1
|
Saad MJA, Santos A. The Microbiota and Evolution of Obesity. Endocr Rev 2025; 46:300-316. [PMID: 39673174 PMCID: PMC11894537 DOI: 10.1210/endrev/bnae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/03/2024] [Accepted: 12/12/2024] [Indexed: 12/16/2024]
Abstract
Obesity is a major global concern and is generally attributed to a combination of genetic and environmental factors. Several hypotheses have been proposed to explain the evolutionary origins of obesity epidemic, including thrifty and drifty genotypes, and changes in thermogenesis. Here, we put forward the hypothesis of metaflammation, which proposes that due to intense selection pressures exerted by environmental pathogens, specific genes that help develop a robust defense mechanism against infectious diseases have had evolutionary advantages and that this may contribute to obesity in modern times due to connections between the immune and energy storage systems. Indeed, incorporating the genetic variations of gut microbiota into the complex genetic framework of obesity makes it more polygenic than previously believed. Thus, uncovering the evolutionary origins of obesity requires a multifaceted approach that considers the complexity of human history, the unique genetic makeup of different populations, and the influence of gut microbiome on host genetics.
Collapse
Affiliation(s)
- Mario J A Saad
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, CEP 13083-887 Campinas, SP, Brazil
| | - Andrey Santos
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, CEP 13083-887 Campinas, SP, Brazil
| |
Collapse
|
2
|
Shao G, Liu Y, Lu L, Wang L, Ji G, Xu H. Therapeutic potential of traditional Chinese medicine in the prevention and treatment of digestive inflammatory cancer transformation: Portulaca oleracea L. as a promising drug. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:117999. [PMID: 38447616 DOI: 10.1016/j.jep.2024.117999] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) has been used for centuries to treat various types of inflammation and tumors of the digestive system. Portulaca oleracea L. (POL), has been used in TCM for thousands of years. The chemical composition of POL is variable and includes flavonoids, alkaloids, terpenoids and organic acids and other classes of natural compounds. Many of these compounds exhibit powerful anti-inflammatory and anti-cancer-transforming effects in the digestive system. AIM OF STUDY In this review, we focus on the potential therapeutic role of POL in NASH, gastritis and colitis and their associated cancers, with a focus on the pharmacological properties and potential mechanisms of action of the main natural active compounds in POL. METHODS The information and data on Portulaca oleracea L. and its main active ingredients were collated from various resources like ethnobotanical textbooks and literature databases such as CNKI, VIP (Chinese literature), PubMed, Science Direct, Elsevier and Google Scholar (English literatures), Wiley, Springer, Tailor and Francis, Scopus, Inflibnet. RESULTS Kaempferol, luteolin, myricetin, quercetin, genistein, EPA, DHA, and melatonin were found to improve NASH and NASH-HCC, while kaempferol, apigenin, luteolin, and quercetin played a therapeutic role in gastritis and gastric cancer. Apigenin, luteolin, myricetin, quercetin, genistein, lupeol, vitamin C and melatonin were found to have therapeutic effects in the treatment of colitis and its associated cancers. The discovery of the beneficial effects of these natural active compounds in POL supports the idea that POL could be a promising novel candidate for the treatment and prevention of inflammation-related cancers of the digestive system. CONCLUSION The discovery of the beneficial effects of these natural active compounds in POL supports the idea that POL could be a promising novel candidate for the treatment and prevention of inflammation-related cancers of the digestive system. However, clinical data describing the mode of action of the naturally active compounds of POL are still lacking. In addition, pharmacokinetic data for POL compounds, such as changes in drug dose and absorption rates, cannot be extrapolated from animal models and need to be measured in patients in clinical trials. On the one hand, a systematic meta-analysis of the existing publications on TCM containing POL still needs to be carried out. On the other hand, studies on the hepatic and renal toxicity of POL are also needed. Additionally, well-designed preclinical and clinical studies to validate the therapeutic effects of TCM need to be performed, thus hopefully providing a basis for the validation of the clinical benefits of POL.
Collapse
Affiliation(s)
- Gaoxuan Shao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China
| | - Ying Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China
| | - Lu Lu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China
| | - Lei Wang
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China.
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China.
| |
Collapse
|
3
|
Wu YC, Yan Q, Yue SQ, Pan LX, Yang DS, Tao LS, Wei ZY, Rong F, Qian C, Han MQ, Zuo FC, Yang JF, Xu JJ, Shi ZR, Du J, Chen ZL, Xu T. NUP85 alleviates lipid metabolism and inflammation by regulating PI3K/AKT signaling pathway in nonalcoholic fatty liver disease. Int J Biol Sci 2024; 20:2219-2235. [PMID: 38617542 PMCID: PMC11008257 DOI: 10.7150/ijbs.92337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/10/2024] [Indexed: 04/16/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the common causes of chronic liver disease in the world. The problem of NAFLD had become increasingly prominent. However, its pathogenesis is still indistinct. As we all know, NAFLD begins with the accumulation of triglyceride (TG), leading to fatty degeneration, inflammation and other liver tissues damage. Notably, structure of nucleoporin 85 (NUP85) is related to lipid metabolism and inflammation of liver diseases. In this study, the results of researches indicated that NUP85 played a critical role in NAFLD. Firstly, the expression level of NUP85 in methionine-choline-deficient (MCD)-induced mice increased distinctly, as well as the levels of fat disorder and inflammation. On the contrary, knockdown of NUP85 had the opposite effects. In vitro, AML-12 cells were stimulated with 2 mm free fatty acids (FFA) for 24 h. Results also proved that NUP85 significantly increased in model group, and increased lipid accumulation and inflammation level. Besides, NUP85 protein could interact with C-C motif chemokine receptor 2 (CCR2). Furthermore, when NUP85 protein expressed at an extremely low level, the expression level of CCR2 protein also decreased, accompanied with an inhibition of phosphorylation of phosphoinositol-3 kinase (PI3K)-protein kinase B (AKT) signaling pathway. What is more, trans isomer (ISRIB), a targeted inhibitor of NUP85, could alleviate NAFLD. In summary, our findings suggested that NUP85 functions as an important regulator in NAFLD through modulation of CCR2.
Collapse
Affiliation(s)
- Yin-cui Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Qi Yan
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Si-qing Yue
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Lin-xin Pan
- College of life sciences, Anhui Medical University, Hefei 230032, China
| | - Da-shuai Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Liang-song Tao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Ze-yuan Wei
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Fan Rong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Cheng Qian
- Research and Experiment center, Anhui Medical University, Hefei 230032, China
| | - Meng-qi Han
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Fu-cheng Zuo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Jun-fa Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| | - Jia-jia Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Zheng-rong Shi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhao-lin Chen
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, Anhui, 230001, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, China
| |
Collapse
|
4
|
Makiuchi N, Takano S, Tada Y, Kasai K, Igarashi N, Kani K, Kato M, Goto K, Matsuura Y, Ichimura-Shimizu M, Furusawa Y, Tsuneyama K, Nagai Y. Dynamics of Liver Macrophage Subsets in a Novel Mouse Model of Non-Alcoholic Steatohepatitis Using C57BL/6 Mice. Biomedicines 2023; 11:2659. [PMID: 37893033 PMCID: PMC10604124 DOI: 10.3390/biomedicines11102659] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Macrophages are critical for the development of non-alcoholic steatohepatitis (NASH). Our previous findings in TSNO mouse livers showed that an iHFC (high-fat/cholesterol/cholate) diet induced liver fibrosis similar to human NASH and led to the accumulation of distinct subsets of macrophage: CD11c+/Ly6C- and CD11c-/Ly6C+ cells. CD11c+/Ly6C- cells were associated with the promotion of advanced liver fibrosis in NASH. On the other hand, CD11c-/Ly6C+ cells exhibited an anti-inflammatory effect and were involved in tissue remodeling processes. This study aimed to elucidate whether an iHFC diet with reduced cholic acid (iHFC#2 diet) induces NASH in C57BL/6 mice and examine the macrophage subsets accumulating in the liver. Histological and quantitative real-time PCR analyses revealed that the iHFC#2 diet promoted inflammation and fibrosis indicative of NASH in the livers of C57BL/6 mice. Cell numbers of Kupffer cells decreased and recruited macrophages were accumulated in the livers of iHFC#2 diet-fed C57BL/6 mice. Notably, the iHFC#2 diet resulted in the accumulation of three macrophage subsets in the livers of C57BL/6 mice: CD11c+/Ly6C-, CD11c-/Ly6C+, and CD11c+/Ly6C+ cells. However, CD11c+/Ly6C+ cells were not distinct populations in the iHFC-fed TSNO mice. Thus, differences in cholic acid content and mouse strain affect the macrophage subsets that accumulate in the liver.
Collapse
Affiliation(s)
- Nana Makiuchi
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Shun Takano
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Yuki Tada
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Kaichi Kasai
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Naoya Igarashi
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Koudai Kani
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Miyuna Kato
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Kana Goto
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Yudai Matsuura
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Mayuko Ichimura-Shimizu
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-8-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.I.-S.); (K.T.)
| | - Yukihiro Furusawa
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-8-15 Kuramoto-cho, Tokushima 770-8503, Japan; (M.I.-S.); (K.T.)
| | - Yoshinori Nagai
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan; (N.M.); (S.T.); (Y.T.); (K.K.); (N.I.); (K.K.); (M.K.); (K.G.); (Y.M.); (Y.F.)
| |
Collapse
|
5
|
Aoki H, Isobe Y, Yoshida M, Kang JX, Maekawa M, Arita M. Enzymatically-epoxidized docosahexaenoic acid, 19,20-EpDPE, suppresses hepatic crown-like structure formation and nonalcoholic steatohepatitis fibrosis through GPR120. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159275. [PMID: 36566874 DOI: 10.1016/j.bbalip.2022.159275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
A hepatic crown-like structure (hCLS) formed by macrophages accumulating around lipid droplets and dead cells in the liver is a unique feature of nonalcoholic steatohepatitis (NASH) that triggers progression of liver fibrosis. As hCLS plays a key role in the progression of NASH fibrosis, hCLS formation has emerged as a potential therapeutic target. n-3 polyunsaturated fatty acids (n-3 PUFAs) have potential suppressive effects on NASH fibrosis; however, the mechanisms underlying this effect are poorly understood. Here, we report that n-3 PUFA-enriched Fat-1 transgenic mice are resistant to hCLS formation and liver fibrosis in a NASH model induced by a combination of high-fat diet, CCl4 and a Liver X receptor (LXR) agonist. Liquid chromatography-tandem mass spectrometry-based mediator lipidomics revealed that the amount of endogenous n-3 PUFA-derived metabolites, such as 17,18-dihydroxyeicosatetraenoic acid (17,18-diHETE), and 19,20-epoxy docosapentaenoic acid (19,20-EpDPE), was significantly elevated in Fat-1 mice, along with hCLS formation. In particular, DHA-derived 19,20-EpDPE produced by Cyp4f18 attenuated the hCLS formation and liver fibrosis in a G protein-coupled receptor 120 (GPR120)-dependent manner. These results indicated that 19,20-EpDPE is an endogenous active metabolite that mediates the preventive effect of n-3 PUFAs against NASH fibrosis.
Collapse
Affiliation(s)
- Hidenori Aoki
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan; Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa 230-0045, Japan
| | - Yosuke Isobe
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan; Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa 230-0045, Japan
| | - Mio Yoshida
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan; Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa 230-0045, Japan
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Massachusetts General Hospital and Harvard Medical School, 02114 Boston, MA, USA
| | - Masashi Maekawa
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan; Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa 230-0045, Japan
| | - Makoto Arita
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan; Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa 230-0045, Japan; Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa 230-0045, Japan.
| |
Collapse
|
6
|
Tada Y, Kasai K, Makiuchi N, Igarashi N, Kani K, Takano S, Honda H, Yanagibashi T, Watanabe Y, Usui-Kawanishi F, Furusawa Y, Ichimura-Shimizu M, Tabuchi Y, Takatsu K, Tsuneyama K, Nagai Y. Roles of Macrophages in Advanced Liver Fibrosis, Identified Using a Newly Established Mouse Model of Diet-Induced Non-Alcoholic Steatohepatitis. Int J Mol Sci 2022; 23:13251. [PMID: 36362037 PMCID: PMC9654696 DOI: 10.3390/ijms232113251] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 10/29/2023] Open
Abstract
Macrophages play critical roles in the pathogenesis of non-alcoholic steatohepatitis (NASH). However, it is unclear which macrophage subsets are critically involved in the development of inflammation and fibrosis in NASH. In TSNO mice fed a high-fat/cholesterol/cholate-based diet, which exhibit advanced liver fibrosis that mimics human NASH, we found that Kupffer cells (KCs) were less abundant and recruited macrophages were more abundant, forming hepatic crown-like structures (hCLS) in the liver. The recruited macrophages comprised two subsets: CD11c+/Ly6C- and CD11c-/Ly6C+ cells. CD11c+ cells were present in a mesh-like pattern around the lipid droplets, constituting the hCLS. In addition, CD11c+ cells colocalized with collagen fibers, suggesting that this subset of recruited macrophages might promote advanced liver fibrosis. In contrast, Ly6C+ cells were present in doughnut-like inflammatory lesions, with a lipid droplet in the center. Finally, RNA sequence analysis indicates that CD11c+/Ly6C- cells promote liver fibrosis and hepatic stellate cell (HSC) activation, whereas CD11c-/Ly6C+ cells are a macrophage subset that play an anti-inflammatory role and promote tissue repair in NASH. Taken together, our data revealed changes in liver macrophage subsets during the development of NASH and shed light on the roles of the recruited macrophages in the pathogenesis of advanced fibrosis in NASH.
Collapse
Affiliation(s)
- Yuki Tada
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Toyama 939-0398, Japan
| | - Kaichi Kasai
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Toyama 939-0398, Japan
| | - Nana Makiuchi
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Toyama 939-0398, Japan
| | - Naoya Igarashi
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Toyama 939-0398, Japan
| | - Koudai Kani
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Toyama 939-0398, Japan
| | - Shun Takano
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Toyama 939-0398, Japan
| | - Hiroe Honda
- Toyama Prefectural Institute for Pharmaceutical Research, 17-1 Nakataikouyama, Toyama 939-0363, Japan
| | - Tsutomu Yanagibashi
- Toyama Prefectural Institute for Pharmaceutical Research, 17-1 Nakataikouyama, Toyama 939-0363, Japan
| | - Yasuharu Watanabe
- Toyama Prefectural Institute for Pharmaceutical Research, 17-1 Nakataikouyama, Toyama 939-0363, Japan
| | - Fumitake Usui-Kawanishi
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Toyama 939-0398, Japan
| | - Yukihiro Furusawa
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Toyama 939-0398, Japan
| | - Mayuko Ichimura-Shimizu
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-8-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Kiyoshi Takatsu
- Toyama Prefectural Institute for Pharmaceutical Research, 17-1 Nakataikouyama, Toyama 939-0363, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, 3-8-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yoshinori Nagai
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Toyama 939-0398, Japan
| |
Collapse
|
7
|
Elebeedy D, Ghanem A, Saleh A, Ibrahim MH, Kamaly OA, Abourehab MAS, Ali MA, Abd El Maksoud AI, El Hassab MA, Eldehna WM. In Vivo and In Silico Investigation of the Anti-Obesity Effects of Lactiplantibacillus plantarum Combined with Chia Seeds, Green Tea, and Chitosan in Alleviating Hyperlipidemia and Inflammation. Int J Mol Sci 2022; 23:12200. [PMID: 36293055 PMCID: PMC9602495 DOI: 10.3390/ijms232012200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022] Open
Abstract
The increasing prevalence of obesity has become a demanding issue in both high-income and low-income countries. Treating obesity is challenging as the treatment options have many limitations. Recently, diet modification has been commonly applied to control or prevent obesity and its risks. In this study, we investigated novel therapeutic approaches using a combination of a potential probiotic source with prebiotics. Forty-eight adult male Sprague-Dawley rats were selected and divided into seven groups (eight rats per group). The first group was fed a high-fat diet, while the second group was a negative control. The other five groups were orally administered with a probiotic, Lactiplantibacillus plantarum (L. plantarum), and potential prebiotics sources (chia seeds, green tea, and chitosan) either individually or in combination for 45 days. We collected blood samples to analyze the biochemical parameters and dissected organs, including the liver, kidney, and pancreas, to evaluate obesity-related injuries. We observed a more significant decrease in the total body weight by combining these approaches than with individual agents. Moreover, treating the obese rats with this combination decreased serum catalase, superoxide dismutase, and liver malondialdehyde levels. A histopathological examination revealed a reduction in obesity-related injuries in the liver, kidney, and pancreas. Further docking studies indicated the potential role of chia seeds and green tea components in modulating obesity and its related problems. Therefore, we suggest that the daily administration of a pre- and probiotic combination may reduce obesity and its related problems.
Collapse
Affiliation(s)
- Dalia Elebeedy
- Pharmaceutical Biotechnology Department, College of Biotechnology, Misr University for Science and Technology (MUST), 6th of October City 12573, Egypt
| | - Aml Ghanem
- School of Biotechnology, Badr University in Cairo, Cairo 11829, Egypt
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mona H. Ibrahim
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo 11754, Egypt
| | - Omkulthom Al Kamaly
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mohammed A. S. Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, P.O. Box 21961, Makkah 24382, Saudi Arabia
| | - Mohamed A. Ali
- School of Biotechnology, Badr University in Cairo, Cairo 11829, Egypt
| | - Ahmed I. Abd El Maksoud
- Industrial Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City 32897, Egypt
| | - Mahmoud A. El Hassab
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai 46612, Egypt
| | - Wagdy M. Eldehna
- School of Biotechnology, Badr University in Cairo, Cairo 11829, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33511, Egypt
| |
Collapse
|
8
|
Paul B, Lewinska M, Andersen JB. Lipid alterations in chronic liver disease and liver cancer. JHEP Rep 2022; 4:100479. [PMID: 35469167 PMCID: PMC9034302 DOI: 10.1016/j.jhepr.2022.100479] [Citation(s) in RCA: 147] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
Lipids are a complex and diverse group of molecules with crucial roles in many physiological processes, as well as in the onset, progression, and maintenance of cancers. Fatty acids and cholesterol are the building blocks of lipids, orchestrating these crucial metabolic processes. In the liver, lipid alterations are prevalent as a cause and consequence of chronic hepatitis B and C virus infections, alcoholic hepatitis, and non-alcoholic fatty liver disease and steatohepatitis. Recent developments in lipidomics have also revealed that dynamic changes in triacylglycerols, phospholipids, sphingolipids, ceramides, fatty acids, and cholesterol are involved in the development and progression of primary liver cancer. Accordingly, the transcriptional landscape of lipid metabolism suggests a carcinogenic role of increasing fatty acids and sterol synthesis. However, limited mechanistic insights into the complex nature of the hepatic lipidome have so far hindered the development of effective therapies.
Collapse
|
9
|
Prikhodko VA, Bezborodkina NN, Okovityi SV. Pharmacotherapy for Non-Alcoholic Fatty Liver Disease: Emerging Targets and Drug Candidates. Biomedicines 2022; 10:274. [PMID: 35203484 PMCID: PMC8869100 DOI: 10.3390/biomedicines10020274] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/08/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), or metabolic (dysfunction)-associated fatty liver disease (MAFLD), is characterized by high global incidence and prevalence, a tight association with common metabolic comorbidities, and a substantial risk of progression and associated mortality. Despite the increasingly high medical and socioeconomic burden of NAFLD, the lack of approved pharmacotherapy regimens remains an unsolved issue. In this paper, we aimed to provide an update on the rapidly changing therapeutic landscape and highlight the major novel approaches to the treatment of this disease. In addition to describing the biomolecules and pathways identified as upcoming pharmacological targets for NAFLD, we reviewed the current status of drug discovery and development pipeline with a special focus on recent evidence from clinical trials.
Collapse
Affiliation(s)
- Veronika A. Prikhodko
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical and Pharmaceutical University, 14A Prof. Popov Str., 197022 St. Petersburg, Russia;
| | - Natalia N. Bezborodkina
- Zoological Institute, Russian Academy of Sciences, 1 Universitetskaya emb., 199034 St. Petersburg, Russia;
| | - Sergey V. Okovityi
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical and Pharmaceutical University, 14A Prof. Popov Str., 197022 St. Petersburg, Russia;
- Scientific, Clinical and Educational Center of Gastroenterology and Hepatology, Saint Petersburg State University, 7/9 Universitetskaya emb., 199034 St. Petersburg, Russia
| |
Collapse
|
10
|
Kizilaslan N, Zekiye Erdem N, Katar M, Gevrek F. The Effects of Probiotics and Omega-3 Fatty Acids in Liver Steatosis Induced in Rats by High-Fructose Corn Syrup. Int J Clin Pract 2022; 2022:7172492. [PMID: 35685520 PMCID: PMC9159191 DOI: 10.1155/2022/7172492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022] Open
Abstract
AIMS This study was designed to reveal the effect of probiotics and omega-3 fatty acids in a fatty liver model in rats induced by high-fructose corn syrup (HFCS). METHODS In the study, 40 male Wistar Albino rats were used, and these rats were divided into five groups. HFCS was added to the drinking water (30% solution) of four groups (Groups 2, 3, 4, and 5) for three weeks, and the animals were fed ad libitum. At the end of three weeks, the rats in Groups 3, 4, and 5 were administered omega-3 fatty acids (400 mg/kg) and probiotics (1.5 × 109 cfu/mL/day) with the gavage method for four weeks. The body weights of rats were weighed and recorded before starting the experiment, at the end of the third week, and before the animals were sacrificed at the last week, all at the same hour. By subtracting the remaining amount of food and water from the daily food and water amount, the amount of food and water consumed was calculated. These values were recorded for seven weeks. At the end of the seven weeks, the rats were sacrificed after blood specimens and tissues were taken. RESULTS Analyzing the changes in the food intake of each group within itself throughout the experiment, it was observed that there was an increase in the food intake in the control group; from the starting week to the last week, the food intake amount of the HFCS group began to decrease particularly after the second week; and it began to decrease after the third week in the groups that were administered probiotics and omega-3 fatty acids. The changes in the sacrifice weights in the HFCS + omega-3 fatty acid, HFCS + probiotic, and HFCS + probiotic + omega-3 fatty acid groups were found to be lower than that in the HFCS group. The maximum levels of glucose, ALT, ALP, serum cholesterol, triglyceride and AST were found to be in the HFCS group. It was determined that the minimum mean steatosis level was in the control group, while the maximum steatosis level was in the HFCS group. CONCLUSIONS As a result, there was a protective effect of probiotic and omega-3 fatty acid.
Collapse
Affiliation(s)
- Nildem Kizilaslan
- Tokat Gaziosmanpasa University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Tokat, Turkey
| | - Nihal Zekiye Erdem
- Istanbul Medipol University, School of Health Sciences, Department of Nutrition and Dietetics, Istanbul, Turkey
| | - Muzaffer Katar
- Tokat Gaziosmanpasa University, Faculty of Medicine, Department of Biochemistry, Tokat, Turkey
| | - Fikret Gevrek
- Tokat Gaziosmanpasa University, Faculty of Medicine, Department of Histology, Tokat, Turkey
| |
Collapse
|
11
|
Tamura YO, Sugama J, Iwasaki S, Sasaki M, Yasuno H, Aoyama K, Watanabe M, Erion DM, Yashiro H. Selective Acetyl-CoA Carboxylase 1 Inhibitor Improves Hepatic Steatosis and Hepatic Fibrosis in a Preclinical Nonalcoholic Steatohepatitis Model. J Pharmacol Exp Ther 2021; 379:280-289. [PMID: 34535562 DOI: 10.1124/jpet.121.000786] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022] Open
Abstract
Acetyl-CoA carboxylase (ACC) 1 and ACC2 are essential rate-limiting enzymes that synthesize malonyl-CoA (M-CoA) from acetyl-CoA. ACC1 is predominantly expressed in lipogenic tissues and regulates the de novo lipogenesis flux. It is upregulated in the liver of patients with nonalcoholic fatty liver disease (NAFLD), which ultimately leads to the formation of fatty liver. Therefore, selective ACC1 inhibitors may prevent the pathophysiology of NAFLD and nonalcoholic steatohepatitis (NASH) by reducing hepatic fat, inflammation, and fibrosis. Many studies have suggested ACC1/2 dual inhibitors for treating NAFLD/NASH; however, reports on selective ACC1 inhibitors are lacking. In this study, we investigated the effects of compound-1, a selective ACC1 inhibitor for treating NAFLD/NASH, using preclinical in vitro and in vivo models. Compound-1 reduced M-CoA content and inhibited the incorporation of [14C] acetate into fatty acids in HepG2 cells. Additionally, it reduced hepatic M-CoA content and inhibited de novo lipogenesis in C57BL/6J mice after a single dose. Furthermore, compound-1 treatment of 8 weeks in Western diet-fed melanocortin 4 receptor knockout mice-NAFLD/NASH mouse model-improved liver hypertrophy and reduced hepatic triglyceride content. The reduction of hepatic M-CoA by the selective ACC1 inhibitor was highly correlated with the reduction in hepatic steatosis and fibrosis. These findings support further investigations of the use of this ACC1 inhibitor as a new treatment of NFLD/NASH. SIGNIFICANCE STATEMENT: This is the first study to demonstrate that a novel selective inhibitor of acetyl-CoA carboxylase (ACC) 1 has anti-nonalcoholic fatty liver disease (NAFLD) and anti-nonalcoholic steatohepatitis (NASH) effects in preclinical models. Treatment with this compound significantly improved hepatic steatosis and fibrosis in a mouse model. These findings support the use of this ACC1 inhibitor as a new treatment for NAFLD/NASH.
Collapse
Affiliation(s)
- Yumiko Okano Tamura
- Cardiovascular and Metabolic Drug Discovery Unit (Y.O.T., J.S., M.W.), Drug Metabolism & Pharmacokinetics Research Laboratories (S.I., K.A.), Biomolecular Research Laboratories (M.S.), and Drug Safety Research Laboratories (H.Yasu.), Takeda Pharmaceutical Company Limited, Kanagawa, Japan; and Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (D.M.E., H.Yash.)
| | - Jun Sugama
- Cardiovascular and Metabolic Drug Discovery Unit (Y.O.T., J.S., M.W.), Drug Metabolism & Pharmacokinetics Research Laboratories (S.I., K.A.), Biomolecular Research Laboratories (M.S.), and Drug Safety Research Laboratories (H.Yasu.), Takeda Pharmaceutical Company Limited, Kanagawa, Japan; and Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (D.M.E., H.Yash.)
| | - Shinji Iwasaki
- Cardiovascular and Metabolic Drug Discovery Unit (Y.O.T., J.S., M.W.), Drug Metabolism & Pharmacokinetics Research Laboratories (S.I., K.A.), Biomolecular Research Laboratories (M.S.), and Drug Safety Research Laboratories (H.Yasu.), Takeda Pharmaceutical Company Limited, Kanagawa, Japan; and Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (D.M.E., H.Yash.)
| | - Masako Sasaki
- Cardiovascular and Metabolic Drug Discovery Unit (Y.O.T., J.S., M.W.), Drug Metabolism & Pharmacokinetics Research Laboratories (S.I., K.A.), Biomolecular Research Laboratories (M.S.), and Drug Safety Research Laboratories (H.Yasu.), Takeda Pharmaceutical Company Limited, Kanagawa, Japan; and Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (D.M.E., H.Yash.)
| | - Hironobu Yasuno
- Cardiovascular and Metabolic Drug Discovery Unit (Y.O.T., J.S., M.W.), Drug Metabolism & Pharmacokinetics Research Laboratories (S.I., K.A.), Biomolecular Research Laboratories (M.S.), and Drug Safety Research Laboratories (H.Yasu.), Takeda Pharmaceutical Company Limited, Kanagawa, Japan; and Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (D.M.E., H.Yash.)
| | - Kazunobu Aoyama
- Cardiovascular and Metabolic Drug Discovery Unit (Y.O.T., J.S., M.W.), Drug Metabolism & Pharmacokinetics Research Laboratories (S.I., K.A.), Biomolecular Research Laboratories (M.S.), and Drug Safety Research Laboratories (H.Yasu.), Takeda Pharmaceutical Company Limited, Kanagawa, Japan; and Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (D.M.E., H.Yash.)
| | - Masanori Watanabe
- Cardiovascular and Metabolic Drug Discovery Unit (Y.O.T., J.S., M.W.), Drug Metabolism & Pharmacokinetics Research Laboratories (S.I., K.A.), Biomolecular Research Laboratories (M.S.), and Drug Safety Research Laboratories (H.Yasu.), Takeda Pharmaceutical Company Limited, Kanagawa, Japan; and Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (D.M.E., H.Yash.)
| | - Derek M Erion
- Cardiovascular and Metabolic Drug Discovery Unit (Y.O.T., J.S., M.W.), Drug Metabolism & Pharmacokinetics Research Laboratories (S.I., K.A.), Biomolecular Research Laboratories (M.S.), and Drug Safety Research Laboratories (H.Yasu.), Takeda Pharmaceutical Company Limited, Kanagawa, Japan; and Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (D.M.E., H.Yash.)
| | - Hiroaki Yashiro
- Cardiovascular and Metabolic Drug Discovery Unit (Y.O.T., J.S., M.W.), Drug Metabolism & Pharmacokinetics Research Laboratories (S.I., K.A.), Biomolecular Research Laboratories (M.S.), and Drug Safety Research Laboratories (H.Yasu.), Takeda Pharmaceutical Company Limited, Kanagawa, Japan; and Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (D.M.E., H.Yash.)
| |
Collapse
|
12
|
Inoue I, Qin XY, Masaki T, Mezaki Y, Matsuura T, Kojima S, Furutani Y. Latency-associated Peptide Degradation Fragments Produced in Stellate Cells and Phagocytosed by Macrophages in Bile Duct-ligated Mouse Liver. J Histochem Cytochem 2021; 69:723-730. [PMID: 34674567 DOI: 10.1369/00221554211053665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transforming growth factor-β (TGF-β) activation is involved in various pathogeneses, such as fibrosis and malignancy. We previously showed that TGF-β was activated by serine protease plasma kallikrein-dependent digestion of latency-associated peptides (LAPs) and developed a method to detect LAP degradation products (LAP-DPs) in the liver and blood using specific monoclonal antibodies. Clinical studies have revealed that blood LAP-DPs are formed in the early stages of liver fibrosis. This study aimed to identify the cell source of LAP-DP formation during liver fibrosis. The N-terminals of LAP-DPs ending at residue Arg58 (R58) were stained in liver sections of a bile duct-ligated liver fibrosis model at 3 and 13 days. R58 LAP-DPs were detected in quiescent hepatic stellate cells at day 3 and in macrophages on day 13 after ligation of the bile duct. We then performed a detailed analysis of the axial localization of R58 signals in a single macrophage, visualized the cell membrane with the anti-CLEC4F antibody, and found R58 LAP-DPs surrounded by the membrane in phagocytosed debris that appeared to be dead cells. These findings suggest that in the early stages of liver fibrosis, TGF-β is activated on the membrane of stellate cells, and then the cells are phagocytosed after cell death.
Collapse
Affiliation(s)
- Ikuyo Inoue
- RIKEN Cluster for Pioneering Research Liver Cancer Prevention Research Unit, Saitama, Japan
| | - Xian-Yang Qin
- RIKEN Cluster for Pioneering Research Liver Cancer Prevention Research Unit, Saitama, Japan
| | - Takahiro Masaki
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshihiro Mezaki
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomokazu Matsuura
- RIKEN Cluster for Pioneering Research Liver Cancer Prevention Research Unit, Saitama, Japan.,Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Soichi Kojima
- RIKEN Cluster for Pioneering Research Liver Cancer Prevention Research Unit, Saitama, Japan
| | - Yutaka Furutani
- RIKEN Cluster for Pioneering Research Liver Cancer Prevention Research Unit, Saitama, Japan
| |
Collapse
|
13
|
Zhang C, Yan Y, Gao X, Ma Y. [Therapeutic mechanism of the Mongolian medicine Qiwei Qinggan Powder against liver fibrosis based on UHPLC-TOF-MS combined with network pharmacological methods]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1131-1141. [PMID: 34549702 DOI: 10.12122/j.issn.1673-4254.2021.08.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To elucidate the chemical composition of the Mongolian medicine Qiwei Qinggan Powder and explore its key targets, related pathways and its therapeutic mechanism for liver fibrosis. METHODS UHPLC-TOF-MS was used to analyze the composition of Qiwei Qinggan Powder. The therapeutic targets of Qiwei Qinggan Powder were screened in Swiss Target Prediction database, and liver fibrosis-related targets were screened in TTD and GeneCards databases to identify the anti-fibrosis targets of Qiwei Qinggan Powder by intersection using Venny.2.1.0. The protein interaction was analyzed using STRING database, the GO functions and KEGG pathways were analyzed on the Metascape platform, and the core targets and active components were verified by molecular docking using AutoDock software. The therapeutic mechanism of Qiwei Qinggan Powder against liver fibrosis was verified in rat models and cell experiment. RESULTS We identified a total of 45 chemical constituents in Qiwei Qinggan Powder, including flavonoids, alkaloids, coumarins, terpenes, phenols and fatty acids. Network pharmacological analysis identified 62 targets of Qiwei Qinggan Powder, including 10 core targets. GO enrichment analysis suggested that the therapeutic effect of Qiwei Qinggan Powder was mediated by biological processes (BP), cell components (CC) and molecular functions (MF). KEGG enrichment results showed that PI3K/Akt, Rap1, MAPK, AMPK and PPAR were all pathways associated with liver fibrosis. Molecular docking showed that quercetin, luteolin and kaempferol could bind to Akt1, PIK3R1 and MAPK1, respectively. In rat models of liver fibrosis, treatment with Qiwei Qinggan Powder significantly suppressed proliferation of fibrous tissues and inflammatory cell infiltration to improve fibrosis in the liver tissue. Western blotting demonstrated that Qiwei Qinggan Powder significantly decreased the expressions of the Liver fibrosis markers including α-SMA, Collagen1, PI3K and Akt (P < 0.01). In vitro cell experiment, Qiwei Qinggan Powder-containing serum obviously promoted apoptosis of HSC-T6 cells. CONCLUSION The therapeutic effect of Qiwei Qinggan Powder against liver fibrosis is mediated by multiple components, targets and channels, and its mechanism may involve the regulation of PI3K, Akt and other key targets and modulation of cell apoptosis and energy metabolism.
Collapse
Affiliation(s)
- C Zhang
- Department of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010059, China
| | - Y Yan
- Department of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010059, China
| | - X Gao
- Department of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010059, China
| | - Y Ma
- Department of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010059, China
| |
Collapse
|
14
|
Valenzuela R, Ortiz M, Hernández-Rodas MC, Echeverría F, Videla LA. Targeting n-3 Polyunsaturated Fatty Acids in Non-Alcoholic Fatty Liver Disease. Curr Med Chem 2020; 27:5250-5272. [PMID: 30968772 DOI: 10.2174/0929867326666190410121716] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/14/2018] [Accepted: 01/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Non-Alcoholic Fatty Liver Disease (NAFLD) is characterized by abnormal hepatic accumulation of triacylglycerides in the absence of alcohol consumption, in association with Oxidative Stress (OS), a pro-inflammatory state and Insulin Resistance (IR), which are attenuated by n-3 long-chain polyunsaturated Fatty Acids (FAs) C20-C22 (LCPUFAs) supplementation. Main causes of NAFLD comprise high caloric intake and a sedentary lifestyle, with high intakes of saturated FAs. METHODS The review includes several searches considering the effects of n-3 LCPUFAs in NAFLD in vivo and in vitro models, using the PubMed database from the National Library of Medicine- National Institutes of Health. RESULT The LCPUFAs eicosapentaenoic acid (C20:5 n-3, EPA) and docosahexaenoic acid (C22:6 n- 3, DHA) have a positive effect in diminishing liver steatosis, OS, and the levels of aspartate aminotransferase, alanine aminotransferase and pro-inflammatory cytokines, with improvement of insulin sensitivity and adiponectin levels. The molecular pathways described for n-3 LCPUFAs in cellular and animal models and humans include peroxisome proliferator-activated receptor-α activation favouring FA oxidation, diminution of lipogenesis due to sterol responsive element binding protein-1c downregulation and inflammation resolution. Besides, nuclear factor erythroid-2-related factor-2 activation is elicited by n-3 LCPUFA-derived oxidation products producing direct and indirect antioxidant responses, with concomitant anti-fibrogenic action. CONCLUSION The discussed effects of n-3 LCPUFA supplementation support its use in NAFLD, although having a limited value in NASH, a contention that may involve n-3 LCPUFA oxygenated derivatives. Clinical trials establishing optimal dosages, intervention times, type of patients and possible synergies with other natural products are needed in future studies.
Collapse
Affiliation(s)
- Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Av. Independencia 1027, Independencia, Santiago 8380453, Chile
| | - Macarena Ortiz
- Nutrition and Dietetics School, Faculty of Health Sciences, Catholic University of Maule, Merced 333, Curicó 3340000, Chile
| | - María Catalina Hernández-Rodas
- Department of Nutrition, Faculty of Medicine, University of Chile, Av. Independencia 1027, Independencia, Santiago 8380453, Chile
| | - Francisca Echeverría
- Department of Nutrition, Faculty of Medicine, University of Chile, Av. Independencia 1027, Independencia, Santiago 8380453, Chile
| | - Luis Alberto Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Av. Independencia 1027, Independencia, Santiago 8380453, Chile
| |
Collapse
|
15
|
Matsumoto M, Yashiro H, Ogino H, Aoyama K, Nambu T, Nakamura S, Nishida M, Wang X, Erion DM, Kaneko M. Acetyl-CoA carboxylase 1 and 2 inhibition ameliorates steatosis and hepatic fibrosis in a MC4R knockout murine model of nonalcoholic steatohepatitis. PLoS One 2020; 15:e0228212. [PMID: 31990961 PMCID: PMC6986730 DOI: 10.1371/journal.pone.0228212] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/10/2020] [Indexed: 12/16/2022] Open
Abstract
Acetyl-CoA carboxylase (ACC) catalyzes the rate-limiting step in de novo lipogenesis, which is increased in the livers of patients with nonalcoholic steatohepatitis. GS-0976 (firsocostat), an inhibitor of isoforms ACC1 and ACC2, reduced hepatic steatosis and serum fibrosis biomarkers such as tissue inhibitor of metalloproteinase 1 in patients with nonalcoholic steatohepatitis in a randomized controlled trial, although the impact of this improvement on fibrosis has not fully been evaluated in preclinical models. Here, we used Western diet-fed melanocortin 4 receptor-deficient mice that have similar phenotypes to nonalcoholic steatohepatitis patients including progressively developed hepatic steatosis as well as fibrosis. We evaluated the effects of ACC1/2 inhibition on hepatic fibrosis. After the confirmation of significant hepatic fibrosis with a 13-week pre-feeding, GS-0976 (4 and 16 mg/kg/day) treatment for 9 weeks lowered malonyl-CoA and triglyceride content in the liver and improved steatosis, histologically. Furthermore, GS-0976 reduced the histological area of hepatic fibrosis, hydroxyproline content, mRNA expression level of type I collagen in the liver, and plasma tissue metalloproteinase inhibitor 1, suggesting an improvement of hepatic fibrosis. The treatment with GS-0976 was also accompanied by reductions of plasma ALT and AST levels. These data demonstrate that improvement of hepatic lipid metabolism by ACC1/2 inhibition could be a new option to suppress fibrosis progression as well as to improve hepatic steatosis in nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Mitsuharu Matsumoto
- Department of Integrated Biology, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
- * E-mail:
| | - Hiroaki Yashiro
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States of America
| | - Hitomi Ogino
- Department of Integrated Biology, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Kazunobu Aoyama
- Department of Drug Disposition & Analysis, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Tadahiro Nambu
- Department of Nonclinical Safety Research, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Sayuri Nakamura
- Department of Nonclinical Safety Research, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Mayumi Nishida
- Department of Integrated Biology, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Xiaolun Wang
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States of America
| | - Derek M. Erion
- Gastroenterology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States of America
| | - Manami Kaneko
- Department of Integrated Biology, Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| |
Collapse
|
16
|
Asakawa M, Itoh M, Suganami T, Sakai T, Kanai S, Shirakawa I, Yuan X, Hatayama T, Shimada S, Akiyama Y, Fujiu K, Inagaki Y, Manabe I, Yamaoka S, Yamada T, Tanaka S, Ogawa Y. Upregulation of cancer-associated gene expression in activated fibroblasts in a mouse model of non-alcoholic steatohepatitis. Sci Rep 2019; 9:19601. [PMID: 31862949 PMCID: PMC6925281 DOI: 10.1038/s41598-019-56039-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/03/2019] [Indexed: 12/24/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH), characterized by chronic inflammation and fibrosis, is predicted to be the leading cause of cirrhosis and hepatocellular carcinoma (HCC) in the next decade. Although recent evidence suggests the importance of fibrosis as the strongest determinant of HCC development, the molecular mechanisms underlying NASH-induced carcinogenesis still remain unclear. Here we performed RNA sequencing analysis to compare gene expression profiles of activated fibroblasts prepared from two distinct liver fibrosis models: carbon tetrachloride-induced fibrosis as a model without obesity and HCC and genetically obese melanocortin 4 receptor-deficient (MC4R-KO) mice fed Western diet, which develop steatosis, NASH, and eventually HCC. Our data showed that activated fibroblasts exhibited distinct gene expression patterns in each etiology, and that the 'pathways in cancer' were selectively upregulated in the activated fibroblasts from MC4R-KO mice. The most upregulated gene in these pathways was fibroblast growth factor 9 (FGF9), which was induced by metabolic stress such as palmitate. FGF9 exerted anti-apoptotic and pro-migratory effects in fibroblasts and hepatoma cells in vitro and accelerated tumor growth in a subcutaneous xenograft model. This study reveals upregulation of cancer-associated gene expression in activated fibroblasts in NASH, which would contribute to the progression from NASH to HCC.
Collapse
Affiliation(s)
- Masahiro Asakawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michiko Itoh
- Department of Organ Network and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
- Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan.
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
- Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Takeru Sakai
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sayaka Kanai
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ibuki Shirakawa
- Department of Organ Network and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Xunmei Yuan
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomomi Hatayama
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shu Shimada
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshimitsu Akiyama
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Katsuhito Fujiu
- Department of Advanced Cardiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Isehara, Japan
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shoji Yamaoka
- Department of Molecular Virology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tetsuya Yamada
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
- Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
- Japan Agency for Medical Research and Development, CREST, Tokyo, Japan.
| |
Collapse
|
17
|
Fernández-Martínez E, Lira-Islas IG, Cariño-Cortés R, Soria-Jasso LE, Pérez-Hernández E, Pérez-Hernández N. Dietary chia seeds (Salvia hispanica) improve acute dyslipidemia and steatohepatitis in rats. J Food Biochem 2019; 43:e12986. [PMID: 31489674 DOI: 10.1111/jfbc.12986] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/27/2019] [Accepted: 06/30/2019] [Indexed: 02/06/2023]
Abstract
Chia seeds (Salvia hispanica L.) are rich in omega fatty acids. Dyslipidemia and steatohepatitis are diseases that require effective treatments in obese and non-obese patients. The aim was to evaluate the effect of chia intake on acute tyloxapol (TI)-induced dyslipidemia, on acute carbon tetrachloride (TC)-induced steatohepatitis, and on mixed damage (TC+TI) in non-obese rats. Four experimental groups were fed for 4 weeks a diet with established rodent food (DE), and four groups were fed a diet with 15% added chia (DC). Plasma samples were analyzed for total cholesterol, triglycerides, glucose, biochemical liver damage markers, and tumor necrosis factor-α (TNF-α). Liver samples were used to quantify glycogen, catalase, lipid peroxidation, and TNF-α. A histopathological analysis was performed. DC intake partially or totally prevented steatohepatitis, and reduced lipids in the dyslipidemic groups. The hypolipidemic and hepatoprotective effects of chia may be correlated to its high content of α-linolenic acid (omega-3) and phenolics. PRACTICAL APPLICATIONS: Metabolic syndrome is associated with non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), which are currently the most common causes of chronic liver disease, cirrhosis, and hepatocellular carcinoma (HCC) worldwide. Dyslipidemia is a significant risk factor for NAFLD and NASH. Non-obese patients may have NAFLD or NASH. Metabolic syndrome and dyslipidemia are more strongly associated with NAFLD in non-obese than in obese patients. This is the first study evaluating the hypolipidemic and hepatoprotective effects of chia seed intake on acute dyslipidemia and/or steatohepatitis caused by the individual or combined administration of the inducers tyloxapol and carbon tetrachloride, respectively, in non-obese rats. The pharmacological effects of dietary chia are correlated to its high content of omega-3 and omega-6 (1:1), protein, dietary fiber, and phenolics. The results suggest that inclusion of chia in diets of non-obese patients with dyslipidemia and/or NAFLD/NASH may improve their health state and preventing cirrhosis or HCC.
Collapse
Affiliation(s)
- Eduardo Fernández-Martínez
- Laboratory of Medicinal Chemistry and Pharmacology, Centro de Investigación en Biología de la Reproducción, Área Académica de Medicina, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, México
| | - Ivet G Lira-Islas
- Laboratory of Medicinal Chemistry and Pharmacology, Centro de Investigación en Biología de la Reproducción, Área Académica de Medicina, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, México
| | - Raquel Cariño-Cortés
- Laboratory of Medicinal Chemistry and Pharmacology, Centro de Investigación en Biología de la Reproducción, Área Académica de Medicina, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, México
| | - Luis E Soria-Jasso
- Laboratory of Medicinal Chemistry and Pharmacology, Centro de Investigación en Biología de la Reproducción, Área Académica de Medicina, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca, México
| | | | - Nury Pérez-Hernández
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
18
|
Protective effect of cultured bear bile powder against dimethylnitrosamine-induced hepatic fibrosis in rats. Biomed Pharmacother 2019; 112:108701. [DOI: 10.1016/j.biopha.2019.108701] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023] Open
|
19
|
Current Models of Fatty Liver Disease; New Insights, Therapeutic Targets and Interventions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1134:33-58. [PMID: 30919331 DOI: 10.1007/978-3-030-12668-1_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of disorders ranging from simple steatosis to steatosis with inflammation and fibrosis. NAFLD is currently the most prevalent chronic liver disease worldwide, with a global prevalence of 25%, and is soon projected to be the leading cause for liver transplantation in the US. Alarmingly, few effective pharmacotherapeutic approaches are currently available to block or attenuate development and progression of NAFLD. Preclinical models are critical for unraveling the complex and multi-factorial etiology of NAFLD and for testing potential therapeutics. Here we review preclinical models that have been instrumental in highlighting molecular and cellular mechanisms underlying the pathogenesis of NAFLD and in facilitating early proof-of-concept investigations into novel intervention strategies.
Collapse
|
20
|
Inoue-Yamauchi A, Itagaki H, Oda H. Eicosapentaenoic acid attenuates obesity-related hepatocellular carcinogenesis. Carcinogenesis 2018; 39:28-35. [PMID: 29040439 PMCID: PMC5862334 DOI: 10.1093/carcin/bgx112] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 10/10/2017] [Indexed: 12/17/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), the hepatic manifestation of obesity, is an emerging risk factor for hepatocellular carcinoma (HCC). Accumulating evidence has shown that chronic inflammation represents a plausible link between obesity and HCC and that the pro-inflammatory cytokine interleukin (IL)-6 contributes to the development of obesity-related HCC. In the present study, we aimed to examine the therapeutic potential of the omega-3 polyunsaturated fatty acid, eicosapentaenoic acid (EPA), which exerts anti-inflammatory effects. The results showed that the development of carcinogen-induced HCC was significantly less in mice fed a high-fat diet (HFD) supplemented with EPA than in those fed HFD only, suggesting that EPA attenuates the development of obesity-related HCC. Although EPA did not appear to affect obesity-linked inflammation, it suppressed the activation of the pro-tumorigenic IL-6 effector STAT3, contributing to the inhibition of tumor growth. These findings suggest a clinical implication of EPA as a treatment for obesity-related HCC.
Collapse
Affiliation(s)
| | - Hiroko Itagaki
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan.,Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hideaki Oda
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
21
|
Li M, Qin XY, Furutani Y, Inoue I, Sekihara S, Kagechika H, Kojima S. Prevention of acute liver injury by suppressing plasma kallikrein-dependent activation of latent TGF-β. Biochem Biophys Res Commun 2018; 504:857-864. [PMID: 30219233 DOI: 10.1016/j.bbrc.2018.09.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/06/2018] [Indexed: 01/30/2023]
Abstract
Acute liver injury (ALI) is highly lethal acute liver failure caused by different etiologies. Transforming growth factor β (TGF-β) is a multifunctional cytokine and a well-recognized inducer of apoptotic and necrotic cell death in hepatocytes. Latent TGF-β is activated partly through proteolytic cleavage by a serine protease plasma kallikrein (PLK) between the R58 and L59 residues of its propeptide region. Recently, we developed a specific monoclonal antibody to detect the N-terminal side LAP degradation products ending at residue R58 (R58 LAP-DPs) that reflect PLK-dependent TGF-β activation. This study aimed to explore the potential roles of PLK-dependent TGF-β activation in the pathogenesis of ALI. We established a mouse ALI model via the injection of anti-Fas antibodies (Jo2) and observed increases in the TGF-β1 mRNA level, Smad3 phosphorylation, TUNEL-positive apoptotic hepatocytes and R58-positive cells in the liver tissues of Jo2-treated mice. The R58 LAP-DPs were observed in/around F4/80-positive macrophages, while macrophage depletion with clodronate liposomes partly alleviated the Jo2-induced liver injury. Blocking PLK-dependent TGF-β activation using either the serine proteinase inhibitor FOY305 or the selective PLK inhibitor PKSI-527 or blocking the TGF-β receptor-mediated signaling pathway using SB431542 significantly prevented Jo2-induced hepatic apoptosis and mortality. Furthermore, similar phenomena were observed in the mouse model of ALI with the administration of acetaminophen (APAP). In summary, R58 LAP-DPs reflecting PLK-dependent TGF-β activation may serve as a biomarker for ALI, and targeting PLK-dependent TGF-β activation has potential as a therapeutic strategy for ALI.
Collapse
Affiliation(s)
- Mengqian Li
- Liver Cancer Prevention Research Unit, RIKEN Center for Integrative Medical Sciences, Saitama, Japan; Graduate School of Medical & Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Xian-Yang Qin
- Liver Cancer Prevention Research Unit, RIKEN Center for Integrative Medical Sciences, Saitama, Japan
| | - Yutaka Furutani
- Liver Cancer Prevention Research Unit, RIKEN Center for Integrative Medical Sciences, Saitama, Japan
| | - Ikuyo Inoue
- Liver Cancer Prevention Research Unit, RIKEN Center for Integrative Medical Sciences, Saitama, Japan
| | - Sanae Sekihara
- Liver Cancer Prevention Research Unit, RIKEN Center for Integrative Medical Sciences, Saitama, Japan
| | - Hiroyuki Kagechika
- Graduate School of Medical & Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Soichi Kojima
- Liver Cancer Prevention Research Unit, RIKEN Center for Integrative Medical Sciences, Saitama, Japan; Graduate School of Medical & Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
22
|
Goto T, Itoh M, Suganami T, Kanai S, Shirakawa I, Sakai T, Asakawa M, Yoneyama T, Kai T, Ogawa Y. Obeticholic acid protects against hepatocyte death and liver fibrosis in a murine model of nonalcoholic steatohepatitis. Sci Rep 2018; 8:8157. [PMID: 29802399 PMCID: PMC5970222 DOI: 10.1038/s41598-018-26383-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 04/17/2018] [Indexed: 02/08/2023] Open
Abstract
Accumulating evidence has suggested that farnesoid X receptor (FXR) agonists, such as obeticholic acid (OCA) are therapeutically useful for non-alcoholic steatohepatitis (NASH). However, it is still unclear how FXR agonists protect against NASH and which cell type is the main target of FXR agonists. In this study, we examined the effects of OCA on the development of NASH using melanocortin 4 receptor-deficient (MC4R-KO) mice that progressively developed hepatic steatosis and NASH on Western diet (WD). Treatment with OCA effectively prevented chronic inflammation and liver fibrosis in WD-fed MC4R-KO mice with only marginal effect on body weight and hepatic steatosis. Hepatic crown-like structure (hCLS) is a unique histological structure characteristic of NASH, which triggers hepatocyte death-induced interstitial fibrosis. Intriguingly, treatment with OCA markedly reduced hCLS formation even after MC4R-KO mice developed NASH, thereby inhibiting the progression of liver fibrosis. As its mechanism of action, OCA suppressed metabolic stress-induced p53 activation and cell death in hepatocytes. Our findings in this study highlight the role of FXR in hepatocytes in the pathogenesis of NASH. Collectively, this study demonstrates the anti-fibrotic effect of OCA in a murine model of NASH with obesity and insulin resistance, which suggests the clinical implication for human NASH.
Collapse
Affiliation(s)
- Toshihiro Goto
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Specialty Medicine Group, Drug Development Research Laboratories, Sumitomo Dainippon Pharma. Co., Ltd, Osaka, Japan
| | - Michiko Itoh
- Department of Organ Network and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
| | - Sayaka Kanai
- Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ibuki Shirakawa
- Department of Organ Network and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeru Sakai
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masahiro Asakawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiro Yoneyama
- Omics Group, Genomic Science Laboratories, Sumitomo Dainippon Pharma. Co., Ltd, Osaka, Japan
| | - Toshihiro Kai
- Omics Group, Genomic Science Laboratories, Sumitomo Dainippon Pharma. Co., Ltd, Osaka, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. .,Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan. .,Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. .,Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. .,Japan Agency for Medical Research and Development, CREST, Tokyo, Japan.
| |
Collapse
|
23
|
Takakura K, Oikawa T, Tomita Y, Mizuno Y, Nakano M, Saeki C, Torisu Y, Saruta M. Mouse models for investigating the underlying mechanisms of nonalcoholic steatohepatitis-derived hepatocellular carcinoma. World J Gastroenterol 2018; 24:1989-1994. [PMID: 29760542 PMCID: PMC5949712 DOI: 10.3748/wjg.v24.i18.1989] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/01/2018] [Accepted: 05/05/2018] [Indexed: 02/06/2023] Open
Abstract
As the incidence of hepatocellular carcinoma (HCC) caused by infection with the hepatotropic viruses hepatitis B and hepatitis C decreases, greater attention has become focused on HCC caused by nonalcoholic steatohepatitis (NASH), an advanced form of nonalcoholic fatty liver disease which has shown increasing prevalence in correspondence with the overall increase in metabolic syndrome over the recent decades. Several clinical population studies have shown a positive relationship between NASH and HCC, while also providing initial insights into the underlying mechanisms of HCC development from NASH. Research into the pathological progression of NASH to HCC has advanced by use of several beneficial rodent models. In this review, we summarize the established mouse models for preclinical research of NASH-associated HCC and discuss the underlying hepatic mechanisms of NASH-related tumorigenesis identified to date that could lead to new targets for treatment and prevention.
Collapse
Affiliation(s)
- Kazuki Takakura
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Tsunekazu Oikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Yoichi Tomita
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Yusuke Mizuno
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Masanori Nakano
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Chisato Saeki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Yuichi Torisu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| |
Collapse
|
24
|
Tanaka M, Itoh M, Ogawa Y, Suganami T. Molecular mechanism of obesity-induced 'metabolic' tissue remodeling. J Diabetes Investig 2018; 9:256-261. [PMID: 29086488 PMCID: PMC5835451 DOI: 10.1111/jdi.12769] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 10/25/2017] [Indexed: 12/26/2022] Open
Abstract
Chronic inflammation is a common molecular basis underlying a variety of chronic diseases. Accumulating evidence has also suggested that chronic inflammation contributes to the pathogenesis of obesity and diabetes, which have been considered as metabolic diseases. For the past several decades, there has been dramatic progress in understanding the underlying mechanism of adipose tissue dysfunction induced by obesity. Tissue remodeling is one of the histological features of chronic inflammation, in which stromal cells dramatically change in number and cell type. Indeed, adipose tissue remodeling is induced by various stromal cells, and results in the impairment of adipose tissue function, such as adipocytokine production and lipid storage, which leads to systemic metabolic disorder. In addition to adipose tissue, the liver is another example of obesity-induced tissue remodeling. In the present review, we discuss how obesity induces interstitial fibrosis in adipose tissue and the liver, particularly focusing on the role of macrophages.
Collapse
Affiliation(s)
- Miyako Tanaka
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
| | - Michiko Itoh
- Department of Organ Network and MetabolismGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Yoshihiro Ogawa
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
- Department of Molecular Endocrinology and MetabolismGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Department of Molecular and Cellular MetabolismGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Department of Medicine and Bioregulatory ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Japan Agency for Medical Research and DevelopmentCRESTTokyoJapan
| | - Takayoshi Suganami
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
| |
Collapse
|
25
|
Wu J. Utilization of animal models to investigate nonalcoholic steatohepatitis-associated hepatocellular carcinoma. Oncotarget 2018; 7:42762-42776. [PMID: 27072576 PMCID: PMC5173170 DOI: 10.18632/oncotarget.8641] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) comprises a spectrum of liver disorders with fat accumulation from simple fatty liver, nonalcoholic steatohepatitis (NASH), fibrosis/cirrhosis and NAFLD/NASH-associated hepatocellular carcinoma (HCC). NASH is a progressive form of NAFLD and requires medical attention. One of 5-10 NASH patients may progress to end-state liver disease (ESLD or cirrhosis) in 5-10 years; meanwhile, life-threatening complications of ESLD and HCC account for major mortality. An increasing burden of NAFLD in clinics, elucidation of its pathogenesis and progression, and assessment of the efficacy of potential therapeutics demand reliable animal models. Most NASH-associated HCC occurs in cirrhotic subjects; however, HCC does appear in NASH patients without cirrhosis. Lipotoxicity, oxidant stress, insulin resistance, endoplasmic reticulum stress, altered adipokine and lymphokine profiles and gut microbiome changes affect NAFLD progression and constitute key pathobiologic interplays. How these factors promote malignant transformation in a microenvironment of steatotic inflammation and fibrosis/cirrhosis, and lead to development of neoplasms is one of critical questions faced in the hepatology field. The present review summarizes the characteristics of emerging rodent NASH-HCC models, and discusses the challenges in utilizing these models to unveil the mysteries of NASH-associated HCC development.
Collapse
Affiliation(s)
- Jian Wu
- Key Laboratory of Molecular Virology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Institute of Liver Diseases, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Itoh M, Suganami T, Kato H, Kanai S, Shirakawa I, Sakai T, Goto T, Asakawa M, Hidaka I, Sakugawa H, Ohnishi K, Komohara Y, Asano K, Sakaida I, Tanaka M, Ogawa Y. CD11c+ resident macrophages drive hepatocyte death-triggered liver fibrosis in a murine model of nonalcoholic steatohepatitis. JCI Insight 2017; 2:92902. [PMID: 29202448 DOI: 10.1172/jci.insight.92902] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 10/17/2017] [Indexed: 12/14/2022] Open
Abstract
Although recent evidence has pointed to the role of organ- and pathogenesis-specific macrophage subsets, it is still unclear which subsets are critically involved in the pathogenesis of nonalcoholic steatohepatitis (NASH). Using melanocortin-4 receptor-deficient (MC4R-KO) mice fed Western diet (WD), which exhibit liver phenotypes similar to those of human NASH, we found a histological structure, termed hepatic crown-like structure (hCLS), in which CD11c+ macrophages surround dead/dying hepatocytes, a prominent feature of NASH. Here, we demonstrate that hCLS-constituting macrophages could be a novel macrophage subset that drives hepatocyte death-triggered liver fibrosis. In an "inducible NASH model," hepatocyte death induces hCLS formation and liver fibrosis sequentially in the short term. In combination with the long-term WD feeding model, we also showed that resident macrophages are a major cellular source of CD11c+ macrophages constituting hCLS, which exhibited gene expression profiles distinct from CD11c- macrophages scattered in the liver. Moreover, depletion of CD11c+ macrophages abolished hCLS formation and fibrogenesis in NASH. Our clinical data suggest the role of CD11c+ macrophages in the disease progression from simple steatosis to NASH. This study sheds light on the role of resident macrophages, in addition to recruited macrophages, in the pathogenesis of NASH.
Collapse
Affiliation(s)
| | - Takayoshi Suganami
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Hideaki Kato
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Drug Discovery & Disease Research Laboratory, Shionogi & Co. Ltd., Osaka, Japan
| | - Sayaka Kanai
- Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Takeru Sakai
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiro Goto
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masahiro Asakawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Isao Hidaka
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | | | - Koji Ohnishi
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Asano
- Laboratory of Immune regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Isao Sakaida
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Masato Tanaka
- Laboratory of Immune regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Japan Agency for Medical Research and Development, CREST, Tokyo, Japan
| |
Collapse
|
27
|
Fujino S, Miyoshi N, Ohue M, Takahashi Y, Yasui M, Sugimura K, Akita H, Takahashi H, Kobayashi S, Yano M, Sakon M. Prediction model and treatment of high-output ileostomy in colorectal cancer surgery. Mol Clin Oncol 2017; 7:468-472. [PMID: 28894582 DOI: 10.3892/mco.2017.1336] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/24/2017] [Indexed: 12/19/2022] Open
Abstract
The aim of the present study was to examine the risk factors of high-output ileostomy (HOI), which is associated with electrolyte abnormalities and/or stoma complications, and to create a prediction model. The medical records of 68 patients who underwent colorectal cancer surgery with ileostomy between 2011 and 2016 were retrospectively investigated. All the patients underwent surgical resection for colorectal cancer at the Osaka Medical Center for Cancer and Cardiovascular Diseases (Osaka, Japan). A total of 7 patients with inadequate data on ileostomy output were excluded. Using a group of 50 patients who underwent surgery between 2011 and 2013, the risk of HOI was classified by a decision tree model using a partition platform. The HOI prediction model was validated in an additional group of 11 patients who underwent surgery between 2014 and 2016. Univariate analysis of clinical factors demonstrated that young age (P=0.003) and high white blood cell (WBC) count (P<0.001) after surgery were significantly correlated with HOI. Operative factors, such as surgical procedure, approach, operative time and blood loss, were not significantly correlated with HOI. Using these clinical factors, the risk of HOI was classified by statistical partition. In this model, three factors (gender, age and WBC on postoperative day 1) were generated for the prediction of HOI. The patients were classified into five groups, and HOI was observed in 0-88% of patients in each group. The area under the curve (AUC) was 0.838. The model was validated by an external dataset in an independent patient group, for which the AUC was 0.792. In conclusion, HOI patients were classified and an HOI prediction model was developed that may help clinicians in postoperative care.
Collapse
Affiliation(s)
- Shiki Fujino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Norikatsu Miyoshi
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Masayuki Ohue
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Yuske Takahashi
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Masayoshi Yasui
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Keijiro Sugimura
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Hirohumi Akita
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Hidenori Takahashi
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Shogo Kobayashi
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Masahiko Yano
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Masato Sakon
- Department of Surgery, Osaka International Cancer Institute, Osaka 541-8567, Japan
| |
Collapse
|
28
|
Jump DB, Lytle KA, Depner CM, Tripathy S. Omega-3 polyunsaturated fatty acids as a treatment strategy for nonalcoholic fatty liver disease. Pharmacol Ther 2017; 181:108-125. [PMID: 28723414 DOI: 10.1016/j.pharmthera.2017.07.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obese and type 2 diabetic (T2DM) patients have a high prevalence of nonalcoholic fatty liver disease (NAFLD). NAFLD is a continuum of chronic liver diseases ranging from benign hepatosteatosis to nonalcoholic steatohepatitis (NASH), cirrhosis and primary hepatocellular cancer (HCC). Because of its strong association with the obesity epidemic, NAFLD is rapidly becoming a major public health concern worldwide. Surprisingly, there are no FDA approved NAFLD therapies; and current therapies focus on the co-morbidities associated with NAFLD, namely, obesity, hyperglycemia, dyslipidemia, and hypertension. The goal of this review is to provide background on the disease process, discuss human studies and preclinical models that have examined treatment options. We also provide an in-depth rationale for the use of dietary ω3 polyunsaturated fatty acid (ω3 PUFA) supplements as a treatment option for NAFLD. This focus is based on recent studies indicating that NASH patients and preclinical mouse models of NASH have low levels of hepatic C20-22 ω3 PUFA. This decline in hepatic PUFA may account for the major phenotypic features associated with NASH, including steatosis, inflammation and fibrosis. Finally, our discussion will address the strengths and limitations of ω3 PUFA supplements use in NAFLD therapy.
Collapse
Affiliation(s)
- Donald B Jump
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, United States.
| | - Kelli A Lytle
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, United States
| | - Christopher M Depner
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, United States
| | - Sasmita Tripathy
- Molecular Nutrition and Diabetes Research Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, United States
| |
Collapse
|
29
|
Nishiyama K, Fujimoto Y, Takeuchi T, Azuma YT. Aggressive Crosstalk Between Fatty Acids and Inflammation in Macrophages and Their Influence on Metabolic Homeostasis. Neurochem Res 2017; 43:19-26. [PMID: 28424949 DOI: 10.1007/s11064-017-2269-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/09/2017] [Accepted: 04/12/2017] [Indexed: 02/08/2023]
Abstract
From the immunological point of view, macrophages are required to maintain metabolic homeostasis. Recently, there has been an increased focus on the influence of macrophage phenotypes in adipose tissue on the maintenance of metabolic homeostasis in healthy conditions because dysregulated metabolic homeostasis causes metabolic syndrome. This review notes several types of inflammatory and anti-inflammatory mediators in metabolic homeostasis. M1 macrophage polarization mediates inflammation, whereas M2 macrophage polarization mediates anti-inflammation. Fatty acids and their related factors mediate both inflammatory and anti-inflammatory responses. Saturated fatty acids and polyunsaturated fatty acids mediate inflammation, whereas marine-derived n-3 fatty acids, such as eicosapentaenoic acid and docosahexaenoic acid, mediate anti-inflammation. In this review, we discuss the current understanding of the crosstalk between fatty acids and inflammation in macrophages and their influence on metabolic homeostasis.
Collapse
Affiliation(s)
- Kazuhiro Nishiyama
- Laboratory of Veterinary Pharmacology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Science, 1-58 Rinku-ohraikita, Izumisano, Osaka, 598-8531, Japan
| | - Yasuyuki Fujimoto
- Laboratory of Veterinary Pharmacology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Science, 1-58 Rinku-ohraikita, Izumisano, Osaka, 598-8531, Japan
| | - Tadayoshi Takeuchi
- Laboratory of Veterinary Pharmacology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Science, 1-58 Rinku-ohraikita, Izumisano, Osaka, 598-8531, Japan
| | - Yasu-Taka Azuma
- Laboratory of Veterinary Pharmacology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Science, 1-58 Rinku-ohraikita, Izumisano, Osaka, 598-8531, Japan.
| |
Collapse
|
30
|
Siddiqui RA, Xu Z, Harvey KA, Pavlina TM, Becker MJ, Zaloga GP. Comparative study of the modulation of fructose/sucrose-induced hepatic steatosis by mixed lipid formulations varying in unsaturated fatty acid content. Nutr Metab (Lond) 2015; 12:41. [PMID: 26583036 PMCID: PMC4650347 DOI: 10.1186/s12986-015-0038-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/08/2015] [Indexed: 12/18/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in developed countries. NAFLD encompasses a spectrum of diseases, ranging from hepatic steatosis to non-alcoholic steatohepatitis (NASH), cirrhosis, and liver failure. The etiology of NAFLD remains unclear but is thought to relate to increased fatty acid flux within the liver that results in toxic fatty acid metabolite production. One source of increased fatty acid flux is fructose/sucrose-induced hepatic lipogenesis. Current treatment for NAFLD encompasses dietary modifications. However, little scientific evidence exists on which to base many dietary recommendations, especially the intake of different types of carbohydrates and fats. We hypothesized that lipid mixtures of unsaturated fatty acids would inhibit lipogenesis and subsequent hepatic steatosis induced by high carbohydrate diets. The aim of this study was to examine the effects of different complex mixtures of fatty acids upon the development of fructose/sucrose-induced hepatic steatosis. Methods C57BL/6 mice were randomized to normocaloric chow-based diets that varied in the type of carbohydrate (starch, sucrose, fructose). Animals in each carbohydrate group were further randomized to diets that varied in lipid type (no additional lipid, soybean oil, fish oil, olive/soybean oil, macadamia nut oil). These oils were chosen based upon their content of omega-6 polyunsaturated fatty acids, omega-3 polyunsaturated fatty acids, omega-9 monounsaturated fatty acids, or omega-7 monounsaturated fatty acids. Fatty acid flux in the liver was determine by assessing hepatic lipid content (steatosis). We also assessed fatty acid levels in the plasma and liver of the animals, hepatic lipogenesis activity, hepatic stearoyl-CoA-1 desaturase activity, and hepatic elongase activity. Results Animals consumed similar amounts of the diets and maintained normal body weights throughout the study. Both sucrose and fructose induced hepatic lipogenesis and steatosis, with fructose being more potent. All mixed lipids similarly inhibited steatosis, limiting lipid content to levels found in the control (starch) animals. Lipogenesis and stearoyl-CoA-1 desaturase activity were increased in the sucrose and fructose groups. Levels of these enzymatic processes remained at baseline in all of the lipid groups. Conclusion This is the first study to compare various complex lipid mixtures, based upon dietary oils with different types of long-chain fatty acids, upon development of sucrose/fructose-induced steatosis. Both carbohydrate source and lipid content appear important for the modulation of steatosis. Moderate intake of complex lipids with high unsaturated to saturated fatty acid ratios inhibited both lipogenesis and steatosis.
Collapse
Affiliation(s)
- Rafat A Siddiqui
- Methodist Research Institute, Indiana University Health, 1800 N. Capitol Ave, Indianapolis, IN 46202 USA
| | - Zhidong Xu
- Methodist Research Institute, Indiana University Health, 1800 N. Capitol Ave, Indianapolis, IN 46202 USA
| | - Kevin A Harvey
- Methodist Research Institute, Indiana University Health, 1800 N. Capitol Ave, Indianapolis, IN 46202 USA
| | | | | | - Gary P Zaloga
- Baxter Healthcare Corporation, Deerfield, IL 60015 USA
| |
Collapse
|
31
|
L(59) TGF-β LAP degradation products serve as a promising blood biomarker for liver fibrogenesis in mice. FIBROGENESIS & TISSUE REPAIR 2015; 8:17. [PMID: 26379781 PMCID: PMC4570586 DOI: 10.1186/s13069-015-0034-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/20/2015] [Indexed: 02/08/2023]
Abstract
Background Hepatic fibrosis, which is the excessive accumulation of extracellular matrices (ECMs) produced mainly from activated hepatic stellate cells (HSCs), develops to cirrhosis over several decades. There are no validated biomarkers that can non-invasively monitor excessive production of ECM (i.e., fibrogenesis). Transforming growth factor (TGF)-β, a key driver of fibrogenesis, is produced as an inactive latent complex, in which active TGF-β is enveloped by its pro-peptide, the latency-associated protein (LAP). Thus, active TGF-β must be released from the complex for binding to its receptor and inducing ECM synthesis. We recently reported that during the pathogenesis of liver fibrosis, plasma kallikrein (PLK) activates TGF-β by cleavage between R58 and L59 residues within LAP and that one of its by-products, the N-terminal side LAP degradation products ending at residue R58 (R58 LAP-DPs), can be detected mainly around activated HSCs by specific antibodies against R58 cleavage edges and functions as a footprint of PLK-dependent TGF-β activation. Here, we describe a sandwich enzyme-linked immunosorbent assay (ELISA) that detects the other by-products, the C-terminal side LAP-DPs starting from residue L59 (L59 LAP-DPs). We demonstrated that the L59 LAP-DPs are a potentially novel blood biomarker reflecting hepatic fibrogenesis. Results We established a specific sandwich ELISA to quantify L59 LAP-DPs as low as 2 pM and measured L59 LAP-DP levels in the culture media of a human activated HSC line, TWNT-4 cells. L59 LAP-DPs could be detected in their media, and after treatment of TWNT-4 cells with a TGF-β receptor kinase inhibitor, SB431542, a simultaneous reduction was observed in both L59 LAP-DP levels in the culture media and the mRNA expression levels of collagen type (I) α1. In carbon tetrachloride- and bile duct ligation-induced liver fibrosis models in mice, plasma L59 LAP-DP levels increased prior to increase of hepatic hydroxyproline (HDP) contents and well correlated with α-smooth muscle actin (αSMA) expression in liver tissues. At this time, αSMA-positive cells as well as R58 LAP-DPs were seen in their liver tissues. Conclusions L59 LAP-DPs reflect PLK-dependent TGF-β activation and the increase in αSMA-positive activated HSCs in liver injury, thereby serving as a novel blood biomarker for liver fibrogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s13069-015-0034-9) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Ogawa Y, Suganami T, Tanaka M, Itoh M. Parenchymal-stromal cell interaction in metabolic diseases. Inflamm Regen 2015. [DOI: 10.2492/inflammregen.35.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Takayoshi Suganami
- Department of Organ Network and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- PREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Miyako Tanaka
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michiko Itoh
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|