, significantly contribute to our understanding of important pathogenic aspects of NASH despite the obvious differences in etiology.
Collapse
|
40
|
Lin J, Zheng S, Attie AD, Keller MP, Bernlohr DA, Blaner WS, Newberry EP, Davidson NO, Chen A. Perilipin 5 and liver fatty acid binding protein function to restore quiescence in mouse hepatic stellate cells. J Lipid Res 2018; 59:416-428. [PMID: 29317465 DOI: 10.1194/jlr.m077487] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 12/07/2017] [Indexed: 12/23/2022] Open
Abstract
Hepatic stellate cell (HSC) activation occurs along with decreased Perilipin5 (Plin5) and liver fatty acid-binding protein (L-Fabp) expression and coincident lipid droplet (LD) depletion. Conversely, the activated phenotype is reversible in WT HSCs upon forced expression of Plin5. Here, we asked if L-Fabp expression is required for Plin5-mediated rescue of the quiescent phenotype. Lentiviral Plin5 transduction of passaged L-Fabp-/- HSCs failed to reverse activation markers or restore lipogenic gene expression and LD formation. However, adenoviral L-Fabp infection of lentiviral Plin5 transduced L-Fabp-/- HSCs restored both the quiescent phenotype and LD formation, an effect also mediated by adenoviral intestine-Fabp or adipocyte-Fabp. Expression of exogenous Plin5 in activated WT HSCs induced a transcriptional program of lipogenic gene expression including endogenous L-Fabp, but none of the other FABPs. We further demonstrated that selective, small molecule inhibition of endogenous L-Fabp also eliminated the ability of exogenous Plin5 to rescue LD formation and reverse activation of WT HSCs. This functional coordination of L-Fabp with Plin5 was 5'-AMP-activated protein kinase (AMPK)-dependent and was eliminated by AMPK inhibition. Taken together, our results indicate that L-Fabp is required for Plin5 to activate a transcriptional program that restores LD formation and reverses HSC activation.
Collapse
Affiliation(s)
- Jianguo Lin
- Department of Pathology, School of Medicine, Saint Louis University, St. Louis, MO.,Department of Neurology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shizhong Zheng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Alan D Attie
- Department of Biochemistry, Molecular Biology and Biophysics, University of Wisconsin, Madison, WI, 53706
| | - Mark P Keller
- Department of Biochemistry, Molecular Biology and Biophysics, University of Wisconsin, Madison, WI, 53706
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | | | - Elizabeth P Newberry
- Gastroenterology Division, Washington University School of Medicine, St. Louis, MO 63110
| | - Nicholas O Davidson
- Gastroenterology Division, Washington University School of Medicine, St. Louis, MO 63110
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St. Louis, MO
| |
Collapse
|
41
|
Milligan S, Martin GG, Landrock D, McIntosh AL, Mackie JT, Schroeder F, Kier AB. Ablating both Fabp1 and Scp2/Scpx (TKO) induces hepatic phospholipid and cholesterol accumulation in high fat-fed mice. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:323-338. [PMID: 29307784 DOI: 10.1016/j.bbalip.2017.12.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/13/2017] [Accepted: 12/31/2017] [Indexed: 01/16/2023]
Abstract
Although singly ablating Fabp1 or Scp2/Scpx genes may exacerbate the impact of high fat diet (HFD) on whole body phenotype and non-alcoholic fatty liver disease (NAFLD), concomitant upregulation of the non-ablated gene, preference for ad libitum fed HFD, and sex differences complicate interpretation. Therefore, these issues were addressed in male and female mice ablated in both genes (Fabp1/Scp2/Scpx null or TKO) and pair-fed HFD. Wild-type (WT) males gained more body weight as fat tissue mass (FTM) and exhibited higher hepatic lipid accumulation than WT females. The greater hepatic lipid accumulation in WT males was associated with higher hepatic expression of enzymes in glyceride synthesis, higher hepatic bile acids, and upregulation of transporters involved in hepatic reuptake of serum bile acids. While TKO had little effect on whole body phenotype and hepatic bile acid accumulation in either sex, TKO increased hepatic accumulation of lipids in both, specifically phospholipid and cholesteryl esters in males and females and free cholesterol in females. TKO-induced increases in glycerides were attributed not only to complete loss of FABP1, SCP2 and SCPx, but also in part to sex-dependent upregulation of hepatic lipogenic enzymes. These data with WT and TKO mice pair-fed HFD indicate that: i) Sex significantly impacted the ability of HFD to increase body weight, induce hepatic lipid accumulation and increase hepatic bile acids; and ii) TKO exacerbated the HFD ability to induce hepatic lipid accumulation, regardless of sex, but did not significantly alter whole body phenotype in either sex.
Collapse
Affiliation(s)
- Sherrelle Milligan
- Department of Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA
| | - Gregory G Martin
- Department of Physiology/Pharmacology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4466, USA
| | - Danilo Landrock
- Department of Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA
| | - Avery L McIntosh
- Department of Physiology/Pharmacology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4466, USA
| | - John T Mackie
- Department of Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA
| | - Friedhelm Schroeder
- Department of Physiology/Pharmacology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4466, USA
| | - Ann B Kier
- Department of Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA.
| |
Collapse
|
42
|
Cui CX, Deng JN, Yan L, Liu YY, Fan JY, Mu HN, Sun HY, Wang YH, Han JY. Silibinin Capsules improves high fat diet-induced nonalcoholic fatty liver disease in hamsters through modifying hepatic de novo lipogenesis and fatty acid oxidation. JOURNAL OF ETHNOPHARMACOLOGY 2017; 208:24-35. [DOI: 10.1016/j.jep.2017.06.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
43
|
Marinho TDS, Kawasaki A, Bryntesson M, Souza-Mello V, Barbosa-da-Silva S, Aguila MB, Mandarim-de-Lacerda CA. Rosuvastatin limits the activation of hepatic stellate cells in diet-induced obese mice. Hepatol Res 2017; 47:928-940. [PMID: 27653239 DOI: 10.1111/hepr.12821] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 09/07/2016] [Accepted: 09/19/2016] [Indexed: 12/14/2022]
Abstract
AIM The aim of this study was to investigate the effects of rosuvastatin in a model of diet-induced obesity and non-alcoholic fatty liver disease, with attention to the activation of hepatic stellate cells (HSCs). METHOD Male C57BL/6 mice received a control diet (C; 10% energy as lipids) or a high-fat diet (HF; 50% energy as lipids) for 12 weeks, followed by 7 weeks of treatment. Group CR received control diet + rosuvastatin; group HFR received high-fat diet + rosuvastatin. RESULTS The HF group showed higher insulin, total cholesterol, triacylglycerol, and leptin levels than the C group, all of which were significantly diminished by rosuvastatin in the HFR group. The HF group had greater steatosis and activated HSCs than the C group, whereas rosuvastatin diminished the steatosis (less 21%, P < 0.001) and significantly inhibited the activation of the HSCs in the HFR group compared to the HF group. The sterol regulatory element-binding protein-1 and the peroxisome proliferator-activated receptor (PPAR)-γ protein expressions were increased in HF animals and reduced after treatment in the HFR group. By contrast, low PPAR-α and carnitine palmitoyltransferase-1 expressions were found in the HF group, and were restored by rosuvastatin treatment in the HFR group. CONCLUSION Rosuvastatin mitigated hepatic steatosis by modulating PPAR balance, favoring PPAR-α over PPAR-γ downstream effects. The effects were accompanied by a diminishing of insulin resistance, the anti-inflammatory adipokine profile, and HSC activation, avoiding non-alcoholic fatty liver disease progression and non-alcoholic steatohepatitis onset in this model.
Collapse
Affiliation(s)
- Thatiany de Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriana Kawasaki
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sandra Barbosa-da-Silva
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia B Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Zheng Z, Zhao Z, Li S, Lu X, Jiang M, Lin J, An Y, Xie Y, Xu M, Shen W, Guo GL, Huang Y, Li S, Zhang X, Xie W. Altenusin, a Nonsteroidal Microbial Metabolite, Attenuates Nonalcoholic Fatty Liver Disease by Activating the Farnesoid X Receptor. Mol Pharmacol 2017; 92:425-436. [PMID: 28739572 DOI: 10.1124/mol.117.108829] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/07/2017] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disease. The incidence of NAFLD has increased steadily due to its close association with the global epidemic of obesity and type 2 diabetes. However, there is no effective pharmacological therapy approved for NAFLD. Farnesoid X receptor (FXR), a member of the nuclear receptor subfamily, plays important roles in maintaining the homeostasis of bile acids, glucose, and lipids. FXR agonists have shown promise for the treatment of NAFLD. In this study, we report altenusin (2076A), a natural nonsteroidal fungal metabolite, as a novel selective agonist of FXR with an EC50 value of 3.2 ± 0.2 μM. Administration of 2076A protected mice from high-fat diet (HFD)-induced obesity by reducing the body weight and fat mass by 22.9% and 50.0%, respectively. Administration of 2076A also decreased the blood glucose level from 178.3 ± 12.4 mg/dl to 116.2 ± 4.1 mg/dl and the serum insulin level from 1.4 ± 0.6 ng/dl to 0.4 ± 0.1 ng/dl. Moreover, 2076A treatment nearly reversed HFD-induced hepatic lipid droplet accumulation and macrovesicular steatosis. These metabolic effects were abolished in FXR knockout mice. Mechanistically, the metabolic benefits of 2076A might have been accounted for by the increased insulin sensitivity and suppression of genes that are involved in hepatic gluconeogenesis and lipogenesis. In summary, we have uncovered a new class of nonsteroidal FXR agonist that shows promise in treating NAFLD and the associated metabolic syndrome.
Collapse
Affiliation(s)
- Zhihui Zheng
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Zanmei Zhao
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Shuqiang Li
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Xinhua Lu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Mengxi Jiang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Jie Lin
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Yunqi An
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Yang Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Wenbin Shen
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Grace L Guo
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Yixian Huang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Song Li
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Xuexia Zhang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences (Zh.Z., Za.Z., M.J., Y.A., Y.X., M.X., Y.H., S.L., W.X.) and Department of Pharmacology and Chemical Biology (W.X.), University of Pittsburgh, Pittsburgh, Pennsylvania; New Drug Research and Development Center, North China Pharmaceutical Group, Shijiazhuang, Hebei, China (Zh.Z., X.L., J.L., W.S., X.Z.); Occupational Disease Department, Peking University Third Hospital, Beijing, China (Za.Z., S.L.); and Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey (G.L.G.)
| |
Collapse
|
45
|
McCommis KS, Hodges WT, Brunt EM, Nalbantoglu ILK, McDonald WG, Holley C, Fujiwara H, Schaffer JE, Colca JR, Finck BN. Targeting the mitochondrial pyruvate carrier attenuates fibrosis in a mouse model of nonalcoholic steatohepatitis. Hepatology 2017; 65:1543-1556. [PMID: 28027586 PMCID: PMC5397348 DOI: 10.1002/hep.29025] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/30/2016] [Accepted: 12/22/2016] [Indexed: 12/30/2022]
Abstract
Diseases of the liver related to metabolic syndrome have emerged as the most common and undertreated hepatic ailments. The cause of nonalcoholic fatty liver disease is the aberrant accumulation of lipid in hepatocytes, though the mechanisms whereby this leads to hepatocyte dysfunction, death, and hepatic fibrosis are still unclear. Insulin-sensitizing thiazolidinediones have shown efficacy in treating nonalcoholic steatohepatitis (NASH), but their widespread use is constrained by dose-limiting side effects thought to be due to activation of the peroxisome proliferator-activated receptor γ. We sought to determine whether a next-generation thiazolidinedione with markedly diminished ability to activate peroxisome proliferator-activated receptor γ (MSDC-0602) would retain its efficacy for treating NASH in a rodent model. We also determined whether some or all of these beneficial effects would be mediated through an inhibitory interaction with the mitochondrial pyruvate carrier 2 (MPC2), which was recently identified as a mitochondrial binding site for thiazolidinediones, including MSDC-0602. We found that MSDC-0602 prevented and reversed liver fibrosis and suppressed expression of markers of stellate cell activation in livers of mice fed a diet rich in trans-fatty acids, fructose, and cholesterol. Moreover, mice with liver-specific deletion of MPC2 were protected from development of NASH on this diet. Finally, MSDC-0602 directly reduced hepatic stellate cell activation in vitro, and MSDC-0602 treatment or hepatocyte MPC2 deletion also limited stellate cell activation indirectly by affecting secretion of exosomes from hepatocytes. CONCLUSION Collectively, these data demonstrate the effectiveness of MSDC-0602 for attenuating NASH in a rodent model and suggest that targeting hepatic MPC2 may be an effective strategy for pharmacologic development. (Hepatology 2017;65:1543-1556).
Collapse
Affiliation(s)
- Kyle S. McCommis
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Wesley T. Hodges
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Elizabeth M. Brunt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - ILKe Nalbantoglu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | | | - Christopher Holley
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Hideji Fujiwara
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jean E. Schaffer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jerry R. Colca
- Metabolic Solutions Development Company, Kalamazoo, MI 49007
| | - Brian N. Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
46
|
Asimakopoulou A, Fülöp A, Borkham-Kamphorst E, de Leur EV, Gassler N, Berger T, Beine B, Meyer HE, Mak TW, Hopf C, Henkel C, Weiskirchen R. Altered mitochondrial and peroxisomal integrity in lipocalin-2-deficient mice with hepatic steatosis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2093-2110. [PMID: 28396286 DOI: 10.1016/j.bbadis.2017.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/04/2017] [Accepted: 04/06/2017] [Indexed: 01/10/2023]
Abstract
Lipocalin-2 (LCN2) is a secreted adipokine that transports small hydrophobic molecules such as fatty acids and steroids. LCN2 limits bacterial growth by sequestering iron-containing siderophores and in mammalian liver protects against inflammation, infection, injury and other stressors. Because LCN2 modulates hepatic fat metabolism and homeostasis, we performed a comparative profiling of proteins and lipids of wild type (WT) and Lcn2-deficient mice fed either standard chow or a methionine- and choline-deficient (MCD) diet. Label-free proteomics and 2D-DIGE protein expression profiling revealed differential expression of BRIT1/MCPH1, FABP5, HMGB1, HBB2, and L-FABP, results confirmed by Western blotting. Gene ontology enrichment analysis identified enrichment for genes associated with mitochondrial membrane permeabilization and metabolic processes involving carboxylic acid. Measurements of mitochondrial membrane potential, mitochondrial chelatable iron pool, intracellular lipid peroxidation, and peroxisome numbers in primary hepatocytes confirmed that LCN2 regulates mitochondrial and peroxisomal integrity. Matrix-Assisted Laser Desorption/Ionization Time-Of-Flight (MALDI-TOF) mass spectrometry imaging identified significant changes to sphingomyelins, triglycerides, and glycerophospholipids in livers of mice fed an MCD diet regardless of LCN2 status. However, two arachidonic acid-containing glycerophospholipids were increased in Lcn2-deficient livers. Thus, LCN2 influences peroxisomal and mitochondrial biology in the liver to maintain triglyceride balance, handle oxidative stress, and control apoptosis.
Collapse
Affiliation(s)
- Anastasia Asimakopoulou
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Annabelle Fülöp
- Applied Research Center in Biomedical Mass Spectrometry (ABIMAS), Instrumental Analysis and Bioanalysis, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Erawan Borkham-Kamphorst
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Eddy Van de Leur
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| | | | - Thorsten Berger
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada
| | - Birte Beine
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany; Medizinisches Proteom-Center, Ruhr-University, Bochum, Germany
| | - Helmut E Meyer
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Tak W Mak
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada; Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Carsten Hopf
- Applied Research Center in Biomedical Mass Spectrometry (ABIMAS), Instrumental Analysis and Bioanalysis, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Corinna Henkel
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany; Medizinisches Proteom-Center, Ruhr-University, Bochum, Germany; Bruker Daltonik GmbH, Bremen
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany.
| |
Collapse
|
47
|
Newberry EP, Xie Y, Kennedy SM, Graham MJ, Crooke RM, Jiang H, Chen A, Ory DS, Davidson NO. Prevention of hepatic fibrosis with liver microsomal triglyceride transfer protein deletion in liver fatty acid binding protein null mice. Hepatology 2017; 65:836-852. [PMID: 27862118 PMCID: PMC5319898 DOI: 10.1002/hep.28941] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/24/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022]
Abstract
UNLABELLED Blocking hepatic very low-density lipoprotein secretion through genetic or pharmacologic inhibition of microsomal triglyceride transfer protein (Mttp) causes hepatic steatosis, yet the risks for developing hepatic fibrosis are poorly understood. We report that liver-specific Mttp knockout mice (Mttp-LKO) exhibit both steatosis and fibrosis, which is exacerbated by a high-transfat/fructose diet. When crossed into germline liver fatty acid (FA) binding protein null mice (Mttp-LKO, i.e., double knockout mice) hepatic steatosis was greatly diminished and fibrosis prevented, on both low-fat and high-fat diets. The mechanisms underlying protection include reduced long chain FA uptake, shifts in FA distribution (lipidomic profiling), and metabolic turnover, specifically decreased hepatic 18:2 FA and triglyceride species and a shift in 18:2 FA use for oxidation versus incorporation into newly synthesized triglyceride. Double knockout mice were protected against fasting-induced hepatic steatosis (a model of enhanced exogenous FA delivery) yet developed steatosis upon induction of hepatic de novo lipogenesis with fructose feeding. Mttp-LKO mice, on either the liver FA binding protein null or Apobec-1 null background (i.e., apolipoprotein B100 only) exhibited only subtle increases in endoplasmic reticulum stress, suggesting that an altered unfolded protein response is unlikely to account for the attenuated phenotype in double knockout mice. Acute, antisense-mediated liver FA binding protein knockdown in Mttp-LKO mice also reduced FA uptake, increased oxidation versus incorporation of 18:2 species with complete reversal of hepatic steatosis, increased hepatic injury, and worsened fibrosis. CONCLUSION Perturbing exogenous hepatic FA use modulates both hepatic steatosis and fibrosis in the setting of hepatic Mttp deletion, adding new insight into the pathophysiological mechanisms and consequences of defective very low-density lipoprotein secretion. (Hepatology 2017;65:836-852).
Collapse
Affiliation(s)
- Elizabeth P. Newberry
- Department of Medicine Washington University School of Medicine Saint Louis, MO 63110
| | - Yan Xie
- Department of Medicine Washington University School of Medicine Saint Louis, MO 63110
| | - Susan M. Kennedy
- Department of Medicine Washington University School of Medicine Saint Louis, MO 63110
| | | | | | - Hui Jiang
- Department of Medicine Washington University School of Medicine Saint Louis, MO 63110
| | - Anping Chen
- Department of Pathology, Saint Louis University Saint Louis, MO
| | - Daniel S. Ory
- Department of Medicine Washington University School of Medicine Saint Louis, MO 63110
| | - Nicholas O. Davidson
- Department of Medicine Washington University School of Medicine Saint Louis, MO 63110,Correspondence:
| |
Collapse
|
48
|
Liver fatty acid-binding protein (L-FABP) promotes cellular angiogenesis and migration in hepatocellular carcinoma. Oncotarget 2017; 7:18229-46. [PMID: 26919097 PMCID: PMC4951284 DOI: 10.18632/oncotarget.7571] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 01/29/2016] [Indexed: 01/18/2023] Open
Abstract
Liver fatty acid-binding protein (L-FABP) is abundant in hepatocytes and known to be involved in lipid metabolism. Overexpression of L-FABP has been reported in various cancers; however, its role in hepatocellular carcinoma (HCC) remains unclear. In this study, we investigated L-FABP and its association with vascular endothelial growth factors (VEGFs) in 90 HCC patients. We found that L-FABP was highly expressed in their HCC tissues, and that this expression was positively correlated with that of VEGF-A. Additionally, L-FABP significantly promoted tumor growth and metastasis in a xenograft mouse model. We also assessed the mechanisms of L-FABP activity in tumorigenesis; L-FABP was found to associate with VEGFR2 on membrane rafts and subsequently activate the Akt/mTOR/P70S6K/4EBP1 and Src/FAK/cdc42 pathways, which resulted in up-regulation of VEGF-A accompanied by an increase in both angiogenic potential and migration activity. Our results thus suggest that L-FABP could be a potential target for HCC chemotherapy.
Collapse
|
49
|
Perilipin 5 restores the formation of lipid droplets in activated hepatic stellate cells and inhibits their activation. J Transl Med 2016; 96:791-806. [PMID: 27135793 DOI: 10.1038/labinvest.2016.53] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 02/10/2016] [Accepted: 03/07/2016] [Indexed: 12/15/2022] Open
Abstract
Hepatic stellate cells (HSC) are major effectors during hepatic fibrogenesis. The activation of HSC is coupled to the loss of lipid droplets (LDs), which are specialized organelles composed of neutral lipids surrounded by perilipins. LDs have emerged as a focal point of interest in understanding the metabolic regulation of intrahepatic lipids during lipid-mediated liver fibrogenesis. Perilipin 5 (Plin5) is a newly identified LD protein in the perilipin family, which plays a key role in regulating aspects of intracellular trafficking, signaling, and cytoskeletal organization in hepatocytes. Recent work in Plin5 knockout mice suggests a role in high fat diet-induced hepatic lipotoxicity. The current report is to evaluate the impact of Plin5 on HSC activation and to elucidate the underlying mechanisms. We now show that high fat diet-induced liver fibrosis is accompanied by an approximate 75% reduction in Plin5 in HSC, and that spontaneous activation of primary HSC produces temporally coincident loss of Plin5 expression and LD depletion. As modulating lipid content in HSC is a suggested strategy for inhibition of HSC activation and treatment of hepatic fibrosis, we asked whether exogenous Plin5 expression in primary HSC would reverse the activation phenotype and promote LD formation. Recombinant lentiviral Plin5 expression in primary mouse HSC restored the formation of LDs, increased lipid content by inducing expression of pro-lipogenic genes and suppressing expression of pro-lipolytic genes, and suppressed HSC activation (~two fold reduction in expression of procollagen and α-smooth muscle actin, two unique biomarkers for activated HSC). In addition, the expression of exogenous Plin5 in HSC attenuated cellular oxidative stress by reducing cellular reactive oxygen species, elevating cellular glutathione, and inducing gene expression of glutamate-cysteine ligase. Taken together, our results indicate that expression of Plin5 plays a critical role in the formation of LDs, the elevation of lipid content in HSC, and the inhibition of the activation of HSC.
Collapse
|
50
|
Noureddin M, Anstee QM, Loomba R. Review article: emerging anti-fibrotic therapies in the treatment of non-alcoholic steatohepatitis. Aliment Pharmacol Ther 2016; 43:1109-23. [PMID: 27061197 PMCID: PMC5906100 DOI: 10.1111/apt.13620] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 02/23/2016] [Accepted: 03/21/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) can lead to non-alcoholic steatohepatitis (NASH) and cirrhosis. Fibrosis predicts worse outcomes and mortality. New treatments targeting fibrosis are being investigated to reverse disease progression. AIM To review the new pipeline therapeutic agents targeting fibrosis in NASH patients, with particular focus on clinical trials in which reversing fibrosis and portal hypertension are the primary outcomes. METHODS The literature was searched in PubMed between January 2000 and January 2016 using search terms non-alcoholic fatty liver disease and NASH, with filters of 'English language'. We focused on fibrosis improvement as the key outcome. We also searched the ClinicalTrials.gov for promising agents that target fibrosis in NASH patients. RESULTS Significant advances have been made on approaches targeting fibrosis in NASH patients. Many therapeutic agents are already in development, some of which have shown promising results in preclinical and phase I studies. Novel therapies have entered phase II and III studies targeting fibrosis reversal and/or improvement in portal hypertension. Innovative studies have also started looking into combining these agents, aiming at different mechanisms to maximise therapeutic outcomes. We found five clinical trials in phase II and one in phase III focusing on fibrosis in NASH patients as key outcomes. One of the phase II trials is using combination therapy to target fibrosis. CONCLUSIONS Ongoing research studies are already investigating new pathways aimed at reversing fibrosis in NASH patients. Novel therapeutic agents are in development and are expected to offer unique options to NASH patients with advanced fibrosis.
Collapse
Affiliation(s)
- M. Noureddin
- Fatty Liver Program, Division of Digestive and Liver Diseases, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Q. M. Anstee
- Liver Research Group, Institute of Cellular Medicine, The Medical School, Newcastle University, Newcastle-upon-Tyne, UK
| | - R. Loomba
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA,Division of Epidemiology, University of California, San Diego, La Jolla, CA, USA,NAFLD Research Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|