1
|
Tao J, Chen Z, Xie Q, Bai H, Li Z, Jia Y, Zhang X, Xiao Y, Bu T, Yuan M, Li Q, Tang Z. Buckwheat resistant starch alleviates hyperlipidaemia in mice by inhibiting lipid accumulation and regulating gut microbiota. Int J Biol Macromol 2025; 310:143446. [PMID: 40274138 DOI: 10.1016/j.ijbiomac.2025.143446] [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: 02/08/2025] [Revised: 04/03/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
Hyperlipidemia, a prevalent metabolic disorder disease, has become a significant global health challenge. In this research, we looked into the effects of buckwheat resistant starch (BRS) supplementation on lipid metabolism and gut microbiota in hyperlipidemic mice. Results showed that HBRS intervention significantly inhibited weight gain in high-fat diet mice, reducing body weight by 2.91 g versus MC group. Serum TC, TG, and LDL-C decreased by 74.25 %, 76.79 %, and 56.25 % respectively, while HDL-C increased by 67.66 %. HBRS mitigated hepatic steatosis and epididymal adipocyte hypertrophy. HBRS downregulated TNF-α and IL-6 by 24.4 % and 18.25 %, respectively, and upregulated IL-10 by 15.35 %. It modulated lipid metabolism genes by suppressing SREBP2, FAS, SCD1 and activating PPARα/CPT1. Moreover, BRS improved gut microbiota composition by lowering the F/B ratio and enhancing populations of SCFA-producing bacteria (e.g., Akkermansia, Bifidobacterium, and Parabacteroides). BRS significantly increased the total levels of SCFAs in the gut. Compared to the MC group, HBRS resulted in increases of 40.2 %, 51.8 %, and 68.3 % in acetate, propionate, and butyrate, respectively. Collectively, these results demonstrate that BRS serves as a beneficial nutritional resource for the management of hyperlipidemia.
Collapse
Affiliation(s)
- Jiwen Tao
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Zhao Chen
- Ya'an People's Hospital, Ya'an 625014, China
| | - Qiqi Xie
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Hongjie Bai
- Chengdu Jinkai Bioengineering Co., Ltd., Chen'du 611100, China; Sichuan Food Fermentation Industry Research and Design Institute Co., Ltd, Chen'du 611100, China
| | - Zhenjiang Li
- Chengdu Jinkai Bioengineering Co., Ltd., Chen'du 611100, China; Sichuan Food Fermentation Industry Research and Design Institute Co., Ltd, Chen'du 611100, China
| | - Yujie Jia
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Xinyu Zhang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Yirong Xiao
- Sichuan Agricultural University Hospital, Ya'an 625014, China
| | - Tongliang Bu
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Ming Yuan
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Qingfeng Li
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China.
| | - Zizhong Tang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China.
| |
Collapse
|
2
|
Shin HJ, Kim KE, An HS, Jeong EA, Oh J, Sun Y, Park DJ, Lee J, Yang J, Roh GS. Carbon tetrachloride does not promote hepatic fibrosis in ob/ob mice via downregulation of lipocalin-2 protein. Redox Biol 2025; 80:103506. [PMID: 39832399 PMCID: PMC11787671 DOI: 10.1016/j.redox.2025.103506] [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: 11/23/2024] [Revised: 01/01/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
Although leptin-deficient ob/ob mice have been investigated to determine whether hepatic steatosis promotes susceptibility to hepatotoxic insults, carbon tetrachloride (CCl4)-induced hepatic fibrosis in ob/ob mice remains largely unknown. In this study, we evaluate the pathogenic mechanisms of hepatic fibrosis in CCl4-treated wild-type (WT) and ob/ob mice and analyze some parameters related to lipogenesis, inflammation, fibrosis, oxidative stress, apoptosis, and autophagy. CCl4 treatment attenuated liver weight and lipogenesis in ob/ob mice. Increased hepatic fibrosis-related proteins were reduced in CCl4-treated ob/ob mice compared with CCl4-treated WT mice. Specifically, the expression of lipocalin-2 (LCN2) was markedly reduced in CCl4-treated ob/ob mice versus CCl4-treated WT mice. Compared with CCl4-treated WT mice, CCl4-treated ob/ob mice had reduced expression of neutrophil-related inflammatory genes and proteins. Hepatic heme oxygenase-1 protein was reduced in CCl4-treated ob/ob mice compared with CCl4-treated WT mice. However, CCl4 did not promote hepatic apoptosis in ob/ob mice. Therefore, these findings highlight LCN2 as a key signaling factor in CCl4-induced hepatic fibrosis.
Collapse
Affiliation(s)
- Hyun Joo Shin
- Department of Anatomy and Convergence Medical Science, College of Medicine, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Kyung Eun Kim
- Department of Anatomy and Convergence Medical Science, College of Medicine, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Hyeong Seok An
- Department of Anatomy and Convergence Medical Science, College of Medicine, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Eun Ae Jeong
- Department of Anatomy and Convergence Medical Science, College of Medicine, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Jiwon Oh
- Department of Anatomy and Convergence Medical Science, College of Medicine, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Yundong Sun
- Department of Anatomy and Convergence Medical Science, College of Medicine, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Dong-Ju Park
- Department of Anatomy and Convergence Medical Science, College of Medicine, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Jaewoong Lee
- Department of Anatomy and Convergence Medical Science, College of Medicine, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Jinsung Yang
- Department of Biochemistry, College of Medicine, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, College of Medicine, Metabolic Dysfunction Liver Disease Research Center, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea.
| |
Collapse
|
3
|
Saijou E, Kamiya Y, Fujiki K, Shirahige K, Nakato R. Modulation of liver cholesterol homeostasis by choline supplementation during fibrosis resolution. Heliyon 2024; 10:e36727. [PMID: 39296030 PMCID: PMC11407984 DOI: 10.1016/j.heliyon.2024.e36727] [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: 06/26/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Liver fibrosis is a critical global health challenge, often leading to severe liver diseases without timely intervention. Choline deficiency has been linked to metabolic dysfunction associated steatohepatitis (MASH) and liver fibrosis, suggesting choline supplementation as a potential therapeutic approach. This study aimed to explore the therapeutic potential of choline supplementation in liver fibrosis resolution and its effects on cholesterol homeostasis using a mouse model with induced liver fibrosis. Our findings reveal that choline supplementation significantly decreases blood lactate dehydrogenase (LDH) and non-high-density lipoprotein cholesterol (non-HDL-C) levels. Transcriptome analysis showed that choline supplementation primarily induces genes related to cholesterol homeostasis, suggesting a significant impact on liver cholesterol synthesis. However, choline supplementation did not significantly alter the expression of fibrosis-related, choline metabolism-related, or epigenetics-related genes. This study provides novel insights into the role of choline in liver health and cholesterol metabolism, potentially informing treatments for liver fibrosis and related conditions.
Collapse
Affiliation(s)
- Eiko Saijou
- Laboratory of Computational Genomics, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Yoshiko Kamiya
- Laboratory of Computational Genomics, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Katsunori Fujiki
- Laboratory of Genome Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Katsuhiko Shirahige
- Laboratory of Genome Structure and Function, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Ryuichiro Nakato
- Laboratory of Computational Genomics, Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| |
Collapse
|
4
|
Li R, Zhao M, Miao C, Shi X, Lu J. Identification and validation of key biomarkers associated with macrophages in nonalcoholic fatty liver disease based on hdWGCNA and machine learning. Aging (Albany NY) 2023; 15:15451-15472. [PMID: 38147020 PMCID: PMC10781485 DOI: 10.18632/aging.205374] [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: 08/19/2023] [Accepted: 11/21/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND NAFLD has attracted increasing attention because of its high prevalence and risk of progression to cirrhosis or even hepatocellular carcinoma. Therefore, research into the root causes and molecular indicators of NAFLD is crucial. METHODS We analyzed scRNA-seq data and RNA-seq data from normal and NAFLD liver samples. We utilized hdWGCNA to find module-related genes associated with the phenotype. Multiple machine learning algorithms were used to validate the model diagnostics and further screen for genes that are characteristic of NAFLD. The NAFLD mouse model was constructed using the MCD diet to validate the diagnostic effect of the genes. RESULTS We identified a specific macrophage population called NASH-macrophages by single-cell sequencing analysis. Cell communication analysis and Pseudo-time trajectory analysis revealed the specific role and temporal distribution of NASH-macrophages in NAFLD. The hdWGCNA screening yielded 30 genes associated with NASH-macrophages, and machine learning algorithms screened and obtained two genes characterizing NAFLD. The immune infiltration indicated that these genes were highly associated with macrophages. Notably, we verified by RT-qPCR, IHC, and WB that MAFB and CX3CR1 are highly expressed in the MCD mouse model and may play important roles. CONCLUSIONS Our study revealed a macrophage population that is closely associated with NAFLD. Using hdWGCNA analysis and multiple machine learning algorithms, we identified two NAFLD signature genes that are highly correlated with macrophages. Our findings may provide potential feature markers and therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Ruowen Li
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Mingjian Zhao
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Chengxu Miao
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Xiaojia Shi
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Jinghui Lu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
5
|
Smiriglia A, Lorito N, Serra M, Perra A, Morandi A, Kowalik MA. Sex difference in liver diseases: How preclinical models help to dissect the sex-related mechanisms sustaining NAFLD and hepatocellular carcinoma. iScience 2023; 26:108363. [PMID: 38034347 PMCID: PMC10682354 DOI: 10.1016/j.isci.2023.108363] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Abstract
Only a few preclinical findings are confirmed in the clinic, posing a critical issue for clinical development. Therefore, identifying the best preclinical models can help to dissect molecular and mechanistic insights into liver disease pathogenesis while being clinically relevant. In this context, the sex relevance of most preclinical models has been only partially considered. This is particularly significant in NAFLD and HCC, which have a higher prevalence in men when compared to pre-menopause women but not to those in post-menopausal status, suggesting a role for sex hormones in the pathogenesis of the diseases. This review gathers the sex-relevant findings and the available preclinical models focusing on both in vitro and in vivo studies and discusses the potential implications and perspectives of introducing the sex effect in the selection of the best preclinical model. This is a critical aspect that would help to tailor personalized therapies based on sex.
Collapse
Affiliation(s)
- Alfredo Smiriglia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Nicla Lorito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Marina Serra
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Andrea Perra
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Marta Anna Kowalik
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| |
Collapse
|
6
|
Cheng Z, Li N, Chen Z, Li K, Qiao D, Zhao S, Zhang B. Ingesting retrograded rice (Oryza sativa) starch relieves high-fat diet induced hyperlipidemia in mice by altering intestinal bacteria. Food Chem 2023; 426:136540. [PMID: 37315419 DOI: 10.1016/j.foodchem.2023.136540] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/06/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
High-fat diet is a risk factor for many chronic diseases, whose symptoms are probably regulated by ingesting food ingredients such as resistant starch. For cooked rice stored in cold-chain, the starch component can retrograde to generate ordered structures (helices and crystallites) and become resistant. However, the role of retrograded starch in managing hyperlipidemia symptoms is insufficiently understood. Here, compared to the normal high-fat diet, ingesting retrograded starch reduced the triglyceride and low-density lipoprotein cholesterol levels of high-fat diet mice by 17.69% and 41.33%, respectively. This relieved hyperlipidemia could be linked to the changes in intestinal bacteria. Retrograded starch intervention increased the relative abundance of Bacteroides (2.30 times higher), which produces propionic acid (increased by 8.26%). Meanwhile, Bacteroides were positively correlated with butyric acid (increased by 98.4%) with strong anti-inflammatory functions. Hence, retrograded starch intervention may regulate the body's health by altering intestinal bacteria.
Collapse
Affiliation(s)
- Zihang Cheng
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070 China; College of Food Science, Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China
| | - Nannan Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070 China
| | - Zhining Chen
- Chinese Cereals and Oils Association, Beijing 100037, China
| | - Kaikai Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070 China
| | - Dongling Qiao
- College of Food Science, Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China
| | - Siming Zhao
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070 China
| | - Binjia Zhang
- College of Food Science, Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China.
| |
Collapse
|
7
|
Ntambi JM, Liu X, Burhans MS, ALjohani A, Selen ES, Kalyesubula M, Assadi-Porter F. Hepatic oleate regulates one-carbon metabolism during high carbohydrate feeding. Biochem Biophys Res Commun 2023; 651:62-69. [PMID: 36791500 PMCID: PMC9992055 DOI: 10.1016/j.bbrc.2023.02.018] [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: 01/02/2023] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
Obesity is a major risk factor for type 2 diabetes, coronary heart disease, and strok. These diseases are associated with profound alterations in gene expression in metabolic tissues. Epigenetic-mediated regulation of gene expression is one mechanism through which environmental factors, such as diet, modify gene expression and disease predisposition. However, epigenetic control of gene expression in obesity and insulin resistance is not fully characterized. We discovered that liver-specific stearoyl-CoA desaturase-1 (Scd1) knockout mice (LKO) fed a high-carbohydrate low-fat diet exhibit dramatic changes in hepatic gene expression and metabolites of the folate cycle and one-carbon metabolism respectively for the synthesis of S-adenosylmethionine (SAM). LKO mice show an increased ratio of S-adenosylmethionine to S-adenosylhomocysteine, a marker for increased cellular methylation capacity. Furthermore, expression of DNA and histone methyltransferase genes is up-regulated while the mRNA and protein levels of the non-DNA methyltransferases including phosphatidylethanolamine methyltransferase (PEMT), Betaine homocysteine methyltransferase (Bhmt), and the SAM-utilizing enzymes such as glycine-N-methyltransferase (Gnmt) and guanidinoacetate methyltransferase (Gamt) are generally down-regulated. Feeding LKO mice a high carbohydrate diet supplemented with triolein, but not tristearin, and increased endogenous hepatic synthesis of oleate but not palmitoleate in Scd1 global knockout mice normalized one carbon gene expression and metabolite levels. Additionally, changes in one carbon gene expression are independent of the PGC-1α-mediated ER stress response previously reported in the LKO mice. Together, these results highlight the important role of oleate in maintaining one-carbon cycle homeostasis and point to observed changes in one-carbon metabolism as a novel mediator of the Scd1 deficiency-induced liver phenotype.
Collapse
Affiliation(s)
- James M Ntambi
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Xueqing Liu
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Maggie S Burhans
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Ahmed ALjohani
- College of Science and Health Professions, King Saudi Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Ebru Selin Selen
- Integrative Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Mugagga Kalyesubula
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Fariba Assadi-Porter
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Integrative Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
8
|
Shedding light on non-alcoholic fatty liver disease: Pathogenesis, molecular mechanisms, models, and emerging therapeutics. Life Sci 2022; 312:121185. [PMID: 36375569 DOI: 10.1016/j.lfs.2022.121185] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder globally impacting an estimated 25% of the population associated with severe consequences such as cirrhosis, hepatocellular carcinoma (HCC), and overall mortality. Fatty liver disease is triggered through multiple pathways, but the most prominent cause is either diabetes or obesity, or a combination of both. Therefore, hepatic glucose, insulin and fatty acid signaling becomes a dire need to understand which is well elaborated in this review. This review summarizes the popular two-hit pathogenesis of NAFLD, the molecular mechanisms underlying hepatic insulin resistance. As fatty liver disease gets advanced, it requires in-vitro as well as in-vivo models closer to disease progression in humans for better understanding the pathological state and identifying a novel therapeutic target. This review summarizes in-vitro (2D cell-culture/co-culture, 3D spheroid/organoid/liver-on-a-chip) models as well as in-vivo (genetically/dietary/chemically induced fatty liver disease) research models. Fatty liver disease research has gathered lots of attention recently since there is no FDA approved therapy available so far. However, there have been numerous promising targets to treat fatty liver disease including potential therapeutic targets under clinical trials are listed in this review.
Collapse
|
9
|
Zhang Y, Jia XB, Liu YC, Yu WQ, Si YH, Guo SD. Fenofibrate enhances lipid deposition via modulating PPARγ, SREBP-1c, and gut microbiota in ob/ob mice fed a high-fat diet. Front Nutr 2022; 9:971581. [PMID: 36172518 PMCID: PMC9511108 DOI: 10.3389/fnut.2022.971581] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity is characterized by lipid accumulation in distinct organs. Presently, fenofibrate is a commonly used triglyceride-lowering drug. This study is designed to investigate whether long-term fenofibrate intervention can attenuate lipid accumulation in ob/ob mouse, a typical model of obesity. Our data demonstrated that fenofibrate intervention significantly decreased plasma triglyceride level by 21.0%, increased liver index and hepatic triglyceride content by 31.7 and 52.1%, respectively, and elevated adipose index by 44.6% compared to the vehicle group. As a PPARα agonist, fenofibrate intervention significantly increased the expression of PPARα protein in the liver by 46.3% and enhanced the expression of LDLR protein by 3.7-fold. However, fenofibrate dramatically increased the expression of PPARγ and SREBP-1c proteins by ~2.1- and 0.9-fold in the liver, respectively. Fenofibrate showed no effects on the expression of genes-related to fatty acid β-oxidation. Of note, it significantly increased the gene expression of FAS and SCD-1. Furthermore, fenofibrate modulated the gut microbiota. Collectively, long-term fenofibrate induces lipid accumulation in liver and adipose tissues in ob/ob mice by enhancing the expression of adipogenesis-related proteins and gut microbiota. These data suggest that fenofibrate may have limited effects on attenuating lipid deposition in obese patients.
Collapse
Affiliation(s)
- Ying Zhang
- College of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiu-Bin Jia
- College of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yun-Chao Liu
- College of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wen-Qian Yu
- Innovative Drug Research Centre, School of Pharmacy, Institute of Lipid Metabolism and Atherosclerosis, Weifang Medical University, Weifang, China
| | - Yan-Hong Si
- College of Pharmacy and Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- College of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Yan-Hong Si
| | - Shou-Dong Guo
- Innovative Drug Research Centre, School of Pharmacy, Institute of Lipid Metabolism and Atherosclerosis, Weifang Medical University, Weifang, China
- *Correspondence: Shou-Dong Guo
| |
Collapse
|
10
|
Rajapaksha IG, Gunarathne LS, Asadi K, Laybutt R, Andrikopoulous S, Alexander IE, Watt MJ, Angus PW, Herath CB. Angiotensin Converting Enzyme-2 Therapy Improves Liver Fibrosis and Glycemic Control in Diabetic Mice With Fatty Liver. Hepatol Commun 2022; 6:1056-1072. [PMID: 34951153 PMCID: PMC9035567 DOI: 10.1002/hep4.1884] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/26/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease and is frequently associated with type 2 diabetes. However, there is no specific medical therapy to treat this condition. Angiotensin-converting enzyme 2 (ACE2) of the protective renin angiotensin system generates the antifibrotic peptide angiotensin-(1-7) from profibrotic angiotensin II peptide. In this study, we investigated the therapeutic potential of ACE2 in diabetic NAFLD mice fed a high-fat (20%), high-cholesterol (2%) diet for 40 weeks. Mice were given a single intraperitoneal injection of ACE2 using an adeno-associated viral vector at 30 weeks of high-fat, high-cholesterol diet (15 weeks after induction of diabetes) and sacrificed 10 weeks later. ACE2 significantly reduced liver injury and fibrosis in diabetic NAFLD mice compared with the control vector injected mice. This was accompanied by reductions in proinflammatory cytokine expressions, hepatic stellate cell activation, and collagen 1 expression. Moreover, ACE2 therapy significantly increased islet numbers, leading to an increased insulin protein content in β-cells and plasma insulin levels with subsequent reduction in plasma glucose levels compared with controls. Conclusion: We conclude that ACE2 gene therapy reduces liver fibrosis and hyperglycemia in diabetic NAFLD mice and has potential as a therapy for patients with NAFLD with diabetes.
Collapse
Affiliation(s)
- Indu G Rajapaksha
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia
| | - Lakmie S Gunarathne
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia
| | | | - Ross Laybutt
- Garvan Institute of Medical ResearchSydneyNSWAustralia.,St. Vincent's Clinical SchoolUniversity of New South WalesSydneyNSWAustralia
| | - Sof Andrikopoulous
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia
| | - Ian E Alexander
- School of MedicineUniversity of SydneyChildren's Medical Research InstituteSydneyNSWAustralia
| | - Mathew J Watt
- Department Anatomy and PhysiologyThe University of MelbourneMelbourneVICAustralia
| | - Peter W Angus
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia.,Department GastroenterologyAustin HealthHeidelbergVICAustralia
| | - Chandana B Herath
- Department of MedicineThe University of MelbourneAustin HealthHeidelbergVICAustralia.,South Western Sydney Clinical SchoolFaculty of MedicineUniversity of New South WalesSydneyNSWAustralia.,Ingham Institute for Applied Medical ResearchLiverpoolNSWAustralia
| |
Collapse
|
11
|
Li CW, Yu K, Shyh-Chang N, Jiang Z, Liu T, Ma S, Luo L, Guang L, Liang K, Ma W, Miao H, Cao W, Liu R, Jiang LJ, Yu SL, Li C, Liu HJ, Xu LY, Liu RJ, Zhang XY, Liu GS. Pathogenesis of sarcopenia and the relationship with fat mass: descriptive review. J Cachexia Sarcopenia Muscle 2022; 13:781-794. [PMID: 35106971 PMCID: PMC8977978 DOI: 10.1002/jcsm.12901] [Citation(s) in RCA: 289] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/26/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
Age-associated obesity and muscle atrophy (sarcopenia) are intimately connected and are reciprocally regulated by adipose tissue and skeletal muscle dysfunction. During ageing, adipose inflammation leads to the redistribution of fat to the intra-abdominal area (visceral fat) and fatty infiltrations in skeletal muscles, resulting in decreased overall strength and functionality. Lipids and their derivatives accumulate both within and between muscle cells, inducing mitochondrial dysfunction, disturbing β-oxidation of fatty acids, and enhancing reactive oxygen species (ROS) production, leading to lipotoxicity and insulin resistance, as well as enhanced secretion of some pro-inflammatory cytokines. In turn, these muscle-secreted cytokines may exacerbate adipose tissue atrophy, support chronic low-grade inflammation, and establish a vicious cycle of local hyperlipidaemia, insulin resistance, and inflammation that spreads systemically, thus promoting the development of sarcopenic obesity (SO). We call this the metabaging cycle. Patients with SO show an increased risk of systemic insulin resistance, systemic inflammation, associated chronic diseases, and the subsequent progression to full-blown sarcopenia and even cachexia. Meanwhile in many cardiometabolic diseases, the ostensibly protective effect of obesity in extremely elderly subjects, also known as the 'obesity paradox', could possibly be explained by our theory that many elderly subjects with normal body mass index might actually harbour SO to various degrees, before it progresses to full-blown severe sarcopenia. Our review outlines current knowledge concerning the possible chain of causation between sarcopenia and obesity, proposes a solution to the obesity paradox, and the role of fat mass in ageing.
Collapse
Affiliation(s)
- Chun-Wei Li
- Department of Clinical Nutrition & Health Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kang Yu
- Department of Clinical Nutrition & Health Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ng Shyh-Chang
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zongmin Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Taoyan Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shilin Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lanfang Luo
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lu Guang
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Kun Liang
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenwu Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hefan Miao
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenhua Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ruirui Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ling-Juan Jiang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Song-Lin Yu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Li
- Department of General Surgery, Tianjin Union Medical Center, The Affiliated Hospital of Nankai University, China (Tianjin Union Medical Center, Tianjin, China
| | - Hui-Jun Liu
- Department of nursing & Clinical Nutrition, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Long-Yu Xu
- Department of Sport Physiatry, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rong-Ji Liu
- Department of Pharmacy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Yuan Zhang
- Department of stomatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gao-Shan Liu
- Department of Health Education, Shijingshan Center for Disease Prevention and Control, Beijing, China
| |
Collapse
|
12
|
Salzman NH, Schwimmer JB. Pediatric nonalcoholic fatty liver disease and the microbiome: Mechanisms contributing to pathogenesis and progression. ACTA ACUST UNITED AC 2021; 19:22-29. [PMID: 34222711 DOI: 10.1016/j.coemr.2021.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common form of pediatric liver disease in the United States, and often associated with obesity and metabolic syndrome. NAFLD comprises a broad spectrum of liver diseases, from hepatic steatosis to steatohepatitis, fibrosis and cirrhosis. Disease progression is considered a multi-modal process of liver injury. The intestinal microbiome has been implicated in several aspects of NAFLD pathophysiology. Pediatric studies associating the intestinal microbiome with NAFLD have been limited in number and complicated by inconsistencies in study design and approach. Nevertheless, they provide support for involvement of the intestinal microbiome in NAFLD development and progression and point to common mechanisms shared by microbiome-associated inflammatory diseases with potential to inform future therapeutic intervention.
Collapse
Affiliation(s)
- Nita H Salzman
- Division of Gastroenterology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin.,Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jeffrey B Schwimmer
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla, California.,Department of Gastroenterology, Rady Children's Hospital San Diego, Dan Diego, California
| |
Collapse
|
13
|
Katoch S, Patial V. Zebrafish: An emerging model system to study liver diseases and related drug discovery. J Appl Toxicol 2021; 41:33-51. [PMID: 32656821 DOI: 10.1002/jat.4031] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/31/2020] [Accepted: 06/11/2020] [Indexed: 01/03/2023]
Abstract
The zebrafish has emerged as a powerful vertebrate model for studying liver-associated disorders. Liver damage is a crucial problem in the process of drug development and zebrafish have proven to be an important tool for the high-throughput screening of drugs for hepatotoxicity. Although the structure of the zebrafish liver differs to that of mammals, the fundamental physiologic processes, genetic mutations and manifestations of pathogenic responses to environmental insults exhibit much similarity. The larval transparency of the zebrafish is a great advantage for real-time imaging in hepatic studies. The zebrafish has a broad spectrum of cytochrome P450 enzymes, which enable the biotransformation of drugs via similar pathways as mammals, including oxidation, reduction and hydrolysis reactions. In the present review, we appraise the various drugs, chemicals and toxins used to study liver toxicity in zebrafish and their similarities to the rodent models for liver-related studies. Interestingly, the zebrafish has also been effectively used to study the pathophysiology of nonalcoholic and alcoholic fatty liver disease. The genetic models of liver disorders and their easy manipulation provide great opportunity in the area of drug development. The zebrafish has proven to be an influential model for the hepatic system due to its invertebrate-like advantages coupled with its vertebrate biology. The present review highlights the pivotal role of zebrafish in bridging the gap between cell-based and mammalian models.
Collapse
Affiliation(s)
- Swati Katoch
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR- Institute of Himalayan Bioresource Technology, Palampur, India
| |
Collapse
|
14
|
Animal Models: A Useful Tool to Unveil Metabolic Changes in Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12113318. [PMID: 33182674 PMCID: PMC7696782 DOI: 10.3390/cancers12113318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) represents an important health problem. At the moment, systemic therapies offered only modest clinical benefits. Thus, HCC represents a cancer extremely difficult to treat, and therapeutic breakthroughs are urgently needed. Metabolic reprogramming of neoplastic cells has been recognized as one of the core hallmarks of cancer. Experimental animal models represent an important tool that allows to investigate metabolic changes underlying HCC development and progression. In the present review, we characterize available rodent models of hepatocarcinogenesis. Moreover, we discuss the possibility that pharmacological targeting of Warburg metabolism may represent an additional tool to improve already available therapeutic approaches for HCC. Abstract Hepatocellular carcinoma (HCC) is one the most frequent and lethal human cancers. At present, no effective treatment for advanced HCC exist; therefore, the overall prognosis for HCC patients remains dismal. In recent years, a better knowledge of the signaling pathways involved in the regulation of HCC development and progression, has led to the identification of novel potential targets for therapeutic strategies. However, the obtained benefits from current therapeutic options are disappointing. Altered cancer metabolism has become a topic of renewed interest in the last decades, and it has been included among the core hallmarks of cancer. In the light of growing evidence for metabolic reprogramming in cancer, a wide number of experimental animal models have been exploited to study metabolic changes characterizing HCC development and progression and to further expand our knowledge of this tumor. In the present review, we discuss several rodent models of hepatocarcinogenesis, that contributed to elucidate the metabolic profile of HCC and the implications of these changes in modulating the aggressiveness of neoplastic cells. We also highlight the apparently contrasting results stemming from different animal models. Finally, we analyze whether these observations could be exploited to improve current therapeutic strategies for HCC.
Collapse
|
15
|
Mohamed DA, Fouda KA, Mohamed RS. <i>In vitro</i> Anticancer Activity of Quinoa and Safflower Seeds and Their Preventive Effects on Non-alcoholic Fatty Liver. Pak J Biol Sci 2020; 22:383-392. [PMID: 31930826 DOI: 10.3923/pjbs.2019.383.392] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is not only the most common cause of liver diseases in humans but also it may complicate and become a risk factor for liver cancer. The present work aimed to evaluate the anticancer activity (in vitro) of quinoa and safflower seeds powder and their beneficial effects against NAFLD (in vivo). MATERIALS AND METHODS Proximate analysis, fatty acids profile, total phenolic and phytic acid of quinoa and safflower seeds were assessed. Also their anticancer activities (in vitro) against liver cancer were evaluated. The preventive effect of both seeds on NAFLD was evaluated using twenty four male rats. NAFLD was induced in rats by high fructose diet (HFD) for 4 weeks. The effects of HFD and HFD supplemented with 20% quinoa or safflower powder on plasma and liver lipids, lipid peroxidation, total protein, albumin as well as liver and kidney functions were determined. RESULTS Quinoa seeds powder was promising in cytotoxicity against hepatocarcinoma cell line HEPG2 (IC50 was 14.6 μg). Feeding rats on HFD produced dyslipidemia and significant increase in liver functions and lipid peroxidation with significant elevation in liver triglycerides and total cholesterol. Quinoa and safflower seeds powder produced improvement in the biochemical parameters with different degrees. CONCLUSION Quinoa and safflower seeds powder possessed cytotoxicity against hepatocarcinoma cell line HEPG2 and afford hepato-protection against NAFLD.
Collapse
|
16
|
Yanagita T, Tsuge K, Koga M, Inoue N, Nagao K. Eicosapentaenoic acid-containing polar lipids from seaweed Susabinori (Pyropia yezoensis) alleviate hepatic steatosis in obese db/db mice. Arch Biochem Biophys 2020; 691:108486. [PMID: 32710880 DOI: 10.1016/j.abb.2020.108486] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/21/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is emerging as the most common liver disease in industrialized countries. Because hepatic steatosis is an early pathogenesis of NAFLD, the discovery of food components that could ameliorate hepatic steatosis is of interest. Susabinori (Pyropia yezoensis) is recognized as one of the most delicious edible brown algae, and we prepared lipid component of susabinori (SNL), which is rich in eicosapentaenoic acid (EPA)-containing polar lipids. In this study, we tested whether feeding SNL to db/db mice protects them from developing obesity-induced hepatic steatosis. After four weeks of feeding, hepatomegaly, hepatic steatosis, and hepatic injury were markedly alleviated in SNL-fed db/db mice. These effects were partly attributable to the suppression of activities and mRNA expressions of lipogenic enzymes and enhanced levels of adiponectin due to the SNL diet. Additionally, mRNA expression of monocyte chemoattractant protein-1, an inflammatory chemokine, was markedly suppressed, and the mRNA levels of PPARδ, the anti-inflammatory transcription factor, were strongly enhanced in the livers of db/db mice by the SNL diet. We speculate that the development and progression of obesity-induced hepatic steatosis was prevented by the suppression of chronic inflammation due to the combination of bioactivities of EPA, phospholipids, and glycolipids in the SNL diet.
Collapse
Affiliation(s)
- Teruyoshi Yanagita
- Department of Biological Resource Science, Saga University, Saga, 840-8502, Japan; Department of Health and Nutrition Sciences, Nishikyushu University, Kanzaki, 842-8585, Japan; Saga Regional Industry Support Center, Saga, 849-0932, Japan
| | - Keisuke Tsuge
- Industrial Technology Center of Saga, Saga, 849-0932, Japan
| | - Misato Koga
- Department of Biological Resource Science, Saga University, Saga, 840-8502, Japan
| | - Nao Inoue
- Faculty of Agriculture, Yamagata University, Tsuruoka, 997-8555, Japan
| | - Koji Nagao
- Department of Biological Resource Science, Saga University, Saga, 840-8502, Japan.
| |
Collapse
|
17
|
Zheng J, Manabe Y, Sugawara T. Siphonaxanthin, a carotenoid from green algae Codium cylindricum, protects Ob/Ob mice fed on a high-fat diet against lipotoxicity by ameliorating somatic stresses and restoring anti-oxidative capacity. Nutr Res 2020; 77:29-42. [PMID: 32315893 DOI: 10.1016/j.nutres.2020.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 12/17/2022]
Abstract
Oxidative stress is implicated in the pathogenesis of many diseases including obesity, non-alcoholic fatty liver disease, and diabetes mellitus. Previously, we reported that siphonaxanthin, a carotenoid from green algae, elicited a potent inhibitory effect on hepatic de novo lipogenesis, and an anti-obesity effect in both 3T3L1 cells and KKAy mice. Thus, we hypothesized that consumption of siphonaxanthin could improve metabolic disorders including hepatic steatosis and systemic adiposity, as well as ameliorate somatic stress under obese conditions. Both the hepatocyte cell line HepG2 and a mouse model of severe obesity, produced by feeding Ob/Ob mice on a high-fat diet (HFD), were used to test this hypothesis. In obese mice, siphonaxanthin intake did not improve liver steatosis or systemic adiposity. However, intake did lower plasma glucose and alanine aminotransferase (ALT) levels and diminished hepatic lipid peroxidation products and antioxidant gene expression, which increased significantly in control group obese mice. Renal protein carbonyl content decreased significantly in the siphonaxanthin group, which might also indicate an ameliorated oxidative stress. Siphonaxanthin restored gene expression related to antioxidant signaling, lipid β-oxidation, and endoplasmic-reticulum-associated protein degradation in the kidney, which decreased significantly in obese mice. Liver and kidney responded to obesity-induced somatic stress in a divergent pattern. In addition, we confirmed that siphonaxanthin potently induced Nrf2-regulated antioxidant signaling in HepG2 cells. In conclusion, our results indicated that siphonaxanthin might protect obesity-leading somatic stress through restoration of Nrf2-regulated antioxidant signaling, and might be a promising nutritional supplement.
Collapse
Affiliation(s)
- Jiawen Zheng
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| | - Yuki Manabe
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| | - Tatsuya Sugawara
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
18
|
Yahaghi L, Ebrahim‐Habibi A, Hayati‐Roodbari N, Irani S, Yaghmaei P. A simple method for inducing nonalcoholic steatohepatitis with fibrosis. Animal Model Exp Med 2019; 2:282-290. [PMID: 31942560 PMCID: PMC6930990 DOI: 10.1002/ame2.12089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is increasingly occurring in sedentary people, and may progress to NASH and hepatocellular carcinoma. It is essential to design affordable animal models for the study of various diseases, including fatty liver, which was the aim of the study. In this study, a high-fat diet was devised that triggers NASH's animal model quickly and easily. High-fat diet (HFD) was used both with intra-mouth oral gavage and in combination with animal pellets. METHODS Twenty-four male C57BL/6J mice were divided into HFD and ND groups, which received a high-fat diet and a normal diet, respectively. At the end of the experiment (fourth week of treatment), body and liver weights, biochemical parameters, PPAR-α gene expression and histopathologic characteristics of the liver were evaluated. RESULTS During 4 weeks, body weight of mice did not show a significant increase in the HFD group compared to the ND group, while weight gain of the liver was significant. Histological assessment of the HFD group's liver confirmed NASH symptoms. In the HFD group, HDL-c, SOD, catalase, FRAP, adiponectin, and PPAR-α decreased significantly, and lipid profiles, hepatic enzymes, MDA, leptin, and TNF-α showed a significant increase compared to the ND group. CONCLUSION Our high-fat diet has successfully induced all aspects of NASH with fibrosis in 4 weeks, and with low cost.
Collapse
Affiliation(s)
- Leyla Yahaghi
- Department of Biology, Science and Research BranchIslamic Azad UniversityTehranIran
| | - Azadeh Ebrahim‐Habibi
- Biosensor Research CenterEndocrinology and Metabolism Molecular‐Cellular Sciences InstituteTehran University of Medical SciencesTehranIran
- Endocrinology and Metabolism Research CenterTehran University of Medical SciencesTehranIran
| | | | - Shiva Irani
- Department of Biology, Science and Research BranchIslamic Azad UniversityTehranIran
| | - Parichehreh Yaghmaei
- Department of Biology, Science and Research BranchIslamic Azad UniversityTehranIran
| |
Collapse
|
19
|
A Mohamed D, S Abdelgayed S, A Essa H, S Mohamed R. Preparation and Evaluation of Functional Foods for Prevention of Non-alcoholic Fatty Liver Disease. Pak J Biol Sci 2019; 21:454-462. [PMID: 30724047 DOI: 10.3923/pjbs.2018.454.462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is a public health problem presenting one of the most important common forms of liver diseases worldwide. This study was carried out to investigate the protective effect of two functional foods in form of bread containing purslane seeds meal and garden cress seeds against NAFLD. MATERIALS AND METHODS High fat and high cholesterol diet were used for induction of NAFLD in rats for 6 weeks. Plasma lipid profile (total cholesterol, triglycerides, high density lipoprotein-cholesterol and low-density lipoprotein-cholesterol, hepatic lipid profile (total fat, cholesterol, triglycerides), malondialdehyde (MDA), as well as liver (AST, ALT, total and direct bilirubin) and kidney (creatinine and urea) functions were assessed. Histological examination of liver tissue was carried out. RESULTS Results revealed that significant elevation in plasma and liver lipid profiles, MDA, liver enzymes (AST and ALT), bilirubin (total and direct) and kidney function (creatinine and urea) were observed in NAFLD control compared to normal control. Feeding rats on diet containing functional food I and II (purslane and garden cress bread, respectively) showed significant improvement in all the studied parameters with remarkable effect regards to functional food I (purslane bread). CONCLUSION Purslane bread and garden cress bread as functional foods prepared in the present study prevent weight gain, improve plasma lipid profile and prevent hepatic lipid accumulation effectively in NAFLD model in rats. Also decreased lipid peroxidation, improve liver and kidney functions and possess hypoglycemic effect. Purslane bread was superior in the prevention of hepatic lipid accumulation.
Collapse
|
20
|
Mediterranean diet: The role of long-chain ω-3 fatty acids in fish; polyphenols in fruits, vegetables, cereals, coffee, tea, cacao and wine; probiotics and vitamins in prevention of stroke, age-related cognitive decline, and Alzheimer disease. Rev Neurol (Paris) 2019; 175:724-741. [PMID: 31521398 DOI: 10.1016/j.neurol.2019.08.005] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/28/2019] [Indexed: 12/18/2022]
Abstract
The mechanisms of action of the dietary components of the Mediterranean diet are reviewed in prevention of cardiovascular disease, stroke, age-associated cognitive decline and Alzheimer disease. A companion article provides a comprehensive review of extra-virgin olive oil. The benefits of consumption of long-chain ω-3 fatty acids are described. Fresh fish provides eicosapentaenoic acid while α-linolenic acid is found in canola and soybean oils, purslane and nuts. These ω-3 fatty acids interact metabolically with ω-6 fatty acids mainly linoleic acid from corn oil, sunflower oil and peanut oil. Diets rich in ω-6 fatty acids inhibit the formation of healthier ω-3 fatty acids. The deleterious effects on lipid metabolism of excessive intake of carbohydrates, in particular high-fructose corn syrup and artificial sweeteners, are explained. The critical role of the ω-3 fatty acid docosahexaenoic acid in the developing and aging brain and in Alzheimer disease is addressed. Nutritional epidemiology studies, prospective population-based surveys, and clinical trials confirm the salutary effects of fish consumption on prevention of coronary artery disease, stroke and dementia. Recent recommendations on fish consumption by pregnant women and potential mercury toxicity are reviewed. The polyphenols and flavonoids of plant origin play a critical role in the Mediterranean diet, because of their antioxidant and anti-inflammatory properties of benefit in type-2 diabetes mellitus, cardiovascular disease, stroke and cancer prevention. Polyphenols from fruits and vegetables modulate tau hyperphosphorylation and beta amyloid aggregation in animal models of Alzheimer disease. From the public health viewpoint worldwide the daily consumption of fruits and vegetables has become the main tool for prevention of cardiovascular disease and stroke. We review the important dietary role of cereal grains in prevention of coronary disease and stroke. Polyphenols from grapes, wine and alcoholic beverages are discussed, in particular their effects on coagulation. The mechanisms of action of probiotics and vitamins are also included.
Collapse
|
21
|
Satheesh Naik K, Gurushanthaiah M, Kavimani M, Prabhu K, Lokanadham S. Hepatoprotective Role of Eclipta alba against High Fatty Diet Treated Experimental Models - A Histopathological Study. MÆDICA 2019; 13:217-222. [PMID: 31490461 DOI: 10.26574/maedica.2018.13.3.217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Background The hepatic injury due to oxidative stress was ameliorated through administration of an aqueous extract of Eclipta alba leaves and suggested that wedelolactone and demethylwedelolactone were the possible components of Eclipta alba behind the protective effect on liver as well as against liver disorders. Objective To study the hepato-protective effects of Eclipta alba on high fatty diet treated experimental models. Material and methods A total of 30 adult albino rats of Wistar strain weighing 165-215 grams, from the animal house of the Basaveshwara Medical College, Hospital and Research Centre, Chitradurga, were used for the present study: group 1 included animals fed with normal diet (control); group 2, animals treated with hyperlipidemic diet for eight weeks; group 3, animals treated with hyperlipidemic diet for eight weeks, followed by one week post treatment of Eclipta alba with normal diet; group 4, animals treated with hyperlipidemic diet for eight weeks, followed by two weeks post-treatment of Eclipta alba with normal diet; and group 5, animals treated with hyperlipidemic diet for eight weeks, followed by three weeks posttreatment of Eclipta alba with normal diet. Results In animals with high fat diet (30%), we observed the deposition of fat in the form of fat lobules in and around the hepatocytes, mononuclear in filtration in the liver parenchyma, dilation of blood vessels, necrotic foci and damaged hepatocytes. Conclusion The components of Eclipta alba like wedelolactone, demethylwedelolactone and saponins reduced fat deposition, mononuclear infiltration, and necrotic foci, and stimulated hepatocyte regeneration in the liver.
Collapse
Affiliation(s)
- K Satheesh Naik
- Department of Anatomy, Bharath University (BIHER), Chennai, Tamilnadu, India
| | - M Gurushanthaiah
- Department of Anatomy, Basaveshwara Medical College, Chitradurga, Karnataka, India
| | - M Kavimani
- Department of Anatomy, Sree Balaji Medical College and Hospital, Chennai, Tamilnadu, India
| | - K Prabhu
- Department of Anatomy, Sree Balaji Medical College and Hospital, Chennai, Tamilnadu, India
| | - S Lokanadham
- Department of Anatomy, Santhiram Medical College and Hospital, Nandyal, Andhra Pradesh, India
| |
Collapse
|
22
|
Rapid Recapitulation of Nonalcoholic Steatohepatitis upon Loss of Host Cell Factor 1 Function in Mouse Hepatocytes. Mol Cell Biol 2019; 39:MCB.00405-18. [PMID: 30559308 PMCID: PMC6379584 DOI: 10.1128/mcb.00405-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Host cell factor 1 (HCF-1), encoded by the ubiquitously expressed X-linked gene Hcfc1, is an epigenetic coregulator important for mouse development and cell proliferation, including during liver regeneration. We used a hepatocyte-specific inducible Hcfc1 knockout allele (called Hcfc1hepKO) to induce HCF-1 loss in hepatocytes of hemizygous Hcfc1hepKO/Y males by 4 days. Host cell factor 1 (HCF-1), encoded by the ubiquitously expressed X-linked gene Hcfc1, is an epigenetic coregulator important for mouse development and cell proliferation, including during liver regeneration. We used a hepatocyte-specific inducible Hcfc1 knockout allele (called Hcfc1hepKO) to induce HCF-1 loss in hepatocytes of hemizygous Hcfc1hepKO/Y males by 4 days. In heterozygous Hcfc1hepKO/+ females, owing to random X-chromosome inactivation, upon Hcfc1hepKO allele induction, a 50/50 mix of HCF-1-positive and -negative hepatocyte clusters is engineered. The livers with Hcfc1hepKO/Y hepatocytes displayed a 21- to 24-day terminal nonalcoholic fatty liver (NAFL), followed by nonalcoholic steatohepatitis (NASH) disease progression typical of severe NAFL disease (NAFLD). In contrast, in livers with heterozygous Hcfc1hepKO/+ hepatocytes, HCF-1-positive hepatocytes replaced HCF-1-negative hepatocytes and revealed only mild NAFL development. Loss of HCF-1 led to loss of PGC1α protein, probably owing to its destabilization, and deregulation of gene expression, particularly of genes involved in mitochondrial structure and function, likely explaining the severe Hcfc1hepKO/Y liver pathology. Thus, HCF-1 is essential for hepatocyte function, likely playing both transcriptional and nontranscriptional roles. These genetically engineered loss-of-HCF-1 mice can be used to study NASH as well as NAFLD resolution.
Collapse
|
23
|
Disease Progression and Pharmacological Intervention in a Nutrient-Deficient Rat Model of Nonalcoholic Steatohepatitis. Dig Dis Sci 2019; 64:1238-1256. [PMID: 30511198 PMCID: PMC6548202 DOI: 10.1007/s10620-018-5395-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/22/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND There is a marked need for improved animal models of nonalcoholic steatohepatitis (NASH) to facilitate the development of more efficacious drug therapies for the disease. METHODS Here, we investigated the development of fibrotic NASH in male Wistar rats fed a choline-deficient L-amino acid-defined (CDAA) diet with or without cholesterol supplementation for subsequent assessment of drug treatment efficacy in NASH biopsy-confirmed rats. The metabolic profile and liver histopathology were evaluated after 4, 8, and 12 weeks of dieting. Subsequently, rats with biopsy-confirmed NASH were selected for pharmacological intervention with vehicle, elafibranor (30 mg/kg/day) or obeticholic acid (OCA, 30 mg/kg/day) for 5 weeks. RESULTS The CDAA diet led to marked hepatomegaly and fibrosis already after 4 weeks of feeding, with further progression of collagen deposition and fibrogenesis-associated gene expression during the 12-week feeding period. Cholesterol supplementation enhanced the stimulatory effect of CDAA on gene transcripts associated with fibrogenesis without significantly increasing collagen deposition. Pharmacological intervention with elafibranor, but not OCA, significantly reduced steatohepatitis scores, and fibrosis-associated gene expression, however, was unable to prevent progression in fibrosis scores. CONCLUSION CDAA-fed rats develop early-onset progressive NASH, which offers the opportunity to probe anti-NASH compounds with potential disease-modifying properties.
Collapse
|
24
|
Vaezi M, Yaghmaei P, Hayati-Roodbari N, Irani S, Ebrahim-Habibi A. Citral effect in male NMRI mice nonalcoholic steatosis model: assessing biochemical and histological parameters and PPARα gene expression. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000317596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | | | | | - Azadeh Ebrahim-Habibi
- Tehran University of Medical Sciences, Iran; Tehran University of Medical Sciences, Iran
| |
Collapse
|
25
|
Columbano A, Chiellini G, Kowalik MA. GC-1: A Thyromimetic With Multiple Therapeutic Applications in Liver Disease. Gene Expr 2017; 17:265-275. [PMID: 28635586 PMCID: PMC5885148 DOI: 10.3727/105221617x14968563796227] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Thyroid hormones (THs), namely, 3,5,3'-triiodo-l-thyronine (T3) and 3,5,3',5'-tetraiodo-l-thyronine (thyroxine or T4), influence a variety of physiological processes that have important implications in fetal development, metabolism, cell growth, and proliferation. While THs elicit several beneficial effects on lipid metabolism and improve myocardial contractility, these therapeutically desirable effects are associated to a thyrotoxic state that severely limits the possible use of THs as therapeutic agents. Therefore, several efforts have been made to develop T3 analogs that could retain the beneficial actions (triglyceride, cholesterol, obesity, and body mass lowering) without the adverse TH-dependent side effects. This goal was achieved by the synthesis of TRβ-selective agonists. In this review, we summarize the current knowledge on the effects of one of the best characterized TH analogs, the TRβ1-selective thyromimetic, GC-1. In particular, we review some of the effects of GC-1 on different liver disorders, with reference to its possible clinical application. A brief comment on the possible therapeutic use of GC-1 in extrahepatic disorders is also included.
Collapse
Affiliation(s)
- Amedeo Columbano
- *Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Grazia Chiellini
- †Department of Surgical, Medical and Molecular Pathology, University of Pisa, Pisa, Italy
| | - Marta Anna Kowalik
- *Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| |
Collapse
|
26
|
Van den Bossche L, Schoonenberg VAC, Burgener IA, Penning LC, Schrall IM, Kruitwagen HS, van Wolferen ME, Grinwis GCM, Kummeling A, Rothuizen J, van Velzen JF, Stathonikos N, Molenaar MR, Helms BJ, Brouwers JFHM, Spee B, van Steenbeek FG. Aberrant hepatic lipid storage and metabolism in canine portosystemic shunts. PLoS One 2017; 12:e0186491. [PMID: 29049355 PMCID: PMC5648188 DOI: 10.1371/journal.pone.0186491] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a poorly understood multifactorial pandemic disorder. One of the hallmarks of NAFLD, hepatic steatosis, is a common feature in canine congenital portosystemic shunts. The aim of this study was to gain detailed insight into the pathogenesis of steatosis in this large animal model. Hepatic lipid accumulation, gene-expression analysis and HPLC-MS of neutral lipids and phospholipids in extrahepatic (EHPSS) and intrahepatic portosystemic shunts (IHPSS) was compared to healthy control dogs. Liver organoids of diseased dogs and healthy control dogs were incubated with palmitic- and oleic-acid, and lipid accumulation was quantified using LD540. In histological slides of shunt livers, a 12-fold increase of lipid content was detected compared to the control dogs (EHPSS P<0.01; IHPSS P = 0.042). Involvement of lipid-related genes to steatosis in portosystemic shunting was corroborated using gene-expression profiling. Lipid analysis demonstrated different triglyceride composition and a shift towards short chain and omega-3 fatty acids in shunt versus healthy dogs, with no difference in lipid species composition between shunt types. All organoids showed a similar increase in triacylglycerols after free fatty acids enrichment. This study demonstrates that steatosis is probably secondary to canine portosystemic shunts. Unravelling the pathogenesis of this hepatic steatosis might contribute to a better understanding of steatosis in NAFLD.
Collapse
Affiliation(s)
- Lindsay Van den Bossche
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Vivien A. C. Schoonenberg
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Iwan A. Burgener
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Louis C. Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Ingrid M. Schrall
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Hedwig S. Kruitwagen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Monique E. van Wolferen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Guy C. M. Grinwis
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Anne Kummeling
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jan Rothuizen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jeroen F. van Velzen
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nikolas Stathonikos
- Department of Pathology, University Medical Center, Utrecht, The Netherlands
| | - Martijn R. Molenaar
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht, The Netherlands
| | - Bernd J. Helms
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht, The Netherlands
| | - Jos F. H. M. Brouwers
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Frank G. van Steenbeek
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
27
|
Hyperpolarized 13C Spectroscopic Evaluation of Oxidative Stress in a Rodent Model of Steatohepatitis. Sci Rep 2017; 7:46014. [PMID: 28425467 PMCID: PMC5397869 DOI: 10.1038/srep46014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 03/09/2017] [Indexed: 12/30/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become highly prevalent, now considered the most common liver disease in the western world. Approximately one-third of patients with NASH develop non-alchoholic steatohepatitis (NASH), histologically defined by lobular and portal inflammation, and accompanied by marked oxidative stress. Patients with NASH are at increased risk for cirrhosis and hepatocellular carcinoma, and diagnosis currently requires invasive biopsy. In animal models of NASH, particularly the methionine-choline deficient (MCD) model, profound changes are seen in redox enzymes and key intracellular antioxidants. To study antioxidant status in NASH non-invasively, we applied the redox probe hyperpolarized [1-13C] dehydroascorbic acid (HP DHA), which is reduced to Vitamin C (VitC) rapidly in the normal liver. In MCD mice, we observed a significant decrease in HP DHA to VitC conversion that accompanied hepatic fat deposition. When these animals were subsequently placed on a normal diet, resonance ratios reverted to those seen in control mice. These findings suggest that HP DHA, a potentially clinically translatable imaging agent, holds special promise in imaging NASH and other metabolic syndromes, to monitor disease progression and response to targeted therapies.
Collapse
|
28
|
Shetty A, Cho W, Alazawi W, Syn WK. Methotrexate Hepatotoxicity and the Impact of Nonalcoholic Fatty Liver Disease. Am J Med Sci 2017; 354:172-181. [PMID: 28864376 DOI: 10.1016/j.amjms.2017.03.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/10/2017] [Accepted: 03/08/2017] [Indexed: 01/01/2023]
Abstract
Methotrexate (MTX) is commonly used to treat individuals with rheumatological and dermatologic disorders. Current American College of Rheumatology (ACR) and American Association of Dermatology (AAD) guidelines identify diabetes and obesity as risk factors for MTX-induced liver injury. Both diabetes and obesity are components of the metabolic syndrome, and are also risk factors for nonalcoholic fatty liver disease (NAFLD). NAFLD affects approximately 40% of the U.S. population, and those with more advanced NAFLD (i.e., nonalcoholic steatohepatitis with or without fibrosis) are likely to develop progressive liver disease. As such, individuals who are treated with MTX may need to be screened for advanced NAFLD, as this may put them at an increased risk of MTX-induced liver injury. In this mini-review, we review the current ACR and AAD guidelines on MTX hepatotoxicity, discuss the evidence (or lack thereof) of the impact of metabolic risk factors on MTX-induced liver injury and highlight the areas that need further research.
Collapse
Affiliation(s)
- Akshay Shetty
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina
| | - WonKyung Cho
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina
| | - William Alazawi
- Department of Hepatology, Barts Health NHS Trust, London, United Kingdom; Centre for Immunobiology, Blizzard Institute, Queen Mary University of London, London, United Kingdom
| | - Wing-Kin Syn
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina; Section of Gastroenterology, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina.
| |
Collapse
|
29
|
Enhanced prevention of progression of non alcoholic fatty liver to steatohepatitis by incorporating pumpkin seed oil in nanoemulsions. J Mol Liq 2017. [DOI: 10.1016/j.molliq.2016.10.138] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
30
|
Takeno A, Yamamoto M, Notsu M, Sugimoto T. Administration of anti-receptor activator of nuclear factor-kappa B ligand (RANKL) antibody for the treatment of osteoporosis was associated with amelioration of hepatitis in a female patient with growth hormone deficiency: a case report. BMC Endocr Disord 2016; 16:66. [PMID: 27881164 PMCID: PMC5122017 DOI: 10.1186/s12902-016-0148-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/18/2016] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Growth hormone deficiency (GHD) is associated with non-alcoholic fatty liver disease (NAFLD). A recent animal study showed that hepatocyte-specific receptor activator of nuclear factor-kappa B (RANK) knockout mice had significantly lower liver fat content compared with control mice concomitant with a decrease in production of inflammatory cytokines such as tumor necrosis factor-α (TNF-α) from hepatocytes and kupffer cells. The role of anti-RANK ligand (RANKL) antibody for osteoporosis on hepatitis in patients with aGHD is still unknown. CASE PRESENTATION A forty-seven-year-old female patient was referred to our hospital to investigate chronic hepatitis caused by unknown etiology. She had past history of craniopharyngioma treated with craniotomy and post-surgical radiotherapy. She was for the first time diagnosed as panhypopituitarism including growth hormone deficiency and osteoporosis by endocrine examinations and bone mineral densitometry, respectively. In addition, non-alcoholic steatohepatitis (NASH) was histologically confirmed by liver biopsy in this time. Sixty mg anti-RANKL antibody, which was subcutaneously injected to treat the osteoporosis every six months after replacement of 5 mg hydrocortisone and 30 μg oral desmopressin, rapidly decreased the levels of her liver enzymes (ALT and γGTP were 133 to 72 U/L and 284 to 99 U/L at 16 months after the beginning of the treatment, respectively). Additional amelioration of liver dysfunction was not observed after growth hormone replacement. CONCLUSIONS The clinical course of the present case suggested that RANKL-RANK signaling may be a key pathological mechanism in establishment or development of NAFLD or NASH in patients with panhypopituitarism including GHD.
Collapse
Affiliation(s)
- Ayumu Takeno
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane 693-8501 Japan
| | - Masahiro Yamamoto
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane 693-8501 Japan
| | - Masakazu Notsu
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane 693-8501 Japan
| | - Toshitsugu Sugimoto
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane 693-8501 Japan
| |
Collapse
|
31
|
Kim SB, Kang OH, Lee YS, Han SH, Ahn YS, Cha SW, Seo YS, Kong R, Kwon DY. Hepatoprotective Effect and Synergism of Bisdemethoycurcumin against MCD Diet-Induced Nonalcoholic Fatty Liver Disease in Mice. PLoS One 2016; 11:e0147745. [PMID: 26881746 PMCID: PMC4755614 DOI: 10.1371/journal.pone.0147745] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 01/07/2016] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), the hepatic manifestation of the metabolic syndrome, has become one of the most common causes of chronic liver disease over the last decade in developed countries. NAFLD includes a spectrum of pathological hepatic changes, such as steatosis, steatohepatitis, advanced fibrosis, and cirrhosis. Bisdemethoxycurcumin (BDMC) is polyphenolic compounds with a diarylheptanoid skeleton, curcumin close analogues, which is derived from the Curcumae Longae Rhizoma. While the rich bioavailability research of curcumin, BDMC is the poor studies. We investigated whether BDMC has the hepatoprotective effect and combinatory preventive effect with silymarin on methionine choline deficient (MCD)-diet-induced NAFLD in C57BL/6J mice. C57BL/6J mice were divided into five groups of normal (normal diet without any treatment), MCD diet (MCD diet only), MCD + silymarin (SIL) 100 mg/kg group, MCD + BDMC 100 mg/kg group, MCD + SIL 50 mg/kg + BDMC 50 mg/kg group. Body weight, liver weight, liver function tests, histological changes were assessed and quantitative real-time polymerase chain reaction and Western blot analyses were conducted after 4 weeks. Mice lost body weight on the MCD-diet, but BDMC did not lose less than the MCD-diet group. Liver weights decreased from BDMC, but they increased significantly in the MCD-diet groups. All liver function test values decreased from the MCD-diet, whereas those from the BDMC increased significantly. The MCD- diet induced severe hepatic fatty accumulation, but the fatty change was reduced in the BDMC. The BDMC showed an inhibitory effect on liver lipogenesis by reducing associated gene expression caused by the MCD-diet. In all experiments, the combinations of BDMC with SIL had a synergistic effect against MCD-diet models. In conclusion, our findings indicate that BDMC has a potential suppressive effect on NAFLD. Therefore, our data suggest that BDMC may act as a novel and potent therapeutic agent against NAFLD.
Collapse
Affiliation(s)
- Sung-Bae Kim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Wonkwang Oriental Medicines Research Institute, Iksan, Jeonbuk, 570–749, Korea
| | - Ok-Hwa Kang
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Wonkwang Oriental Medicines Research Institute, Iksan, Jeonbuk, 570–749, Korea
| | - Young-Seob Lee
- BK21 Plus Team, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk, 570–749, Korea
| | - Sin-Hee Han
- Department of Herbal Crop Research, National Institute of Horticultural & Herbal Science, RDA, 92 Bisanro, Eumsung, Chungbuk, 369–873, Korea
| | - Young-Sup Ahn
- Department of Herbal Crop Research, National Institute of Horticultural & Herbal Science, RDA, 92 Bisanro, Eumsung, Chungbuk, 369–873, Korea
| | - Seon-Woo Cha
- Department of Herbal Crop Research, National Institute of Horticultural & Herbal Science, RDA, 92 Bisanro, Eumsung, Chungbuk, 369–873, Korea
| | - Yun-Soo Seo
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Wonkwang Oriental Medicines Research Institute, Iksan, Jeonbuk, 570–749, Korea
| | - Ryong Kong
- BK21 Plus Team, Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk, 570–749, Korea
| | - Dong-Yeul Kwon
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Wonkwang Oriental Medicines Research Institute, Iksan, Jeonbuk, 570–749, Korea
- * E-mail:
| |
Collapse
|
32
|
In Situ Evaluation of Oxidative Stress in Rat Fatty Liver Induced by a Methionine- and Choline-Deficient Diet. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9307064. [PMID: 26881047 PMCID: PMC4736780 DOI: 10.1155/2016/9307064] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/01/2015] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a serious health problem in developed countries. We documented the effects of feeding with a NAFLD-inducing, methionine- and choline-deficient (MCD) diet, for 1-4 weeks on rat liver oxidative stress, with respect to a control diet. Glycogen, neutral lipids, ROS, peroxidated proteins, and SOD2 were investigated using histochemical procedures; ATP, GSH, and TBARS concentrations were investigated by biochemical dosages, and SOD2 expression was investigated by Western Blotting. In the 4-week-diet period, glycogen stores decreased whereas lipid droplets, ROS, and peroxidated proteins expression (especially around lipid droplets of hepatocytes) increased. SOD2 immunostaining decreased in poorly steatotic hepatocytes but increased in the thin cytoplasm of macrosteatotic cells; a trend towards a quantitative decrease of SOD expression in homogenates occurred after 3 weeks. ATP and GSH values were significantly lower for rats fed with the MCD diet with respect to the controls. An increase of TBARS in the last period of the diet is in keeping with the high ROS production and low antioxidant defense; these TBARS may promote protein peroxidation around lipid droplets. Since these proteins play key roles in lipid mobilization, storage, and metabolism, this last information appears significant, as it points towards a previously misconsidered target of NAFLD-associated oxidative stress that might be responsible for lipid dysfunction.
Collapse
|
33
|
Khoury T, Ben Ya'acov A, Shabat Y, Zolotarovya L, Snir R, Ilan Y. Altered distribution of regulatory lymphocytes by oral administration of soy-extracts exerts a hepatoprotective effect alleviating immune mediated liver injury, non-alcoholic steatohepatitis and insulin resistance. World J Gastroenterol 2015; 21:7443-7456. [PMID: 26139990 PMCID: PMC4481439 DOI: 10.3748/wjg.v21.i24.7443] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/08/2015] [Accepted: 03/30/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To determine the immune-modulatory and the hepatoprotective effects of oral administration of two soy extracts in immune mediated liver injury and non-alcoholic steatohepatitis (NASH). METHODS Two soy extracts, M1 and OS, were orally administered to mice with concanavalin A (ConA) immune-mediated hepatitis, to high-fat diet (HFD) mice and to methionine and choline reduced diet combined with HFD mice. Animals were followed for disease and immune biomarkers. RESULTS Oral administration of OS and M1 had an additive effect in alleviating ConA hepatitis manifested by a decrease in alanine aminotransferase and aspartate aminotransferase serum levels. Oral administration of the OS and M1 soy derived fractions, ameliorated liver injury in the high fat diet model of NASH, manifested by a decrease in hepatic triglyceride levels, improvement in liver histology, decreased serum cholesterol and triglycerides and improved insulin resistance. In the methionine and choline reduced diet combined with the high fat diet model, we noted a decrease in hepatic triglycerides and improvement in blood glucose levels and liver histology. The effects were associated with reduced serum tumor necrosis factor alpha and alteration of regulatory T cell distribution. CONCLUSION Oral administration of the combination of OS and M1 soy derived extracts exerted an adjuvant effect in the gut-immune system, altering the distribution of regulatory T cells, and alleviating immune mediated liver injury, hyperlipidemia and insulin resistance.
Collapse
|
34
|
Choline and Cystine Deficient Diets in Animal Models with Hepatocellular Injury: Evaluation of Oxidative Stress and Expression of RAGE, TNF-α, and IL-1β. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:121925. [PMID: 26137185 PMCID: PMC4468296 DOI: 10.1155/2015/121925] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 05/15/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023]
Abstract
This study aims to evaluate the effects of diets deficient in choline and/or cystine on hepatocellular injury in animal models (young male Wistar rats, aged 21 days), by monitoring some of the oxidative stress biomarkers and the expression of RAGE, TNF-α, and IL-1β. The animals were divided into 6 groups (n = 10) and submitted to different diets over 30 days: AIN-93 diet (standard, St), AIN-93 choline deficient (CD) diet and AIN-93 choline and cystine deficient (CCD) diet, in the pellet (pl) and powder (pw) diet forms. Independently of the diet form, AIN-93 diet already led to hepatic steatosis and CD/CCD diets provoked hepatic damage. The increase of lipid peroxidation, represented by the evaluation of thiobarbituric acid reactive species, associated with the decrease of levels of antioxidant enzymes, were the parameters with higher significance toward redox profile in this model of hepatic injury. Regarding inflammation, in relation to TNF-α, higher levels were evidenced in CD(pl), while, for IL-1β, no significant alteration was detected. RAGE expression was practically the same in all groups, with exception of CCD(pw) versus CCD(pl). These results together confirm that AIN-93 causes hepatic steatosis and choline and/or cysteine deficiencies produce important hepatic injury associated with oxidative stress and inflammatory profiles.
Collapse
|
35
|
Choi HN, Jeong SM, Huh GH, Kim JI. Quercetin ameliorates insulin sensitivity and liver steatosis partly by increasing adiponectin expression in ob/ob mice. Food Sci Biotechnol 2015. [DOI: 10.1007/s10068-015-0036-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
|
36
|
Tzeng TF, Liou SS, Chang CJ, Liu IM. 6-gingerol protects against nutritional steatohepatitis by regulating key genes related to inflammation and lipid metabolism. Nutrients 2015; 7:999-1020. [PMID: 25658238 PMCID: PMC4344571 DOI: 10.3390/nu7020999] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 12/26/2014] [Accepted: 01/16/2015] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease, including non-alcoholic steatohepatitis (NASH), appears to be increasingly common worldwide. The aim of the study was to investigate the effects of 6-gingerol ((S)-5-hydroxy-1-(4-hydroxy-3-methoxyphenyl)-3-decanone), a bioactive ingredient of plants belonging to the Zingiberaceae family, on experimental models of NASH. In HepG2 cells, 6-gingerol (100 μmol/L) treatment inhibited free fatty acids mixture (0.33 mmol/L palmitate and 0.66 mmol/L oleate)-induced triglyceride and inflammatory marker accumulations. Male C57BL/6 mice were fed with a methionine and choline-deficient (MCD) diet to induce steatohepatitis. After four weeks of MCD diet feeding, the mice were dosed orally with 6-gingerol (25, 50 or 100 mg/kg/day) once daily for another four weeks. 6-Gingerol (100 mg/kg/day) attenuated liver steatosis and necro-inflammation in MCD diet-fed mice. The expressions of inflammatory cytokine genes, including those for monocyte chemoattractant protein-1, tumor necrosis factor-α, and interleukin-6, and nuclear transcription factor (NF-κB), which were increased in the livers of MCD diet-fed mice, were attenuated by 6-gingerol. 6-Gingerol possesses a repressive property on hepatic steatosis, which is associated with induction of peroxisome proliferator-activated receptor α. Our study demonstrated the protective role of 6-gingerol in ameliorating nutritional steatohepatitis. The effect was mediated through regulating key genes related to lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Thing-Fong Tzeng
- Department of Pharmacy & Graduate Institute of Pharmaceutical Technology, Tajen University, Yanpu Township, 90741 Pingtung County, Taiwan.
| | - Shorong-Shii Liou
- Department of Pharmacy & Graduate Institute of Pharmaceutical Technology, Tajen University, Yanpu Township, 90741 Pingtung County, Taiwan.
| | - Chia Ju Chang
- Department of Pharmacy & Graduate Institute of Pharmaceutical Technology, Tajen University, Yanpu Township, 90741 Pingtung County, Taiwan.
| | - I-Min Liu
- Department of Pharmacy & Graduate Institute of Pharmaceutical Technology, Tajen University, Yanpu Township, 90741 Pingtung County, Taiwan.
| |
Collapse
|
37
|
Tzeng TF, Liou SS, Liu IM. 6-Gingerol mitigates nutritional steatohepatitis through regulating key genes related to oxidative stress, inflammation and fibrogenesis. RSC Adv 2014. [DOI: 10.1039/c4ra12030b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
38
|
Chiu WC, Yang HH, Chiang SC, Chou YX, Yang HT. Auricularia polytricha aqueous extract supplementation decreases hepatic lipid accumulation and improves antioxidative status in animal model of nonalcoholic fatty liver. Biomedicine (Taipei) 2014; 4:12. [PMID: 25520925 PMCID: PMC4265006 DOI: 10.7603/s40681-014-0012-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 04/20/2014] [Indexed: 01/12/2023] Open
Abstract
Background: Amelioration effect of Auricularia polytricha water extract (AP) on hepatic injury in an animal model of NAFLD was investigated. Methods: Forty six-week-old Wistar rats were housed and thirty-two fed ten percent lard high-fat diet to induce NAFLD. After eight weeks of induction, animals were divided into five groups of eight rats each: normal control, high-fat diet, RN (reversion to a normal diet), 1× AP (normal diet plus 0.75% AP, w/w), and 2×AP (normal diet plus 1.5% AP). Animals were sacrificed four weeks later. Results: Rats receiving either 0.75% or 1.5% AP exhibited effective interruption of NAFLD progression, as evidenced by decreased lipid accumulation and elevated antioxidative status. Histological examination proved AP anti-inflammatory function and lower level of related markers for tumor necrosis factor-α and interleukin-6. Besides abundant polysaccharides against lipid accumulation, AP had a specific high level of phenolic compounds and tannins thus may be a potent anti-inflammatory and antioxidative agent. Conclusion: Findings suggest that under normal diet recovery, AP supplement may represent novel, protective material against NAFLD by attenuating inflammatory response, oxidative stress and lipid deposition.
Collapse
Affiliation(s)
- Wan-Chun Chiu
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Hsu-Hui Yang
- Council of Agriculture, Executive Yuan, Fengshan Tropical Horticultural Experimental Branch, Kaohsiung, Taiwan
| | - Shu-Chi Chiang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Yu-Xuan Chou
- Department of Nutrition, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taichung, Taiwan
| | - Hui-Ting Yang
- Department of Nutrition, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taichung, Taiwan
| |
Collapse
|
39
|
Reed C, Hutcheson J, Mayhew CN, Witkiewicz AK, Knudsen ES. RB tumor suppressive function in response to xenobiotic hepatocarcinogens. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1853-9. [PMID: 24726645 DOI: 10.1016/j.ajpath.2014.02.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/27/2014] [Accepted: 02/04/2014] [Indexed: 12/30/2022]
Abstract
Diverse etiologic events are associated with the development of hepatocellular carcinoma. During hepatocarcinogenesis, genetic events likely occur that subsequently cooperate with long-term exposures to further drive the progression of hepatocellular carcinoma. In this study, the frequent loss of the retinoblastoma (RB) tumor suppressor in hepatocellular carcinoma was modeled in response to diverse hepatic stresses. Loss of RB did not significantly affect the response to a steatotic stress as driven by a methionine- and choline-deficient diet. In addition, RB status did not significantly influence the response to peroxisome proliferators that can drive hepatomegaly and tumor development in rodents. However, RB loss exhibited a highly significant effect on the response to the xenobiotic1,4-Bis-[2-(3,5-dichloropyridyloxy)] benzene. Loss of RB yielded a unique proliferative response to this agent, which was distinct from both regenerative stresses and genotoxic carcinogens. Long-term exposure to 1,4-Bis-[2-(3,5-dichloropyridyloxy)] benzene yielded profound tumor development in RB-deficient livers that was principally absent in RB-sufficient tissue. These data demonstrate the context specificity of RB and the key role RB plays in the suppression of hepatocellular carcinoma driven by xenobiotic stress.
Collapse
Affiliation(s)
- Christopher Reed
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jack Hutcheson
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Agnieszka K Witkiewicz
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Erik S Knudsen
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
40
|
Choi HN, Jang YH, Kim MJ, Seo MJ, Kang BW, Jeong YK, Kim JI. Cordyceps militaris alleviates non-alcoholic fatty liver disease in ob/ob mice. Nutr Res Pract 2014; 8:172-6. [PMID: 24741401 PMCID: PMC3988506 DOI: 10.4162/nrp.2014.8.2.172] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 08/03/2013] [Accepted: 08/16/2013] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND/OBJECTIVES Non-alcoholic fatty liver disease (NAFLD) is becoming an important public health problem as metabolic syndrome and type 2 diabetes have become epidemic. In this study we investigated the protective effect of Cordyceps militaris (C. militaris) against NAFLD in an obese mouse model. MATERIALS/METHODS Four-week-old male ob/ob mice were fed an AIN-93G diet or a diet containing 1% C. militaris water extract for 10 weeks after 1 week of adaptation. Serum glucose, insulin, free fatty acid (FFA), alanine transaminase (ALT), and proinflammatory cytokines were measured. Hepatic levels of lipids, glutathione (GSH), and lipid peroxide were determined. RESULTS Consumption of C. militaris significantly decreased serum glucose, as well as homeostasis model assessment for insulin resistance (HOMA-IR), in ob/ob mice. In addition to lowering serum FFA levels, C. militaris also significantly decreased hepatic total lipids and triglyceride contents. Serum ALT activities and tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels were reduced by C. militaris. Consumption of C. militaris increased hepatic GSH and reduced lipid peroxide levels. CONCLUSIONS These results indicate that C. militaris can exert protective effects against development of NAFLD, partly by reducing inflammatory cytokines and improving hepatic antioxidant status in ob/ob mice.
Collapse
Affiliation(s)
- Ha-Neul Choi
- Department of Smart Food and Drugs, School of Food and Life Science, Inje University, 197 Inje-ro, Gimhae, Gyeongnam 621-749, Korea
| | - Yang-Hee Jang
- Department of Smart Food and Drugs, School of Food and Life Science, Inje University, 197 Inje-ro, Gimhae, Gyeongnam 621-749, Korea
| | - Min-Joo Kim
- Laboratory of Nutritional Analysis, Hurom Co., Ltd., Gyeongnam 660-701, Korea
| | - Min Jeong Seo
- Department of Biotechnology, Dong-A University, Busan 604-714, Korea. ; Medi-Farm Industrialization Research Center, Dong-A University, Busan 604-714, Korea
| | - Byoung Won Kang
- Medi-Farm Industrialization Research Center, Dong-A University, Busan 604-714, Korea
| | - Yong Kee Jeong
- Department of Biotechnology, Dong-A University, Busan 604-714, Korea. ; Medi-Farm Industrialization Research Center, Dong-A University, Busan 604-714, Korea
| | - Jung-In Kim
- Department of Smart Food and Drugs, School of Food and Life Science, Inje University, 197 Inje-ro, Gimhae, Gyeongnam 621-749, Korea
| |
Collapse
|
41
|
Narayan S, Flask CA, Kalhan SC, Wilson DL. Hepatic fat during fasting and refeeding by MRI fat quantification. J Magn Reson Imaging 2014; 41:347-53. [PMID: 24590550 DOI: 10.1002/jmri.24616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 12/03/2013] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To explore the sensitivity of high-field small animal magnetic resonance imaging to dynamic changes in fat content in the liver and to characterize the effect of prandial state on imaging studies of hepatic fat. MATERIALS AND METHODS A total of three timepoints were acquired using asymmetric spin-echo acquisitions for 12 mice with 24-hour spacing. After the first scan, half of the cohort was placed on a water-only diet. The second half of the cohort continued to have access to their high-fat chow. The scans were repeated after 24 hours. All animals were then returned to the high-fat diet, and the scans were again repeated after 24 hours. Fat fraction maps were computed using previously described methods. Regions of interests were manually drawn in the livers and the patterns of the two groups over time were compared. RESULTS Five out of six of the animals in the starved group showed an increase in hepatic fat fraction during the fasting period (average increase 0.54 ± 0.48), which decreased on refeeding. Analysis of variance indicated that the results significantly depended on both the group and the timepoint (P = 0.003). CONCLUSION Fat-water imaging methods are able to measure hepatic fat changes caused by short-term dietary perturbations.
Collapse
Affiliation(s)
- Sreenath Narayan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | |
Collapse
|
42
|
Bhatia H, Verma G, Datta M. miR-107 orchestrates ER stress induction and lipid accumulation by post-transcriptional regulation of fatty acid synthase in hepatocytes. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:334-43. [PMID: 24560669 DOI: 10.1016/j.bbagrm.2014.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/27/2014] [Accepted: 02/13/2014] [Indexed: 12/13/2022]
Abstract
MicroRNAs, a class of small non-coding RNAs, are believed to regulate several biological pathways and processes and are implicated in several diseases. They mostly regulate the levels of their target genes at the post transcriptional stage by primarily binding to the 3' UTR. Elevated hepatic levels of miR-107 are a consistent feature associated with several obese and diabetic models. Here, we show that miR-107 post-transcriptionally regulates fatty acid synthase (FASN) by binding to its 3' UTR and reduces its protein levels and the 3'UTR luciferase reporter activity, which are blunted by the miR-107 inhibitor and mutation in the miR-107 binding site in the 3' UTR. Knock-down of endogenous miR-107 levels increased FASN levels in a dose-dependent manner. Overexpression of miR-107 led to significant accumulation of malonyl CoA, accompanied by ER stress induction. All these events were prevented in the presence of the miR-107 inhibitor. While overexpression of FASN could attenuate miR-107 mediated ER stress markers' induction; the ER stress inhibitor, 4-phenyl-butyric acid did not rescue miR-107 induced FASN inhibition. This was followed by increased triglyceride formation and lipid accumulation in the presence of miR-107. These indicate that miR-107 inhibits FASN levels by binding to its 3' UTR and this interaction promotes ER stress induction and malonyl CoA and lipid accumulation in HepG2 cells and primary hepatocytes. Our results suggest that increased levels of miR-107 are critical in promoting lipid accumulation in hepatocytes and this might form the basis of diverse etiologies encountered in a fatty liver.
Collapse
Affiliation(s)
- Himanshi Bhatia
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110 007, India; AcSIR, Anusandhan Bhavan, Rafi Marg, New Delhi 110 001, India
| | - Gaurav Verma
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110 007, India; AcSIR, Anusandhan Bhavan, Rafi Marg, New Delhi 110 001, India
| | - Malabika Datta
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110 007, India.
| |
Collapse
|
43
|
'The way to a man's heart is through his gut microbiota'--dietary pro- and prebiotics for the management of cardiovascular risk. Proc Nutr Soc 2014; 73:172-85. [PMID: 24495527 DOI: 10.1017/s0029665113003911] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human gut microbiota has been identified as a possible novel CVD risk factor. This review aims to summarise recent insights connecting human gut microbiome activities with CVD and how such activities may be modulated by diet. Aberrant gut microbiota profiles have been associated with obesity, type 1 and type 2 diabetes and non-alcoholic fatty liver disease. Transfer of microbiota from obese animals induces metabolic disease and obesity in germ-free animals. Conversely, transfer of pathogen-free microbiota from lean healthy human donors to patients with metabolic disease can increase insulin sensitivity. Not only are aberrant microbiota profiles associated with metabolic disease, but the flux of metabolites derived from gut microbial metabolism of choline, phosphatidylcholine and l-carnitine has been shown to contribute directly to CVD pathology, providing one explanation for increased disease risk of eating too much red meat. Diet, especially high intake of fermentable fibres and plant polyphenols, appears to regulate microbial activities within the gut, supporting regulatory guidelines encouraging increased consumption of whole-plant foods (fruit, vegetables and whole-grain cereals), and providing the scientific rationale for the design of efficacious prebiotics. Similarly, recent human studies with carefully selected probiotic strains show that ingestion of viable microorganisms with the ability to hydrolyse bile salts can lower blood cholesterol, a recognised risk factor in CVD. Taken together such observations raise the intriguing possibility that gut microbiome modulation by whole-plant foods, probiotics and prebiotics may be at the base of healthy eating pyramids advised by regulatory agencies across the globe. In conclusion, dietary strategies which modulate the gut microbiota or their metabolic activities are emerging as efficacious tools for reducing CVD risk and indicate that indeed, the way to a healthy heart may be through a healthy gut microbiota.
Collapse
|
44
|
Park HJ, Han JM, Kim HG, Choi MK, Lee JS, Lee HW, Son CG. Chunggan extract (CGX), methionine-and choline-deficient (MCD) diet-induced hepatosteatosis and oxidative stress in C57BL/6 mice. Hum Exp Toxicol 2013; 32:1258-1269. [PMID: 23970447 DOI: 10.1177/0960327113485253] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In the present study, we aimed to evaluate the hepatoprotective and antioxidant effects of Chunggan extract (CGX) in an animal model of hepatosteatosis. The C57BL/6N mice were fed either methionine- and choline-sufficient (MCS) diet (n = 10) or a methionine- and choline-deficient (MCD) diet (n = 50) for 4 weeks, and then they were treated orally with CGX (100 or 200 mg/kg), ursodeoxycholic acid (80 mg/kg, as a positive control), or distilled water (DW, MCS diet group, and MCD diet group) for the final 2 weeks (once per day). The MCD diet induced severe hepatic injury with the typical features of hepatosteatosis in both serum and hepatic tissues. CGX treatment significantly attenuated these alterations in the serum levels including triglyceride (TG), aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, and total bilirubin. Moreover, CGX also efficiently prevented from the hepatic TG accumulation in the hepatic tissue, evidenced by histopathological findings, compared with the MCD diet. In addition, CGX treatment significantly ameliorated the excessive oxidative stress and antioxidant markers in the serum as well as the hepatic levels of reactive oxygen species, the levels of malondialdehyde, the protein carbonyl, and total antioxidant capacity, and the activities of superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase. In conclusion, our results indicate the experimental relevance of CGX for potential clinical application in patients with hepatosteatotic disorders and a possible mechanism related to its antioxidant properties.
Collapse
Affiliation(s)
- H-J Park
- 1Department of Liver and Immunology Research Center, Daejeon Oriental Hospital of Oriental Medical College of Daejeon University, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
45
|
Boeykens N, Ponsaerts P, Van der Linden A, Berneman Z, Ysebaert D, De Greef K. Injury-dependent retention of intraportally administered mesenchymal stromal cells following partial hepatectomy of steatotic liver does not lead to improved liver recovery. PLoS One 2013; 8:e69092. [PMID: 23874878 PMCID: PMC3715456 DOI: 10.1371/journal.pone.0069092] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/04/2013] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to evaluate the effect of bone marrow-derived mesenchymal stromal cell (BM-MSC) administration on liver function following partial hepatectomy (PHx) of methionine/choline-deficient (MCD) diet induced steatotic livers in rodents. Here we identified and validated serum cholinesterase (CHE) and triglyceride (TG) levels as non-invasive markers to longitudinally monitor rat liver function. Using in vivo bioluminescence imaging, retention of BM-MSC in the liver was observed following intraportal administration, but not after intravenous administration. Therefore, BM-MSC were intraportally delivered to investigate the effect on liver recovery and/or regeneration after PHx. However, despite recovery to normal body weight, liver weight and NAS score, both serum CHE and TG levels of non-treated and cell-treated rats with PHx after MCD diet remained significantly lower as compared to those of control rats. Importantly, serum CHE levels, but not TG levels, of cell-treated rats remained significantly lower as compared to those of non-treated rats, thereby warranting that certain caution should be considered for future clinical application of IP BM-MSC administration in order to promote liver regeneration and/or function.
Collapse
Affiliation(s)
- Nele Boeykens
- Laboratory of Experimental Surgery, Antwerp Surgical Training and Research Centre, University of Antwerp/University Hospital of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | | | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Dirk Ysebaert
- Laboratory of Experimental Surgery, Antwerp Surgical Training and Research Centre, University of Antwerp/University Hospital of Antwerp, Antwerp, Belgium
- * E-mail:
| | - Kathleen De Greef
- Laboratory of Experimental Surgery, Antwerp Surgical Training and Research Centre, University of Antwerp/University Hospital of Antwerp, Antwerp, Belgium
| |
Collapse
|
46
|
Fernando H, Wiktorowicz JE, Soman KV, Kaphalia BS, Khan MF, Ansari GAS. Liver proteomics in progressive alcoholic steatosis. Toxicol Appl Pharmacol 2013; 266:470-80. [PMID: 23200777 PMCID: PMC3565568 DOI: 10.1016/j.taap.2012.11.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/15/2012] [Accepted: 11/16/2012] [Indexed: 02/08/2023]
Abstract
Fatty liver is an early stage of alcoholic and nonalcoholic liver disease (ALD and NALD) that progresses to steatohepatitis and other irreversible conditions. In this study, we identified proteins that were differentially expressed in the livers of rats fed 5% ethanol in a Lieber-DeCarli diet daily for 1 and 3 months by discovery proteomics (two-dimensional gel electrophoresis and mass spectrometry) and non-parametric modeling (Multivariate Adaptive Regression Splines). Hepatic fatty infiltration was significantly higher in ethanol-fed animals as compared to controls, and more pronounced at 3 months of ethanol feeding. Discovery proteomics identified changes in the expression of proteins involved in alcohol, lipid, and amino acid metabolism after ethanol feeding. At 1 and 3 months, 12 and 15 different proteins were differentially expressed. Of the identified proteins, down regulation of alcohol dehydrogenase (-1.6) at 1 month and up regulation of aldehyde dehydrogenase (2.1) at 3 months could be a protective/adaptive mechanism against ethanol toxicity. In addition, betaine-homocysteine S-methyltransferase 2 a protein responsible for methionine metabolism and previously implicated in fatty liver development was significantly up regulated (1.4) at ethanol-induced fatty liver stage (1 month) while peroxiredoxin-1 was down regulated (-1.5) at late fatty liver stage (3 months). Nonparametric analysis of the protein spots yielded fewer proteins and narrowed the list of possible markers and identified d-dopachrome tautomerase (-1.7, at 3 months) as a possible marker for ethanol-induced early steatohepatitis. The observed differential regulation of proteins have potential to serve as biomarker signature for the detection of steatosis and its progression to steatohepatitis once validated in plasma/serum.
Collapse
Affiliation(s)
- Harshica Fernando
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555
| | - John E. Wiktorowicz
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555
| | - Kizhake V. Soman
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555
| | - Bhupendra S. Kaphalia
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555
| | - M. Firoze Khan
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555
| | - G. A. Shakeel Ansari
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555
| |
Collapse
|
47
|
Yang J, Lv F, Chen XQ, Cui WX, Chen LH, Wen XD, Wang Q. Pharmacokinetic study of major bioactive components in rats after oral administration of extract of Ilex hainanensis by high-performance liquid chromatography/electrospray ionization mass spectrometry. J Pharm Biomed Anal 2013; 77:21-8. [PMID: 23384548 DOI: 10.1016/j.jpba.2013.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Revised: 01/04/2013] [Accepted: 01/05/2013] [Indexed: 12/21/2022]
Abstract
Ilex hainanensis Merr. is commonly used as a folk remedy for treating hypertension, dyslipidemia and inflammation in Traditional Chinese Medicine (TCMs) and it also has great potential to treat non-alcoholic fatty liver disease (NAFLD). Chlorogenic acid, kaempferol-7-O-β-d-glucoside, and ilexgenin A are three major bioactive components in I. hainanensis extract. In this study, a rapid, sensitive and convenient LC-MS method was developed for their simultaneous determination in the plasma of normal and NAFLD rats. The method was validated in terms of selectivity, linearity and sensitivity, and shows advantages in monitoring the pharmacokinetic behaviors of these three compounds. Results revealed the pharmacokinetic behaviors of chlorogenic acid, kaempferol-7-O-β-d-glucoside, and ilexgenin A could be significantly changed in NAFLD rats after oral administration of I. hainanensis extract compared with normal rats. The areas under the plasma concentration-time curve (AUC) and maximum plasma concentration (Cmax) of the three analytes were greatly decreased and the plasma clearance (CL) for kaempferol-7-O-β-d-glucoside, Ilexgenin A were greatly increased in NAFLD rats. Meanwhile, the mean residence time (MRT) of kaempferol-7-O-β-d-glucoside and Ilexgenin A were increased in the NAFLD rats. This is the first report on the determination of the major bioactive components in rat plasma after oral administration of I. hainanensis extract. These results provided a meaningful basis for evaluating the clinical application of this medicine.
Collapse
Affiliation(s)
- Jie Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | | | | | | | | | | | | |
Collapse
|
48
|
Dandelion Leaf Extract Protects Against Liver Injury Induced by Methionine- and Choline-Deficient Diet in Mice. J Med Food 2013; 16:26-33. [DOI: 10.1089/jmf.2012.2226] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
49
|
Wang SD, Xie ZQ, Chen J, Wang K, Wei T, Zhao AH, Zhang QH. Inhibitory effect of Ginkgo biloba extract on fatty liver: regulation of carnitine palmitoyltransferase 1a and fatty acid metabolism. J Dig Dis 2012; 13:525-35. [PMID: 22988926 DOI: 10.1111/j.1751-2980.2012.00627.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate the potential effect of Ginkgo biloba extract (GBE) on the prevention and treatment of nonalcoholic fatty liver disease (NAFLD). METHODS Male Wistar rats were divided into 4 groups (the control group, GBE group, high-fat diet [HFD] group and HFD + GBE group). The human hepatocellular carcinoma cell line (HepG2) was treated with GBE and its flavonoid ingredients. The fatty acid composition of the rat liver was analyzed with gas chromatography/time-of-flight mass spectrometry (GC/TOFMS). Triglyceride contents of both the rat liver and HepG2 cells were measured by enzymatic colorimetric method. The expressions of fatty acid metabolism-related genes were analyzed with real-time reverse transcription-polymerase chain reaction (RT-PCR). The protein expression and enzymatic activity were subsequently measured. RESULTS In rat livers, GBE reduced the elevations of hepatic triglyceride contents caused by HFD and the increased hepatic fatty acids were differentially affected by GBE. Notably, the expression and total activity of the fatty acid β-oxidation rate-limiting enzyme, carnitine palmitoyltransferase 1a (CPT1A), were also promoted with GBE ingestion. In HepG2 cells, GBE and its ingredients, quercetin, kaempferol and isorhamnetin, could decrease the cellular triglyceride content and upregulate the expression and total activity of CPT1A, respectively. CONCLUSIONS The triglyceride-lowering effect of GBE on the HFD rat liver is closely associated with the increased expression and activity of CPT1A, and the flavonoid ingredients are the major contributors of GBE.
Collapse
Affiliation(s)
- Shi Dong Wang
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Hardwick RN, Cherrington NJ. Measuring altered disposition of xenobiotics in experimental models of liver disease. ACTA ACUST UNITED AC 2012; Chapter 23:Unit 23.1.. [PMID: 22549269 DOI: 10.1002/0471140856.tx2301s52] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Understanding the metabolic pathway and excretion mechanisms governing the disposition of a compound is essential to the safe use of pharmaceutical agents. Because the liver is the primary organ responsible for the metabolism and elimination of xenobiotics, chronic liver disease can have a significant effect on the disposition of many xenobiotics due to changes in the expression or function of drug metabolizing enzymes and transporters. Liver disease can result in increased retention of a xenobiotic within the body, causing greater exposure of the individual to a potentially harmful compound, which may lead to toxicity. On the other hand, liver disease may also up-regulate the elimination processes of a xenobiotic, accelerating its removal from the body. With regard to a pharmaceutical agent, enhanced elimination may result in a decreased pharmacologic effect. Such alterations may necessitate dosage adjustments to achieve the desired therapeutic outcome.
Collapse
Affiliation(s)
- Rhiannon N Hardwick
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA
| | | |
Collapse
|