1
|
Wang W, Xu Y, Zhou J, Zang Y. Effects of omega-3 supplementation on lipid metabolism, inflammation, and disease activity in rheumatoid arthritis: a meta-analysis of randomized controlled trials. Clin Rheumatol 2024; 43:2479-2488. [PMID: 38922552 DOI: 10.1007/s10067-024-07040-0] [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: 03/04/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION Omega-3 possesses anti-inflammatory and lipid metabolism modifying effects in rheumatoid arthritis (RA), but inconsistency exists among previous studies. This meta-analysis intended to explore the effects of omega-3 supplementation on fatty acid distribution, blood lipid profiles, inflammation, and disease activity in RA patients. METHODS This meta-analysis followed the Preferred Reporting Item for Systematic Reviews and Meta-Analyses (PRISMA) protocol. PubMed, Web of Science, and Embase databases were searched until August 31, 2023. RESULTS Eighteen randomized controlled trials with 1018 RA patients were included. Regarding fatty acid distribution, omega-3 supplementation increased eicosapentaenoic acid (EPA) [standardized mean difference (SMD): 0.74; 95% confidence interval (CI): 0.46, 1.01; P < 0.001] and docosahexanoic acid (DHA) (SMD: 0.62; 95% CI: 0.35, 0.89; P < 0.001), but reduced omega-6:omega-3 ratio (SMD: -1.06; 95% CI: -1.39, -0.73; P < 0.001) in RA patients. Regarding blood lipid, omega-3 supplementation decreased triglyceride (TG) in RA patients (SMD: -0.47; 95% CI: -0.78, -0.16; P = 0.003). Regarding clinical symptoms, omega-3 supplementation reduced tender joint count (TJC) in RA patients (SMD: -0.59; 95% CI: -0.79, -0.39; P < 0.001). Notably, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), and disease activity score on 28 joints (DAS28) score were slightly decreased by omega-3 supplementation but without statistical significance (all P > 0.05). Publication bias was low, and stability assessed by sensitivity analysis was good. CONCLUSION Omega-3 supplementation increases EPA and DHA, but reduces the omega-6:omega-3 ratio, TG, and TJC in RA patients.
Collapse
Affiliation(s)
- Wen Wang
- Department of Rheumatology & Immunology, The Suqian Clinical College of Xuzhou Medical University, Suqian, 223800, China
- Department of Rheumatology & Immunology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, China
| | - Yan Xu
- Department of Rheumatology & Immunology, The Suqian Clinical College of Xuzhou Medical University, Suqian, 223800, China
- Department of Rheumatology & Immunology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, China
| | - Jun Zhou
- Department of Rheumatology & Immunology, The Suqian Clinical College of Xuzhou Medical University, Suqian, 223800, China
| | - Yinshan Zang
- Department of Rheumatology & Immunology, The Suqian Clinical College of Xuzhou Medical University, Suqian, 223800, China.
- Department of Rheumatology & Immunology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, China.
| |
Collapse
|
2
|
Youn I, Han S, Jung HJ, Noh SG, Chung HY, Koo YK, Shin S, Seo EK. Anti-Inflammatory Activity of the Constituents from the Leaves of Perilla frutescens var. acuta. Pharmaceuticals (Basel) 2023; 16:1655. [PMID: 38139782 PMCID: PMC10747482 DOI: 10.3390/ph16121655] [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: 10/30/2023] [Revised: 11/18/2023] [Accepted: 11/26/2023] [Indexed: 12/24/2023] Open
Abstract
Perilla frutense var. acuta (Lamiaceae) has been used to treat indigestion, asthma, and allergies in traditional medicine. In this study, luteolin 7-O-diglucuronide (1), apigenin 7-O-diglucuronide (2), and rosmarinic acid (3) were isolated from the leaves of P. frutescens var. acuta through various chromatographic purification techniques. Several approaches were used to investigate the anti-inflammatory activity of the constituents (1-3) and their working mechanisms. In silico docking simulation demonstrated that 1-3 would work as a PPAR-α/δ/γ agonist, and in vitro PPAR-α/δ/γ transcriptional assay showed that the Perilla water extract (PWE) and 3 increased PPAR-α luciferase activity (1.71 and 1.61 times of the control (PPAR-α + PPRE, p < 0.001)). In the NF-κB luciferase assay, 1 suppressed NF-κB activity the most (56.83% at 5 µM; 74.96% at 10 µM; 79.86% at 50 µM). In addition, 1 and 2 inhibited the mRNA expression of NF-κB target genes, including Il6, Mcp1, and Tnfa, at 50 µM, and 3 suppressed the genes at the mRNA level in a dose-dependent manner. We report that 1 and 2 exert anti-inflammatory effects through NF-κB inhibition, and the PPAR-α/NF-κB signaling pathway is related to the anti-inflammatory activity of 3.
Collapse
Affiliation(s)
- Isoo Youn
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (I.Y.); (S.H.)
| | - Sujin Han
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (I.Y.); (S.H.)
| | - Hee Jin Jung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea; (H.J.J.); (S.G.N.); (H.Y.C.)
| | - Sang Gyun Noh
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea; (H.J.J.); (S.G.N.); (H.Y.C.)
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea; (H.J.J.); (S.G.N.); (H.Y.C.)
| | - Yean Kyoung Koo
- Department of R&I Center, COSMAXBIO, Seongnam 13487, Republic of Korea;
| | - Sunhye Shin
- Major of Food and Nutrition, Division of Applied Food System, Seoul Women’s University, Seoul 01797, Republic of Korea
| | - Eun Kyoung Seo
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (I.Y.); (S.H.)
| |
Collapse
|
3
|
Peng Z, Chen L, Wang M, Yue X, Wei H, Xu F, Hou W, Li Y. SREBP inhibitors: an updated patent review for 2008-present. Expert Opin Ther Pat 2023; 33:669-680. [PMID: 38054657 DOI: 10.1080/13543776.2023.2291393] [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: 07/18/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
INTRODUCTION Sterol regulatory element-binding proteins (SREBPs) are a family of membrane-binding transcription factors that activate genes encoding enzymes required for cholesterol and unsaturated fatty acid synthesis. Overactivation of SREBP is related to the occurrence and development of diabetes, nonalcoholic fatty liver, tumor, and other diseases. In the past period, many SREBP inhibitors have been found. AREAS COVERED This manuscript is a patent review of SREBP inhibitors. We searched 2008 to date for all data from the US patent database (https://www.uspto.gov/) and the European patent database (https://www.epo.org/) with 'SREBP' and 'inhibitor' as keywords and analyzed the search results. EXPERT OPINION Both synthetic and natural SREBP inhibitors have been reported. Despite the lack of cocrystal structure of SREBP inhibitor, the mechanisms of several compounds have been clarified. Importantly, some SREBP inhibitors have been proved to have good activity in preclinical studies. As the characteristics of lipid metabolism reprogramming in cardio-cerebrovascular diseases and tumors are gradually revealed, more and more attention will be focused on SREBP.
Collapse
Affiliation(s)
- Zhenyu Peng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Leyuan Chen
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Manjiang Wang
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Xufan Yue
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huiqiang Wei
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Feifei Xu
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Wenbin Hou
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Yiliang Li
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| |
Collapse
|
4
|
Gunasekar SK, Heebink J, Carpenter DH, Kumar A, Xie L, Zhang H, Schilling JD, Sah R. Adipose-targeted SWELL1 deletion exacerbates obesity- and age-related nonalcoholic fatty liver disease. JCI Insight 2023; 8:e154940. [PMID: 36749637 PMCID: PMC10077479 DOI: 10.1172/jci.insight.154940] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/27/2023] [Indexed: 02/08/2023] Open
Abstract
Healthy expansion of adipose tissue is critical for the maintenance of metabolic health, providing an optimized reservoir for energy storage in the form of triacylglycerol-rich lipoproteins. Dysfunctional adipocytes that are unable to efficiently store lipid can result in lipodystrophy and contribute to nonalcoholic fatty liver disease (NAFLD) and metabolic syndrome. Leucine-rich repeat containing protein 8a/SWELL1 functionally encodes the volume-regulated anion channel complex in adipocytes, is induced in early obesity, and is required for normal adipocyte expansion during high-fat feeding. Adipose-specific SWELL1 ablation (Adipo KO) leads to insulin resistance and hyperglycemia during caloric excess, both of which are associated with NAFLD. Here, we show that Adipo-KO mice exhibited impaired adipose depot expansion and excess lipolysis when raised on a variety of high-fat diets, resulting in increased diacylglycerides and hepatic steatosis, thereby driving liver injury. Liver lipidomic analysis revealed increases in oleic acid-containing hepatic triacylglycerides and injurious hepatic diacylglyceride species, with reductions in hepatocyte-protective phospholipids and antiinflammatory free fatty acids. Aged Adipo-KO mice developed hepatic steatosis on a regular chow diet, and Adipo-KO male mice developed spontaneous, aggressive hepatocellular carcinomas (HCCs). These data highlight the importance of adipocyte SWELL1 for healthy adipocyte expansion to protect against NAFLD and HCC in the setting of overnutrition and with aging.
Collapse
Affiliation(s)
- Susheel K. Gunasekar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John Heebink
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Danielle H. Carpenter
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Ashutosh Kumar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Litao Xie
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Haixia Zhang
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joel D. Schilling
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| |
Collapse
|
5
|
Calder PC. Omega-3 fatty acids and metabolic partitioning of fatty acids within the liver in the context of nonalcoholic fatty liver disease. Curr Opin Clin Nutr Metab Care 2022; 25:248-255. [PMID: 35762160 DOI: 10.1097/mco.0000000000000845] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Nonalcoholic fatty liver disease (NAFLD) is now the most prevalent form of liver disease globally, affecting about 25% of the world's adult population. It is more common in those living with obesity, where it may affect as many as 80% of individuals. The aim of this article is to describe recent human studies evaluating the influence of omega-3 fatty acids on de novo lipogenesis (DNL) and hepatic fatty acid partitioning between incorporation into triacylglycerols (TAGs) and β-oxidation, to discuss the relevance of these effects in the context of NAFLD, and to provide an overview of the mechanisms that might be involved. RECENT FINDINGS The omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) decrease hepatic DNL and partition fatty acids away from TAG synthesis and toward β-oxidation. EPA and DHA affect multiple hepatic transcription factors resulting in down-regulation of the DNL pathway and upregulation of β-oxidation. The net result is decreased accumulation of hepatic TAG and lowering of circulating TAG concentrations. Human trials demonstrate that EPA and DHA can decrease liver fat in patients with NAFLD. SUMMARY Increased intake of EPA and DHA may reduce the likelihood of hepatic TAG accumulation and could be used to reduce liver fat in patients with NAFLD.
Collapse
Affiliation(s)
- Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| |
Collapse
|
6
|
Quispe R, Alfaddagh A, Kazzi B, Zghyer F, Marvel FA, Blumenthal RS, Sharma G, Martin SS. Controversies in the Use of Omega-3 Fatty Acids to Prevent Atherosclerosis. Curr Atheroscler Rep 2022; 24:571-581. [PMID: 35499805 DOI: 10.1007/s11883-022-01031-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW We discuss current controversies in the clinical use of omega-3 fatty acids (FA), primarily eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), and examine discrepancies between recent trials. Furthermore, we discuss potential side effects reported in these studies and the role of mixed omega-3 FA dietary supplements and concerns about their use. RECENT FINDINGS REDUCE-IT showed that addition of icosapent ethyl, a highly purified form of EPA, can reduce risk of cardiovascular events among statin-treated individuals with high triglycerides. Additional supportive evidence for EPA has come from other trials and meta-analyses of omega-3 FA therapy. In contrast, trials of mixed EPA/DHA products have consistently failed to improve cardiovascular outcomes. Discrepancies in results reported in RCTs could be explained by differences in omega-3 FA products, dosing, study populations, and study designs including the placebo control formulation. Evidence obtained from highly purified forms should not be extrapolated to other mixed formulations, including "over-the-counter" omega-3 supplements. Targeting TG-rich lipoproteins represents a new frontier for mitigating ASCVD risk. Clinical and basic research evidence suggests that the use of omega-3 FA, specifically EPA, appears to slow atherosclerosis by reducing triglyceride-rich lipoproteins and/or inflammation, therefore addressing residual risk of clinical ASCVD.
Collapse
Affiliation(s)
- Renato Quispe
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Abdulhamied Alfaddagh
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Brigitte Kazzi
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Fawzi Zghyer
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Francoise A Marvel
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Roger S Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Garima Sharma
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA
| | - Seth S Martin
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Carnegie 591, Baltimore, MD, 21287, USA.
| |
Collapse
|
7
|
Omega-3 Fatty Acids and Their Interaction with the Gut Microbiome in the Prevention and Amelioration of Type-2 Diabetes. Nutrients 2022; 14:nu14091723. [PMID: 35565691 PMCID: PMC9104474 DOI: 10.3390/nu14091723] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 01/27/2023] Open
Abstract
Type-2 diabetes mellitus (T2DM) is often linked with hyperglycemia, disturbed lipid profiles, inflammation, and gut dysbiosis. Omega-3 fatty acid supplementation has a vital role in the management of T2DM. As a result, a better understanding of the potential role of omega-3 fatty acids in the development and progression of T2DM by influencing the intestinal microflora will help to improve the therapeutic intervention for T2DM and related complications. Focusing on the molecular mechanisms and signaling pathways induced by omega-3 fatty acids, this paper attempts to comprehensively review and discuss the putative associations between omega-3 fatty acids, gut dysbiosis, and the pathophysiology of T2DM and its related comorbidities. In addition, we contemplate the importance of gut microbiota in T2DM prevention and treatment and ponder the role of omega-3 fatty acids in T2DM by positively modulating gut microbiota, which may lead to discovery of novel targets and therapeutic strategies thereby paving way for further comprehensive, mechanistic, and clinical studies.
Collapse
|
8
|
Tzanaki I, Agouridis AP, Kostapanos MS. Is there a role of lipid-lowering therapies in the management of fatty liver disease? World J Hepatol 2022; 14:119-139. [PMID: 35126843 PMCID: PMC8790403 DOI: 10.4254/wjh.v14.i1.119] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/30/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Atherogenic dyslipidemia is characterized by increased triglyceride-rich lipoproteins and low high-density lipoprotein cholesterol concentrations. It is highly prevalent in non-alcoholic fatty liver disease (NAFLD) and contributes to the increased cardiovascular risk associated with this condition. Alongside insulin resistance it plays an important pathogenetic role in NAFLD/non-alcoholic steatohepatitis (NASH) development and progression. It has been shown that cholesterol-lowering reduces cardiovascular risk more in NAFLD vs non-NAFLD high-risk individuals. This evidence highlights the importance of effective lipid modulation in NAFLD. In this narrative review the effects of the most commonly used lipid-lowering therapies on liver outcomes alongside their therapeutic implications in NAFLD/NASH are critically discussed. Preclinical and clinical evidence suggests that statins reduce hepatic steatosis, inflammation and fibrosis in patients with NAFLD/NASH. Most data are derived from observational and small prospective clinical studies using changes in liver enzyme activities, steatosis/fibrosis scores, and imaging evidence of steatosis as surrogates. Also, relevant histologic benefits were noted in small biopsy studies. Atorvastatin and rosuvastatin showed greater benefits, whereas data for other statins are scarce and sometimes conflicting. Similar studies to those of statins showed efficacy of ezetimibe against hepatic steatosis. However, no significant anti-inflammatory and anti-fibrotic actions of ezetimibe have been shown. Preclinical studies showed that fibrates through peroxisome proliferator-activated receptor (PPAR)α activation may have a role in NAFLD prevention and management. Nevertheless, no relevant benefits have been noted in human studies. Species-related differences in PPARα expression and its activation responsiveness may help explain this discrepancy. Omega-3 fatty acids reduced hepatic steatosis in numerous heterogeneous studies, but their benefits on hepatic inflammation and fibrosis have not been established. Promising preliminary data for the highly purified eicosapentaenoic acid require further confirmation. Observational studies suggest that proprotein convertase subtilisin/kexin9 inhibitors may also have a role in the management of NAFLD, though this needs to be established by future prospective studies.
Collapse
Affiliation(s)
- Ismini Tzanaki
- School of Medicine, European University Cyprus, Nicosia, Cyprus, Nicosia 2404, Cyprus
| | - Aris P Agouridis
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus
| | - Michael S Kostapanos
- General Medicine, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge CB20QQ, United Kingdom
| |
Collapse
|
9
|
Khorshidi M, Sayyari A, Olang B, Alaei MR, Shab-Bidar S, Khalili M, Salehi M, Aryaeian N. Study protocol of a randomized controlled clinical trial investigating the effects of omega-3 supplementation on endothelial function, vascular structure, and metabolic parameters in adolescents with type 1 diabetes. Trials 2021; 22:953. [PMID: 34961564 PMCID: PMC8710929 DOI: 10.1186/s13063-021-05930-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 12/09/2021] [Indexed: 01/09/2023] Open
Abstract
Background Type 1 diabetes is a main health burden with several related comorbidities. It has been shown that endothelial function, vascular structure, and metabolic parameters are considerably disrupted in patients with type 1 diabetes. Omega-3 as an adjuvant therapy may exert profitable effects on type 1 diabetes and its complications by improving inflammation, oxidative stress, immune responses, and metabolic status. Because no randomized clinical trial has examined the effects of omega-3 consumption in children and adolescents with type 1 diabetes; the present study aims to close this gap. Methods This investigation is a randomized clinical trial, in which sixty adolescents with type 1 diabetes will be randomly assigned to receive either omega-3 (600 mg/day) or placebo capsules for 12 weeks. Evaluation of anthropometric parameters, flow-mediated dilation (FMD) as an endothelial function marker, carotid intima-media thickness (CIMT) as a vascular structure marker, proteinuria, biochemical factors including glycemic and lipid profile, blood urea nitrogen (BUN), creatinine, high-sensitivity C-reactive protein (hs-CRP), and erythrocyte sedimentation rate (ESR), as well as blood pressure will be done at the baseline and end of the trial. Also, dietary intake and physical activity will be assessed throughout the study. Statistical analysis will be performed using the SPSS software (Version 24), and P < 0.05 will be considered statistically meaningful. Discussion It is hypothesized that omega-3 supplementation may be beneficial for the management of type 1 diabetes and its complications by reducing inflammation and oxidative stress and also modulating immune responses and glucose and lipid metabolism through various mechanisms. The present study aims to investigate any effect of omega-3 on patients with type 1 diabetes. Ethical aspects This trial received approval from Medical Ethics Committee of Iran University of Medical Sciences, Tehran, Iran (IR.IUMS.REC.1400.070). Trial registration Iranian Registry of Clinical Trials IRCT20210419051010N1. Registered on 29 April 2021 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-021-05930-1.
Collapse
Affiliation(s)
- Masoud Khorshidi
- Department of Community Nutrition, School of Public Health, Iran University of Medical Sciences (IUMS), Hemmat Broadway, Tehran, 1449614535, Iran
| | - Aliakbar Sayyari
- Pediatric Gastroenterology, Hepatology and Nutrition Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Beheshteh Olang
- Pediatric Gastroenterology, Hepatology and Nutrition Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Community Medicine, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Alaei
- Department of Pediatric, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sakineh Shab-Bidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Khalili
- Department of Radiology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Salehi
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Naheed Aryaeian
- Department of Community Nutrition, School of Public Health, Iran University of Medical Sciences (IUMS), Hemmat Broadway, Tehran, 1449614535, Iran.
| |
Collapse
|
10
|
Al Rijjal D, Liu Y, Lai M, Song Y, Danaei Z, Wu A, Mohan H, Wei L, Schopfer FJ, Dai FF, Wheeler MB. Vascepa protects against high-fat diet-induced glucose intolerance, insulin resistance, and impaired β-cell function. iScience 2021; 24:102909. [PMID: 34458694 PMCID: PMC8379293 DOI: 10.1016/j.isci.2021.102909] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/28/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Omega-3 fatty acid prescription drugs, Vascepa (≥96% eicosapentaenoic acid [EPA] ethyl ester) and Lovaza (46.5% EPA and 37.5% docosahexaenoic acid ethyl ester) are known therapeutic regimens to treat hypertriglyceridemia. However, their impact on glucose homeostasis, progression to type 2 diabetes, and pancreatic beta cell function are not well understood. In the present study, mice were treated with Vascepa or Lovaza for one week prior to six weeks of high-fat diet feeding. Vascepa but not Lovaza led to reduced insulin resistance, reduced fasting insulin and glucose, and improved glucose intolerance. Vascepa improved beta cell function, reduced liver triglycerides with enhanced expression of hepatic fatty acid oxidation genes, and altered microbiota composition. Vascepa has protective effects on diet-induced insulin resistance and glucose intolerance in mice.
Collapse
Affiliation(s)
- Dana Al Rijjal
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Ying Liu
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
- Division of Advanced Diagnostics, Metabolism, Toronto General Research Institute, ON, Canada
| | - Mi Lai
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
- Division of Advanced Diagnostics, Metabolism, Toronto General Research Institute, ON, Canada
| | - Youchen Song
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Zahra Danaei
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Anne Wu
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Haneesha Mohan
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Li Wei
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Francisco J. Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Feihan F. Dai
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Michael B. Wheeler
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
- Division of Advanced Diagnostics, Metabolism, Toronto General Research Institute, ON, Canada
| |
Collapse
|
11
|
Ma APY, Yeung CLS, Tey SK, Mao X, Wong SWK, Ng TH, Ko FCF, Kwong EML, Tang AHN, Ng IOL, Cai SH, Yun JP, Yam JWP. Suppression of ACADM-Mediated Fatty Acid Oxidation Promotes Hepatocellular Carcinoma via Aberrant CAV1/SREBP1 Signaling. Cancer Res 2021; 81:3679-3692. [PMID: 33975883 DOI: 10.1158/0008-5472.can-20-3944] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/24/2021] [Accepted: 04/27/2021] [Indexed: 12/24/2022]
Abstract
Lipid accumulation exacerbates tumor development, as it fuels the proliferative growth of cancer cells. The role of medium-chain acyl-CoA dehydrogenase (ACADM), an enzyme that catalyzes the first step of mitochondrial fatty acid oxidation, in tumor biology remains elusive. Therefore, investigating its mode of dysregulation can shed light on metabolic dependencies in cancer development. In hepatocellular carcinoma (HCC), ACADM was significantly underexpressed, correlating with several aggressive clinicopathologic features observed in patients. Functionally, suppression of ACADM promoted HCC cell motility with elevated triglyceride, phospholipid, and cellular lipid droplet levels, indicating the tumor suppressive ability of ACADM in HCC. Sterol regulatory element-binding protein-1 (SREBP1) was identified as a negative transcriptional regulator of ACADM. Subsequently, high levels of caveolin-1 (CAV1) were observed to inhibit fatty acid oxidation, which revealed its role in regulating lipid metabolism. CAV1 expression negatively correlated with ACADM and its upregulation enhanced nuclear accumulation of SREBP1, resulting in suppressed ACADM activity and contributing to increased HCC cell aggressiveness. Administration of an SREBP1 inhibitor in combination with sorafenib elicited a synergistic antitumor effect and significantly reduced HCC tumor growth in vivo. These findings indicate that deregulation of fatty acid oxidation mediated by the CAV1/SREBP1/ACADM axis results in HCC progression, which implicates targeting fatty acid metabolism to improve HCC treatment. SIGNIFICANCE: This study identifies tumor suppressive effects of ACADM in hepatocellular carcinoma and suggests promotion of β-oxidation to diminish fatty acid availability to cancer cells could be used as a therapeutic strategy.
Collapse
Affiliation(s)
- Angel P Y Ma
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cherlie L S Yeung
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sze Keong Tey
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaowen Mao
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Samuel W K Wong
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Tung Him Ng
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Frankie C F Ko
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ernest M L Kwong
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Alexander H N Tang
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Irene Oi-Lin Ng
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Shao Hang Cai
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Ping Yun
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Judy W P Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China. .,State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
12
|
Picklo M, Vallée Marcotte B, Bukowski M, de Toro-Martín J, Rust BM, Guénard F, Vohl MC. Identification of Phenotypic Lipidomic Signatures in Response to Long Chain n-3 Polyunsaturated Fatty Acid Supplementation in Humans. J Am Heart Assoc 2021; 10:e018126. [PMID: 33461307 PMCID: PMC7955441 DOI: 10.1161/jaha.120.018126] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Supplementation with long chain n‐3 polyunsaturated fatty acids is used to reduce total circulating triacylglycerol (TAG) concentrations. However, in about 30% of people, supplementation with long chain n‐3 polyunsaturated fatty acids does not result in decreased plasma TAG. Lipidomic analysis may provide insight into this inter‐individual variability. Methods Lipidomic analyses using targeted, mass spectrometry were performed on plasma samples obtained from a clinical study in which participants were supplemented with 3 g/day of long chain n‐3 in the form of fish oil capsules over a 6‐week period. TAG species and cholesteryl esters (CE) were quantified for 130 participants pre‐ and post‐supplementation. Participants were segregated into 3 potential responder phenotypes: (1) positive responder (Rpos; TAG decrease), (2) non‐responder (Rnon; lacking TAG change), and (3) negative responder (Rneg; TAG increase) representing 67%, 18%, and 15% of the study participants, respectively. Separation of the 3 phenotypes was attributed to differential responses in TAG with 50 to 54 carbons with 1 to 4 desaturations. Elevated TAG with higher carbon number and desaturation were common to all phenotypes following supplementation. Using the TAG responder phenotype for grouping, decreases in total CE and specific CE occurred in the Rpos phenotype versus the Rneg phenotype with intermediate responses in the Rnon phenotype. CE 20:5, containing eicosapentaenoic acid (20:5n‐3), was elevated in all phenotypes. A classifier combining lipidomic and genomic features was built to discriminate triacylglycerol response phenotypes and reached a high predictive performance with a balanced accuracy of 75%. Conclusions These data identify lipidomic signatures, TAG and CE, associated with long chain n‐3 response p henotypes and identify a novel phenotype based upon CE changes. Registration URL: https://www.ClinicalTrials.gov; Unique Identifier: NCT01343342.
Collapse
Affiliation(s)
- Matthew Picklo
- USDA-ARS Grand Forks Human Nutrition Research Center Grand Forks ND
| | - Bastien Vallée Marcotte
- Centre Nutrition Santé et Société (NUTRISS) Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Université Laval Québec City QC Canada
| | - Michael Bukowski
- USDA-ARS Grand Forks Human Nutrition Research Center Grand Forks ND
| | - Juan de Toro-Martín
- Centre Nutrition Santé et Société (NUTRISS) Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Université Laval Québec City QC Canada
| | - Bret M Rust
- USDA-ARS Grand Forks Human Nutrition Research Center Grand Forks ND
| | - Frédéric Guénard
- Centre Nutrition Santé et Société (NUTRISS) Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Université Laval Québec City QC Canada
| | - Marie-Claude Vohl
- Centre Nutrition Santé et Société (NUTRISS) Institut sur la Nutrition et les Aliments Fonctionnels (INAF) Université Laval Québec City QC Canada
| |
Collapse
|
13
|
Antunes MM, Godoy G, Fernandes IDL, Manin LP, Zappielo C, Masi LN, de Oliveira VAB, Visentainer JV, Curi R, Bazotte RB. The Dietary Replacement of Soybean Oil by Canola Oil Does Not Prevent Liver Fatty Acid Accumulation and Liver Inflammation in Mice. Nutrients 2020; 12:E3667. [PMID: 33260679 PMCID: PMC7760057 DOI: 10.3390/nu12123667] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/16/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022] Open
Abstract
A high-carbohydrate diet (HCD) is a well-established experimental model of accelerated liver fatty acid (FA) deposition and inflammation. In this study, we evaluated whether canola oil can prevent these physiopathological changes. We evaluated hepatic FA accumulation and inflammation in mice fed with a HCD (72.1% carbohydrates) and either canola oil (C group) or soybean oil (S group) as a lipid source for 0, 7, 14, 28, or 56 days. Liver FA compositions were analyzed by gas chromatography. The mRNA expression of acetyl-CoA carboxylase 1 (ACC1) was measured as an indicator of lipogenesis. The mRNA expression of F4/80, tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, and IL-10, as mediators of liver inflammation, were also measured. The C group stored less n-6 polyunsaturated FAs (n-6 PUFAs) and had more intense lipid deposition of monounsaturated FAs (MUFAs), n-3 PUFAs, and total FAs. The C group also showed higher ACC1 expression. Moreover, on day 56, the C group showed higher expressions of the inflammatory genes F4/80, TNF-α, IL-1β, and IL-6, as well as the anti-inflammatory IL-10. In conclusion, a diet containing canola oil as a lipid source does not prevent the fatty acid accumulation and inflammation induced by a HCD.
Collapse
Affiliation(s)
- Marina Masetto Antunes
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020-900, Brazil; (M.M.A.); (G.G.)
| | - Guilherme Godoy
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020-900, Brazil; (M.M.A.); (G.G.)
| | - Ingrid de Lima Fernandes
- Department of Chemistry, State University of Maringá, Maringá 87020-900, Brazil; (I.d.L.F.); (L.P.M.); (C.Z.); (J.V.V.)
| | - Luciana Pelissari Manin
- Department of Chemistry, State University of Maringá, Maringá 87020-900, Brazil; (I.d.L.F.); (L.P.M.); (C.Z.); (J.V.V.)
| | - Caroline Zappielo
- Department of Chemistry, State University of Maringá, Maringá 87020-900, Brazil; (I.d.L.F.); (L.P.M.); (C.Z.); (J.V.V.)
| | - Laureane Nunes Masi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 03342-000, Brazil; (L.N.M.); (V.A.B.d.O.); (R.C.)
| | - Vivian Araújo Barbosa de Oliveira
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 03342-000, Brazil; (L.N.M.); (V.A.B.d.O.); (R.C.)
| | - Jesuí Vergílio Visentainer
- Department of Chemistry, State University of Maringá, Maringá 87020-900, Brazil; (I.d.L.F.); (L.P.M.); (C.Z.); (J.V.V.)
| | - Rui Curi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 03342-000, Brazil; (L.N.M.); (V.A.B.d.O.); (R.C.)
| | - Roberto Barbosa Bazotte
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020-900, Brazil; (M.M.A.); (G.G.)
| |
Collapse
|
14
|
APOC3 rs2070667 Associates with Serum Triglyceride Profile and Hepatic Inflammation in Nonalcoholic Fatty Liver Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8869674. [PMID: 33294458 PMCID: PMC7718051 DOI: 10.1155/2020/8869674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
Single-nucleotide polymorphisms (SNPs) of apolipoprotein C3 (APOC3) play important role in lipid metabolism, and dyslipidemia underlies nonalcoholic fatty liver disease (NAFLD). But the correlation of serum lipidomics, APOC3 SNPs, and NAFLD remains limited understood. Enrolling thirty-four biopsy-proven NAFLD patients from Tianjin, Shanghai, Fujian, we investigated their APOC3 genotype and serum lipid profile by DNA sequencing and ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), respectively. Scoring of hepatocyte steatosis, ballooning, lobular inflammation, and liver fibrosis was then performed to reveal the role of lipidomics-affecting APOC3 SNPs in NAFLD-specific pathological alterations. Here, we reported that APOC3 SNPs (rs4225, rs4520, rs5128, rs2070666, and rs2070667) intimately correlated to serum lipidomics in NAFLD patients. A allele instead of G allele at rs2070667, which dominated the SNPs underlying lipidomic alteration, exhibited downregulatory effect on triacylglycerols (TGs: TG 54 : 7, TG 54 : 8, and TG 56 : 9) containing polyunsaturated fatty acid (PUFA). Moreover, subjects with low-level PUFA-containing TGs were predisposed to high-grade lobular inflammation (TG 54 : 7, rho = -0.454 and P = 0.007; TG 54 : 8, rho = -0.411 and P =0.016; TG 56 : 9, rho = -0.481 and P = 0.004). The significant correlation of APOC3 rs2070667 and inflammation grading [G/G vs. G/A+A/A: 0.00 (0.00 and 1.00) vs. 1.50 (0.75 and 2.00), P = 0.022] further confirmed its pathological action on the basis of lipidomics-impacting activity. These findings suggest an inhibitory effect of A allele at APOC3 rs2070667 on serum levels of PUFA-containing TGs, which are associated with high-grade lobular inflammation in NAFLD patients.
Collapse
|
15
|
Omega-3 polyunsaturated fatty acids: anti-inflammatory and anti-hypertriglyceridemia mechanisms in cardiovascular disease. Mol Cell Biochem 2020; 476:993-1003. [PMID: 33179122 DOI: 10.1007/s11010-020-03965-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) is the world's most recognized and notorious cause of death. It is known that increased triglyceride-rich lipoproteins (TRLs) and remnants of triglyceride-rich lipoproteins (RLP) are the major risk factor for CVD. Furthermore, hypertriglyceridemia commonly leads to a reduction in HDL and an increase in atherogenic small dense low-density lipoprotein (sdLDL or LDL-III) levels. Thus, the evidence shows that Ω-3 fatty acids (eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have a beneficial effect on CVD through reprogramming of TRL metabolism, reducing inflammatory mediators (cytokines and leukotrienes), and modulation of cell adhesion molecules. Therefore, the purpose of this review is to provide the molecular mechanism related to the beneficial effect of Ω-3 PUFA on the lowering of plasma TAG levels and other atherogenic lipoproteins. Taking this into account, this study also provides the TRL lowering and anti-inflammatory mechanism of Ω-3 PUFA metabolites such as RvE1 and RvD2 as a cardioprotective function.
Collapse
|
16
|
Wu H, Xu L, Ballantyne CM. Dietary and Pharmacological Fatty Acids and Cardiovascular Health. J Clin Endocrinol Metab 2020; 105:dgz174. [PMID: 31678992 PMCID: PMC7174038 DOI: 10.1210/clinem/dgz174] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 11/01/2019] [Indexed: 12/30/2022]
Abstract
CONTEXT The effects of dietary intake of different fatty acids and pharmacological use of fatty acids, specifically long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFAs), on cardiovascular health and atherosclerotic cardiovascular disease (ASCVD) prevention have been examined in a large number of observational studies and clinical trials. This review summarizes recent data and discusses potential mechanisms. EVIDENCE ACQUISITION The review is based on the authors' knowledge of the field supplemented by a PubMed search using the terms seafood, fish oil, saturated fatty acids, omega-3 fatty acids, eicosapentaenoic acid, docosahexaenoic acid, polyunsaturated fatty acids, monounsaturated fatty acids, and ASCVD. EVIDENCE SYNTHESIS We mainly discuss the recent clinical trials that examine the effects of different types of dietary fatty acids and pharmacological use of n-3 PUFA products on ASCVD prevention and the potential mechanisms. CONCLUSIONS While replacement of dietary saturated fat with unsaturated fat, polyunsaturated fat in particular, or intake of LC n-3 PUFA-rich seafood has generally shown benefit for ASCVD prevention and is recommended for cardiovascular benefits, data on effects of n-3 PUFA products on ASCVD health are inconsistent. However, recent clinical trials support benefits of prescription EPA in ASCVD prevention. n-3 PUFAs may contribute to ASCVD prevention through multiple mechanisms, including lowering plasma triglyceride levels, anti-inflammatory effects, antithrombotic effects, and effects on endothelial function.
Collapse
Affiliation(s)
- Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Lu Xu
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | |
Collapse
|
17
|
Akhlaghi M, Ghasemi-Nasab M, Riasatian M. Mediterranean diet for patients with non-alcoholic fatty liver disease, a systematic review and meta-analysis of observational and clinical investigations. J Diabetes Metab Disord 2020; 19:575-584. [PMID: 32550210 DOI: 10.1007/s40200-019-00475-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/04/2019] [Indexed: 02/07/2023]
Abstract
Aim Mediterranean diet (MD) is a healthful dietary pattern with benefits for prevention of metabolic diseases including non-alcoholic fatty liver disease (NAFLD). In the current meta-analysis, we assessed the association between MD and liver steatosis and cardiometabolic risk factors in patients with NAFLD. Methods PubMed, Scopus, and Embase were searched to find observational and clinical studies on the issue. No restriction on date and language was made. Outcomes included body mass index (BMI), waist circumference, blood pressure, triglycerides (TG), cholesterol fractions, glucose, insulin, insulin resistance, and liver transaminases. Results Seven observational reports and 6 trials met our inclusion criteria and entered in the meta-analysis. In observational studies, there was an inverse association between MD and NAFLD (effect size (ES) = 0.95; 95% CI: 0.90, 1.00; P = 0.05). In trials, subgroup analysis based on the method of liver examination showed effectiveness of MD on steatosis examined by magnetic resonance spectroscopy (P < 0.002; n = 2) but not by ultrasound (P = 0.08; n = 2). MD also showed a significant decreasing effect on BMI (ES = -1.23 kg/m2; 95% CI: -2.38, -0.09), plasma triglycerides (ES = -33.01 mg/dL; 95% CI: -52.84, -13.18), and HOMA-IR (ES = -0.94; 95% CI: -1.29, -0.58) but no significant effect was observed in waist circumference, cholesterol fractions, glucose and insulin, and liver transferases. Conclusion Overall, available data from observational and clinical studies indicated a trend for the relationship between MD and hepatic steatosis. Improvement in the most important risk factors of NAFLD, i.e. BMI, serum triglycerides, and insulin resistance, may be involved in such relationship.
Collapse
Affiliation(s)
- Masoumeh Akhlaghi
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Razi Blvd, Shiraz, Iran
| | - Maryam Ghasemi-Nasab
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Razi Blvd, Shiraz, Iran
| | - Maryamsadat Riasatian
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Razi Blvd, Shiraz, Iran
| |
Collapse
|
18
|
Rodríguez M, G Rebollar P, Mattioli S, Castellini C. n-3 PUFA Sources (Precursor/Products): A Review of Current Knowledge on Rabbit. Animals (Basel) 2019; 9:ani9100806. [PMID: 31618904 PMCID: PMC6827073 DOI: 10.3390/ani9100806] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 09/27/2019] [Accepted: 10/09/2019] [Indexed: 01/01/2023] Open
Abstract
This review compares the effects of different n-3 polyunsaturated fatty acid (PUFA) sources on biological activity, physiological/reproductive endpoints, and health implications with a special emphasis on a rabbit case study. Linoleic acid (LA) and α-linolenic acid (ALA) are members of two classes of PUFAs, namely the n-6 and n-3 series, which are required for normal human health. Both are considered precursors of a cascade of molecules (eicosanoids), which take part in many biological processes (inflammation, vasoconstriction/vasodilation, thromboregulation, etc.). However, their biological functions are opposite and are mainly related to the form (precursor or long-chain products) in which they were administered and to the enzyme-substrate preference. ALA is widely present in common vegetable oils and foods, marine algae, and natural herbs, whereas its long-chain PUFA derivatives are available mainly in fish and animal product origins. Recent studies have shown that the accumulation of n-3 PUFAs seems mostly to be tissue-dependent and acts in a tissue-selective manner. Furthermore, dietary n-3 PUFAs widely affect the lipid oxidation susceptibility of all tissues. In conclusion, sustainable sources of n-3 PUFAs are limited and exert a different effect about (1) the form in which they are administered, precursor or derivatives; (2) their antioxidant protections; and (3) the purpose to be achieved (health improvement, physiological and reproductive traits, metabolic pathways, etc.).
Collapse
Affiliation(s)
- María Rodríguez
- Departamento de Producción Agraria, Escuela Técnica Superior de Ingeniería Agronómica Alimentaria y de Biosistemas, Universidad Politécnica de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain.
| | - Pilar G Rebollar
- Departamento de Producción Agraria, Escuela Técnica Superior de Ingeniería Agronómica Alimentaria y de Biosistemas, Universidad Politécnica de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain.
| | - Simona Mattioli
- Department of Agricultural, Environmental and Food Science, University of Perugia, Borgo XX Giugno, 74, 06121 Perugia, Italy.
| | - Cesare Castellini
- Department of Agricultural, Environmental and Food Science, University of Perugia, Borgo XX Giugno, 74, 06121 Perugia, Italy.
| |
Collapse
|
19
|
Skulas-Ray AC, Wilson PWF, Harris WS, Brinton EA, Kris-Etherton PM, Richter CK, Jacobson TA, Engler MB, Miller M, Robinson JG, Blum CB, Rodriguez-Leyva D, de Ferranti SD, Welty FK. Omega-3 Fatty Acids for the Management of Hypertriglyceridemia: A Science Advisory From the American Heart Association. Circulation 2019; 140:e673-e691. [PMID: 31422671 DOI: 10.1161/cir.0000000000000709] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hypertriglyceridemia (triglycerides 200-499 mg/dL) is relatively common in the United States, whereas more severe triglyceride elevations (very high triglycerides, ≥500 mg/dL) are far less frequently observed. Both are becoming increasingly prevalent in the United States and elsewhere, likely driven in large part by growing rates of obesity and diabetes mellitus. In a 2002 American Heart Association scientific statement, the omega-3 fatty acids (n-3 FAs) eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) were recommended (at a dose of 2-4 g/d) for reducing triglycerides in patients with elevated triglycerides. Since 2002, prescription agents containing EPA+DHA or EPA alone have been approved by the US Food and Drug Administration for treating very high triglycerides; these agents are also widely used for hypertriglyceridemia. The purpose of this advisory is to summarize the lipid and lipoprotein effects resulting from pharmacological doses of n-3 FAs (>3 g/d total EPA+DHA) on the basis of new scientific data and availability of n-3 FA agents. In treatment of very high triglycerides with 4 g/d, EPA+DHA agents reduce triglycerides by ≥30% with concurrent increases in low-density lipoprotein cholesterol, whereas EPA-only did not raise low-density lipoprotein cholesterol in very high triglycerides. When used to treat hypertriglyceridemia, n-3 FAs with EPA+DHA or with EPA-only appear roughly comparable for triglyceride lowering and do not increase low-density lipoprotein cholesterol when used as monotherapy or in combination with a statin. In the largest trials of 4 g/d prescription n-3 FA, non-high-density lipoprotein cholesterol and apolipoprotein B were modestly decreased, indicating reductions in total atherogenic lipoproteins. The use of n-3 FA (4 g/d) for improving atherosclerotic cardiovascular disease risk in patients with hypertriglyceridemia is supported by a 25% reduction in major adverse cardiovascular events in REDUCE-IT (Reduction of Cardiovascular Events With EPA Intervention Trial), a randomized placebo-controlled trial of EPA-only in high-risk patients treated with a statin. The results of a trial of 4 g/d prescription EPA+DHA in hypertriglyceridemia are anticipated in 2020. We conclude that prescription n-3 FAs (EPA+DHA or EPA-only) at a dose of 4 g/d (>3 g/d total EPA+DHA) are an effective and safe option for reducing triglycerides as monotherapy or as an adjunct to other lipid-lowering agents.
Collapse
|
20
|
Zhou Q, Zhang Z, Wang P, Zhang B, Chen C, Zhang C, Su Y. EPA+DHA, but not ALA, Improved Lipids and Inflammation Status in Hypercholesterolemic Adults: A Randomized, Double‐Blind, Placebo‐Controlled Trial. Mol Nutr Food Res 2019; 63:e1801157. [DOI: 10.1002/mnfr.201801157] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/30/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Quan Zhou
- Department of Nutrition and Food HygieneSchool of Public HealthGuangzhou Medical University Guangzhou 510182 P. R. China
- Guangdong Provincial Key Laboratory of FoodSchool of Public HealthSun Yat‐Sen University Guangzhou 510080 P. R. China
| | - Zheqing Zhang
- Department of Nutrition and Food HygieneGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical University Guangzhou 510515 P. R. China
- Guangdong Provincial Key Laboratory of FoodSchool of Public HealthSun Yat‐Sen University Guangzhou 510080 P. R. China
| | - Ping Wang
- Guangdong Provincial Institute of Public Health Guangzhou 511430 P. R. China
- Guangdong Provincial Key Laboratory of FoodSchool of Public HealthSun Yat‐Sen University Guangzhou 510080 P. R. China
| | - Bo Zhang
- Food Safety and Health Research CenterGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical University
- Guangdong Provincial Key Laboratory of FoodSchool of Public HealthSun Yat‐Sen University Guangzhou 510080 P. R. China
| | - Chaogang Chen
- Department of Clinical NutritionSun Yat‐Sen Memorial HospitalSun Yat‐Sen University Guangzhou 510120 Guangdong P. R. China
| | - Caixia Zhang
- Department of Medical Statistics and EpidemiologySchool of Public HealthSun Yat‐Sen University Guangzhou 510080 P. R. China
| | - Yixiang Su
- Guangdong Provincial Key Laboratory of FoodSchool of Public HealthSun Yat‐Sen University Guangzhou 510080 P. R. China
| |
Collapse
|
21
|
Yeung S, Soliternik J, Mazzola N. Nutritional supplements for the prevention of diabetes mellitus and its complications. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
22
|
Dietary Flaxseed Oil Prevents Western-Type Diet-Induced Nonalcoholic Fatty Liver Disease in Apolipoprotein-E Knockout Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3256241. [PMID: 29081885 PMCID: PMC5610846 DOI: 10.1155/2017/3256241] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 06/28/2017] [Accepted: 07/05/2017] [Indexed: 02/08/2023]
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) has dramatically increased globally during recent decades. Intake of n-3 polyunsaturated fatty acids (PUFAs), mainly eicosapentaenoic acid (EPA, C20:5n-3) and docosahexaenoic acid (DHA, C22:6n-3), is believed to be beneficial to the development of NAFLD. However, little information is available with regard to the effect of flaxseed oil rich in α-linolenic acid (ALA, C18:3n-3), a plant-derived n-3 PUFA, in improving NAFLD. This study was to gain the effect of flaxseed oil on NAFLD and further investigate the underlying mechanisms. Apolipoprotein-E knockout (apoE-KO) mice were given a normal chow diet, a western-type high-fat and high-cholesterol diet (WTD), or a WTD diet containing 10% flaxseed oil (WTD + FO) for 12 weeks. Our data showed that consumption of flaxseed oil significantly improved WTD-induced NAFLD, as well as ameliorated impaired lipid homeostasis, attenuated oxidative stress, and inhibited inflammation. These data were associated with the modification effects on expression levels of genes involved in de novo fat synthesis (SREBP-1c, ACC), triacylglycerol catabolism (PPARα, CPT1A, and ACOX1), inflammation (NF-κB, IL-6, TNF-α, and MCP-1), and oxidative stress (ROS, MDA, GSH, and SOD).
Collapse
|
23
|
Zimmer M, Bista P, Benson EL, Lee DY, Liu F, Picarella D, Vega RB, Vu CB, Yeager M, Ding M, Liang G, Horton JD, Kleemann R, Kooistra T, Morrison MC, Wielinga PY, Milne JC, Jirousek MR, Nichols AJ. CAT-2003: A novel sterol regulatory element-binding protein inhibitor that reduces steatohepatitis, plasma lipids, and atherosclerosis in apolipoprotein E*3-Leiden mice. Hepatol Commun 2017; 1:311-325. [PMID: 29404461 PMCID: PMC5721391 DOI: 10.1002/hep4.1042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022] Open
Abstract
CAT‐2003 is a novel conjugate of eicosapentaenoic acid (EPA) and niacin designed to be hydrolyzed by fatty acid amide hydrolase to release EPA inside cells at the endoplasmic reticulum. In cultured liver cells, CAT‐2003 blocked the maturation of sterol regulatory element‐binding protein (SREBP)‐1 and SREBP‐2 proteins and decreased the expression of multiple SREBP target genes, including HMGCR and PCSK9. Consistent with proprotein convertase subtilisin/kexin type 9 (PCSK9) reduction, both low‐density lipoprotein receptor protein at the cell surface and low‐density lipoprotein particle uptake were increased. In apolipoprotein E*3‐Leiden mice fed a cholesterol‐containing western diet, CAT‐2003 decreased hepatic inflammation and steatosis as evidenced by fewer inflammatory cell aggregates in histopathologic sections, decreased nuclear factor kappa B activity in liver lysates, reduced inflammatory gene expression, reduced intrahepatic cholesteryl ester and triglyceride levels, and decreased liver mass. Plasma PCSK9 was reduced and hepatic low‐density lipoprotein receptor protein expression was increased; plasma cholesterol and triglyceride levels were lowered. Aortic root segments showed reduction of several atherosclerotic markers, including lesion size, number, and severity. CAT‐2003, when dosed in combination with atorvastatin, further lowered plasma cholesterol levels and decreased hepatic expression of SREBP target genes. Conclusion: SREBP inhibition is a promising new strategy for the prevention and treatment of diseases associated with abnormal lipid metabolism, such as atherosclerosis and nonalcoholic steatohepatitis. (Hepatology Communications 2017;1:311–325)
Collapse
Affiliation(s)
| | | | | | | | - Feng Liu
- Catabasis Pharmaceuticals Cambridge MA
| | | | | | - Chi B Vu
- Catabasis Pharmaceuticals Cambridge MA
| | | | - Min Ding
- Department of Molecular Genetics University of Texas Southwestern Medical Center at Dallas Dallas TX
| | - Guosheng Liang
- Department of Molecular Genetics University of Texas Southwestern Medical Center at Dallas Dallas TX
| | - Jay D Horton
- Department of Molecular Genetics University of Texas Southwestern Medical Center at Dallas Dallas TX.,Department of Internal Medicine University of Texas Southwestern Medical Center at Dallas Dallas TX.,Center for Human Nutrition University of Texas Southwestern Medical Center at Dallas Dallas TX
| | - Robert Kleemann
- Netherlands Organization for Applied Scientific Research, Department of Metabolic Health Research Leiden the Netherlands
| | - Teake Kooistra
- Netherlands Organization for Applied Scientific Research, Department of Metabolic Health Research Leiden the Netherlands
| | - Martine C Morrison
- Netherlands Organization for Applied Scientific Research, Department of Metabolic Health Research Leiden the Netherlands
| | - Peter Y Wielinga
- Netherlands Organization for Applied Scientific Research, Department of Metabolic Health Research Leiden the Netherlands
| | | | | | | |
Collapse
|
24
|
Tajima-Shirasaki N, Ishii KA, Takayama H, Shirasaki T, Iwama H, Chikamoto K, Saito Y, Iwasaki Y, Teraguchi A, Lan F, Kikuchi A, Takeshita Y, Murao K, Matsugo S, Kaneko S, Misu H, Takamura T. Eicosapentaenoic acid down-regulates expression of the selenoprotein P gene by inhibiting SREBP-1c protein independently of the AMP-activated protein kinase pathway in H4IIEC3 hepatocytes. J Biol Chem 2017; 292:10791-10800. [PMID: 28465347 DOI: 10.1074/jbc.m116.747006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 04/27/2017] [Indexed: 02/05/2023] Open
Abstract
Selenoprotein P (encoded by SELENOP in humans, Selenop in rat), a liver-derived secretory protein, induces resistance to insulin and vascular endothelial growth factor (VEGF) in type 2 diabetes. Suppression of selenoprotein P may provide a novel therapeutic approach to treating type 2 diabetes; however, few drugs inhibiting SELENOP expression in hepatocytes have been identified. The present findings demonstrate that eicosapentaenoic acid (EPA) suppresses SELENOP expression by inactivating sterol regulatory element-binding protein-1c (SREBP-1c, encoded by Srebf1 in rat) in H4IIEC3 hepatocytes. Treatment with EPA caused concentration- and time-dependent reduction in SELENOP promoter activity. EPA activated AMP-activated protein kinase (AMPK); however, the inhibitory effect of EPA on SELENOP promoter activity was not canceled with an AMPK inhibitor compound C and dominant-negative AMPK transfection. Deletion mutant promoter assays and computational analysis of transcription factor-binding sites conserved among the species resulted in identification of a sterol regulatory element (SRE)-like site in the SELENOP promoter. A chromatin immunoprecipitation (ChIP) assay revealed that EPA decreases binding of SREBP-1c to the SELENOP promoter. Knockdown of Srebf1 resulted in a significant down-regulation of Selenop expression. Conversely, SREBP-1c overexpression inhibited the suppressive effect of EPA. These data provide a novel mechanism of action for EPA involving improvement of systemic insulin sensitivity through the regulation of selenoprotein P production independently of the AMPK pathway and suggest an additional approach to developing anti-diabetic drugs.
Collapse
Affiliation(s)
| | - Kiyo-Aki Ishii
- From the Departments of Endocrinology and Metabolism.,System Biology, and
| | - Hiroaki Takayama
- From the Departments of Endocrinology and Metabolism.,System Biology, and
| | - Takayoshi Shirasaki
- System Biology, and.,Advanced Medical Technology, Kanazawa University Graduate School of Health Medicine, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8641
| | - Hisakazu Iwama
- the Life Science Research Center, Kagawa University, Ikenobe 1750-1, Miki-cho, Kita-gun, Kagawa 761-0793
| | - Keita Chikamoto
- From the Departments of Endocrinology and Metabolism.,the Division of Natural System, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192
| | - Yoshiro Saito
- the Department of Medical Life Systems, Faculty of Medical and Life Sciences, Systems Life Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394
| | | | | | - Fei Lan
- From the Departments of Endocrinology and Metabolism
| | - Akihiro Kikuchi
- From the Departments of Endocrinology and Metabolism.,System Biology, and
| | | | - Koji Murao
- the Department of Advanced Medicine, Kagawa University, Ikenobe 1750-1, Miki-cho, Kita-gun, Kagawa 761-0793, and
| | - Seiichi Matsugo
- the Division of Natural System, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192
| | | | - Hirofumi Misu
- From the Departments of Endocrinology and Metabolism.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | | |
Collapse
|
25
|
Saraswathi V, Perriotte-Olson C, Ganesan M, Desouza CV, Alnouti Y, Duryee MJ, Thiele GM, Nordgren TM, Clemens DL. A combination of dietary N-3 fatty acids and a cyclooxygenase-1 inhibitor attenuates nonalcoholic fatty liver disease in mice. J Nutr Biochem 2017; 42:149-159. [PMID: 28187366 DOI: 10.1016/j.jnutbio.2017.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/13/2016] [Accepted: 01/19/2017] [Indexed: 12/12/2022]
Abstract
We sought to determine whether a combination of purified n-3 fatty acids (n-3) and SC-560 (SC), a cyclooxygenase-1-specific inhibitor, is effective in ameliorating nonalcoholic fatty liver disease in obesity. Female wild-type mice were fed a high-fat and high-cholesterol diet (HF) supplemented with n-3 in the presence or absence of SC. Mice treated with SC alone exhibited no change in liver lipids, whereas n-3-fed mice tended to have lower hepatic lipids. Mice given n-3+SC had significantly lower liver lipids compared with HF controls indicating enhanced lipid clearance. Total and sulfated bile acids were significantly higher only in n-3+SC-treated mice compared with chow diet (CD) controls. Regarding mechanisms, the level of pregnane X receptor (PXR), a nuclear receptor regulating drug/bile detoxification, was significantly higher in mice given n-3 or n-3+SC. Studies in precision-cut liver slices and in cultured hepatoma cells showed that n-3+SC enhanced not only the expression/activation of PXR and its target genes but also the expression of farnesoid X receptor (FXR), another regulator of bile synthesis/clearance, indicating that n-3+SC can induce both PXR and FXR. The mRNA level of FGFR4 which inhibits bile formation showed a significant reduction in Huh 7 cells upon n-3 and n-3+SC treatment. PXR overexpression in hepatoma cells confirmed that n-3 or SC each induced the expression of PXR target genes and in combination had an enhanced effect. Our findings suggest that combining SC with n-3 potentiates its lipid-lowering effect, in part, by enhanced PXR and/or altered FXR/FGFR4 signaling.
Collapse
Affiliation(s)
- Viswanathan Saraswathi
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Curtis Perriotte-Olson
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA
| | - Murali Ganesan
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cyrus V Desouza
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Yazen Alnouti
- Department of Pharmaceutical Science, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael J Duryee
- Department of Internal Medicine, Division of Rheumatology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pharmaceutical Science, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Tara M Nordgren
- Department of Internal Medicine, Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dahn L Clemens
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
26
|
da Silva-Santi LG, Antunes MM, Caparroz-Assef SM, Carbonera F, Masi LN, Curi R, Visentainer JV, Bazotte RB. Liver Fatty Acid Composition and Inflammation in Mice Fed with High-Carbohydrate Diet or High-Fat Diet. Nutrients 2016; 8:E682. [PMID: 27801862 PMCID: PMC5133070 DOI: 10.3390/nu8110682] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/17/2016] [Accepted: 10/24/2016] [Indexed: 02/06/2023] Open
Abstract
Both high-carbohydrate diet (HCD) and high-fat diet (HFD) modulate liver fat accumulation and inflammation, however, there is a lack of data on the potential contribution of carbohydrates and lipids separately. For this reason, the changes in liver fatty acid (FA) composition in male Swiss mice fed with HCD or HFD were compared, at the time points 0 (before starting the diets), and after 7, 14, 28 or 56 days. Activities of stearoyl-CoA desaturase-1 (SCD-1), ∆-6 desaturase (D6D), elongases and de novo lipogenesis (DNL) were estimated. Liver mRNA expression of acetyl-CoA carboxylase 1 (ACC1) was evaluated as an additional indicator of the de novo lipogenesis. Myeloperoxidase activity, nitric oxide (NO) production, and mRNA expressions of F4/80, type I collagen, interleukin (IL)-6, IL-1β, IL-10, and tumor necrosis factor-α (TNF-α) were measured as indication of the liver inflammatory state. The HCD group had more intense lipid deposition, particularly of saturated fatty acids (SFAs) and monounsaturated fatty acids (MUFAs). This group also showed higher DNL, SCD-1, and D6D activities associated with increased NO concentration, as well as myeloperoxidase activity. Livers from the HFD group showed higher elongase activity, stored more polyunsaturated fatty acids (PUFAs) and had a lower omega-6/omega-3 fatty acid (n-6/n-3) ratio. In conclusion, liver lipid accumulation, fatty acids (FA) composition and inflammation were modulated by the dietary composition of lipids and carbohydrates. The HCD group had more potent lipogenic and inflammatory effects in comparison with HFD.
Collapse
Affiliation(s)
| | - Marina Masetto Antunes
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020-900, Paraná, Brazil.
| | | | - Fabiana Carbonera
- Department of Chemistry, State University of Maringá, Maringá 87020-900, Paraná, Brazil.
| | - Laureane Nunes Masi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil.
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-900, Brazil.
| | | | - Roberto Barbosa Bazotte
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020-900, Paraná, Brazil.
| |
Collapse
|
27
|
Kim M, Pichiah PBT, Kim DK, Cha YS. Black adzuki bean (Vigna angularis) extract exerts phenotypic effects on white adipose tissue and reverses liver steatosis in diet-induced obese mice. J Food Biochem 2016. [DOI: 10.1111/jfbc.12333] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Mina Kim
- Department of Food Science and Human Nutrition, and Research Institute of Human Ecology; Chonbuk National University; Jeonju 561-756 South Korea
| | | | - Dae Keun Kim
- College of Pharmacy; Woosuk University; Jeonju 565-701 South Korea
| | - Youn-Soo Cha
- Department of Food Science and Human Nutrition, and Research Institute of Human Ecology; Chonbuk National University; Jeonju 561-756 South Korea
| |
Collapse
|
28
|
Vu CB, Bemis JE, Benson E, Bista P, Carney D, Fahrner R, Lee D, Liu F, Lonkar P, Milne JC, Nichols AJ, Picarella D, Shoelson A, Smith J, Ting A, Wensley A, Yeager M, Zimmer M, Jirousek MR. Synthesis and Characterization of Fatty Acid Conjugates of Niacin and Salicylic Acid. J Med Chem 2016; 59:1217-31. [PMID: 26784936 DOI: 10.1021/acs.jmedchem.5b01961] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
This report describes the synthesis and preliminary biological characterization of novel fatty acid niacin conjugates and fatty acid salicylate conjugates. These molecular entities were created by covalently linking two bioactive molecules, either niacin or salicylic acid, to an omega-3 fatty acid. This methodology allows the simultaneous intracellular delivery of two bioactives in order to elicit a pharmacological response that could not be replicated by administering the bioactives individually or in combination. The fatty acid niacin conjugate 5 has been shown to be an inhibitor of the sterol regulatory element binding protein (SREBP), a key regulator of cholesterol metabolism proteins such as PCSK9, HMG-CoA reductase, ATP citrate lyase, and NPC1L1. On the other hand, the fatty acid salicylate conjugate 11 has been shown to have a unique anti-inflammatory profile based on its ability to modulate the NF-κB pathway through the intracellular release of the two bioactives.
Collapse
Affiliation(s)
- Chi B Vu
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Jean E Bemis
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Ericka Benson
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Pradeep Bista
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - David Carney
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Richard Fahrner
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Diana Lee
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Feng Liu
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Pallavi Lonkar
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Jill C Milne
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Andrew J Nichols
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Dominic Picarella
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Adam Shoelson
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Jesse Smith
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Amal Ting
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Allison Wensley
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Maisy Yeager
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Michael Zimmer
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Michael R Jirousek
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
29
|
Nanthirudjanar T, Furumoto H, Zheng J, Kim YI, Goto T, Takahashi N, Kawada T, Park SB, Hirata A, Kitamura N, Kishino S, Ogawa J, Hirata T, Sugawara T. Gut Microbial Fatty Acid Metabolites Reduce Triacylglycerol Levels in Hepatocytes. Lipids 2015; 50:1093-102. [PMID: 26399511 DOI: 10.1007/s11745-015-4067-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 08/31/2015] [Indexed: 12/27/2022]
Abstract
Hydroxy and oxo fatty acids were recently found to be produced as intermediates during gut microbial fatty acid metabolism. Lactobacillus plantarum produces these fatty acids from unsaturated fatty acids such as linoleic acid. In this study, we investigated the effects of these gut microbial fatty acid metabolites on the lipogenesis in liver cells. We screened their effect on sterol regulatory element binding protein-1c (SREBP-1c) expression in HepG2 cells treated with a synthetic liver X receptor α (LXRα) agonist (T0901317). The results showed that 10-hydroxy-12(Z)-octadecenoic acid (18:1) (HYA), 10-hydroxy-6(Z),12(Z)-octadecadienoic acid (18:2) (γHYA), 10-oxo-12(Z)-18:1 (KetoA), and 10-oxo-6(Z),12(Z)-18:2 (γKetoA) significantly decreased SREBP-1c mRNA expression induced by T0901317. These fatty acids also downregulated the mRNA expression of lipogenic genes by suppressing LXRα activity and inhibiting SREBP-1 maturation. Oral administration of KetoA, which effectively reduced triacylglycerol accumulation and acetyl-CoA carboxylase 2 (ACC2) expression in HepG2 cells, for 2 weeks significantly decreased Srebp-1c, Scd-1, and Acc2 expression in the liver of mice fed a high-sucrose diet. Our findings suggest that the hypolipidemic effect of the fatty acid metabolites produced by L. plantarum can be exploited in the treatment of cardiovascular diseases or dyslipidemia.
Collapse
Affiliation(s)
- Tharnath Nanthirudjanar
- Laboratory of Marine Bioproducts Technology, Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Hidehiro Furumoto
- Laboratory of Marine Bioproducts Technology, Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Jiawen Zheng
- Laboratory of Marine Bioproducts Technology, Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Young-Il Kim
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, 611-0011, Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, 611-0011, Japan
| | - Nobuyuki Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, 611-0011, Japan
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, 611-0011, Japan
| | - Si-Bum Park
- Laboratory of Industrial Microbiology, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Akiko Hirata
- Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Nahoko Kitamura
- Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Shigenobu Kishino
- Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Jun Ogawa
- Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Takashi Hirata
- Laboratory of Marine Bioproducts Technology, Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
- Shijonawate Gakuen University, Daito, 574-0011, Japan
| | - Tatsuya Sugawara
- Laboratory of Marine Bioproducts Technology, Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan.
| |
Collapse
|
30
|
Targher G, Byrne CD. A Perspective on Metabolic Syndrome and Nonalcoholic Fatty Liver Disease. Metab Syndr Relat Disord 2015; 13:235-8. [DOI: 10.1089/met.2015.1502] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Giovanni Targher
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Christopher D. Byrne
- Nutrition and Metabolism, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Southampton National Institute for Health Research Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| |
Collapse
|
31
|
Flaxseed Oil Containing α -Linolenic Acid Ester of Plant Sterol Improved Atherosclerosis in ApoE Deficient Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:958217. [PMID: 26180602 PMCID: PMC4477243 DOI: 10.1155/2015/958217] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/05/2015] [Indexed: 01/04/2023]
Abstract
Plant sterols (PS) have potential preventive function in atherosclerosis due to their cholesterol-lowering ability. Dietary α-linolenic acid in flaxseed oil is associated with a reduction in cardiovascular events through its hypolipidemic and anti-inflammation properties. This study was designed to evaluate the effects of flaxseed oil containing α-linolenic acid ester of PS (ALA-PS) on atherosclerosis and investigate the underlying mechanisms. C57BL/6 mice were administered a regular diet and apoE knockout (apoE-KO) mice were given a high fat diet alone or supplemented with 5% flaxseed oil with or without 3.3% ALA-PS for 18 weeks. Results demonstrated that flaxseed oil containing ALA-PS was synergistically interaction in ameliorating atherosclerosis as well as optimizing overall lipid levels, inhibiting inflammation and reducing oxidative stress. These data were associated with the modification effects on expression levels of genes involved in lipid metabolism (PPARα, HMGCR, and SREBPs), inflammation (IL-6, TNF, MCP-1, and VCAM-1), and oxidative stress (NADPH oxidase).
Collapse
|
32
|
Komatsu M, Kimura T, Yazaki M, Tanaka N, Yang Y, Nakajima T, Horiuchi A, Fang ZZ, Joshita S, Matsumoto A, Umemura T, Tanaka E, Gonzalez FJ, Ikeda SI, Aoyama T. Steatogenesis in adult-onset type II citrullinemia is associated with down-regulation of PPARα. Biochim Biophys Acta Mol Basis Dis 2014; 1852:473-81. [PMID: 25533124 DOI: 10.1016/j.bbadis.2014.12.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/09/2014] [Accepted: 12/15/2014] [Indexed: 12/13/2022]
Abstract
SLC25A13 (citrin or aspartate-glutamate carrier 2) is located in the mitochondrial membrane in the liver and its genetic deficiency causes adult-onset type II citrullinemia (CTLN2). CTLN2 is one of the urea cycle disorders characterized by sudden-onset hyperammonemia due to reduced argininosuccinate synthase activity. This disorder is frequently accompanied with hepatosteatosis in the absence of obesity and ethanol consumption. However, the precise mechanism of steatogenesis remains unclear. The expression of genes associated with fatty acid (FA) and triglyceride (TG) metabolism was examined using liver samples obtained from 16 CTLN2 patients and compared with 7 healthy individuals. Although expression of hepatic genes associated with lipogenesis and TG hydrolysis was not changed, the mRNAs encoding enzymes/proteins involved in FA oxidation (carnitine palmitoyl-CoA transferase 1α, medium- and very-long-chain acyl-CoA dehydrogenases, and acyl-CoA oxidase 1), very-low-density lipoprotein secretion (microsomal TG transfer protein), and FA transport (CD36 and FA-binding protein 1), were markedly suppressed in CTLN2 patients. Serum concentrations of ketone bodies were also decreased in these patients, suggesting reduced mitochondrial β-oxidation activity. Consistent with these findings, the expression of peroxisome proliferator-activated receptor α (PPARα), a master regulator of hepatic lipid metabolism, was significantly down-regulated. Hepatic PPARα expression was inversely correlated with severity of steatosis and circulating ammonia and citrulline levels. Additionally, phosphorylation of c-Jun-N-terminal kinase was enhanced in CTLN2 livers, which was likely associated with lower hepatic PPARα. Collectively, down-regulation of PPARα is associated with steatogenesis in CTLN2 patients. These findings provide a novel link between urea cycle disorder, lipid metabolism, and PPARα.
Collapse
Affiliation(s)
- Michiharu Komatsu
- Department of Medicine (Gastroenterology), Shinshu University School of Medicine, Japan
| | - Takefumi Kimura
- Department of Medicine (Gastroenterology), Shinshu University School of Medicine, Japan; Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Japan
| | - Masahide Yazaki
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Japan; Department of Biological Sciences for Intractable Neurological Diseases, Institute for Biomedical Sciences, Shinshu University, Japan
| | - Naoki Tanaka
- Department of Medicine (Gastroenterology), Shinshu University School of Medicine, Japan; Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Japan.
| | - Yang Yang
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Japan
| | - Takero Nakajima
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Japan
| | - Akira Horiuchi
- Digestive Disease Center, Showa Inan General Hospital, Japan
| | - Zhong-Ze Fang
- Department of Toxicology, School of Public Health, Tianjin Medical University, China
| | - Satoru Joshita
- Department of Medicine (Gastroenterology), Shinshu University School of Medicine, Japan
| | - Akihiro Matsumoto
- Department of Medicine (Gastroenterology), Shinshu University School of Medicine, Japan
| | - Takeji Umemura
- Department of Medicine (Gastroenterology), Shinshu University School of Medicine, Japan
| | - Eiji Tanaka
- Department of Medicine (Gastroenterology), Shinshu University School of Medicine, Japan
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, USA
| | - Shu-Ichi Ikeda
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Japan
| | - Toshifumi Aoyama
- Department of Metabolic Regulation, Shinshu University Graduate School of Medicine, Japan
| |
Collapse
|
33
|
Bouchard-Mercier A, Rudkowska I, Lemieux S, Couture P, Pérusse L, Vohl MC. SREBF1 gene variations modulate insulin sensitivity in response to a fish oil supplementation. Lipids Health Dis 2014; 13:152. [PMID: 25270430 PMCID: PMC4196000 DOI: 10.1186/1476-511x-13-152] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/27/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND An important inter-individual variability in the response of insulin sensitivity following a fish oil supplementation has been observed. The objective was to examine the associations between single nucleotide polymorphisms (SNPs) within sterol regulatory element binding transcription factor 1 (SREBF1) gene and the response of insulin sensitivity to a fish oil supplementation. METHODS Participants (n = 210) were recruited in the greater Quebec City area and followed a 6-week fish oil supplementation protocol (5 g/day: 1.9-2.2 g EPA; 1.1 g DHA). Insulin sensitivity was assessed by the quantitative insulin sensitivity check index (QUICKI). Three tag SNPs (tSNPs) within SREBF1 gene were genotyped according to TAQMAN methodology. RESULTS Three tSNPs (rs12953299, rs4925118 and rs4925115) covered 100% of the known genetic variability within SREBF1 gene. None of the three tSNPs was associated with either baseline fasting insulin concentrations (rs12953299, rs4925118 and rs4925115) (p = 0.29, p = 0.20 and p = 0.70, respectively) or QUICKI (p = 0.20, p = 0.18 and p = 0.76, respectively). The three tSNPs (rs12953299, rs4925118 and rs4925115) were associated with differences in the response of plasma insulin levels (p = 0.01, p = 0.005 and p = 0.004, respectively) and rs12953299 as well as rs4925115 were associated with the insulin sensitivity response (p = 0.009 and p = 0.01, respectively) to the fish oil supplementation, independently of the effects of age, sex and BMI. CONCLUSIONS The genetic variability within SREBF1 gene has an impact on the insulin sensitivity in response to a fish oil supplementation. TRIAL REGISTRATION clinicaltrials.gov: NCT01343342.
Collapse
Affiliation(s)
| | | | | | | | | | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 Hochelaga Blvd,, Quebec G1V 0A6, Canada.
| |
Collapse
|
34
|
Omega-3 fatty acids protect renal functions by increasing docosahexaenoic acid-derived metabolite levels in SHR.Cg-Lepr(cp)/NDmcr rats, a metabolic syndrome model. Molecules 2014; 19:3247-63. [PMID: 24642910 PMCID: PMC6271130 DOI: 10.3390/molecules19033247] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 03/07/2014] [Accepted: 03/07/2014] [Indexed: 01/01/2023] Open
Abstract
The omega-3 polyunsaturated fatty acids (ω-3 PUFAs) docosahexaenoic acid (DHA) and/or eicosapentaenoic acid (EPA) protect against diabetic nephropathy by inhibiting inflammation. The aim of this study was to assess the effects of highly purified DHA and EPA or EPA only administration on renal function and renal eicosanoid and docosanoid levels in an animal model of metabolic syndrome, SHR.Cg-Leprcp/NDmcr (SHRcp) rats. Male SHRcp rats were divided into 3 groups. Control (5% arabic gum), TAK-085 (300 mg/kg/day, containing 467 mg/g EPA and 365 mg/g DHA), or EPA (300 mg/kg/day) was orally administered for 20 weeks. The urinary albumin to creatinine ratio in the TAK-085-administered group was significantly lower than that in other groups. The glomerular sclerosis score in the TAK-085-administered group was significantly lower than that in the other groups. Although DHA levels were increased in total kidney fatty acids, the levels of nonesterified DHA were not significantly different among the 3 groups, whereas the levels of protectin D1, resolvin D1, and resolvin D2 were significantly increased in the TAK-085-administered group. The results show that the use of combination therapy with DHA and EPA in SHRcp rats improved or prevented renal failure associate with metabolic syndrome with decreasing triglyceride levels and increasing ω-3 PUFA lipid mediators.
Collapse
|
35
|
Bargut TCL, Frantz EDC, Mandarim-de-Lacerda CA, Aguila MB. Effects of a diet rich in n-3 polyunsaturated fatty acids on hepatic lipogenesis and beta-oxidation in mice. Lipids 2014; 49:431-44. [PMID: 24627299 DOI: 10.1007/s11745-014-3892-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 02/24/2014] [Indexed: 12/12/2022]
Abstract
Here, we investigate whether a diet rich in fish oil can lead to the development of hepatic alterations associated with non-alcoholic fatty liver disease (NAFLD). To achieve this goal, we provided, for 8 weeks, four different diets to 3-month-old C57BL/6 mice: (a) standard-chow diet (SC; 40 g soybean oil/kg diet, 10 % of the total energy content from lipids), (b) fish oil diet (FO; 4 g soybean oil and 36 g fish oil/kg diet, 10 % of the total energy content from lipids), (c) high-fat diet (HF; 40 g soybean oil and 238 g lard/kg diet, 50 % of the total energy content from lipids), and (d) high-fish oil diet (HFO; 40 g soybean oil and 238 g fish oil/kg diet, 50 % of the total energy content from lipids). Biochemical analyses, stereology, western-blotting and RT-qPCR were used. In the HF group, we found evidence of obesity, metabolic syndrome, and liver damage, along with hypertriglyceridemia, hepatic insulin resistance, and steatosis. On the other hand, the HFO group did not present these alterations and remained similar to the controls. The changes observed in the animals fed the HF diet were accompanied by an increase in hepatic lipogenesis and a decrease in beta-oxidation; meanwhile, in the HFO group, the opposite results were found, that is, reduced lipogenesis and elevated beta-oxidation, were most likely responsible for the prevention of deleterious hepatic alterations and liver damage. In conclusion, a diet rich in fish oil has beneficial effects on hepatic insulin resistance, lipogenesis and beta-oxidation and prevents hepatic tissue from liver damage and NAFLD.
Collapse
Affiliation(s)
- Thereza C Lonzetti Bargut
- Laboratorio de Morfometria, Metabolismo e Doença Cardiovascular, Centro Biomedico Instituto de Biologia, Universidade Do Estado Do Rio de Janeiro, Av 28 de Setembro 87 Fds, Rio de Janeiro, RJ, Brazil
| | | | | | | |
Collapse
|
36
|
DiNicolantonio JJ, Niazi AK, McCarty MF, O'Keefe JH, Meier P, Lavie CJ. Omega-3s and cardiovascular health. Ochsner J 2014; 14:399-412. [PMID: 25249807 PMCID: PMC4171799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND Omega-3 (n-3) fatty acids have previously been shown to reduce the risk of cardiac events, cardiac death, and all-cause mortality in randomized controlled trials. However, recent data have challenged the benefits of n-3 fatty acids in the current era of optimal medical therapy. METHODS We performed a literature review indicating important limitations that must be considered when interpreting the recent negative n-3 fatty acids trials. RESULTS Our review found relative strengths and weaknesses of both the older and more recent studies, along with many possible explanations for the disparate results. The principal difference between the older and the more recent n-3 studies was a greater use of background optimal medical therapy that may have reduced the benefit from n-3s. Additionally, some of the more recent n-3 trials used relatively low doses or tested n-3 supplementation on top of a relatively high baseline intake of n-3s. CONCLUSION Despite the recent negative data about n-3 fatty acids, the overall evidence still supports the American Heart Association recommendation of 1 gram of eicosapentaenoic acid/docosahexaenoic acid per day for patients with coronary heart disease.
Collapse
Affiliation(s)
- James J. DiNicolantonio
- Department of Cardiology, Mid America Heart Institute, Saint Luke's Health System, Kansas City, MO
| | | | | | - James H. O'Keefe
- Mid America Heart Institute, University of Missouri-Kansas City/Saint Luke's Health System, Kansas City, MO
| | - Pascal Meier
- The Heart Hospital, University College London Hospital, London, UK and Yale Medical School, New Haven, CT
| | - Carl J. Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinic Foundation, New Orleans, LA
- The University of Queensland School of Medicine, Ochsner Clinical School, New Orleans, LA
- Department of Preventive Medicine, Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
37
|
Hussein M, Harvatine KJ, Boisclair YR, Bauman DE. Supplementation with fish oil as a source of n-3 fatty acids does not downregulate mammary lipogenesis in lactating mice. J Nutr 2013; 143:1913-9. [PMID: 24132572 DOI: 10.3945/jn.113.181149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The very long chain n-3 (ω-3) polyunsaturated fatty acids (VLCn3PUFAs) are potent regulators of hepatic lipid synthesis, but their effect on lipid synthesis in the lactating mammary gland is less well investigated. The objective of the present study was to examine effects of fish oil (FO) supplementation on mammary lipogenesis and the expression of lipogenic genes in mammary and hepatic tissues of lactating mice. Beginning on day 6 of lactation and continuing for 7 d, female C57BL/6J mice (n = 8/diet) were fed 1 of 3 dietary treatments: a 5%-fat diet containing mainly saturated fatty acids (FAs) (low-fat control) or 2 10%-fat diets, 1 enriched with FO as a source of VLCn3PUFAs and the other enriched with a safflower/palm oil mixture (high-fat control) as a source of oleic acid. Mammary lipogenic capacity, measured by (14)C-glucose incorporation into FAs by mammary explants, was similar among treatments, and there were no treatment effects on the proportion of de novo synthesized FAs in milk fat or on litter weight gain, a proxy for milk energy secretion. Also, there were no treatment effects on mammary mRNA abundance for key lipogenic enzymes and proteins involved in the regulation of milk lipid synthesis. In contrast, there was a treatment effect on hepatic lipogenesis, with FO resulting in a decrease of ~50% in hepatic lipid content and a similar downregulation of lipogenic gene expression compared with the 2 control diets. Overall, there were tissue-specific differences in dietary VLCn3PUFA effects on lipid synthesis with no observed effects for mammary lipogenic variables but marked reductions occurring in hepatic lipogenesis.
Collapse
Affiliation(s)
- Mahmoud Hussein
- Department of Animal Science, Cornell University, Ithaca, NY
| | | | | | | |
Collapse
|
38
|
Bouzianas DG, Bouziana SD, Hatzitolios AI. Potential treatment of human nonalcoholic fatty liver disease with long-chain omega-3 polyunsaturated fatty acids. Nutr Rev 2013; 71:753-71. [PMID: 24148001 DOI: 10.1111/nure.12073] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder in the Western world. Its prevalence has increased with the growing obesity epidemic, yet no definitive treatment has been developed, and optimal management remains a clinical challenge. Long-chain omega-3 polyunsaturated fatty acids (PUFAs) have recently been proposed as a potential treatment for liver inflammation associated with fat accumulation. PubMed literature and the ClinicalTrials.gov database were reviewed for the effects of omega-3 PUFA treatment on NAFLD, from mechanisms to the results of preclinical studies, human studies, and unreported ongoing clinical trials, using terms such as NAFLD, nonalcoholic steatohepatitis, omega-3 fatty acids, and fish oil. Articles published over the last 3-4 years were emphasized, and relevancy was ensured by scanning their abstracts. Preliminary studies have confirmed an ameliorative effect, yet the translation of promising early data into therapeutic interventions will have to await the results of larger, properly conducted, ongoing clinical trials.
Collapse
Affiliation(s)
- Dimitrios G Bouzianas
- First Propedeutic Department of Internal Medicine, Medical School, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | | | | |
Collapse
|
39
|
Kanbe H, Kamijo Y, Nakajima T, Tanaka N, Sugiyama E, Wang L, Fang ZZ, Hara A, Gonzalez FJ, Aoyama T. Chronic ethanol consumption decreases serum sulfatide levels by suppressing hepatic cerebroside sulfotransferase expression in mice. Arch Toxicol 2013; 88:367-79. [PMID: 24065054 DOI: 10.1007/s00204-013-1132-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 09/11/2013] [Indexed: 12/20/2022]
Abstract
Epidemiological studies demonstrate a possible relationship between chronic ethanol drinking and thrombotic diseases, such as myocardial infarction and stroke. However, the precise mechanism for this association remains unclear. Sulfatides are endogenous glycosphingolipids composed of ceramide, galactose, and sulfate, known to have anti-thrombotic properties. Low (0.5 g/kg/day), middle (1.5 g/kg/day), and high (3.0 g/kg/day) doses of ethanol were administered for 21 days intraperitoneally to female wild-type mice, and serum/liver sulfatide levels were measured. No significant changes in cholesterol and triglycerides were seen in serum and liver by ethanol treatment. However, serum/liver sulfatide levels were significantly decreased by middle- and high-dose ethanol treatment, likely due to downregulation of hepatic cerebroside sulfotransferase (CST) levels. Marked decreases in the expression of catalase and superoxide dismutases and ensuing increases in lipid peroxides were also observed in the livers of mice with middle- and high-dose ethanol treatment, suggesting the association between the suppression of hepatic CST expression and enhancement of oxidative stress. Furthermore, serum levels of tissue factor, a typical pro-coagulant molecule, were significantly increased in the mice with middle- and high-dose ethanol treatment showing decreases in serum sulfatide levels. Collectively, these results demonstrate that chronic ethanol consumption reduces serum sulfatide levels by increasing oxidative stress and decreasing the expression of CST in the liver. These findings could provide a mechanism by which chronic ethanol drinking increases thrombotic events.
Collapse
Affiliation(s)
- Hiroki Kanbe
- Department of Metabolic Regulation, Institute on Aging and Adaptation, Shinshu University Graduate School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bouchard-Mercier A, Rudkowska I, Lemieux S, Couture P, Vohl MC. Polymorphisms, de novo lipogenesis, and plasma triglyceride response following fish oil supplementation. J Lipid Res 2013; 54:2866-73. [PMID: 23886516 DOI: 10.1194/jlr.m041590] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interindividual variability in the response of plasma triglyceride concentrations (TG) following fish oil consumption has been observed. Our objective was to examine the associations between single-nucleotide polymorphisms (SNPs) within genes encoding proteins involved in de novo lipogenesis and the relative change in plasma TG levels following a fish oil supplementation. Two hundred and eight participants were recruited in the greater Quebec City area. The participants completed a six-week fish oil supplementation (5 g fish oil/day: 1.9-2.2 g eicosapentaenoic acid and 1.1 g docosahexaenoic acid. SNPs within SREBF1, ACLY, and ACACA genes were genotyped using TAQMAN methodology. After correction for multiple comparison, only two SNPs, rs8071753 (ACLY) and rs1714987 (ACACA), were associated with the relative change in plasma TG concentrations (P = 0.004 and P = 0.005, respectively). These two SNPs explained 7.73% of the variance in plasma TG relative change following fish oil consumption. Genotype frequencies of rs8071753 according to the TG response groups (responders versus nonresponders) were different (P = 0.02). We conclude that the presence of certain SNPs within genes, such as ACLY and ACACA, encoding proteins involved in de novo lipogenesis seem to influence the plasma TG response following fish oil consumption.
Collapse
Affiliation(s)
- Annie Bouchard-Mercier
- Institute of Nutrition and Functional Foods (INAF) Laval University, Quebec, G1V 0A6, Canada
| | | | | | | | | |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW Fatty acids influence human health and diseases in various ways. In recent years, much work has been carried out to elucidate the mechanisms by which dietary fatty acids control short-term and long-term cellular functions. We have reviewed herein the most recent studies on modulation of gene expression by fatty acids. A number of genes respond to transcription factors and present a transcription factor response element in their promoter regions. Fatty acids may exert their effects on metabolism by regulating gene transcription via transcription factors. Understanding how fatty acids control expression of metabolic genes is a promising strategy to be investigated by aiming to treat metabolic diseases such as insulin resistance, obesity, and type 2 diabetes mellitus. RECENT FINDINGS Fatty acids exert many of their biological effects through the modulation of the activity of transcription factors, such as sterol regulatory element-binding proteins, peroxisome proliferator-activated receptors, and liver X receptors. SUMMARY Fatty acid action through transcription factors controls the expression of several inflammatory and metabolic genes.
Collapse
Affiliation(s)
- Laureane N Masi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
42
|
Nanthirudjanar T, Furumoto H, Hirata T, Sugawara T. Oxidized eicosapentaenoic acids more potently reduce LXRα-induced cellular triacylglycerol via suppression of SREBP-1c, PGC-1β and GPA than its intact form. Lipids Health Dis 2013; 12:73. [PMID: 23680128 PMCID: PMC3680052 DOI: 10.1186/1476-511x-12-73] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/13/2013] [Indexed: 11/19/2022] Open
Abstract
Dietary polyunsaturated fatty acids (PUFA), especially eicosapentaenoic acid (EPA), improve lipid metabolism and contribute to the prevention of vascular diseases such as atherosclerosis. However, EPA in the diet is easily oxidized at room temperature and several types of oxidized EPA (OEPA) derivatives are generated. To compare the efficiencies of OEPAs on lipid metabolism with EPA, human hepatocellular liver carcinoma cell line (HepG2) was treated with EPA or OEPAs and their effects on lipid metabolism related genes were studied. OEPAs more potently suppressed the expression of sterol-responsive element-binding protein (SREBP)-1c, a major transcription factor that activates the expression of lipogenic genes, and its downstream target genes than did EPA under conditions of lipid synthesis enhanced by T0901317, a synthetic liver X receptor (LXR) agonist. Furthermore, PGC-1β, a coactivator of both LXRα and SREBP-1, was markedly down-regulated by OEPAs compared with EPA. The treatment of OEPAs also significantly down-regulated the expression of glycerol-3-phosphate acyltransferase (GPA), the initiating enzyme in triacylglycerol (TG) synthesis, more than EPA. Therefore, the advantageous effects of OEPAs on cardiovascular diseases might be due to their SREBP-1c, PGC-1β and GPA mediated ameliorating effects.
Collapse
Affiliation(s)
- Tharnath Nanthirudjanar
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | | | | | | |
Collapse
|
43
|
Hirako S, Kim HJ, Iizuka Y, Nakasatomi M, Matsumoto A. Fish oil prevents excessive hepatic lipid accumulation without inducing oxidative stress. Prostaglandins Leukot Essent Fatty Acids 2013; 88:365-71. [PMID: 23478025 DOI: 10.1016/j.plefa.2013.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 01/28/2013] [Accepted: 02/08/2013] [Indexed: 11/16/2022]
Abstract
We examined the effects of fish oil (FO) on high-cholesterol diet-induced hepatic lipid accumulation and oxidative stress. Female C57BL/6J mice were fed diets consisting of safflower oil (SO), 1 en% FO (1FO), 2 en% FO (2FO), or 20 en% FO (20FO) with or without 2 weight% (wt%) cholesterol (SO/CH, 1FO/CH, 2FO/CH, and 20FO/CH groups, respectively) for 8 weeks. The hepatic triacylglyceride levels were significantly lower in the 2FO/CH and 20FO/CH groups than in the SO/CH group. The hepatic mRNAs of fatty acid oxidation-related genes were upregulated and the fatty acid synthesis-related genes were downregulated by the FO feeding. Adverse effects were not observed in the plasma levels of indicators of oxidative stress in response to the consumption of FO up to 20 en%. These results suggest that FO consumption in the range of 2-20 en% prevents hepatic lipid accumulation, thus improving lipid metabolism without causing oxidative stress.
Collapse
Affiliation(s)
- Satoshi Hirako
- Department of Clinical Dietetics & Human Nutrition, Faculty of Pharmaceutical Sciences, Josai University, Japan
| | | | | | | | | |
Collapse
|
44
|
Midtbø LK, Ibrahim MM, Myrmel LS, Aune UL, Alvheim AR, Liland NS, Torstensen BE, Rosenlund G, Liaset B, Brattelid T, Kristiansen K, Madsen L. Intake of farmed Atlantic salmon fed soybean oil increases insulin resistance and hepatic lipid accumulation in mice. PLoS One 2013; 8:e53094. [PMID: 23301026 PMCID: PMC3534660 DOI: 10.1371/journal.pone.0053094] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 11/27/2012] [Indexed: 12/12/2022] Open
Abstract
Background To ensure sustainable aquaculture, fish derived raw materials are replaced by vegetable ingredients. Fatty acid composition and contaminant status of farmed Atlantic salmon (Salmo salar L.) are affected by the use of plant ingredients and a spillover effect on consumers is thus expected. Here we aimed to compare the effects of intake of Atlantic salmon fed fish oil (FO) with intake of Atlantic salmon fed a high proportion of vegetable oils (VOs) on development of insulin resistance and obesity in mice. Methodology/principal findings Atlantic salmon were fed diets where FO was partly (80%) replaced with three different VOs; rapeseed oil (RO), olive oil (OO) or soy bean oil (SO). Fillets from Atlantic salmon were subsequently used to prepare Western diets (WD) for a mouse feeding trial. Partial replacement of FO with VOs reduced the levels of polychlorinated biphenyls (PCB) and dichloro-diphenyl-tricloroethanes (DDT) with more than 50% in salmon fillets, in WDs containing the fillets, and in white adipose tissue from mice consuming the WDs. Replacement with VOs, SO in particular, lowered the n−3 polyunsaturated fatty acid (PUFA) content and increased n−6 PUFA levels in the salmon fillets, in the prepared WDs, and in red blood cells collected from mice consuming the WDs. Replacing FO with VO did not influence obesity development in the mice, but replacement of FO with RO improved glucose tolerance. Compared with WD-FO fed mice, feeding mice WD-SO containing lower PCB and DDT levels but high levels of linoleic acid (LA), exaggerated insulin resistance and increased accumulation of fat in the liver. Conclusion/Significance Replacement of FO with VOs in aqua feed for farmed salmon had markedly different spillover effects on metabolism in mice. Our results suggest that the content of LA in VOs may be a matter of concern that warrants further investigation.
Collapse
Affiliation(s)
- Lisa Kolden Midtbø
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Mohammad Madani Ibrahim
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- Institute of Biomedicine, University of Bergen, Bergen, NorwayStavanger, Norway
| | - Lene Secher Myrmel
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Ulrike Liisberg Aune
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | | | - Nina S. Liland
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | | | | | - Bjørn Liaset
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Trond Brattelid
- National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Karsten Kristiansen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (LM); (KK)
| | - Lise Madsen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- National Institute of Nutrition and Seafood Research, Bergen, Norway
- * E-mail: (LM); (KK)
| |
Collapse
|
45
|
Watanabe S, Tsuneyama K. Eicosapentaenoic acid attenuates hepatic accumulation of cholesterol esters but aggravates liver injury and inflammation in mice fed a cholate-supplemented high-fat diet. J Toxicol Sci 2013; 38:379-90. [PMID: 23665937 DOI: 10.2131/jts.38.379] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The administration of a sodium cholate-supplemented high-fat (CAHF) diet in mice induced the predominant accumulation of cholesterol esters (CE) in the liver and biochemical and histological features of liver injury. Cholesteryl oleate was the most abundant CE found in the liver of the mice fed the CAHF diet. We examined the effect of ethyl eicosapentaenoate (EPA) on hepatic CE accumulation and liver injury in the mice fed the CAHF diet. The EPA supplementation suppressed the elevation in the level of cholesteryl oleate in the liver. The expression levels of sterol O-acyltransferase-2 and stearoyl-CoA desaturase-1 mRNA in the liver were elevated in the mice fed the CAHF diet, but they were normalized by the EPA supplementation. However, the elevation in serum transaminase activity, the sign of inflammatory cell exudation and inflammatory gene responses in the liver of the mice fed the EPA-supplemented diet were enhanced compared with those of the mice fed the CAHF diet. We demonstrated that EPA supplementation attenuated CE accumulation but aggravated liver injury and liver inflammation in the mice fed the CAHF diet.
Collapse
Affiliation(s)
- Shiro Watanabe
- Division of Nutritional Biochemistry, Institute of Natural Medicine, University of Toyama, Japan.
| | | |
Collapse
|
46
|
Watanabe M, Uesugi M. Small-molecule inhibitors of SREBP activation – potential for new treatment of metabolic disorders. MEDCHEMCOMM 2013. [DOI: 10.1039/c3md00177f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
47
|
Sheng X, Nakajima T, Wang L, Zhang X, Kamijo Y, Takahashi K, Tanaka N, Sugiyama E, Kyogashima M, Aoyama T, Hara A. Attenuation of kidney injuries maintains serum sulfatide levels dependent on hepatic synthetic ability: a possible involvement of oxidative stress. TOHOKU J EXP MED 2012; 227:1-12. [PMID: 22499158 DOI: 10.1620/tjem.227.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Serum sulfatides are the major glycosphingolipids in lipoproteins. Although serum sulfatides are mainly synthesized and secreted by the liver, they are significantly decreased when the kidneys are impaired. Our recent experimental study using a murine protein-overload nephropathy model suggested a hypothetical mechanism whereby serum sulfatides were reduced due to kidney dysfunction. This was the result of decreased hepatic expression of a sulfatide synthetic enzyme, cerebroside sulfotransferase (CST), which is associated with systemic enhancement of oxidative stress. However, there is a possibility that the experimental process, protein-overload itself, directly affected the sulfatide metabolism and oxidative stress in the liver. To determine whether kidney dysfunction actually reduces the hepatic synthesis of sulfatides via oxidative stress, we examined sulfatide levels, the hepatic content of metabolic sulfatide enzymes, and the degree of oxidative stress in protein-overload mice subjected to renoprotective therapy using clofibrate, a representative hypolipidemic medicine. Protein-overload mice exhibited marked kidney injuries, enhancement of hepatic oxidative stress, decreased levels of serum and hepatic sulfatides, and decreased expression of hepatic CST. The clofibrate treatment attenuated kidney damage and hepatic oxidative stress while maintaining serum/hepatic sulfatide levels and hepatic CST content in the mice. Because clofibrate monotherapy without protein-overload treatment only minimally affected these hepatic parameters, the hepatic synthesis of sulfatides appeared to be strongly influenced by kidney dysfunction and subsequent oxidative stress. This study suggests that the crosstalk between kidney dysfunction and hepatic sulfatide metabolism is mediated by oxidative stress. These results should help to understand the phenomenon in patients with end-stage kidney disease.
Collapse
Affiliation(s)
- Xiaona Sheng
- Department of Metabolic Regulation, Institute on Aging and Adaptation, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Omega-3 fatty acids and incident type 2 diabetes: a systematic review and meta-analysis. Br J Nutr 2012; 107 Suppl 2:S214-27. [PMID: 22591895 DOI: 10.1017/s0007114512001602] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The relationship between omega-3 polyunsaturated fatty acids (n-3 PUFA) from seafood sources (eicosapentaenoic acid, EPA; docosahexaenoic acid, DHA) or plant sources (alpha-linolenic acid, ALA) and risk of type 2 diabetes mellitus (DM) remains unclear. We systematically searched multiple literature databases through June 2011 to identify prospective studies examining relations of dietary n-3 PUFA, dietary fish and/or seafood, and circulating n-3 PUFA biomarkers with incidence of DM. Data were independently extracted in duplicate by 2 investigators, including multivariate-adjusted relative risk (RR) estimates and corresponding 95 % CI. Generalized least-squares trend estimation was used to assess dose-response relationships, with pooled summary estimates calculated by both fixed-effect and random-effect models. From 288 identified abstracts, 16 studies met inclusion criteria, including 18 separate cohorts comprising 540,184 individuals and 25,670 cases of incident DM. Consumption of fish and/or seafood was not significantly associated with DM (n = 13 studies; RR per 100 g/d = 1·12, 95 % CI = 0·94, 1·34); nor were consumption of EPA+DHA (n = 16 cohorts; RR per 250 mg/d = 1·04, 95 % CI = 0·97, 1·10) nor circulating levels of EPA+DHA biomarkers (n = 5 cohorts; RR per 3 % of total fatty acids = 0·94, 95 % CI = 0·75, 1·17). Both dietary ALA (n = 7 studies; RR per 0·5 g/d = 0·93, 95 % CI = 0·83, 1·04) and circulating ALA biomarker levels (n = 6 studies; RR per 0·1 % of total fatty acid = 0·90, 95 % CI = 0·80, 1·00, P = 0·06) were associated with non-significant trend towards lower risk of DM. Substantial heterogeneity (I²~80 %) was observed among studies of fish/seafood or EPA+DHA and DM; moderate heterogeneity ( < 55 %) was seen for dietary and biomarker ALA and DM. In unadjusted meta-regressions, study location (Asia vs. North America/Europe), mean BMI, and duration of follow-up each modified the association between fish/seafood and EPA+DHA consumption and DM risk (P-interaction ≤ 0·02 each). We had limited statistical power to determine the independent effect of these sources of heterogeneity due to their high collinearity. The overall pooled findings do not support either major harms or benefits of fish/seafood or EPA+DHA on development of DM, and suggest that ALA may be associated with modestly lower risk. Reasons for potential heterogeneity of effects, which could include true biologic heterogeneity, publication bias, or chance, deserve further investigation.
Collapse
|
49
|
Tanaka N, Matsubara T, Krausz KW, Patterson AD, Gonzalez FJ. Disruption of phospholipid and bile acid homeostasis in mice with nonalcoholic steatohepatitis. Hepatology 2012; 56:118-29. [PMID: 22290395 PMCID: PMC6371056 DOI: 10.1002/hep.25630] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 01/20/2012] [Indexed: 02/06/2023]
Abstract
UNLABELLED Nonalcoholic steatohepatitis (NASH) is a progressive form of nonalcoholic fatty liver disease that can develop into cirrhosis, hepatic failure, and hepatocellular carcinoma. Although several metabolic pathways are disrupted and endogenous metabolites may change in NASH, the alterations in serum metabolites during NASH development remain unclear. To gain insight into the disease mechanism, serum metabolite changes were assessed using metabolomics with ultraperformance liquid chromatography-electrospray ionization-quadrupole time-of-flight mass spectrometry and a conventional mouse NASH model induced by a methionine- and choline-deficient (MCD) diet. Significant decreases in serum palmitoyl-, stearoyl-, and oleoyl-lysophosphatidylcholine (LPC) and marked increases in tauro-β-muricholate, taurocholate and 12-hydroxyeicosatetraenoic acid (12-HETE) were detected in mice with NASH. In agreement with these metabolite changes, hepatic mRNAs encoding enzymes and proteins involved in LPC degradation (lysophosphatidylcholine acyltransferase [Lpcat] 1-4), basolateral bile acid excretion (ATP-binding cassette subfamily C member [Abcc] 1/4/5 and organic solute transporter β), and 12-HETE synthesis (arachidonate 12-lipoxygenase) were significantly up-regulated. In contrast, the expression of solute carrier family 10 member 1 (Slc10a1) and solute carrier organic anion transporter family member (Slco) 1a1 and 1b2, responsible for transporting bile acids into hepatocytes, were markedly suppressed. Supplementation of the MCD diet with methionine revealed that the changes in serum metabolites and the related gene expression were derived from steatohepatitis, but not dietary choline deficiency or steatosis. Furthermore, tumor necrosis factor-α and transforming growth factor-β1 induced the expression of Lpcat2/4 and Abcc1/4 and down-regulated Slc10a1 and Slco1a1 in primary hepatocytes, suggesting an association between the changes in serum LPC and bile acids and proinflammatory cytokines. Finally, induction of hepatitis in ob/ob mice by D-galactosamine injection led to similar changes in serum metabolites and related gene expression. CONCLUSION Phospholipid and bile acid metabolism is disrupted in NASH, likely due to enhanced hepatic inflammatory signaling.
Collapse
Affiliation(s)
- Naoki Tanaka
- laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tsutomu Matsubara
- laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kristopher W. Krausz
- laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Andrew D. Patterson
- laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD;,Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA
| | - Frank J. Gonzalez
- laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
50
|
Hepatic Cerebroside Sulfotransferase Is Induced by PPARα Activation in Mice. PPAR Res 2012; 2012:174932. [PMID: 22645601 PMCID: PMC3356938 DOI: 10.1155/2012/174932] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 02/16/2012] [Indexed: 11/23/2022] Open
Abstract
Sulfatides are one of the major sphingoglycolipids in mammalian serum and are synthesized and secreted mainly from the liver as a component of lipoproteins. Recent studies revealed a protective role for serum sulfatides against arteriosclerosis and hypercoagulation. Although peroxisome proliferator-activated receptor (PPAR) α has important functions in hepatic lipoprotein metabolism, its association with sulfatides has not been investigated. In this study, sulfatide levels and the expression of enzymes related to sulfatide metabolism were examined using wild-type (+/+), Ppara-heterozygous (+/−), and Ppara-null (−/−) mice given a control diet or one containing 0.1% fenofibrate, a clinically used hypolipidemic drug and PPARα activator. Fenofibrate treatment increased serum and hepatic sulfatides in Ppara (+/+) and (+/−) mice through a marked induction of hepatic cerebroside sulfotransferase (CST), a key enzyme in sulfatide synthesis, in a PPARα-dependent manner. Furthermore, increases in CST mRNA levels were correlated with mRNA elevations of several known PPARα target genes, and such changes were not observed for other sulfatide-metabolism enzymes in the liver. These results suggest that PPARα activation enhances hepatic sulfatide synthesis via CST induction and implicate CST as a novel PPARα target gene.
Collapse
|