1
|
Wang D, Zhang M, Zhang Y, Yin Z, Zhang S, Zhao Z, Duan Y. Hepatoprotective effects of polysaccharide from Morchella esculenta are associated with activation of the AMPK/Sirt1 signaling pathway in mice with NAFLD. Int J Biol Macromol 2025; 301:140444. [PMID: 39884630 DOI: 10.1016/j.ijbiomac.2025.140444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The functional food application of edible fungus polysaccharides has been widely studied based on their variety of potential pharmacological activities. However, the hepatoprotective effects and mechanisms of Morchella esculenta polysaccharide against nonalcoholic fatty liver disease (NAFLD) remain unknown. A high-fat diet (HFD) fed C57BL/6 J mice for 8 weeks was employed to establish NAFLD with simple steatosis, methionine choline deficiency (MCD) diet for 4 weeks induced hepatic steatohepatitis and fibrosis. The M. esculenta polysaccharide (MCP) or saline was administered intragastrically. MCP markedly reduced hepatic and serum triglyceride (TG) and cholesterol contents in HFD-fed mice. Moreover, treatment with MCP ameliorated nonalcoholic steatohepatitis (NASH) progression in MCD-fed mice, as evidenced by ameliorated hepatic steatosis, inflammatory response, and fibrosis. Mechanistically, MCP suppressed the expression of lipogenic genes and inflammatory cytokines and upregulated peroxisome proliferator-activated receptor (PPAR)-α expression to induce fatty acid β-oxidation. These beneficial effects were attributed to activating the AMP-activated kinase (AMPK)/Sirtuin 1 (Sirt1) signaling pathway. Therefore, we provided evidence that MCP might be an effective dietary supplement to ameliorate NAFLD.
Collapse
Affiliation(s)
- Dandan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230011, China; College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230601, China
| | - Menglian Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230011, China
| | - Yaowen Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230011, China
| | - Zequn Yin
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Shuang Zhang
- College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230601, China
| | - Zhiwei Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Yajun Duan
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
2
|
Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther 2025; 10:33. [PMID: 39904973 PMCID: PMC11794951 DOI: 10.1038/s41392-024-02072-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025] Open
Abstract
As a highly complex organ with digestive, endocrine, and immune-regulatory functions, the liver is pivotal in maintaining physiological homeostasis through its roles in metabolism, detoxification, and immune response. Various factors including viruses, alcohol, metabolites, toxins, and other pathogenic agents can compromise liver function, leading to acute or chronic injury that may progress to end-stage liver diseases. While sharing common features, liver diseases exhibit distinct pathophysiological, clinical, and therapeutic profiles. Currently, liver diseases contribute to approximately 2 million deaths globally each year, imposing significant economic and social burdens worldwide. However, there is no cure for many kinds of liver diseases, partly due to a lack of thorough understanding of the development of these liver diseases. Therefore, this review provides a comprehensive examination of the epidemiology and characteristics of liver diseases, covering a spectrum from acute and chronic conditions to end-stage manifestations. We also highlight the multifaceted mechanisms underlying the initiation and progression of liver diseases, spanning molecular and cellular levels to organ networks. Additionally, this review offers updates on innovative diagnostic techniques, current treatments, and potential therapeutic targets presently under clinical evaluation. Recent advances in understanding the pathogenesis of liver diseases hold critical implications and translational value for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Can Gan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yuan
- Aier Institute of Ophthalmology, Central South University, Changsha, China
| | - Haiyuan Shen
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangxin Kong
- Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yangkun Guo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| | - Erdan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
- Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
3
|
Maiese K. Cardiovascular and nonalcoholic fatty liver disease: Sharing common ground through SIRT1 pathways. World J Cardiol 2024; 16:632-643. [PMID: 39600987 PMCID: PMC11586725 DOI: 10.4330/wjc.v16.i11.632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/27/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
As a non-communicable disease, cardiovascular disorders have become the leading cause of death for men and women. Of additional concern is that cardiovascular disease is linked to chronic comorbidity disorders that include nonalcoholic fatty liver disease (NAFLD). NAFLD, also termed metabolic-dysfunction-associated steatotic liver disease, is the greatest cause of liver disease throughout the world, increasing in prevalence concurrently with diabetes mellitus (DM), and can progress to nonalcoholic steatohepatitis that leads to cirrhosis and liver fibrosis. Individuals with metabolic disorders, such as DM, are more than two times likely to experience cardiac disease, stroke, and liver disease that includes NAFLD when compared individuals without metabolic disorders. Interestingly, cardiovascular disorders and NAFLD share a common underlying cellular mechanism for disease pathology, namely the silent mating type information regulation 2 homolog 1 (SIRT1; Saccharomyces cerevisiae). SIRT1, a histone deacetylase, is linked to metabolic pathways through nicotinamide adenine dinucleotide and can offer cellular protection though multiple avenues, including trophic factors such as erythropoietin, stem cells, and AMP-activated protein kinase. Translating SIRT1 pathways into clinical care for cardiovascular and hepatic disease can offer significant hope for patients, but further insights into the complexity of SIRT1 pathways are necessary for effective treatment regimens.
Collapse
Affiliation(s)
- Kenneth Maiese
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20810, United States.
| |
Collapse
|
4
|
Wang D, Wang J, Yin Z, Gong K, Zhang S, Zha Z, Duan Y. Polyoxometalates Ameliorate Metabolic Dysfunction-Associated Steatotic Liver Disease by Activating the AMPK Signaling Pathway. Int J Nanomedicine 2024; 19:10839-10856. [PMID: 39479173 PMCID: PMC11522013 DOI: 10.2147/ijn.s485084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction Metabolic dysfunction-associated steatotic liver disease (MASLD), the most prevalent chronic liver disorder, has garnered increasing attention globally owing to its associated health complications. However, the lack of available therapeutic medications and inadequate management of complications in metabolic dysfunction-associated steatohepatitis (MASH) present significant challenges. There are little studies evaluating the effectiveness of POM in treating MASLD. In this study, we synthesized polyoxometalates (POM) for potential treatment of MASLD. Methods We induced liver disease in mice using two approaches: feeding a high-fat diet (HFD) to establish MASLD or feeding a methionine-choline deficient (MCD) diet to induce hepatic lipotoxicity and MASH. Various metabolic parameters were detected, and biochemical and histological evaluations were conducted on MASLD. Western blotting, qRT-PCR and immunofluorescence assays were used to elucidate the molecular mechanism of POM in the treatment of MASLD. Results POM therapy resulted in significant improvements in weight gain, dyslipidemia, liver injury, and hepatic steatosis in mice fed a HFD. Notably, in a more severe dietary-induced MASH model with MCD diet, POM significantly attenuated hepatic lipid accumulation, inflammation, and fibrosis. POM treatment effectively attenuated palmitic acid and oleic acid-induced lipid accumulation in HepG2 and Huh7 cells by targeting the AMPK pathway to regulate lipid metabolism, which was confirmed by AMPK inhibitor. Additionally, the activation of AMPK signaling by POM suppressed the expression of lipid synthesis genes, including sterol regulatory element-binding protein 1c (SREBP1c) and SREBP2, while concurrently upregulating the expression of sirtuin 1 (SIRT1) to promote fatty acid oxidation. Conclusion These findings suggest that POM is a promising therapeutic strategy with high efficacy in multiple MASLD models.
Collapse
Affiliation(s)
- Dandan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230011, People’s Republic of China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, People’s Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230601, People’s Republic of China
| | - Jingguo Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230601, People’s Republic of China
| | - Zequn Yin
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, People’s Republic of China
| | - Ke Gong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230601, People’s Republic of China
| | - Shuang Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230601, People’s Republic of China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230601, People’s Republic of China
| | - Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, People’s Republic of China
| |
Collapse
|
5
|
Jiang Z, Huang B, Cui Z, Lu Z, Ma H. Synergistic effect of genistein and adiponectin reduces fat deposition in chicken hepatocytes by activating the ERβ-mediated SIRT1-AMPK signaling pathway. Poult Sci 2024; 103:103734. [PMID: 38636201 PMCID: PMC11040169 DOI: 10.1016/j.psj.2024.103734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024] Open
Abstract
Dietary supplementation with bioactive substances that can regulate lipid metabolism is an effective approach for reducing excessive fat deposition in chickens. Genistein (GEN) has the potential to alleviate fat deposition; however, the underlying mechanism of GEN's fat-reduction action in chickens remains unclear. Therefore, the present study aimed to explore the underlying mechanism of GEN on the reduction of fat deposition from a novel perspective: intercellular transmission of adipokine between adipocytes and hepatocytes. The findings showed that GEN enhanced the secretion of adiponectin (APN) in chicken adipocytes, and the enhancement effect of GEN was completely blocked when the cells were pretreated with inhibitors targeting estrogen receptor β (ERβ) or proliferator-activated receptor γ (PPARγ) signals, respectively. Furthermore, the results demonstrated that both co-treatment with GEN and APN or treatment with the medium supernatant (Med SUP) derived from chicken adipocytes treated with GEN significantly decreased the content of triglyceride and increased the protein levels of ERβ, Sirtuin 1 (SIRT1) and phosphor-AMP-activated protein kinase (p-AMPK) in chicken hepatocytes compared to the cells treated with GEN or APN alone. Moreover, the increase in the protein levels of SIRT1 and p-AMPK induced by GEN and APN co-treatment or Med SUP treatment were blocked in chicken hepatocytes pretreated with the inhibitor of ERβ signals. Importantly, the up-regulatory effect of GEN and APN co-treatment or Med SUP treatment on the protein level of p-AMPK was also blocked in chicken hepatocytes pretreated with a SIRT1 inhibitor; however, the increase in the protein level of SIRT1 induced by GEN and APN co-treatment or Med SUP treatment was not reversed when the hepatocytes were pretreated with an AMPK inhibitor. In conclusion, the present study demonstrated that GEN enhanced APN secretion by activating the ERβ-Erk-PPARγ signaling pathway in chicken adipocytes. Subsequently, adipocyte-derived APN synergized with GEN to activate the ERβ-mediated SIRT1-AMPK signaling pathway in chicken hepatocytes, ultimately reducing fat deposition. These findings provide substantial evidence from a novel perspective, supporting the potential use of GEN as a dietary supplement to prevent excessive fat deposition in poultry.
Collapse
Affiliation(s)
- Zhihao Jiang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Benzeng Huang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ziyi Cui
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ze Lu
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
6
|
Li L, Yao Y, Wang Y, Cao J, Jiang Z, Yang Y, Wang H, Ma H. G protein-coupled estrogen receptor 1 ameliorates nonalcoholic steatohepatitis through targeting AMPK-dependent signaling. J Biol Chem 2024; 300:105661. [PMID: 38246352 PMCID: PMC10876613 DOI: 10.1016/j.jbc.2024.105661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/22/2023] [Accepted: 01/01/2024] [Indexed: 01/23/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), especially nonalcoholic steatohepatitis (NASH), has emerged as a prevalent cause of liver cirrhosis and hepatocellular carcinoma, posing severe public health challenges worldwide. The incidence of NASH is highly correlated with an increased prevalence of obesity, insulin resistance, diabetes, and other metabolic diseases. Currently, no approved drugs specifically targeted for the therapies of NASH partially due to the unclear pathophysiological mechanisms. G protein-coupled estrogen receptor 1 (GPER1) is a membrane estrogen receptor involved in the development of metabolic diseases such as obesity and diabetes. However, the function of GPER1 in NAFLD/NASH progression remains unknown. Here, we show that GPER1 exerts a beneficial role in insulin resistance, hepatic lipid accumulation, oxidative stress, or inflammation in vivo and in vitro. In particular, we observed that the lipid accumulation, inflammatory response, fibrosis, or insulin resistance in mouse NAFLD/NASH models were exacerbated by hepatocyte-specific GPER1 knockout but obviously mitigated by hepatic GPER1 activation in female and male mice. Mechanistically, hepatic GPER1 activates AMP-activated protein kinase signaling by inducing cyclic AMP release, thereby exerting its protective effect. These data suggest that GPER1 may be a promising therapeutic target for NASH.
Collapse
Affiliation(s)
- Longlong Li
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yao Yao
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yulei Wang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ji Cao
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Zhihao Jiang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ying Yang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Huihui Wang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
7
|
Martínez-Fernández L, Burgos M, Sáinz N, Laiglesia LM, Arbones-Mainar JM, González-Muniesa P, Moreno-Aliaga MJ. Maresin 1 Exerts a Tissue-Specific Regulation of Adipo-Hepato-Myokines in Diet-Induced Obese Mice and Modulates Adipokine Expression in Cultured Human Adipocytes in Basal and Inflammatory Conditions. Biomolecules 2023; 13:919. [PMID: 37371501 DOI: 10.3390/biom13060919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
This study analyses the effects of Maresin 1 (MaR1), a docosahexaenoic acid (DHA)-derived specialized proresolving lipid mediator with anti-inflammatory and insulin-sensitizing actions, on the expression of adipokines, including adiponectin, leptin, dipeptidyl peptidase 4 (DPP-4), cardiotrophin-1 (CT-1), and irisin (FNDC5), both in vitro and in in vivo models of obesity. The in vivo effects of MaR1 (50 μg/kg, 10 days, oral gavage) were evaluated in epididymal adipose tissue (eWAT), liver and muscle of diet-induced obese (DIO) mice. Moreover, two models of human differentiated primary adipocytes were incubated with MaR1 (1 and 10 nM, 24 h) or with a combination of tumor necrosis factor-α (TNF-α, 100 ng/mL) and MaR1 (1-200 nM, 24 h) and the expression and secretion of adipokines were measured in both models. MaR1-treated DIO mice exhibited an increased expression of adiponectin and Ct-1 in eWAT, increased expression of Fndc5 and Ct-1 in muscle and a decreased expression of hepatic Dpp-4. In human differentiated adipocytes, MaR1 increased the expression of ADIPONECTIN, LEPTIN, DPP4, CT-1 and FNDC5. Moreover, MaR1 counteracted the downregulation of ADIPONECTIN and the upregulation of DPP-4 and LEPTIN observed in adipocytes treated with TNF-α. Differential effects for TNF-α and MaR1 on the expression of CT-1 and FNDC5 were observed between both models of human adipocytes. In conclusion, MaR1 reverses the expression of specific adipomyokines and hepatokines altered in obese mice in a tissue-dependent manner. Moreover, MaR1 regulates the basal expression of adipokines in human adipocytes and counteracts the alterations of adipokines expression induced by TNF-α in vitro. These actions could contribute to the metabolic benefits of this lipid mediator.
Collapse
Affiliation(s)
- Leyre Martínez-Fernández
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
| | - Miguel Burgos
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
- IDISNA-Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Neira Sáinz
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
| | - Laura M Laiglesia
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
| | - José Miguel Arbones-Mainar
- Adipocyte and Fat Biology Laboratory (AdipoFat), Unidad de Investigación Traslacional, Instituto Aragonés de Ciencias de la Salud (IACS), Instituto de Investigación Sanitaria (IIS) Aragón, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Pedro González-Muniesa
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
- IDISNA-Navarra Institute for Health Research, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
- IDISNA-Navarra Institute for Health Research, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
8
|
Liao YC, Wu JS, Chou HW, Kuo HY, Lee CT, Wu HT, Li CH, Ou HY. Serum Cardiotrophin-1 Concentration Is Negatively Associated with Controlled Attenuation Parameters in Subjects with Non-Alcoholic Fatty Liver Disease. J Clin Med 2023; 12:jcm12072741. [PMID: 37048824 PMCID: PMC10095180 DOI: 10.3390/jcm12072741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2023] [Accepted: 04/04/2023] [Indexed: 04/09/2023] Open
Abstract
Background: Since non-alcoholic fatty liver disease (NAFLD) is highly associated with obesity, cardiovascular disease, and diabetes, biomarkers for the diagnosis of NAFLD have become an important issue. Although cardiotrophin-1 (CT-1) has a protective effect on the liver in NAFLD animal models, the serum levels of CT-1 in human subjects with NAFLD were still unknown. Objective: The present study aimed to investigate the relationship between the circulating concentration of CT-1 and the severity of hepatic steatosis graded by the value of the controlled attenuation parameter (CAP) in humans. Design and Methods: The study was designed as a cross-sectional study, and a total of 182 subjects were enrolled. Hepatic steatosis measurement was carried out with a Firoscan® device and recorded by CAP. The enrolled study subjects were categorized into CAP < 238 dB/m, 238 ≤ CAP ≤ 259 dB/m, 260 ≤ CAP ≤ 290 dB/m, and CAP > 290 dB/m. Serum CT-1 concentrations were determined by enzyme-linked immunosorbent assay. The association between the serum CT-1 concentration and NAFLD was examined by multivariate linear regression analysis. Results: Body mass index, percentage of body fat, systolic and diastolic blood pressure, alanine aminotransferase (ALT), cholesterol, triglyceride, hemoglobin A1c and homeostatic model assessment for insulin resistance (HOMA-IR) were significantly increased in groups with higher CAP value, whereas high-density lipoprotein cholesterol was significantly decreased. In addition, serum CT-1 concentrations were significantly decreased in subjects with higher CAP values. In multivariate linear regression models, including age, sex, body fat percentage, CAP, high sensitivity- C reactive protein, uric acid, creatinine, ALT, total cholesterol, and HOMA-IR, only age, CAP and uric acid independently associated with CT-1 levels. Moreover, having NAFLD was independently associated with CT-1 after adjustment for sex, obesity and type 2 diabetes. Conclusions: Serum CT-1 concentrations are decreased in subjects with NAFLD and negatively associated with CAP.
Collapse
Affiliation(s)
- Yi-Chun Liao
- Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Juei-Seng Wu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 703, Taiwan
| | - Hsuan-Wen Chou
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 703, Taiwan
| | - Hsin-Yu Kuo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 703, Taiwan
| | - Chun-Te Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 703, Taiwan
| | - Hung-Tsung Wu
- Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chung-Hao Li
- Department of Family Medicine, An Nan Hospital, China Medical University, Tainan 709, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Horng-Yih Ou
- Department of Internal Medicine, School of Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 703, Taiwan
| |
Collapse
|
9
|
Tian R, Yang J, Wang X, Liu S, Dong R, Wang Z, Yang Z, Zhang Y, Cai Z, Yang H, Hu Y, She ZG, Li H, Zhou J, Zhang XJ. Honokiol acts as an AMPK complex agonist therapeutic in non-alcoholic fatty liver disease and metabolic syndrome. Chin Med 2023; 18:30. [PMID: 36932412 PMCID: PMC10024454 DOI: 10.1186/s13020-023-00729-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/15/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver (NAFLD) and its related metabolic syndrome have become major threats to human health, but there is still a need for effective and safe drugs to treat these conditions. Here we aimed to identify potential drug candidates for NAFLD and the underlying molecular mechanisms. METHODS A drug repositioning strategy was used to screen an FDA-approved drug library with approximately 3000 compounds in an in vitro hepatocyte model of lipid accumulation, with honokiol identified as an effective anti-NAFLD candidate. We systematically examined the therapeutic effect of honokiol in NAFLD and metabolic syndrome in multiple in vitro and in vivo models. Transcriptomic examination and biotin-streptavidin binding assays were used to explore the underlying molecular mechanisms, confirmed by rescue experiments. RESULTS Honokiol significantly inhibited metabolic syndrome and NAFLD progression as evidenced by improved hepatic steatosis, liver fibrosis, adipose inflammation, and insulin resistance. Mechanistically, the beneficial effects of honokiol were largely through AMPK activation. Rather than acting on the classical upstream regulators of AMPK, honokiol directly bound to the AMPKγ1 subunit to robustly activate AMPK signaling. Mutation of honokiol-binding sites of AMPKγ1 largely abolished the protective capacity of honokiol against NAFLD. CONCLUSION These findings clearly demonstrate the beneficial effects of honokiol in multiple models and reveal a previously unappreciated signaling mechanism of honokiol in NAFLD and metabolic syndrome. This study also provides new insights into metabolic disease treatment by targeting AMPKγ1 subunit-mediated signaling activation.
Collapse
Affiliation(s)
- Ruifeng Tian
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Jinjie Yang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Xiaoming Wang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Shuaiyang Liu
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Ruixiang Dong
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Zhenya Wang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Zifeng Yang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Yingping Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, 233030, China
| | - Zhiwei Cai
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Hailong Yang
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China
| | - Yufeng Hu
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China
| | - Zhi-Gang She
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China
| | - Hongliang Li
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China.
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China.
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China.
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Junjie Zhou
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, 341000, China.
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, China.
| | - Xiao-Jing Zhang
- Department of Cardiology,Renmin Hospital; School of Basic Medical Science, Wuhan University, Wuhan, 430060, China.
- Institute of Model Animal of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
10
|
Jiang Z, Wang H, Yang Y, Yao Y, Ma H. Genistein activated SIRT1-AMPK signaling pathway mediated by ERβ-FOXO1-Nampt to reduce fat accumulation in chicken hepatocytes. Life Sci 2023; 312:121259. [PMID: 36463943 DOI: 10.1016/j.lfs.2022.121259] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022]
Abstract
Excessive fat accumulation in broiler chickens would seriously threaten the poultry industry. It leads to lower feed conversion rate and worse meat quality. Even worse, it harms the consumers' health due to the intake of high-fat chicken products. Dietary supplements with bioactive ingredients have been considered an effective way to solve this problem. Genistein is the primary phytoestrogen in soybean. Its fat-reduction effect has been reported, but the molecular mechanism is unclear. The present study found that genistein reduced lipid droplets accumulation by regulating lipid metabolism-related factors expression in chicken hepatocytes. The research showed that genistein significantly increased phosphor (p)-AMP-activated protein kinase (p-AMPK) and Sirtuin 1 (SIRT1) protein expressions. The effect of genistein on reducing lipid droplets accumulation and upregulating p-AMPK protein level was blocked entirely when pretreated with SIRT1 inhibitor. These results implied that SIRT1 is required to activate AMPK. Furthermore, genistein treatment significantly upregulated the SIRT1 protein level when pretreated with AMPK inhibitor. We demonstrated that the activation of estrogen receptor β-Forkhead box O1-Nicotinamide phosphoribosyl transferase (ERβ-FOXO1-Nampt) signaling pathway upregulated the NAD+ concentration in hepatocytes, and activated SIRT1 ultimately. In summary, we demonstrated that genistein suppressed lipid droplets accumulation in chicken hepatocytes by activating SIRT1-AMPK. The SIRT1-AMPK signaling pathway was mediated by ERβ-FOXO1-Nampt. These findings increase our understanding of the mechanisms of genistein on fat reduction, and provide compelling evidence for it as a nutritional supplement to prevent excessive fat deposition and lipid metabolism-related diseases in animals and even humans.
Collapse
Affiliation(s)
- Zhihao Jiang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Huihui Wang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ying Yang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yao Yao
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
11
|
New insights into IL-6 family cytokines in metabolism, hepatology and gastroenterology. Nat Rev Gastroenterol Hepatol 2021; 18:787-803. [PMID: 34211157 DOI: 10.1038/s41575-021-00473-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
IL-6 family cytokines are defined by the common use of the signal-transducing receptor chain glycoprotein 130 (gp130). Increasing evidence indicates that these cytokines are essential in the regulation of metabolic homeostasis as well as in the pathophysiology of multiple gastrointestinal and liver disorders, thus making them attractive therapeutic targets. Over the past few years, therapies modulating gp130 signalling have grown exponentially in several clinical settings including obesity, cancer and inflammatory bowel disease. A newly engineered gp130 cytokine, IC7Fc, has shown promising preclinical results for the treatment of type 2 diabetes, obesity and liver steatosis. Moreover, drugs that modulate gp130 signalling have shown promise in refractory inflammatory bowel disease in clinical trials. A deeper understanding of the main roles of the IL-6 family of cytokines during homeostatic and pathological conditions, their signalling pathways, sources of production and target cells will be crucial to the development of improved treatments. Here, we review the current state of the role of these cytokines in hepatology and gastroenterology and discuss the progress achieved in translating therapeutics targeting gp130 signalling into clinical practice.
Collapse
|
12
|
Qian X, Wang T, Gong J, Wang L, Chen X, Lin H, Tu W, Jiang S, Li S. Exercise in mice ameliorates high-fat diet-induced nonalcoholic fatty liver disease by lowering HMGCS2. Aging (Albany NY) 2021; 13:8960-8974. [PMID: 33647884 PMCID: PMC8034885 DOI: 10.18632/aging.202717] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/14/2021] [Indexed: 12/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease worldwide. Exercise is a therapeutic strategy for preventing NAFLD. However, the underlying molecular mechanisms by which NAFLD can be ameliorated through exercise are still not clear. This study investigates the mechanisms by which exercise suppresses NAFLD development induced by a high-fat diet (HFD) in mice. Male 6-week-old C57BL/6J mice were fed a normal diet or HFD for 12 weeks and then induced to swim or remain sedentary for 8 weeks. Histomorphology, inflammatory factors, fat metabolizing enzymes, fibrosis, and steatosis were determined in HFD-fed mouse liver, and levels of hepatic enzymes and molecules in the related pathways were analyzed. NAFLD mice showed evident steatosis, fibrosis, and liver injury, and an increased expression of HMGCS2, Wnt3a/ β-catenin, and phosphorylated (p)-AMPK in the liver. Exercise significantly attenuated these symptoms and downregulated the level of Wnt3a/β-catenin in lipotoxic liver tissue. Inhibition of HMGCS2 expression decreased the activation of the Wnt3a/β-catenin pathway and lowered p-AMPK in palmitate-treated HepG2. Our results suggest that exercise prevents NAFLD-associated liver injury, steatosis, and fibrosis. Exercise-mediated hepatoprotection was achieved partly via the blocking of the upregulation of HMGCS2 and the attenuation of the Wnt3a/β-catenin pathway.
Collapse
Affiliation(s)
- Xiaoli Qian
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.,Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Ting Wang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.,Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jiahong Gong
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.,Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Li Wang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.,Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Xuyan Chen
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.,Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Haiyan Lin
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.,Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Wenzhan Tu
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.,Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Songhe Jiang
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.,Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Shengcun Li
- Rehabilitation Medicine Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.,Integrative and Optimized Medicine Research Center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| |
Collapse
|
13
|
Carneros D, Medina-Gómez G, Giralt M, León-Camacho M, Campbell M, Moreno-Aliaga MJ, Villarroya F, Bustos M. Cardiotrophin-1 contributes to metabolic adaptations through the regulation of lipid metabolism and to the fasting-induced fatty acid mobilization. FASEB J 2020; 34:15875-15887. [PMID: 33047392 DOI: 10.1096/fj.202000109r] [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: 01/15/2020] [Revised: 09/03/2020] [Accepted: 09/16/2020] [Indexed: 11/11/2022]
Abstract
It is becoming clear that several human pathologies are caused by altered metabolic adaptations. During liver development, there are physiological changes, from the predominant utilization of glucose (fetal life) to the use of lipids (postnatal life). Fasting is another physiological stress that elicits well-known metabolic adjustments. We have reported the metabolic properties of cardiotrophin-1 (CT-1), a member of the interleukin-6 family of cytokines. Here, we aimed at analyzing the role of CT-1 in response to these metabolic changes. We used different in vivo models. Furthermore, a differential study was carried out with wild-type and CT-1 null mice in fed (ad libitum) and food-restricted conditions. We demonstrated that Ct-1 is a metabolic gene induced in the liver via PPARα in response to lipids in mice (neonates- and food-restricted adults). We found that Ct-1 mRNA expression in white adipose tissue directly involved PPARα and PPARγ. Finally, the physiological role of CT-1 in fasting is confirmed by the impaired food restriction-induced adipose tissue lipid mobilization in CT-1 null mice. Our findings support a previously unrecognized physiological role of CT-1 in metabolic adaptations, through the regulation of lipid metabolism and contributes to fasting-induced free fatty acid mobilization.
Collapse
Affiliation(s)
- David Carneros
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, Virgen del Rocio University Hospital, Seville, Spain
| | - Gema Medina-Gómez
- Department of Basic Sciences of Health, Area of Biochemistry and Molecular Biology, Rey Juan Carlos University, Madrid, Spain
| | - Marta Giralt
- Department of Biochemistry and Molecular Biomedicine, Barcelona University, Barcelona, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Manuel León-Camacho
- Department of Lipid Characterization and Quality, Instituto de la Grasa (CSIC), Seville, Spain
| | - Mark Campbell
- MRC MDU, Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Maria J Moreno-Aliaga
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Madrid, Spain.,Centre for Nutrition Research and Department of Nutrition, Food Science and Physiology School of Pharmacy and Nutrition, University of Navarra, Navarra's Health Research Institute (IdiSNA), Pamplona, Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, Barcelona University, Barcelona, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Matilde Bustos
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, Virgen del Rocio University Hospital, Seville, Spain
| |
Collapse
|
14
|
Catalpol Attenuates Hepatic Steatosis by Regulating Lipid Metabolism via AMP-Activated Protein Kinase Activation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6708061. [PMID: 32420361 PMCID: PMC7201822 DOI: 10.1155/2020/6708061] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/12/2020] [Accepted: 04/04/2020] [Indexed: 12/18/2022]
Abstract
The increased prevalence of nonalcoholic fatty liver disease (NAFLD), which develops from hepatic steatosis, represents a public health challenge. Catalpol, a natural component extracted from the roots of Radix Rehmanniae, has several pharmacological activities. The present study is aimed at examining whether catalpol prevents hepatic steatosis in cell and animal experiments and elucidating the possible mechanisms. HepG2 cells were treated with 300 μM palmitate (PA) and/or catalpol for 24 h in vitro, and male C57BL/6J mice fed a high-fat diet (HFD) were administered catalpol for 18 weeks in vivo. The results revealed that catalpol significantly decreased lipid accumulation in PA-treated HepG2 cells. Moreover, catalpol drastically reduced body weight and lipid accumulation in the liver, whereas it ameliorated hepatocyte steatosis in HFD-fed mice. Notably, catalpol remarkably promoted the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase. Subsequently, catalpol repressed the expressions of lipogenesis-associated genes such as sterol regulatory element-binding protein 1c and fatty acid synthase but promoted the expressions of genes associated with fatty acid β-oxidation such as peroxisome proliferator-activated receptor α together with its target genes carnitine palmitoyltransferase 1 and acyl-CoA oxidase 1 (ACOX1). However, the preincubation of the HepG2 cells with compound C (10 μM), an AMPK inhibitor, prevented catalpol-mediated beneficial effects. These findings suggest that catalpol ameliorates hepatic steatosis by suppressing lipogenesis and enhancing fatty acid β-oxidation in an AMPK-dependent manner. Therefore, catalpol has potential as a novel agent in the treatment of NAFLD.
Collapse
|
15
|
Sabouri M, Norouzi J, Zarei Y, Sangani MH, Hooshmand Moghadam B. Comparing High-Intensity Interval Training (HIIT) and Continuous Training on Apelin, APJ, NO, and Cardiotrophin-1 in Cardiac Tissue of Diabetic Rats. J Diabetes Res 2020; 2020:1472514. [PMID: 32908933 PMCID: PMC7474749 DOI: 10.1155/2020/1472514] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND AIMS Exercise activity is an important method for managing type 2 diabetes. This investigation examined the HIIT and continuous training on apelin, APJ receptor, NO, and cardiotrophin-1 in the cardiac tissue of diabetic rats. METHODS The animals were categorized into 3 groups of HIIT, continuous (CO), and control (C) (all animals were sacrificed immediately and 2 days after exercise training period). Rats underwent the treadmill exercise program either HIIT (12 bouts at 90-95% of VO2 max with 60 s rest at 50% of VO2 max) or CO (60-65% VO2 max for 40 min). Protocols performed 5 days per week for 8 weeks. Apelin, APJ receptor, NO, and cardiotrophin-1 protein expressions were measured using the Western blotting method in the left ventricle. RESULTS Immediately after HIIT and CO exercise protocols, apelin and CT-1 protein showed a significant difference in contrast by the C-0 group (p < 0.01). However, NO values were substantially higher in HIIT-0 compared to C-0 and CO-0 groups rats (p < 0.01). After two days of exercise protocols, apelin and NO protein showed a significant increase in HIIT and CO groups in contrast to the C animals (p < 0.01). Moreover, APJ and CT-1 protein significantly upregulated in CO-2 and HIIT-2 compared to the other groups (p < 0.01). CONCLUSIONS This study indicates that exercise training, despite the type, is an efficient method to modify apelin, APJ receptor, NO, and cardiotrophin-1 values in animals with type 2 diabetes.
Collapse
Affiliation(s)
- Mostafa Sabouri
- Department of Exercise Physiology & Health Science, University of Tehran, Tehran, Iran
- Oxygen Sports Medical Center, Tehran, Iran
| | - Javad Norouzi
- Department of Exercise Physiology & Health Science, University of Tehran, Tehran, Iran
- Oxygen Sports Medical Center, Tehran, Iran
| | - Yashar Zarei
- Department of Exercise Physiology & Health Science, University of Tehran, Tehran, Iran
| | | | | |
Collapse
|
16
|
Pan X, Kaminga AC, Wen SW, Acheampong K, Liu A. Omentin-1 in diabetes mellitus: A systematic review and meta-analysis. PLoS One 2019; 14:e0226292. [PMID: 31821362 PMCID: PMC6903756 DOI: 10.1371/journal.pone.0226292] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Previous studies found inconsistent results on the relationship between diabetes mellitus and concentrations of omentin-1. This study performed a systematic review and meta-analysis to summarize previous findings on this relationship. METHODS Studies related to this outcome were obtained using a systematic search in the electronic databases of Cochrane Library, PubMed, Embase, SCOPUS, Google Scholar, gray literature and Web of Science in September 2019. The random effects model was used to measure the strength of the association between diabetes mellitus and concentrations of omentin-1, using standardized mean difference. RESULTS Forty-two eligible studies were included in the final meta-analysis. There was no significant difference in omentin-1 concentration between patients with type 1 diabetes mellitus and the controls. On the other hand, lower concentration levels of omentin-1 were observed in patients with gestational diabetes mellitus (standardized mean difference:-0.44, 95% confidence interval:-0.76; -0.12, p = 0.007), or type 2 diabetes mellitus (standardized mean difference: -1.74, 95% confidence interval: -2.31; -1.16, p< 0.001) than in the controls. CONCLUSION Decreased omentin-1 concentrations may be an important indicator for gestational diabetes mellitus and type 2 diabetes mellitus. More studies are needed to validate this hypothesis and evaluate the role of omentin-1 concentrations in type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Xiongfeng Pan
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Atipatsa C. Kaminga
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Department of Mathematics and Statistics, Mzuzu University, Mzuzu, Malawi
| | - Shi Wu Wen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Department of Obstetrics and Gynaecology and Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Kwabena Acheampong
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Department of Public, School of Postgraduate Studies, Adventist University of Africa, Nairobi, Kenya
| | - Aizhong Liu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- * E-mail:
| |
Collapse
|
17
|
Han X, Cui ZY, Song J, Piao HQ, Lian LH, Hou LS, Wang G, Zheng S, Dong XX, Nan JX, Wu YL. Acanthoic acid modulates lipogenesis in nonalcoholic fatty liver disease via FXR/LXRs-dependent manner. Chem Biol Interact 2019; 311:108794. [DOI: 10.1016/j.cbi.2019.108794] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/24/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023]
|
18
|
Li T, Wen L, Cheng B. Cordycepin alleviates hepatic lipid accumulation by inducing protective autophagy via PKA/mTOR pathway. Biochem Biophys Res Commun 2019; 516:632-638. [PMID: 31242974 DOI: 10.1016/j.bbrc.2019.06.108] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 06/19/2019] [Indexed: 11/24/2022]
Abstract
As the major active ingredient of Cordyceps militaris, cordycepin (3'-deoxyadenosine) has been well documented to possess lipid-lowering and anti-oxidative activities, making it a promising candidate for treatment of NAFLD. Autophagy was recently identified as a critical protective mechanism during NAFLD development. Therefore, this study aims to elucidate the mechanism of cordycepin regulating autophagy and lipid metabolism. Here, we found that cordycepin decreased palmitate-induced lipid accumulation by Oil Red O staining, Nile Red staining assays, triglyceride and total cholesterol measurements. Based on Western blot assay and immunocytochemistry, we found that cordycepin induced autophagy in PA-induced steatotic HepG2 cells. Whereas pretreatment with CQ, an autophagy inhibitor, substantially deteriorated the mitigative effects of cordycepin on PA-induced hepatic lipid accumulation. These data taken together indicate that cordycepin protects against PA-induced hepatic lipid accumulation via autophagy induction. Further, cordycepin remarkably increased the expression of P-PKA and decreased P-mTOR, whereas pretreatment with H89, a PKA inhibitor, abolished the ability of cordycepin to activate autophagy via mTOR activation. These data suggested that cordycepin protects against PA-induced hepatic lipid accumulation through the promotion of autophagy. The underlying mechanism might be associated with the PKA/mTOR pathway.
Collapse
Affiliation(s)
- Tianjiao Li
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin Province, 130118, People's Republic of China; College of Food Engineering, Jilin Agricultural Science and Technology University, Jilin, Jilin Province, 132101, People's Republic of China
| | - Liankui Wen
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin Province, 130118, People's Republic of China.
| | - Bijun Cheng
- College of Food Engineering, Jilin Agricultural Science and Technology University, Jilin, Jilin Province, 132101, People's Republic of China.
| |
Collapse
|
19
|
Stephens J, Ravussin E, White U. The Expression of Adipose Tissue-Derived Cardiotrophin-1 in Humans with Obesity. BIOLOGY 2019; 8:biology8020024. [PMID: 31013924 PMCID: PMC6627495 DOI: 10.3390/biology8020024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 01/23/2023]
Abstract
Cardiotrophin-1 (CT-1) is a gp130 cytokine that was previously characterized for its effects on cardiomyocytes and identified as a marker of heart failure. More recent studies reported elevated circulating levels of CT-1 in humans with obesity and metabolic syndrome (MetS). However, a subsequent rodent study implicated CT-1 as a potential therapeutic target for obesity and MetS. Adipose tissue (AT) is broadly acknowledged as an endocrine organ and is a substantial source of CT-1. However, no study has examined the expression of adipose-derived CT-1 in humans. We present the first analysis of CT-1 mRNA expression in subcutaneous AT and its association with clinical variables in 22 women with obesity and 15 men who were 40% overfed for 8-weeks. We observed that CT-1 expression was higher in the subcutaneous abdominal (scABD) than the femoral (scFEM) depot. Importantly, we reveal that scFEM but not scABD, CT-1 expression was negatively associated with visceral adiposity and intrahepatic lipid, while positively correlated with insulin sensitivity in obese women. Also, men with higher CT-1 levels at baseline had less of a decline in insulin sensitivity in response to overfeeding. Our data provide new knowledge on the regulation of adipose-derived CT-1 in obesity and during weight gain in response to overfeeding in humans and suggest that CT-1 may play a protective role in obesity and related disorders.
Collapse
Affiliation(s)
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Ursula White
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| |
Collapse
|
20
|
Carneros D, Santamaría EM, Larequi E, Vélez-Ortiz JM, Reboredo M, Mancheño U, Perugorria MJ, Navas P, Romero-Gómez M, Prieto J, Hervás-Stubbs S, Bustos M. Cardiotrophin-1 is an anti-inflammatory cytokine and promotes IL-4-induced M2 macrophage polarization. FASEB J 2019; 33:7578-7587. [PMID: 30892966 DOI: 10.1096/fj.201801563r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Macrophages play a central role in tissue remodeling, repair, and resolution of inflammation. Macrophage polarization to M1 or M2 activation status may determine the progression or resolution of the inflammatory response. We have previously reported that cardiotrophin-1 (CT-1) displays both cytoprotective and metabolic activities. The role of CT-1 in inflammation remains poorly understood. Here, we employed recombinant CT-1 (rCT-1) and used CT-1-null mice and myeloid-specific CT-1 transgenic mice to investigate whether CT-1 might play a role in the modulation of the inflammatory response. We observed that CT-1 deficiency was associated with enhanced release of inflammatory mediators and with stronger activation of NF-κB in response to LPS, whereas the inflammatory response was attenuated in CT-1 transgenic mice or by administering rCT-1 to wild-type animals prior to LPS challenge. We found that CT-1 promoted IL-6 expression only by nonhematopoietic cells, whereas LPS up-regulated IL-6 expression in both hematopoietic and nonhematopoietic cells. Notably, rCT-1 inhibited LPS-mediated soluble IL-6R induction. Using IL-6-/- mice, we showed that rCT-1 inhibited LPS-induced TNF-α and IFN-γ response in an IL-6-independent manner. Importantly, we demonstrated that CT-1 primes macrophages for IL-4-dependent M2 polarization by inducing IL-4 receptor expression. Mechanistic analyses showed that the signal transducer and activator of transcription 3-suppressor of cytokine signaling 3 axis mediates this effect. Our findings support the notion that CT-1 is a critical regulator of inflammation and suggest that rCT-1 could be a molecule with potential therapeutic application in inflammatory conditions.-Carneros, D., Santamaría, E. M., Larequi, E., Vélez-Ortiz, J. M., Reboredo, M., Mancheño, U., Perugorria, M. J., Navas, P., Romero-Gómez, M., Prieto, J., Hervás-Stubbs, S., Bustos, M. Cardiotrophin-1 is an anti-inflammatory cytokine and promotes IL-4-induced M2 macrophage polarization.
Collapse
Affiliation(s)
- David Carneros
- Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville-Virgen del Rocio University Hospital, Seville, Spain
| | - Eva M Santamaría
- Division of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Eduardo Larequi
- Division of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Jose Miguel Vélez-Ortiz
- Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville-Virgen del Rocio University Hospital, Seville, Spain
| | - Mercedes Reboredo
- Division of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Uxua Mancheño
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - María Jesús Perugorria
- Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain.,Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo (CABD), University of Pablo de Olavide-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain.,Centro de Investigación Biomédica en Red Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Seville, Spain
| | - Manuel Romero-Gómez
- Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville-Virgen del Rocio University Hospital, Seville, Spain.,Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Prieto
- Division of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Hervás-Stubbs
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Matilde Bustos
- Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville-Virgen del Rocio University Hospital, Seville, Spain
| |
Collapse
|
21
|
Potential clinical treatment of colitis with cardiotrophin-1. Clin Sci (Lond) 2018; 132:2169-2174. [PMID: 30341227 DOI: 10.1042/cs20171626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/05/2018] [Accepted: 09/26/2018] [Indexed: 11/17/2022]
Abstract
In a recent issue of Clinical Science, Prieto-Vicente et al. [Clin. Sci. (2018) 132, 985-1001] have smartly demonstrated a potential new use of cardiotrophin-1 (CT-1) to treat and palliate an inflammatory bowel disease such as ulcerative colitis. In that work, authors report that in ulcerative colitic mice, administration of exogenous recombinant CT-1 (rCT-1) promotes lower colon damage and lower disease activity index, reducing systemic levels of tumor necrosis factor α (TNF-α) and also diminishing TNF-α expression in colon together with the reduction in other common inflammation markers. Besides, in vivo rCT-1 administration induces activation of several molecular pathways, including nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator of transcription (STAT)-3, and abolishes bacterial translocation from intestine to other organs, including mesenteric ganglia, lungs, and spleen. Additionally, these results were nicely corroborated in CT-1 depleted mice; in which colon damage and ulcerative colitis severity were greater compared with the wild-type counterparts. All together, these results suggested that CT-1 could be a promising new therapeutic approach for treating inflammatory bowel disease, particularly ulcerative colitis. However, further studies are required to determine its major mechanisms of action and the potential efficacy of CT-1 in human inflammatory bowel diseases.
Collapse
|
22
|
Bae UJ, Park J, Park IW, Chae BM, Oh MR, Jung SJ, Ryu GS, Chae SW, Park BH. Epigallocatechin-3-Gallate-Rich Green Tea Extract Ameliorates Fatty Liver and Weight Gain in Mice Fed a High Fat Diet by Activating the Sirtuin 1 and AMP Activating Protein Kinase Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:617-632. [PMID: 29595075 DOI: 10.1142/s0192415x18500325] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The prevalence of metabolic diseases has risen globally in parallel with the obesity epidemic over the past few decades. Green tea has been reported to have metabolically beneficial effects on obesity; however, the mechanism by which green tea regulates lipid metabolism is not clearly understood. Male c57BL/6 mice were fed a normal chow diet, a high-fat diet (HFD), or an HFD supplemented with various doses of epigallocatechin gallate-rich green tea extract (GTE) for 12 weeks. GTE supplementation reduced body weight gain, prevented hepatic fat accumulation, decreased hypertriglyceridemia, and improved hyperglycemia and insulin resistance in HFD-fed mice. The underlying mechanisms of these beneficial effects of GTE might involve the upregulation of sirtuin 1 and AMP activated protein kinase (AMPK) and the downregulation of enzymes related to de novo lipogenesis. Consistent with the in vivo findings, GTE increased the expression and activity of sirtuin 1, enhanced the binding of sirtuin 1 to liver kinase B1 (LKB1) and subsequent deacetylation of LKB1, and reduced triglyceride accumulation in HepG2 cells. These results suggest the possible therapeutic potential of dietary epigallocatechin gallate-rich GTE supplementation for preventing the development and progression of hepatic steatosis and obesity.
Collapse
Affiliation(s)
- Ui-Jin Bae
- * Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea.,‡ Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Jeonbuk 54907, South Korea
| | - John Park
- § Department of Chemistry, Chonbuk National University, Jeonju, Jeonbuk 54896, South Korea
| | - Il Woon Park
- ¶ Department of Cognitive Science, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Byung Min Chae
- ∥ Division of Biotechnology, College of Environmental and Bioresource Sciences, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Mi-Ra Oh
- ‡ Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Jeonbuk 54907, South Korea
| | - Su-Jin Jung
- ‡ Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Jeonbuk 54907, South Korea
| | | | - Soo-Wan Chae
- † Department of Pharmacology, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea.,‡ Clinical Trial Center for Functional Foods, Chonbuk National University Hospital, Jeonju, Jeonbuk 54907, South Korea
| | - Byung-Hyun Park
- * Department of Biochemistry, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea
| |
Collapse
|
23
|
Protein inhibitor of activated STAT 4 (PIAS4) regulates pro-inflammatory transcription in hepatocytes by repressing SIRT1. Oncotarget 2018; 7:42892-42903. [PMID: 27285989 PMCID: PMC5189995 DOI: 10.18632/oncotarget.9864] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/06/2016] [Indexed: 01/07/2023] Open
Abstract
Excessive nutrition promotes the pathogenesis of non-alcoholic steatohepatitis (NASH), characterized by the accumulation of pro-inflammation mediators in the liver. In the present study we investigated the regulation of pro-inflammatory transcription in hepatocytes by protein inhibitor of activated STAT 4 (PIAS4) in this process and the underlying mechanisms. We report that expression of the class III deacetylase SIRT1 was down-regulated in the livers of NASH mice accompanied by a simultaneous increase in the expression and binding activity of PIAS4. Exposure to high glucose stimulated the expression PIAS4 in cultured hepatocytes paralleling SIRT1 repression. Estrogen, a known NASH-protective hormone, ameliorated SIRT1 trans-repression by targeting PIAS4. Over-expression of PIAS4 enhanced, while PIAS4 knockdown alleviated, repression of SIRT1 transcription by high glucose. Lentiviral delivery of short hairpin RNA (shRNA) targeting PIAS4 attenuated hepatic inflammation in NASH mice by restoring SIRT1 expression. Mechanistically, PIAS4 promoted NF-κB-mediated pro-inflammatory transcription in a SIRT1 dependent manner. In conclusion, our study indicates that PIAS4 mediated SIRT1 repression in response to nutrient surplus contributes to the pathogenesis of NASH. Therefore, targeting PIAS4 might provide novel therapeutic strategies in the intervention of NASH.
Collapse
|
24
|
Wang X, Zhang ZF, Zheng GH, Wang AM, Sun CH, Qin SP, Zhuang J, Lu J, Ma DF, Zheng YL. Attenuation of hepatic steatosis by purple sweet potato colour is associated with blocking Src/ERK/C/EBPβ signalling in high-fat-diet–treated mice. Appl Physiol Nutr Metab 2017. [DOI: 10.1139/apnm-2016-0635] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Our previous work showed that purple sweet potato colour (PSPC), a class of naturally occurring anthocyanins, effectively improved hepatic glucose metabolic dysfunction in high-fat-diet (HFD)–treated mice. This study investigated the effects of PSPC on HFD-induced hepatic steatosis and the signalling events associated with these effects. Mice were divided into 4 groups: control group, HFD group, HFD+PSPC group, and PSPC group. PSPC was administered daily for 20 weeks at oral doses of 700 mg/(kg·day)−1). Our results showed that PSPC significantly improved obesity and related metabolic parameters, as well as liver injury in HFD-treated mice. Moreover, PSPC dramatically attenuated hepatic steatosis in HFD-treated mice. PSPC markedly prevented oxidative stress-mediated Src activation in HFD-treated mouse livers. Furthermore, PSPC feeding remarkably suppressed mitogen-activated protein kinase kinase/extracellular-signal-regulated kinase (MEK/ERK) signalling and consequent CCAAT/enhancer binding protein β (C/EBPβ) activation and restored AMPK activation in HFD-treated mouse livers, which was confirmed by U0126 treatment. Ultimately, PSPC feeding dramatically reduced protein expression of FAS and CD36 and the activation of ACC, and increased the protein expression of CPT1A in the livers of HFD-treated mice, indicating decreased lipogenesis and fatty acid uptake and enhanced fatty acid oxidation. In conclusion, PSPC exhibited beneficial effects on hepatic steatosis, which were associated with blocking Src and C/EBPβ activation.
Collapse
Affiliation(s)
- Xin Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province 221116, PR China
- Key Laboratory of Biology and Genetic Improvement of Sweetpotato, Ministry of Agriculture, Jiangsu Xuzhou Sweetpotato Research Center, Xuzhou 221131, Jiangsu Province, PR China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province 221116, PR China
| | - Gui-Hong Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province 221116, PR China
| | - Ai-Min Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province 221116, PR China
| | - Chun-Hui Sun
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province 221116, PR China
| | - Su-Ping Qin
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province 221116, PR China
| | - Juan Zhuang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province 221116, PR China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province 221116, PR China
| | - Dai-Fu Ma
- Key Laboratory of Biology and Genetic Improvement of Sweetpotato, Ministry of Agriculture, Jiangsu Xuzhou Sweetpotato Research Center, Xuzhou 221131, Jiangsu Province, PR China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu Province 221116, PR China
| |
Collapse
|
25
|
Laiglesia LM, Lorente-Cebrián S, López-Yoldi M, Lanas R, Sáinz N, Martínez JA, Moreno-Aliaga MJ. Maresin 1 inhibits TNF-alpha-induced lipolysis and autophagy in 3T3-L1 adipocytes. J Cell Physiol 2017; 233:2238-2246. [PMID: 28703289 DOI: 10.1002/jcp.26096] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 07/11/2017] [Indexed: 12/30/2022]
Abstract
Obesity is associated with high levels of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), which promotes inflammation in adipose tissue. The omega-3 PUFAs, and their derived lipid mediators, such as Maresin 1 (MaR1) have anti-inflammatory effects on adipose tissue. This study aimed to analyze if MaR1 may counteract alterations induced by TNF-α on lipolysis and autophagy in mature 3T3-L1 adipocytes. Our data revealed that MaR1 (1-100 nM) inhibited the TNF-α-induced glycerol release after 48 hr, which may be related to MaR1 ability of preventing the decrease in lipid droplet-coating protein perilipin and G0/G1 Switch 2 protein expression. MaR1 also reversed the decrease in total hormone sensitive lipase (total HSL), and the ratio of phosphoHSL at Ser-565/total HSL, while preventing the increased ratio of phosphoHSL at Ser-660/total HSL and phosphorylation of extracellular signal-regulated kinase 1/2 induced by TNF-α. Moreover, MaR1 counteracted the cytokine-induced decrease of p62 protein, a key autophagy indicator, and also prevented the induction of LC3II/LC3I, an important autophagosome formation marker. Current data suggest that MaR1 may ameliorate TNF-α-induced alterations on lipolysis and autophagy in adipocytes. This may also contribute to the beneficial actions of MaR1 on adipose tissue and insulin sensitivity in obesity.
Collapse
Affiliation(s)
- Laura M Laiglesia
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Silvia Lorente-Cebrián
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Miguel López-Yoldi
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Raquel Lanas
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain
| | - Neira Sáinz
- Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Jose Alfredo Martínez
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Maria J Moreno-Aliaga
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
26
|
Escoté X, Gómez-Zorita S, López-Yoldi M, Milton-Laskibar I, Fernández-Quintela A, Martínez JA, Moreno-Aliaga MJ, Portillo MP. Role of Omentin, Vaspin, Cardiotrophin-1, TWEAK and NOV/CCN3 in Obesity and Diabetes Development. Int J Mol Sci 2017; 18:ijms18081770. [PMID: 28809783 PMCID: PMC5578159 DOI: 10.3390/ijms18081770] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 01/22/2023] Open
Abstract
Adipose tissue releases bioactive mediators called adipokines. This review focuses on the effects of omentin, vaspin, cardiotrophin-1, Tumor necrosis factor-like Weak Inducer of Apoptosis (TWEAK) and nephroblastoma overexpressed (NOV/CCN3) on obesity and diabetes. Omentin is produced by the stromal-vascular fraction of visceral adipose tissue. Obesity reduces omentin serum concentrations and adipose tissue secretion in adults and adolescents. This adipokine regulates insulin sensitivity, but its clinical relevance has to be confirmed. Vaspin is produced by visceral and subcutaneous adipose tissues. Vaspin levels are higher in obese subjects, as well as in subjects showing insulin resistance or type 2 diabetes. Cardiotrophin-1 is an adipokine with a similar structure as cytokines from interleukin-6 family. There is some controversy regarding the regulation of cardiotrophin-1 levels in obese -subjects, but gene expression levels of cardiotrophin-1 are down-regulated in white adipose tissue from diet-induced obese mice. It also shows anti-obesity and hypoglycemic properties. TWEAK is a potential regulator of the low-grade chronic inflammation characteristic of obesity. TWEAK levels seem not to be directly related to adiposity, and metabolic factors play a critical role in its regulation. Finally, a strong correlation has been found between plasma NOV/CCN3 concentration and fat mass. This adipokine improves insulin actions.
Collapse
Affiliation(s)
- Xavier Escoté
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain.
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain.
| | - Saioa Gómez-Zorita
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
| | - Miguel López-Yoldi
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain.
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain.
| | - Iñaki Milton-Laskibar
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
| | - Alfredo Fernández-Quintela
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
| | - J Alfredo Martínez
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain.
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
- Navarra Institute for Health Research (IdiSNa), 31008 Pamplona, Spain.
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, 31008 Pamplona, Spain.
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
- Navarra Institute for Health Research (IdiSNa), 31008 Pamplona, Spain.
| | - María P Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, 01006 Vitoria, Spain.
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 01006 Vitoria, Spain.
| |
Collapse
|
27
|
Wang D, Liu X, Liu Y, Li S, Wang C. The Effects of Cardiotrophin-1 on Early Synaptic Mitochondrial Dysfunction and Synaptic Pathology in APPswe/PS1dE9 Mice. J Alzheimers Dis 2017; 59:1255-1267. [DOI: 10.3233/jad-170100] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Dongmei Wang
- Department of Pathogen Biology, Medical College, Henan University of Science and Technology, Luolong District, Luoyang, China
| | - Xiaozhuan Liu
- Department of Immunology, Medical College, Henan University of Science and Technology, Luolong District, Luoyang, China
| | - Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luolong District, Luoyang, China
| | - Sanqiang Li
- Department of Biochemistry and Molecular Biology, Medical College, Henan University of Science and Technology, Luolong District, Luoyang, China
| | - Chenying Wang
- Department of Pathogen Biology, Medical College, Henan University of Science and Technology, Luolong District, Luoyang, China
| |
Collapse
|
28
|
Ding RB, Bao J, Deng CX. Emerging roles of SIRT1 in fatty liver diseases. Int J Biol Sci 2017; 13:852-867. [PMID: 28808418 PMCID: PMC5555103 DOI: 10.7150/ijbs.19370] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/19/2017] [Indexed: 12/11/2022] Open
Abstract
Fatty liver diseases, which are commonly associated with high-fat/calorie diet, heavy alcohol consumption and/or other metabolic disorder causes, lead to serious medical concerns worldwide in recent years. It has been demonstrated that metabolic homeostasis disruption is most likely to be responsible for this global epidemic. Sirtuins are a group of conserved nicotinamide adenine dinucleotide (NAD+) dependent histone and/or protein deacetylases belonging to the silent information regulator 2 (Sir2) family. Among seven mammalian sirtuins, sirtuin 1 (SIRT 1) is the most extensively studied one and is involved in both alcoholic and nonalcoholic fatty liver diseases. SIRT1 plays beneficial roles in regulating hepatic lipid metabolism, controlling hepatic oxidative stress and mediating hepatic inflammation through deacetylating some transcriptional regulators against the progression of fatty liver diseases. Here we summarize the latest advances of the biological roles of SIRT1 in regulating lipid metabolism, oxidative stress and inflammation in the liver, and discuss the potential of SIRT1 as a therapeutic target for treating alcoholic and nonalcoholic fatty liver diseases.
Collapse
Affiliation(s)
- Ren-Bo Ding
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Jiaolin Bao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Chu-Xia Deng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| |
Collapse
|
29
|
López-Yoldi M, Marcos-Gomez B, Romero-Lozano MA, Sáinz N, Prieto J, Martínez JA, Bustos M, Moreno-Aliaga MJ. Cardiotrophin-1 Regulates Adipokine Production in 3T3-L1 Adipocytes and Adipose Tissue From Obese Mice. J Cell Physiol 2017; 232:2469-2477. [PMID: 27608275 DOI: 10.1002/jcp.25590] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 09/06/2016] [Indexed: 12/19/2022]
Abstract
Cardiotrophin-1 (CT-1) belongs to the IL-6 family of cytokines. Previous studies of our group revealed that CT-1 is a key regulator of glucose and lipid metabolism. The aim of the present study was to analyze the in vitro and in vivo effects of CT-1 on the production of several adipokines involved in body weight regulation, nutrient metabolism, and inflammation. For this purpose, 3T3-L1 adipocytes were incubated with recombinant protein CT-1 (rCT-1) (1-40 ng/ml) for 1 and 18 h. Moreover, the acute effects of rCT-1 administration (0.2 mg/kg, i.v.) for 30 min and 3 h on adipokines levels were also evaluated in high-fat fed obese mice. In 3T3-L1 adipocytes, rCT-1 treatment downregulated the expression and secretion of leptin, resistin, and visfatin. However, rCT-1 significantly stimulated apelin mRNA and secretion. rCT-1 (18 h) also promoted the activation by phosphorylation of AKT, ERK 1/2, and STAT3. Interestingly, pre-treatment with the PI3K inhibitor LY294002 reversed the stimulatory effects of rCT-1 on apelin expression, suggesting that this pathway could be mediating the effects of rCT-1 on apelin production. In contrast, acute administration of rCT-1 (30 min and 3 h) to diet-induced obese mice downregulated leptin and resistin, without significantly modifying apelin or visfatin mRNA in adipose tissue. Furthermore, CT-1 null mice exhibited altered expression of adipokines in adipose tissue. The present study demonstrates that rCT-1 modulates the production of adipokines in vitro and in vivo, suggesting that the regulation of the secretory function of adipocytes could be involved in the metabolic actions of this cytokine. J. Cell. Physiol. 232: 2469-2477, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Miguel López-Yoldi
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain
| | - Beatriz Marcos-Gomez
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain
| | | | - Neira Sáinz
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain
| | - Jesús Prieto
- Department of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Navarra, Spain.,CIBERehd, Institute of Health Carlos III, Madrid, Spain
| | - Jose Alfredo Martínez
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain.,IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
| | - Matilde Bustos
- Department of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Navarra, Spain
| | - Maria J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Navarra, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain.,IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
| |
Collapse
|
30
|
Wang D, Liu X, Liu Y, Shen G, Zhu X, Li S. Treatment effects of Cardiotrophin-1 (CT-1) on streptozotocin-induced memory deficits in mice. Exp Gerontol 2017; 92:42-45. [PMID: 28285145 DOI: 10.1016/j.exger.2017.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 10/20/2022]
Abstract
Increasing evidence has shown that diabetes-associated cognitive impairment is correlated with mitochondrial dysfunction and resultant synaptic injury as well as brain insulin resistance. Cardiotrophin-1 (CT-1), a regulator of energy metabolism, has been shown to exhibit impressive neuroprotective effects. In this study, we evaluated the effects of CT-1 on brain pathological features in intracerebroventrical-streptozotocin (ICV-STZ)-treated mouse model, and explored its potential mechanisms. STZ was injected twice (3mg/kg, ICV) on alternate days (day 1 and day 3) in mice. Daily treatment with CT-1 (1μg/day, ICV) starting from the first dose of STZ for 14days showed that CT-1 significantly improved learning and memory deficits, alleviated mitochondrial dysfunction, and increased synaptic density in the CA1 region of the hippocampus in ICV-STZ-treated mice. Moreover, CT-1 significantly enhanced insulin signaling pathway in the hippocampus of ICV-STZ-treated mice when compared with the control. However, all the protective effects including biochemistry, pathological changes and cognitive function could be blocked by an ICV injection of Compound C, a specific AMPK inhibitor. Taken together, these results suggested that CT-1 improves pathological changes and cognitive impairments via AMPK activation in ICV-STZ mice.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Pathogen Biology, Medical College, Henan University of Science and Technology, Luoyang, China.
| | - Xiaozhuan Liu
- Department of Immunology, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Guomin Shen
- Department of Biochemistry and Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Xiaoying Zhu
- Department of Pathogen Biology, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Sanqiang Li
- Department of Biochemistry and Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
31
|
Kim M, Yoo G, Randy A, Kim HS, Nho CW. Chicoric acid attenuate a nonalcoholic steatohepatitis by inhibiting key regulators of lipid metabolism, fibrosis, oxidation, and inflammation in mice with methionine and choline deficiency. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201600632] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/29/2016] [Accepted: 12/05/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Myungsuk Kim
- Natural Products Research Center; Korea Institute of Science and Technology; Gangneung Republic of Korea
- Convergence Research Center for Smart Farm Solution; Korea Institute of Science and Technology; Gangneung Republic of Korea
| | - GyHye Yoo
- Natural Products Research Center; Korea Institute of Science and Technology; Gangneung Republic of Korea
- Convergence Research Center for Smart Farm Solution; Korea Institute of Science and Technology; Gangneung Republic of Korea
| | - Ahmad Randy
- Natural Products Research Center; Korea Institute of Science and Technology; Gangneung Republic of Korea
- Department of Biological Chemistry, Korea; University of Science and Technology; Daejeon Republic of Korea
| | - Hyoung Seok Kim
- Natural Products Research Center; Korea Institute of Science and Technology; Gangneung Republic of Korea
- Convergence Research Center for Smart Farm Solution; Korea Institute of Science and Technology; Gangneung Republic of Korea
| | - Chu Won Nho
- Natural Products Research Center; Korea Institute of Science and Technology; Gangneung Republic of Korea
- Convergence Research Center for Smart Farm Solution; Korea Institute of Science and Technology; Gangneung Republic of Korea
| |
Collapse
|
32
|
Kong Q, Zhang H, Zhao T, Zhang W, Yan M, Dong X, Li P. Tangshen formula attenuates hepatic steatosis by inhibiting hepatic lipogenesis and augmenting fatty acid oxidation in db/db mice. Int J Mol Med 2016; 38:1715-1726. [PMID: 27840945 PMCID: PMC5117754 DOI: 10.3892/ijmm.2016.2799] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 11/03/2016] [Indexed: 12/24/2022] Open
Abstract
Tangshen formula (TSF), a well-prescribed traditional Chinese formula, has been used in the treatment of diabetic nephropathy. However, whether TSF ameliorates dyslipidemia and liver injury associated with diabetes remains unclear. In this study, we examined the effects of TSF on lipid profiles and hepatic steatosis in db/db mice. For this purpose, 8‑week-old db/db mice were treated with TSF or saline for 12 weeks via gavage and db/m mice were used as controls. Body weight and blood glucose levels were monitored weekly and bi-weekly, respectively. Blood samples were obtained for the analysis of lipids and enzymes related to hepatic function, and liver tissues were analyzed by histology, immunohistochemistry and molecular examination. The results revealed that TSF markedly reduced body weight, liver index [liver/body weight (LW/BW)] and improved lipid profiles, hepatic function and steatosis in db/db mice. TSF induced the phosphoralation of AMP-activated protein kinase and inhibited the activity of sterol regulatory element-binding protein 1 together with the inhibition of the expression of genes involved in de novo lipogenesis (DNL) and gluconeogenesis, such as fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), stearoyl CoA desaturase 1 (SCD1), glucose-6-phosphatase (G6pc) and phosphoenolpyruvate carboxykinase 1 (Pck1). Additionally, the silent mating type information regulation 2 homolog 1 (Sirt1)/peroxisome proliferator-activated receptor α (PPARα)/malonyl-CoA decarboxylase (MLYCD) cascade was potently activated by TSF in the liver and skeletal muscle of db/db mice, which led to enhanced fatty acid oxidation. These findings demonstrated that TSF attenuated hepatic fat accumulation and steatosis in db/db mice by inhibiting lipogenesis and augmenting fatty acid oxidation.
Collapse
Affiliation(s)
- Qin Kong
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China
| | - Haojun Zhang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Tingting Zhao
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Weiku Zhang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Meihua Yan
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Xi Dong
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Ping Li
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
33
|
Jia L, Li W, Li J, Li Y, Song H, Luan Y, Qi H, Ma L, Lu X, Yang Y. Lycium barbarum polysaccharide attenuates high-fat diet-induced hepatic steatosis by up-regulating SIRT1 expression and deacetylase activity. Sci Rep 2016; 6:36209. [PMID: 27824080 PMCID: PMC5099939 DOI: 10.1038/srep36209] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 10/12/2016] [Indexed: 01/26/2023] Open
Abstract
In this study, we aimed to investigate the protective effects and underlying mechanism of Lycium barbarum polysaccharide (LBP) on high-fat-induced nonalcoholic fatty liver disease (NAFLD). Recently, sirtuin 1 (SIRT1) has been shown to play an important role in the regulation of hepatocellular lipid metabolism. Here, we demonstrated that LBP up-regulates SIRT1 deacetylase activity and protein expression by enhancing the NAD+/NADH ratio. Subsequently, LBP promoted LKB1 deacetylation and AMPK phosphorylation via SIRT1-dependent signalling. We also found that LBP increases acetyl-CoA carboxylase (ACC) phosphorylation and adipose triglyceride lipase (ATGL) protein expression and decreases fatty acid synthase (FAS) by activating the SIRT1/LKB1/AMPK pathway in vitro and in vivo. However, SIRT1 small interfering RNA (siRNA)-mediated knockdown reversed the LBP-mediated effects on ACC, FAS and ATGL. Moreover, LBP elevated carnitine palmitoyltransferase-1 alpha (CPT-1α) expression by suppressing malonyl-CoA accumulation. Taken together, our data indicate that LBP plays a vital role in the regulation of hepatic lipid metabolism and that pharmacological activation of SIRT1 by LBP may be a strategy for the prevention of NAFLD.
Collapse
Affiliation(s)
- Li Jia
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China.,Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| | - Wang Li
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China.,Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| | - Jianning Li
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China.,Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| | - Yan Li
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China.,Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| | - Hui Song
- Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| | - Yansong Luan
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China.,Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| | - Hui Qi
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China.,Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| | - Lirong Ma
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China.,Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| | - Xiaohong Lu
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China.,Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| | - Yi Yang
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China.,Institute of Endocrinology, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
34
|
Smith BK, Marcinko K, Desjardins EM, Lally JS, Ford RJ, Steinberg GR. Treatment of nonalcoholic fatty liver disease: role of AMPK. Am J Physiol Endocrinol Metab 2016; 311:E730-E740. [PMID: 27577854 DOI: 10.1152/ajpendo.00225.2016] [Citation(s) in RCA: 371] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/28/2016] [Indexed: 01/15/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing worldwide epidemic and an important risk factor for the development of insulin resistance, type 2 diabetes, nonalcoholic steatohepatitis (NASH), and hepatic cellular carcinoma (HCC). Despite the prevalence of NAFLD, lifestyle interventions involving exercise and weight loss are the only accepted treatments for this disease. Over the last decade, numerous experimental compounds have been shown to improve NAFLD in preclinical animal models, and many of these therapeutics have been shown to increase the activity of the cellular energy sensor AMP-activated protein kinase (AMPK). Because AMPK activity is reduced by inflammation, obesity, and diabetes, increasing AMPK activity has been viewed as a viable therapeutic strategy to improve NAFLD. In this review, we propose three primary mechanisms by which AMPK activation may improve NAFLD. In addition, we examine the mechanisms by which AMPK is activated. Finally, we identify 27 studies that have used AMPK activators to reduce NAFLD. Future considerations for studies examining the relationship between AMPK and NAFLD are highlighted.
Collapse
Affiliation(s)
- Brennan K Smith
- Division of Endocrinology and Metabolism, Department of Medicine; and
| | - Katarina Marcinko
- Division of Endocrinology and Metabolism, Department of Medicine; and
| | - Eric M Desjardins
- Division of Endocrinology and Metabolism, Department of Medicine; and
| | - James S Lally
- Division of Endocrinology and Metabolism, Department of Medicine; and
| | - Rebecca J Ford
- Division of Endocrinology and Metabolism, Department of Medicine; and
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine; and Department of Biochemistry, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
35
|
López-Yoldi M, Castilla-Madrigal R, Lostao MP, Barber A, Prieto J, Martínez JA, Bustos M, Moreno-Aliaga MJ. Cardiotrophin-1 decreases intestinal sugar uptake in mice and in Caco-2 cells. Acta Physiol (Oxf) 2016; 217:217-26. [PMID: 26972986 DOI: 10.1111/apha.12674] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/12/2015] [Accepted: 03/04/2016] [Indexed: 12/15/2022]
Abstract
AIM Cardiotrophin-1 (CT-1) is a member of the IL-6 family of cytokines with a key role in glucose and lipid metabolism. In the current investigation, we examined the in vivo and in vitro effects of CT-1 treatment on intestinal sugar absorption in different experimental models. METHODS rCT-1 effects on α-Methyl-D-glucoside uptake were assessed in everted intestinal rings from wild-type and CT-1(-/-) mice and in Caco-2 cells. rCT-1 actions on SGLT-1 expression in brush border membrane vesicles and the identification of the potential signalling pathways involved were determined by Western blot. RESULTS In vivo administration (0.2 mg kg(-1) ) of rCT-1 caused a significant decrease on α-Methyl-D-glucoside uptake in everted intestinal rings from wild-type and CT-1(-/-) mice after short-term and long-term treatments. Similarly, in vitro treatment (1-50 ng mL(-1) ) with rCT-1 reduced α-Methyl-D-glucoside uptake in everted intestinal rings. In Caco-2 cells, rCT-1 treatment (20 ng mL(-1) , 1 and 24 h) lowered apical uptake of α-Methyl-D-glucoside in parallel with a decrease on SGLT-1 protein expression. rCT-1 promoted the phosphorylation of STAT-3 after 5 and 15 min treatment, but inhibited the activation by phosphorylation of AMPK after 30 and 60 min. Interestingly, pre-treatment with the JAK/STAT inhibitor (AG490) and with the AMPK activator (AICAR) reversed the inhibitory effects of rCT-1 on α-Methyl-D-glucoside uptake. AICAR also prevented the inhibition of SGLT-1 observed in rCT-1-treated cells. CONCLUSIONS CT-1 inhibits intestinal sugar absorption by the reduction of SGLT-1 levels through the AMPK pathway, which could also contribute to explain the hypoglycaemic and anti-obesity properties of CT-1.
Collapse
Affiliation(s)
- M. López-Yoldi
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
| | - R. Castilla-Madrigal
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
| | - M. P. Lostao
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
- IdiSNA; Navarra Institute for Health Research; Pamplona Spain
| | - A. Barber
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
| | - J. Prieto
- Department of Gene Therapy and Hepatology; CIMA; University of Navarra; Pamplona Navarra Spain
- CIBERehd; Institute of Health Carlos III; Madrid Spain
| | - J. A. Martínez
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
- IdiSNA; Navarra Institute for Health Research; Pamplona Spain
- CIBERobn; Physiopathology of Obesity and Nutrition; Institute of Health Carlos III; Madrid Spain
| | - M. Bustos
- Department of Gene Therapy and Hepatology; CIMA; University of Navarra; Pamplona Navarra Spain
| | - M. J. Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology; University of Navarra; Pamplona Navarra Spain
- Centre for Nutrition Research; University of Navarra; Pamplona Navarra Spain
- IdiSNA; Navarra Institute for Health Research; Pamplona Spain
- CIBERobn; Physiopathology of Obesity and Nutrition; Institute of Health Carlos III; Madrid Spain
| |
Collapse
|
36
|
Pasquin S, Sharma M, Gauchat JF. Cytokines of the LIF/CNTF family and metabolism. Cytokine 2016; 82:122-4. [DOI: 10.1016/j.cyto.2015.12.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/24/2015] [Indexed: 12/17/2022]
|
37
|
Musso G, Cassader M, Gambino R. Non-alcoholic steatohepatitis: emerging molecular targets and therapeutic strategies. Nat Rev Drug Discov 2016; 15:249-74. [PMID: 26794269 DOI: 10.1038/nrd.2015.3] [Citation(s) in RCA: 337] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Non-alcoholic fatty liver disease - the most common chronic liver disease - encompasses a histological spectrum ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). Over the next decade, NASH is projected to be the most common indication for liver transplantation. The absence of an effective pharmacological therapy for NASH is a major incentive for research into novel therapeutic approaches for this condition. The current focus areas for research include the modulation of nuclear transcription factors; agents that target lipotoxicity and oxidative stress; and the modulation of cellular energy homeostasis, metabolism and the inflammatory response. Strategies to enhance resolution of inflammation and fibrosis also show promise to reverse the advanced stages of liver disease.
Collapse
Affiliation(s)
- Giovanni Musso
- Gradenigo Hospital, Corso Regina Margherita 8, 10132 Turin, Italy
| | - Maurizio Cassader
- Department of Medical Sciences, University of Turin, Corso A.M. Dogliotti 14, 10126, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, University of Turin, Corso A.M. Dogliotti 14, 10126, Turin, Italy
| |
Collapse
|
38
|
Cardiotrophin-1 is inversely associated with obesity in non-diabetic individuals. Sci Rep 2015; 5:17438. [PMID: 26621340 PMCID: PMC4664929 DOI: 10.1038/srep17438] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/29/2015] [Indexed: 12/11/2022] Open
Abstract
Cardiotrophin-1 is known to be a key regulator of energy homeostasis, as well as glucose and lipid metabolism in vivo. However, there are inconsistent results of the association between cardiotrophin-1 and obesity in humans, possibly confounded by hyperglycemia. Therefore, the aim of this study was to investigate the relationships among cardiotrophin-1 levels, overweight and obese individuals without diabetes in a Chinese population. The median (inter-quarter range) serum cardiotrophin-1 levels were 447.9 (230.9, 913.9), 350.6 (201.1, 666.5), and 288.1 (162.3, 572.4) pg/ml in non-diabetic subjects who were of normal weight (n = 522), overweight (n = 203), and obese (n = 93), respectively (trend test p < 0.001). Subjects who were overweight and obese had significantly lower cardiotrophin-1 levels than those with normal weight. The multivariate linear regression analyses showed that overweight (beta = −338.718, 95% CI = −552.786 ~ −124.651, p < 0.01), obese (beta = −530.275, 95% CI = −832.967 ~ −227.583, p < 0.01), and smoking (beta = −377.375, 95% CI = −654.353 ~ −100.397, p < 0.01) were negatively related to cardiotrophin-1 after adjusting for age, gender, HOMA-IR, hypertension, total cholesterol, HDL, triglyceride, eGFR, ALT, and alcohol drinking. The results of this study provided epidemiological evidence that non-diabetic subjects who were overweight or obesity had significantly lower cardiotrophin-1 concentrations than those with normal weight, and both obesity and being overweight were inversely associated with cardiotrophin-1 levels.
Collapse
|
39
|
Maiese K. Erythropoietin and diabetes mellitus. World J Diabetes 2015; 6:1259-1273. [PMID: 26516410 PMCID: PMC4620106 DOI: 10.4239/wjd.v6.i14.1259] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/25/2015] [Accepted: 09/28/2015] [Indexed: 02/05/2023] Open
Abstract
Erythropoietin (EPO) is a 30.4 kDa growth factor and cytokine that governs cell proliferation, immune modulation, metabolic homeostasis, vascular function, and cytoprotection. EPO is under investigation for the treatment of variety of diseases, but appears especially suited for the treatment of disorders of metabolism that include diabetes mellitus (DM). DM and the complications of this disease impact a significant portion of the global population leading to disability and death with currently limited therapeutic options. In addition to its utility for the treatment of anemia, EPO can improve cardiac function, reduce fatigue, and improve cognition in patients with DM as well as regulate cellular energy metabolism, obesity, tissue repair and regeneration, apoptosis, and autophagy in experimental models of DM. Yet, EPO can have adverse effects that involve the vasculature system and unchecked cellular proliferation. Critical to the cytoprotective capacity and the potential for a positive clinical outcome with EPO are the control of signal transduction pathways that include protein kinase B, the mechanistic target of rapamycin, Wnt signaling, mammalian forkhead transcription factors of the O class, silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae), and AMP activated protein kinase. Therapeutic strategies that can specifically target and control EPO and its signaling pathways hold great promise for the development of new and effective clinical treatments for DM and the complications of this disorder.
Collapse
|
40
|
López-Yoldi M, Moreno-Aliaga MJ, Bustos M. Cardiotrophin-1: A multifaceted cytokine. Cytokine Growth Factor Rev 2015; 26:523-32. [PMID: 26188636 DOI: 10.1016/j.cytogfr.2015.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
Abstract
Cardiotrophin-1 (CT-1) is a member of the gp130 family of cytokines that have pleiotropic functions on different tissues and cell types. Although many effects of CT-1 have been described on the heart, there is an extensive research showing important protective effects in other organs such as liver, kidney or nervous system. Recently, several studies have pointed out that CT-1 might also play a key role in the regulation of body weight and intermediate metabolism. This paper will review many aspects of CT-1 physiological role in several organs and discuss data for consideration in therapeutic approaches.
Collapse
Affiliation(s)
- Miguel López-Yoldi
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, University of Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Matilde Bustos
- Area of Hepatology and Gene Therapy, CIMA (Center for Applied Medical Research) University of Navarra, Pamplona, Spain.
| |
Collapse
|
41
|
Li YH, Yang LH, Sha KH, Liu TG, Zhang LG, Liu XX. Efficacy of poly-unsaturated fatty acid therapy on patients with nonalcoholic steatohepatitis. World J Gastroenterol 2015; 21:7008-7013. [PMID: 26078579 PMCID: PMC4462743 DOI: 10.3748/wjg.v21.i22.7008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 02/20/2015] [Accepted: 04/09/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To examine whether poly-unsaturated fatty acid (PUFA) therapy is beneficial for improving nonalcoholic steatohepatitis (NASH).
METHODS: In total, 78 patients pathologically diagnosed with NASH were enrolled and were randomly assigned into the control group and the PUFA therapy group (added 50 mL PUFA with 1:1 ratio of EHA and DHA into daily diet). At the initial analysis and after 6 mo of PUFA therapy, parameters of interest including liver enzymes, lipid profiles, markers of inflammation and oxidation, and histological changes were evaluated and compared between these two groups.
RESULTS: At the initial analysis, in patients with NASH, serum levels of alanine aminotransferase (ALT) and aspartase aminotransferase (AST) were slightly elevated. Triglyceride (TG), total cholesterol (TC) and low-density lipoprotein cholesterol levels, markers of systemic inflammation [C-reactive protein (CRP)] and oxidation [malondialdehyde (MDA)], as well as fibrosis parameters of type IV collagen and pro-collagen type III pro-peptide were also increased beyond the normal range. Six months later, ALT and AST levels were significantly reduced in the PUFA group compared with the control group. In addition, serum levels of TG and TC, CRP and MDA, and type IV collagen and pro-collagen type III pro-peptide were also simultaneously and significantly reduced. Of note, histological evaluation showed that steatosis grade, necro-inflammatory grade, fibrosis stage, and ballooning score were all profoundly improved in comparison to the control group, strongly suggesting that increased PUFA consumption was a potential way to offset NASH progression.
CONCLUSION: Increased PUFA consumption is a potential promising approach for NASH prevention and reversal.
Collapse
|
42
|
Hua X, Shan Y, Li D, Xu D, Zhang J, Yang T, Han L, Shen C, Xia Y, Chen Q, Ma X, Zhang J, Xia Q. A Potential Profibrogenic Role of Biliary Epithelium-Derived Cardiotrophin-1 in Pediatric Cholestatic Liver Disease. J Interferon Cytokine Res 2015; 35:606-12. [PMID: 25919795 DOI: 10.1089/jir.2014.0128] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
As a cytokine of the interleukin-6 family, cardiotrophin-1 (CT-1) has been shown to be an important endogenous protector in liver injury. Our study aimed to investigate the role of CT-1 in liver fibrosis in pediatric cholestatic liver disease (PCLD). CT-1 mRNA and protein expression levels were upregulated in PCLD liver biopsy tissues compared with controls. Immunohistochemistry and confocal microscopy of liver sections showed that CT-1 was predominantly expressed by biliary epithelium cells. Serum CT-1 was elevated significantly in the children with PCLD compared with controls. Serum CT-1 levels exhibited a moderate positive correlation with the Scheuer stage of hepatic fibrosis and serum TB levels and a weak correlation with serum ALP levels. In vitro analysis indicated that LX-2 cells preconditioned with CT-1 exhibited significant increments in proliferation and accumulation of extracellular matrix components, while also positively regulating the STAT3 and p38MAPK pathways. In conclusion, biliary epithelium-derived CT-1 may exert a profibrogenic potential in PCLD.
Collapse
Affiliation(s)
- Xiangwei Hua
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Yuhua Shan
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Dawei Li
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Dongwei Xu
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Jiang Zhang
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Taihua Yang
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Longzhi Han
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Conghuan Shen
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Yun Xia
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Qimin Chen
- 2 Department of Urology, Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai, People's Republic of China
| | - Xiong Ma
- 3 Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai Institute of Digestive Disease, Shanghai, People's Republic of China .,4 Key Laboratory of Gastroenterology & Hepatology, Ministry of Health (Shanghai Jiao-Tong University) , Shanghai, People's Republic of China
| | - Jianjun Zhang
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| | - Qiang Xia
- 1 Department of Liver Surgery and Liver Transplantation Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, People's Republic of China
| |
Collapse
|
43
|
Arias N, Macarulla MT, Aguirre L, Miranda J, Portillo MP. Liver delipidating effect of a combination of resveratrol and quercetin in rats fed an obesogenic diet. J Physiol Biochem 2015; 71:569-76. [PMID: 25827944 DOI: 10.1007/s13105-015-0403-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/18/2015] [Indexed: 01/14/2023]
Abstract
Liver steatosis is characterized by an abnormal accumulation of triacylglycerols in this organ. This metabolic disorder is closely associated with obesity. In the present study, we aimed to analyse the effect of a combination of resveratrol and quercetin on liver steatosis in an animal model of dietetic obesity, and to compare it with one induced by the administration of each polyphenol separately. Rats were divided into four dietary groups of nine animals each and fed a high-fat, high-sucrose diet: an untreated control group and three groups treated either with resveratrol (RSV; 15 mg/kg/day), with quercetin (Q; 30 mg/kg/day), or with both (RSV + Q; 15 mg resveratrol/kg/day and 30 mg quercetin/kg/day) for 6 weeks. Liver weight and triacylglycerol content decreased only in the RSV + Q group. A significant reduction in acetyl-CoA carboxylase activity was observed in RSV and RSV + Q groups, without changes in fatty acid synthase activity. A significant increase in carnitine palmitoyltransferase-1a activity was observed only in rats treated with the combination of resveratrol and quercetin, suggesting increased fatty acid oxidation. Citrate synthase, a marker of mitochondrial density, remained unchanged in all groups. No significant changes were observed in the expression of peroxisome proliferator-activated receptor α (PPARα), nuclear respiratory factor 1 (NRF-1) and transcription factor A mitochondrial (TFAM). In conclusion, resveratrol and quercetin together, combining two doses which were shown to be ineffective singly, is an interesting tool to prevent liver steatosis associated with high-fat high-sucrose feeding. The delipidating effect seems to be mediated by increased fatty acid oxidation not associated with increased mitochondriogenesis, and by reduced de novo lipogenesis.
Collapse
Affiliation(s)
- Noemí Arias
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Faculty of Pharmacy and Lucio Lascaray Research Center, University of the Basque Country (UPV/EHU), Vitoria, Spain
| | | | | | | | | |
Collapse
|
44
|
Cardiotrophin-1 (CT-1) Improves High Fat Diet-Induced Cognitive Deficits in Mice. Neurochem Res 2015; 40:843-53. [DOI: 10.1007/s11064-015-1535-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 01/29/2015] [Accepted: 02/04/2015] [Indexed: 01/12/2023]
|
45
|
Maiese K. FoxO Transcription Factors and Regenerative Pathways in Diabetes Mellitus. Curr Neurovasc Res 2015; 12:404-13. [PMID: 26256004 PMCID: PMC4567483 DOI: 10.2174/1567202612666150807112524] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 02/07/2023]
Abstract
Mammalian forkhead transcription factors of the O class (FoxO) are exciting targets under consideration for the development of new clinical entities to treat metabolic disorders and diabetes mellitus (DM). DM, a disorder that currently affects greater than 350 million individuals globally, can become a devastating disease that leads to cellular injury through oxidative stress pathways and affects multiple systems of the body. FoxO proteins can regulate insulin signaling, gluconeogenesis, insulin resistance, immune cell migration, and cell senescence. FoxO proteins also control cell fate through oxidative stress and pathways of autophagy and apoptosis that either lead to tissue regeneration or cell demise. Furthermore, FoxO signaling can be dependent upon signal transduction pathways that include silent mating type information regulation 2 homolog 1 (S. cerevisiae) (SIRT1), Wnt, and Wnt1 inducible signaling pathway protein 1 (WISP1). Cellular metabolic pathways driven by FoxO proteins are complex, can lead to variable clinical outcomes, and require in-depth analysis of the epigenetic and post-translation protein modifications that drive FoxO protein activation and degradation.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101, USA.
| |
Collapse
|
46
|
Maiese K. Programming apoptosis and autophagy with novel approaches for diabetes mellitus. Curr Neurovasc Res 2015; 12:173-88. [PMID: 25742566 PMCID: PMC4380829 DOI: 10.2174/1567202612666150305110929] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/16/2015] [Accepted: 02/19/2015] [Indexed: 12/13/2022]
Abstract
According to the World Health Organization, diabetes mellitus (DM) in the year 2030 will be ranked the seventh leading cause of death in the world. DM impacts all systems of the body with oxidant stress controlling cell fate through endoplasmic reticulum stress, mitochondrial dysfunction, alterations in uncoupling proteins, and the induction of apoptosis and autophagy. Multiple treatment approaches are being entertained for DM with Wnt1 inducible signaling pathway protein 1 (WISP1), mechanistic target of rapamycin (mTOR), and silent mating type information regulation 2 homolog) 1 (S. cerevisiae) (SIRT1) generating significant interest as target pathways that can address maintenance of glucose homeostasis as well as prevention of cellular pathology by controlling insulin resistance, stem cell proliferation, and the programmed cell death pathways of apoptosis and autophagy. WISP1, mTOR, and SIRT1 can rely upon similar pathways such as AMP activated protein kinase as well as govern cellular metabolism through cytokines such as EPO and oral hypoglycemics such as metformin. Yet, these pathways require precise biological control to exclude potentially detrimental clinical outcomes. Further elucidation of the ability to translate the roles of WISP1, mTOR, and SIRT1 into effective clinical avenues offers compelling prospects for new therapies against DM that can benefit hundreds of millions of individuals throughout the globe.
Collapse
Affiliation(s)
- Kenneth Maiese
- MD, Cellular and Molecular Signaling, Newark, New Jersey 07101, USA.
| |
Collapse
|
47
|
Hematopoietic tissue factor-protease-activated receptor 2 signaling promotes hepatic inflammation and contributes to pathways of gluconeogenesis and steatosis in obese mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:524-35. [PMID: 25476527 DOI: 10.1016/j.ajpath.2014.10.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 10/08/2014] [Accepted: 10/14/2014] [Indexed: 12/19/2022]
Abstract
Failure to inhibit hepatic gluconeogenesis is a major mechanism contributing to fasting hyperglycemia in type 2 diabetes and, along with steatosis, is the hallmark of hepatic insulin resistance. Obesity is associated with chronic inflammation in multiple tissues, and hepatic inflammation is mechanistically linked to both steatosis and hepatic insulin resistance. Here, we delineate a role for coagulation signaling via tissue factor (TF) and proteinase-activated receptor 2 (PAR2) in obesity-mediated hepatic inflammation, steatosis, and gluconeogenesis. In diet-induced obese mice, TF tail signaling independent of PAR2 drives CD11b(+)CD11c(+) hepatic macrophage recruitment, and TF-PAR2 signaling contributes to the accumulation of hepatic CD8(+) T cells. Transcripts of key pathways of gluconeogenesis, lipogenesis, and inflammatory cytokines were reduced in high-fat diet-fed mice that lack the cytoplasmic domain of TF (F3) (TF(ΔCT)) or that are deficient in PAR2 (F2rl1), as well as by pharmacological inhibition of TF-PAR2 signaling in diet-induced obese mice. These gluconeogenic, lipogenic, and inflammatory pathway transcripts were similarly reduced in response to genetic ablation or pharmacological inhibition of TF-PAR2 signaling in hematopoietic cells and were mechanistically associated with activation of AMP-activated protein kinase (AMPK). These findings indicate that hematopoietic TF-PAR2 signaling plays a pivotal role in the hepatic inflammatory responses, steatosis, and hepatic insulin resistance that lead to systemic insulin resistance and type 2 diabetes in obesity.
Collapse
|
48
|
López-Yoldi M, Fernández-Galilea M, Laiglesia LM, Larequi E, Prieto J, Martínez JA, Bustos M, Moreno-Aliaga MJ. Cardiotrophin-1 stimulates lipolysis through the regulation of main adipose tissue lipases. J Lipid Res 2014; 55:2634-43. [PMID: 25351614 DOI: 10.1194/jlr.m055335] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiotrophin-1 (CT-1) is a cytokine with antiobesity properties and with a role in lipid metabolism regulation and adipose tissue function. The aim of this study was to analyze the molecular mechanisms involved in the lipolytic actions of CT-1 in adipocytes. Recombinant CT-1 (rCT-1) effects on the main proteins and signaling pathways involved in the regulation of lipolysis were evaluated in 3T3-L1 adipocytes and in mice. rCT-1 treatment stimulated basal glycerol release in a concentration- and time-dependent manner in 3T3-L1 adipocytes. rCT-1 (20 ng/ml for 24 h) raised cAMP levels, and in parallel increased protein kinase (PK)A-mediated phosphorylation of perilipin and hormone sensitive lipase (HSL) at Ser660. siRNA knock-down of HSL or PKA, as well as pretreatment with the PKA inhibitor H89, blunted the CT-1-induced lipolysis, suggesting that the lipolytic action of CT-1 in adipocytes is mainly mediated by activation of HSL through the PKA pathway. In ob/ob mice, acute rCT-1 treatment also promoted PKA-mediated phosphorylation of perilipin and HSL at Ser660 and Ser563, and increased adipose triglyceride lipase (desnutrin) content in adipose tissue. These results showed that the ability of CT-1 to regulate the activity of the main lipases underlies the lipolytic action of this cytokine in vitro and in vivo, and could contribute to CT-1 antiobesity effects.
Collapse
Affiliation(s)
- Miguel López-Yoldi
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain
| | - Marta Fernández-Galilea
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain
| | - Laura M Laiglesia
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain
| | - Eduardo Larequi
- Gene Therapy and Hepatology, CIMA, University of Navarra, Pamplona, Navarra, Spain
| | - Jesús Prieto
- Gene Therapy and Hepatology, CIMA, University of Navarra, Pamplona, Navarra, Spain CIBERehd Institute of Health Carlos III, Madrid, Spain
| | - J Alfredo Martínez
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain CIBERobn, Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Matilde Bustos
- Gene Therapy and Hepatology, CIMA, University of Navarra, Pamplona, Navarra, Spain
| | - Maria J Moreno-Aliaga
- Departments of Nutrition, Food Science, and Physiology University of Navarra, Pamplona, Navarra, Spain Centre for Nutrition Research, University of Navarra, Pamplona, Navarra, Spain CIBERobn, Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
49
|
Nonalcoholic Fatty liver disease: pathogenesis and therapeutics from a mitochondria-centric perspective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:637027. [PMID: 25371775 PMCID: PMC4211163 DOI: 10.1155/2014/637027] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/31/2014] [Accepted: 07/31/2014] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) describes a spectrum of disorders characterized by the accumulation of triglycerides within the liver. The global prevalence of NAFLD has been increasing as the obesity epidemic shows no sign of relenting. Mitochondria play a central role in hepatic lipid metabolism and also are affected by upstream signaling pathways involved in hepatic metabolism. This review will focus on the role of mitochondria in the pathophysiology of NAFLD and touch on some of the therapeutic approaches targeting mitochondria as well as metabolically important signaling pathways. Mitochondria are able to adapt to lipid accumulation in hepatocytes by increasing rates of beta-oxidation; however increased substrate delivery to the mitochondrial electron transport chain (ETC) leads to increased reactive oxygen species (ROS) production and eventually ETC dysfunction. Decreased ETC function combined with increased rates of fatty acid beta-oxidation leads to the accumulation of incomplete products of beta-oxidation, which combined with increased levels of ROS contribute to insulin resistance. Several related signaling pathways, nuclear receptors, and transcription factors also regulate hepatic lipid metabolism, many of which are redox sensitive and regulated by ROS.
Collapse
|
50
|
Guo P, Pi H, Xu S, Zhang L, Li Y, Li M, Cao Z, Tian L, Xie J, Li R, He M, Lu Y, Liu C, Duan W, Yu Z, Zhou Z. Melatonin Improves mitochondrial function by promoting MT1/SIRT1/PGC-1 alpha-dependent mitochondrial biogenesis in cadmium-induced hepatotoxicity in vitro. Toxicol Sci 2014; 142:182-95. [PMID: 25159133 PMCID: PMC4226765 DOI: 10.1093/toxsci/kfu164] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Melatonin is an indolamine synthesized in the pineal gland that has a wide range of physiological functions, and it has been under clinical investigation for expanded applications. Increasing evidence demonstrates that melatonin can ameliorate cadmium-induced hepatotoxicity. However, the potentially protective effects of melatonin against cadmium-induced hepatotoxicity and the underlying mechanisms of this protection remain unclear. This study investigates the protective effects of melatonin pretreatment on cadmium-induced hepatotoxicity and elucidates the potential mechanism of melatonin-mediated protection. We exposed HepG2 cells to different concentrations of cadmium chloride (2.5, 5, and 10μM) for 12 h. We found that Cd stimulated cytotoxicity, disrupted the mitochondrial membrane potential, increased reactive oxygen species production, and decreased mitochondrial mass and mitochondrial DNA content. Consistent with this finding, Cd exposure was associated with decreased Sirtuin 1 (SIRT1) protein expression and activity, thus promoted acetylation of PGC-1 alpha, a key enzyme involved in mitochondrial biogenesis and function, although Cd did not disrupt the interaction between SIRT1 and PGC-1 alpha. However, all cadmium-induced mitochondrial oxidative injuries were efficiently attenuated by melatonin pretreatment. Moreover, Sirtinol and SIRT1 siRNA each blocked the melatonin-mediated elevation in mitochondrial function by inhibiting SIRT1/ PGC-1 alpha signaling. Luzindole, a melatonin receptor antagonist, was found to partially block the ability of melatonin to promote SIRT1/ PGC-1 alpha signaling. In summary, our results indicate that SIRT1 plays an essential role in the ability of moderate melatonin to stimulate PGC-1 alpha and improve mitochondrial biogenesis and function at least partially through melatonin receptors in cadmium-induced hepatotoxicity.
Collapse
Affiliation(s)
- Pan Guo
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Huifeng Pi
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Shangcheng Xu
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Lei Zhang
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Yuming Li
- Institute of Hepatobiliary Surgery, XinQiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Min Li
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Zhengwang Cao
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Li Tian
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Jia Xie
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Renyan Li
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Mindi He
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Yonghui Lu
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Chuan Liu
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Weixia Duan
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Zhengping Yu
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Zhou Zhou
- Department of occupational health, Third Military Medical University, Chongqing 400038, People's Republic of China
| |
Collapse
|