1
|
Peng B, Wang Y, Zhang H. Mitonuclear Communication in Stem Cell Function. Cell Prolif 2025; 58:e13796. [PMID: 39726221 PMCID: PMC12099226 DOI: 10.1111/cpr.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Mitochondria perform multiple functions within the cell, including the production of ATP and a great deal of metabolic intermediates, while also contributing to the cellular stress response. The majority of mitochondrial proteins are encoded by nuclear genomes, highlighting the importance of mitonuclear communication for sustaining mitochondrial homeostasis and functional. As a crucial part of the intracellular signalling network, mitochondria can impact stem cell fate determinations. Considering the essential function of stem cells in tissue maintenance, regeneration and aging, it is important to understand how mitochondria influence stem cell fate. This review explores the significant roles of mitonuclear communication and mitochondrial proteostasis, highlighting their influence on stem cells. We also examine how mitonuclear interactions contribute to cellular homeostasis, stem cell therapies, and the potential for extending lifespan.
Collapse
Affiliation(s)
- Baozhou Peng
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- The Department of Histology and Embryology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yaning Wang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- The Department of Histology and Embryology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Hongbo Zhang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- The Department of Histology and Embryology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
2
|
Uniyal P, Panwar S, Bhatt A, Marianesan AB, Kumar R, Singh TG, Tyagi Y, Bushi G, Gaidhane AM, Kumar B. An update on current type 2 diabetes mellitus (T2DM) druggable targets and drugs targeting them. Mol Divers 2025:10.1007/s11030-025-11149-y. [PMID: 40080341 DOI: 10.1007/s11030-025-11149-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by hyperglycemia and affects millions of people globally. Even after advancement and development in medical science, it is a big task to achieve victory over type 2 diabetes mellitus (T2DM). T2DM can be a reason for fatal events like stroke, cardiac failure, nephropathy, and retinopathy. Many advanced antidiabetic drugs have been introduced in the market in the past two decades, leading researchers to hunt for new target proteins and their potential modulators that can help develop newer antidiabetic drugs. This review article comprises a broad literature of the latest developments in the management of T2DM concerning new target proteins, their inhibitors, or drugs from the clinical arena employed for the successful management of symptoms of T2DM using mono, dual, or triple combination medication therapy. The review categorizes antidiabetic drugs into three general classes that include conventional drug targets, currently explored targets, and upcoming emerging targets. The review aims to merge information on the medicines affecting these targets, their mechanisms, followed by the chemical structures, and recent advancements.
Collapse
Affiliation(s)
- Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Bell Road, Clement Town, Dehradun, Uttarakhand, India
| | - Surbhi Panwar
- School of Pharmacy, Graphic Era Hill University, Bell Road, Clement Town, Dehradun, Uttarakhand, India
| | - Akanksha Bhatt
- School of Pharmacy, Graphic Era Hill University, Bell Road, Clement Town, Dehradun, Uttarakhand, India
| | - Arockia Babu Marianesan
- Institute of Pharmaceutical Research, GLA University, 17, Km Stone, National Highway #2, Delhi-Mathura Road, Mathura, India
| | - Roshan Kumar
- Department of Microbiology, Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
| | - Thakur Gurjeet Singh
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Yogita Tyagi
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Premanagar, Dehradun, Uttarakhand, 248007, India
| | - Ganesh Bushi
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abhay M Gaidhane
- School of Epidemiology and Public Health, Jawaharlal Nehru Medical College, and Global Health Academy, Datta Meghe Institute of Higher Education, Wardha, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal University (Central University), Dist. Garhwal, Srinagar, Uttarakhand, 246174, India.
| |
Collapse
|
3
|
Park JE, Han JS. ( E)-5-hydroxy-7-methoxy-3-(2-hydroxybenzyl)-4-chromanone, a Major Homoisoflavonoid, Attenuates Free Fatty Acid-Induced Hepatic Steatosis by Activating AMPK and PPARα Pathways in HepG2 Cells. Nutrients 2024; 16:3475. [PMID: 39458470 PMCID: PMC11510552 DOI: 10.3390/nu16203475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND (E)-5-hydroxy-7-methoxy-3-(2-hydroxybenzyl)-4-chromanone (HMC), a homoisoflavonoid isolated from Portulaca oleracea, has significant anti-adipogenesis potential; it regulates adipogenic transcription factors. However, whether HMC improves hepatic steatosis in hepatocytes remains vague. This study investigated whether HMC ameliorates hepatic steatosis in free fatty acid-treated human hepatocellular carcinoma (HepG2) cells, and if so, its mechanism of action was analyzed. METHODS Hepatic steatosis was induced by a free fatty acid mixture in HepG2 cells. Thereafter, different HMC concentrations (10, 30, and 50 µM) or fenofibrate (10 µM, a PPARα agonist, positive control) was treated in HepG2 cells. RESULTS HMC markedly decreased lipid accumulation and triglyceride content in free fatty acid-treated HepG2 cell; it (10 and 50 μM) markedly upregulated protein expressions of pAMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase. HMC (10 and 50 μM) markedly inhibited the expression of sterol regulatory element-binding protein-1c, fatty acid synthase, and stearoyl-coA desaturase 1, which are the enzymes involved in lipid synthesis. Furthermore, HMC (10 and 50 μM) markedly upregulated the protein expression of peroxisome proliferator-activated receptor alpha (PPARα) and enhanced the protein expressions of carnitine palmitoyl transferase 1 and acyl-CoA oxidase 1. CONCLUSION HMC inhibits lipid accumulation and promotes fatty acid oxidation by AMPK and PPARα pathways in free fatty acid-treated HepG2 cells, thereby attenuating hepatic steatosis.
Collapse
Affiliation(s)
- Jae-Eun Park
- Department of Hotel Baking Technology, Busan Health University, Busan 49318, Republic of Korea;
| | - Ji-Sook Han
- Department of Food Science and Nutrition, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
4
|
Sharma S, Sharma D, Dhobi M, Wang D, Tewari D. An insight to treat cardiovascular diseases through phytochemicals targeting PPAR-α. Mol Cell Biochem 2024; 479:707-732. [PMID: 37171724 DOI: 10.1007/s11010-023-04755-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
Peroxisome proliferator-activated receptor-α (PPAR-α) belonging to the nuclear hormone receptor superfamily is a promising target for CVDs which mechanistically improves the production of high-density lipid as well as inhibit vascular smooth muscle cell proliferation. PPAR-α mainly interferes with adenosine monophosphate-activated protein kinase, transforming growth factor-β-activated kinase, and nuclear factor-κB pathways to protect against cardiac complications. Natural products/extracts could serve as a potential therapeutic strategy in CVDs for targeting PPAR-α with broad safety margins. In recent years, the understanding of naturally derived PPAR-α agonists has considerably improved; however, the information is scattered. In vitro and in vivo studies on acacetin, apigenin, arjunolic acid, astaxanthin, berberine, resveratrol, vaticanol C, hispidulin, ginsenoside Rb3, and genistein showed significant effects in CVDs complications by targeting PPAR-α. With the aim of demonstrating the tremendous chemical variety of natural products targeting PPAR-α in CVDs, this review provides insight into various natural products that can work to prevent CVDs by targeting the PPAR-α receptor along with their detailed mechanism.
Collapse
Affiliation(s)
- Supriya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Divya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Mahaveer Dhobi
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| |
Collapse
|
5
|
Evans WA, Eccles-Miller JA, Anderson E, Farrell H, Baldwin WS. 9-HODE and 9-HOTrE alter mitochondrial metabolism, increase triglycerides, and perturb fatty acid uptake and synthesis associated gene expression in HepG2 cells. Prostaglandins Leukot Essent Fatty Acids 2024; 202:102635. [PMID: 39142221 PMCID: PMC11404490 DOI: 10.1016/j.plefa.2024.102635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/17/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) prevalence is rising and can lead to detrimental health outcomes such as Non-Alcoholic Steatohepatitis (NASH), cirrhosis, and cancer. Recent studies have indicated that Cytochrome P450 2B6 (CYP2B6) is an anti-obesity CYP in humans and mice. Cyp2b-null mice are diet-induced obese, and human CYP2B6-transgenic (hCYP2B6-Tg) mice reverse the obesity or diabetes progression, but with increased liver triglyceride accumulation in association with an increase of several oxylipins. Notably, 9-hydroxyoctadecadienoic acid (9-HODE) produced from linoleic acid (LA, 18:2, ω-6) is the most prominent of these and 9-hydroxyoctadecatrienoic acid (9-HOTrE) from alpha-linolenic acid (ALA, 18:3, ω-3) is the most preferentially produced when controlling for substrate concentrations in vitro. Transactivation assays indicate that 9-HODE and 9-HOTrE activate PPARα and PPARγ. In Seahorse assays performed in HepG2 cells, 9-HOTrE increased spare respiratory capacity, slightly decreased palmitate metabolism, and increased non-glycolytic acidification in a manner consistent with slightly increased glutamine utilization; however, 9-HODE exhibited no effect on metabolism. Both compounds increased triglyceride and pyruvate concentrations, most strongly by 9-HOTrE, consistent with increased spare respiratory capacity. qPCR analysis revealed several perturbations in fatty acid uptake and metabolism gene expression. 9-HODE increased expression of CD36, FASN, PPARγ, and FoxA2 that are involved in lipid uptake and production. 9-HOTrE decreased ANGPTL4 expression and increased FASN expression consistent with increased fatty acid uptake, fatty acid production, and AMPK activation. Our findings support the hypothesis that 9-HODE and 9-HOTrE promote steatosis, but through different mechanisms as 9-HODE is directly involved in fatty acid uptake and synthesis; 9-HOTrE weakly inhibits mitochondrial fatty acid metabolism while increasing glutamine use.
Collapse
Affiliation(s)
- William A Evans
- Clemson University, Biological Sciences, Clemson, SC 29634, USA
| | | | | | - Hannah Farrell
- Clemson University, Biological Sciences, Clemson, SC 29634, USA
| | | |
Collapse
|
6
|
Li Y, Pan Y, Zhao X, Wu S, Li F, Wang Y, Liu B, Zhang Y, Gao X, Wang Y, Zhou H. Peroxisome proliferator-activated receptors: A key link between lipid metabolism and cancer progression. Clin Nutr 2024; 43:332-345. [PMID: 38142478 DOI: 10.1016/j.clnu.2023.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
Lipids represent the essential components of membranes, serve as fuels for high-energy processes, and play crucial roles in signaling and cellular function. One of the key hallmarks of cancer is the reprogramming of metabolic pathways, especially abnormal lipid metabolism. Alterations in lipid uptake, lipid desaturation, de novo lipogenesis, lipid droplets, and fatty acid oxidation in cancer cells all contribute to cell survival in a changing microenvironment by regulating feedforward oncogenic signals, key oncogenic functions, oxidative and other stresses, immune responses, or intercellular communication. Peroxisome proliferator-activated receptors (PPARs) are transcription factors activated by fatty acids and act as core lipid sensors involved in the regulation of lipid homeostasis and cell fate. In addition to regulating whole-body energy homeostasis in physiological states, PPARs play a key role in lipid metabolism in cancer, which is receiving increasing research attention, especially the fundamental molecular mechanisms and cancer therapies targeting PPARs. In this review, we discuss how cancer cells alter metabolic patterns and regulate lipid metabolism to promote their own survival and progression through PPARs. Finally, we discuss potential therapeutic strategies for targeting PPARs in cancer based on recent studies from the last five years.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yujie Pan
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Xin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
7
|
Pu Y, Cheng CK, Zhang H, Luo JY, Wang L, Tomlinson B, Huang Y. Molecular mechanisms and therapeutic perspectives of peroxisome proliferator-activated receptor α agonists in cardiovascular health and disease. Med Res Rev 2023; 43:2086-2114. [PMID: 37119045 DOI: 10.1002/med.21970] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 03/10/2023] [Accepted: 04/12/2023] [Indexed: 04/30/2023]
Abstract
The prevalence of cardiovascular disease (CVD) has been rising due to sedentary lifestyles and unhealthy dietary patterns. Peroxisome proliferator-activated receptor α (PPARα) is a nuclear receptor regulating multiple biological processes, such as lipid metabolism and inflammatory response critical to cardiovascular homeostasis. Healthy endothelial cells (ECs) lining the lumen of blood vessels maintains vascular homeostasis, where endothelial dysfunction associated with increased oxidative stress and inflammation triggers the pathogenesis of CVD. PPARα activation decreases endothelial inflammation and senescence, contributing to improved vascular function and reduced risk of atherosclerosis. Phenotypic switch and inflammation of vascular smooth muscle cells (VSMCs) exacerbate vascular dysfunction and atherogenesis, in which PPARα activation improves VSMC homeostasis. Different immune cells participate in the progression of vascular inflammation and atherosclerosis. PPARα in immune cells plays a critical role in immunological events, such as monocyte/macrophage adhesion and infiltration, macrophage polarization, dendritic cell (DC) embedment, T cell activation, and B cell differentiation. Cardiomyocyte dysfunction, a major risk factor for heart failure, can also be alleviated by PPARα activation through maintaining cardiac mitochondrial stability and inhibiting cardiac lipid accumulation, oxidative stress, inflammation, and fibrosis. This review discusses the current understanding and future perspectives on the role of PPARα in the regulation of the cardiovascular system as well as the clinical application of PPARα ligands.
Collapse
Affiliation(s)
- Yujie Pu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Hongsong Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jiang-Yun Luo
- Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Brian Tomlinson
- Faculty of Medicine, Macau University of Science & Technology, Macau, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
8
|
Sommerauer C, Kutter C. Noncoding RNAs in liver physiology and metabolic diseases. Am J Physiol Cell Physiol 2022; 323:C1003-C1017. [PMID: 35968891 DOI: 10.1152/ajpcell.00232.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The liver holds central roles in detoxification, energy metabolism and whole-body homeostasis but can develop malignant phenotypes when being chronically overwhelmed with fatty acids and glucose. The global rise of metabolic-associated fatty liver disease (MAFLD) is already affecting a quarter of the global population. Pharmaceutical treatment options against different stages of MAFLD do not yet exist and several clinical trials against hepatic transcription factors and other proteins have failed. However, emerging roles of noncoding RNAs, including long (lncRNA) and short noncoding RNAs (sRNA), in various cellular processes pose exciting new avenues for treatment interventions. Actions of noncoding RNAs mostly rely on interactions with proteins, whereby the noncoding RNA fine-tunes protein function in a process termed riboregulation. The developmental stage-, disease stage- and cell type-specific nature of noncoding RNAs harbors enormous potential to precisely target certain cellular pathways in a spatio-temporally defined manner. Proteins interacting with RNAs can be categorized into canonical or non-canonical RNA binding proteins (RBPs) depending on the existence of classical RNA binding domains. Both, RNA- and RBP-centric methods have generated new knowledge of the RNA-RBP interface and added an additional regulatory layer. In this review, we summarize recent advances of how of RBP-lncRNA interactions and various sRNAs shape cellular physiology and the development of liver diseases such as MAFLD and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Christian Sommerauer
- Science for Life Laboratory, Department of Microbiology, Tumor and Cell Biology, grid.4714.6Karolinska Institute, Stockholm, Sweden
| | - Claudia Kutter
- Science for Life Laboratory, Department of Microbiology, Tumor and Cell Biology, grid.4714.6Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
9
|
Lin Y, Wang Y, Li PF. PPARα: An emerging target of metabolic syndrome, neurodegenerative and cardiovascular diseases. Front Endocrinol (Lausanne) 2022; 13:1074911. [PMID: 36589809 PMCID: PMC9800994 DOI: 10.3389/fendo.2022.1074911] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a ligand-activated transcription factor that is involved in lipid metabolism of various tissues. Different metabolites of fatty acids and agonists like fibrates activate PPARα for its transactivative or repressive function. PPARα is known to affect diverse human diseases, and we focus on advanced studies of its transcriptional regulation in these diseases. In MAFLD, PPARα shows a protective function with its upregulation of lipid oxidation and mitochondrial biogenesis and transcriptional repression of inflammatory genes, which is similar in Alzheimer's disease and cardiovascular disease. Activation of PPARα also prevents the progress of diabetes complications; however, its role in diabetes and cancers remains uncertain. Some PPARα-specific agonists, such as Wy14643 and fenofibrate, have been applied in metabolic syndrome treatment, which might own potential in wider application. Future studies may further explore the functions and interventions of PPARα in cancer, diabetes, immunological diseases, and neurodegenerative disease.
Collapse
Affiliation(s)
- Yijun Lin
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, China
- *Correspondence: Yijun Lin, ; Yan Wang, ; Pei-feng Li,
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, China
- *Correspondence: Yijun Lin, ; Yan Wang, ; Pei-feng Li,
| | - Pei-feng Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen, China
- *Correspondence: Yijun Lin, ; Yan Wang, ; Pei-feng Li,
| |
Collapse
|
10
|
Dixit G, Prabhu A. The pleiotropic peroxisome proliferator activated receptors: Regulation and therapeutics. Exp Mol Pathol 2021; 124:104723. [PMID: 34822814 DOI: 10.1016/j.yexmp.2021.104723] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
The Peroxisome proliferator-activated receptors (PPARs) are key regulators of metabolic events in our body. Owing to their implication in maintenance of homeostasis, both PPAR agonists and antagonists assume therapeutic significance. Understanding the molecular mechanisms of each of the PPAR isotypes in the healthy body and during disease is crucial to exploiting their full therapeutic potential. This article is an attempt to present a rational analysis of the multifaceted therapeutic effects and underlying mechanisms of isotype-specific PPAR agonists, dual PPAR agonists, pan PPAR agonists as well as PPAR antagonists. A holistic understanding of the mechanistic dimensions of these key metabolic regulators will guide future efforts to identify novel molecules in the realm of metabolic, inflammatory and immunotherapeutic diseases.
Collapse
Affiliation(s)
- Gargi Dixit
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
11
|
Lee K, Yu H, Shouse S, Kong B, Lee J, Lee SH, Ko KS. RNA-Seq Reveals Different Gene Expression in Liver-Specific Prohibitin 1 Knock-Out Mice. Front Physiol 2021; 12:717911. [PMID: 34539442 PMCID: PMC8446661 DOI: 10.3389/fphys.2021.717911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022] Open
Abstract
Prohibitin 1 (PHB1) is an evolutionarily conserved and ubiquitously expressed protein that stabilizes mitochondrial chaperone. Our previous studies showed that liver-specific Phb1 deficiency induced liver injuries and aggravated lipopolysaccharide (LPS)-induced innate immune responses. In this study, we performed RNA-sequencing (RNA-seq) analysis with liver tissues to investigate global gene expression among liver-specific Phb1−/−, Phb1+/−, and WT mice, focusing on the differentially expressed (DE) genes between Phb1+/− and WT. When 78 DE genes were analyzed for biological functions, using ingenuity pathway analysis (IPA) tool, lipid metabolism-related genes, including insulin receptor (Insr), sterol regulatory element-binding transcription factor 1 (Srebf1), Srebf2, and SREBP cleavage-activating protein (Scap) appeared to be downregulated in liver-specific Phb1+/− compared with WT. Diseases and biofunctions analyses conducted by IPA verified that hepatic system diseases, including liver fibrosis, liver hyperplasia/hyperproliferation, and liver necrosis/cell death, which may be caused by hepatotoxicity, were highly associated with liver-specific Phb1 deficiency in mice. Interestingly, of liver disease-related 5 DE genes between Phb1+/− and WT, the mRNA expressions of forkhead box M1 (Foxm1) and TIMP inhibitor of metalloproteinase (Timp1) were matched with validation for RNA-seq in liver tissues and AML12 cells transfected with Phb1 siRNA. The results in this study provide additional insights into molecular mechanisms responsible for increasing susceptibility of liver injuries associated with hepatic Phb1.
Collapse
Affiliation(s)
- Kyuwon Lee
- Department of Nutritional Science and Food Management, College of Science and Industry Convergence, Ewha Womans University, Seoul, South Korea
| | - Hyeonju Yu
- Department of Nutritional Science and Food Management, College of Science and Industry Convergence, Ewha Womans University, Seoul, South Korea
| | - Stephanie Shouse
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, AR, United States
| | - Byungwhi Kong
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, AR, United States
| | - Jihye Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, MD, United States
| | - Seong-Ho Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, MD, United States
| | - Kwang Suk Ko
- Department of Nutritional Science and Food Management, College of Science and Industry Convergence, Ewha Womans University, Seoul, South Korea.,Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Beverly Hills, CA, United States
| |
Collapse
|
12
|
Goodarzi R, Jafarirad S, Mohammadtaghvaei N, Dastoorpoor M, Alavinejad P. The effect of pomegranate extract on anthropometric indices, serum lipids, glycemic indicators, and blood pressure in patients with nonalcoholic fatty liver disease: A randomized double-blind clinical trial. Phytother Res 2021; 35:5871-5882. [PMID: 34498307 DOI: 10.1002/ptr.7249] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/25/2021] [Accepted: 08/06/2021] [Indexed: 12/16/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. The beneficial effects of pomegranate have been shown on insulin resistance and obesity, which are linked to NAFLD pathogenesis. The aim of this study was to investigate the efficacy of pomegranate extract in patients with NAFLD. Forty-four NAFLD patients were randomly assigned to receive two pomegranate extract tablets or placebo for 12 weeks. Anthropometric measurements, serum lipids, glycemic indicators, and blood pressure were assessed at baseline and the end of the study. Pomegranate was associated with a reduction in the total cholesterol (p ˂ .001), triglyceride (p ˂ .001), low-density lipoprotein cholesterol (LDL-C)-to-high-density lipoprotein cholesterol (HDL-C) ratio (p ˂ .003), fasting blood sugar (p ˂ .001), homeostatic model assessment of insulin resistance (p = .02), diastolic blood pressure (p = .04), weight (p ˂ .001), body mass index (p ˂ .001), and waist circumference (p = .002), as compared to placebo. A significant increase was observed in serum HDL-C (p ˂ .001) after intervention with the pomegranate extract. However, no significant difference was shown between the two groups in serum insulin and LDL-C. The pomegranate extract supplement could be used as a complementary therapy along with existing therapies to improve glycemic indicators, serum lipids, anthropometric indices, and blood pressure in patients with nonalcoholic fatty liver.
Collapse
Affiliation(s)
- Reza Goodarzi
- Nutrition and Metabolic Diseases Research Center, Clinical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sima Jafarirad
- Nutrition and Metabolic Diseases Research Center, Clinical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Narges Mohammadtaghvaei
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Dastoorpoor
- Department of Biostatistics and Epidemiology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Pejman Alavinejad
- Research Institute for Infectious Disease of Digestive System, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
13
|
Islam SMT, Won J, Khan M, Chavin KD, Singh I. Peroxisomal footprint in the pathogenesis of nonalcoholic steatohepatitis. Ann Hepatol 2021; 19:466-471. [PMID: 31870746 DOI: 10.1016/j.aohep.2019.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/08/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a form of fatty liver disease where benign hepatic steatosis leads to chronic inflammation in the steatotic liver of a patient without any history of alcohol abuse. Mechanisms underlying the progression of hepatic steatosis to NASH have long been investigated. This review outlines the potential role of peroxisomal dysfunctions in exacerbating the disease in NASH. Loss of peroxisomes as well as impaired peroxisomal functions have been demonstrated to occur in inflammatory conditions including NASH. Because peroxisomes and mitochondria co-operatively perform many metabolic functions including O2 and lipid metabolisms, a compromised peroxisomal biogenesis and function can potentially contribute to defective lipid and reactive oxygen species metabolism which in turn can lead the progression of disease in NASH. Impaired peroxisomal biogenesis and function may be due to the decreased expression of peroxisomal proliferator-activated receptor-α (PPAR-α), the major transcription factor of peroxisomal biogenesis. Recent studies indicate that the reduced expression of PPAR-α in NASH is correlated with the activation of the toll-like receptor-4 pathway (TLR-4). Further investigations are required to establish the mechanistic connection between the TLR-4 pathway and PPAR-α-dependent impaired biogenesis/function of peroxisomes in NASH.
Collapse
Affiliation(s)
- S M Touhidul Islam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Jeseong Won
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Kenneth D Chavin
- Department of Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
14
|
Orabi D, Berger NA, Brown JM. Abnormal Metabolism in the Progression of Nonalcoholic Fatty Liver Disease to Hepatocellular Carcinoma: Mechanistic Insights to Chemoprevention. Cancers (Basel) 2021; 13:3473. [PMID: 34298687 PMCID: PMC8307710 DOI: 10.3390/cancers13143473] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is on the rise and becoming a major contributor to the development of hepatocellular carcinoma (HCC). Reasons for this include the rise in obesity and metabolic syndrome in contrast to the marked advances in prevention and treatment strategies of viral HCC. These shifts are expected to rapidly propel this trend even further in the coming decades, with NAFLD on course to become the leading etiology of end-stage liver disease and HCC. No Food and Drug Administration (FDA)-approved medications are currently available for the treatment of NAFLD, and advances are desperately needed. Numerous medications with varying mechanisms of action targeting liver steatosis and fibrosis are being investigated including peroxisome proliferator-activated receptor (PPAR) agonists and farnesoid X receptor (FXR) agonists. Additionally, drugs targeting components of metabolic syndrome, such as antihyperglycemics, have been found to affect NAFLD progression and are now being considered in the treatment of these patients. As NAFLD drug discovery continues, special attention should be given to their relationship to HCC. Several mechanisms in the pathogenesis of NAFLD have been implicated in hepatocarcinogenesis, and therapies aimed at NAFLD may additionally harbor independent antitumorigenic potential. This approach may provide novel prevention and treatment strategies.
Collapse
Affiliation(s)
- Danny Orabi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44106, USA;
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA;
- Department of General Surgery, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nathan A. Berger
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA;
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - J. Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44106, USA;
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA;
| |
Collapse
|
15
|
Valente V, Izzo R, Manzi MV, De Luca MR, Barbato E, Morisco C. Modulation of insulin resistance by renin angiotensin system inhibitors: implications for cardiovascular prevention. Monaldi Arch Chest Dis 2021; 91. [PMID: 33792231 DOI: 10.4081/monaldi.2021.1602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/28/2020] [Indexed: 11/23/2022] Open
Abstract
Insulin resistance (IR) and the related hyperinsulinamia play a key role in the genesis and progression of the continuum of cardiovascular (CV) disease. Thus, it is reasonable to pursue in primary and secondary CV prevention, the pharmacological strategies that are capable to interfere with the development of IR. The renin-angiotensin-aldosterone system (RAAS) plays an important role in the pathogenesis of IR. In particular, angiotensin II (Ang II) through the generation of reactive oxygen species, induces a low grade of inflammation, which impairs the insulin signal transduction. The angiotensin converting enzyme (ACE) inhibitors are effective not only as blood pressure-lowering agents, but also as modulators of metabolic abnormalities. Indeed, experimental evidence indicates that in animal models of IR, ACE inhibitors are capable to ameliorate the insulin sensitivity. The Ang II receptor blockers (ARBs) modulate the peroxisome proliferator-activated receptor (PPAR)-γ activity. PPARâ€"γ is a transcription factor that controls the gene expression of several key enzymes of glucose metabolism. A further mechanism that accounts for the favorable metabolic properties of ARBs is the capability to modulate the hypothalamicâ€"pituitary-adrenal (HPA) axis. The available clinical evidence is consistent with the concept that both ACE inhibitors and ARBs are able to interfere with the development of IR and its consequences like type 2 diabetes. In addition, pharmacological inhibition of the RAAS has favourable effects on dyslipidaemias, metabolic syndrome and obesity. Therefore, the pharmacological antagonism of the RAAS, nowadays, represents the first choice in the prevention of cardio-metabolic diseases.
Collapse
Affiliation(s)
- Valeria Valente
- Department of Translational Medicine, Federico II University of Naples, Italy.
| | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Italy.
| | - Maria Virginia Manzi
- Department of Advanced Biomedical Sciences, Federico II University of Naples, Italy.
| | | | - Emanuele Barbato
- Department of Translational Medicine, Federico II University of Naples, Italy.
| | - Carmine Morisco
- Department of Translational Medicine, Federico II University of Naples, Italy.
| |
Collapse
|
16
|
Su X, Wang W, Fang C, Ni C, Zhou J, Wang X, Zhang L, Xu X, Cao R, Lang H, Wang F. Vitamin K2 Alleviates Insulin Resistance in Skeletal Muscle by Improving Mitochondrial Function Via SIRT1 Signaling. Antioxid Redox Signal 2021; 34:99-117. [PMID: 32253917 DOI: 10.1089/ars.2019.7908] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aims: High-fat diet (HFD)-induced insulin resistance (IR) impairs skeletal muscle mitochondrial biogenesis and functions, adversely affecting human health and lifespan. Vitamin K2 (VK2) has a beneficial role in improving insulin sensitivity and glucose metabolism. However, the underlying molecular mechanisms of VK2 on insulin sensitivity have not been well established. We investigated VK2's modulation of mitochondrial function to protect against IR in mice and cell models. Results: VK2 supplementation could effectively ameliorate the development of IR by improving mitochondrial function in both HFD-fed mice and palmitate acid-exposed cells. We revealed for the first time that HFD-caused mitochondrial dysfunction could be reversed by VK2 treatment. VK2 enhanced the mitochondrial function by improving mitochondrial respiratory capacity, increasing mitochondrial biogenesis and the enzymatic activities of mitochondrial complexes through SIRT1 signaling. The benefits of VK2 were abrogated in C2C12 transfected with SIRT1 siRNA but not in C2C12 transfected with AMPK siRNA. VK2 and SRT1720, a specific agonist of SIRT1, had the same effect on improving mitochondrial function via SIRT1 signaling. Thus, SIRT1 is required for VK2 improvement in skeletal muscle. Further, the beneficial effects of both VK2 and geranylgeraniol contribute to inhibited IR in skeletal muscle via SIRT1. Innovation and Conclusion: These studies demonstrated a previously undiscovered mechanism by which VK2 alleviates IR in skeletal muscle by improving mitochondrial function via SIRT1. Naturally occurring VK2 prevents IR by improving mitochondrial function through SIRT1 signaling. These results could provide a foundation to identify new VK2-based preventive and therapeutic strategies for IR.
Collapse
Affiliation(s)
- Xiangni Su
- Department of Nutrition and Food Hygiene, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Wenchen Wang
- Department of Thoracic Surgery, and The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Congwen Fang
- Department of Nutrition and Food Hygiene, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Chunping Ni
- Department of Nursing, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Jian Zhou
- Department of Nutrition and Food Hygiene, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Xiaohui Wang
- Department of Nutrition and Food Hygiene, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Lei Zhang
- Department of Nutrition and Food Hygiene, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Xiaona Xu
- Department of Nutrition and Food Hygiene, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Rui Cao
- Department of Nutrition and Food Hygiene, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Hongjuan Lang
- Department of Nursing, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| | - Feng Wang
- Department of Nutrition and Food Hygiene, The Fourth Military Medical University of People's Liberation Army, Xi'an, China
| |
Collapse
|
17
|
Tutunchi H, Ostadrahimi A, Saghafi-Asl M, Maleki V. The effects of oleoylethanolamide, an endogenous PPAR-α agonist, on risk factors for NAFLD: A systematic review. Obes Rev 2019; 20:1057-1069. [PMID: 31111657 DOI: 10.1111/obr.12853] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/04/2019] [Accepted: 03/04/2019] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease. Recently, some novel compounds have been investigated for the prevention and treatment of NAFLD. Oleoylethanolamide (OEA), an endogenous PPAR-α agonist, has exhibited a plethora of pharmacological properties for the treatment of obesity and other obesity-associated metabolic complications. This systematic review was performed with a focus on the effects of OEA on the risk factors for NAFLD. PubMed, Scopus, Embase, ProQuest, and Google Scholar databases were searched up to December 2018 using relevant keywords. All articles written in English evaluating the effects of OEA on the risk factors for NAFLD were eligible for the review. The evidence reviewed in this article illustrates that OEA regulates multiple biological processes associated with NAFLD, including lipid metabolism, inflammation, oxidative stress, and energy homeostasis through different mechanisms. In summary, many beneficial effects of OEA have led to the understanding that OEA may be an effective therapeutic strategy for the management of NAFLD. Although a wide range of studies have demonstrated the most useful effects of OEA on NAFLD and the associated risk factors, further clinical trials, from both in vivo studies and in vitro experiments, are warranted to verify these outcomes.
Collapse
Affiliation(s)
- Helda Tutunchi
- Student Research Committee, Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Saghafi-Asl
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Maleki
- Student Research Committee, Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Kim SH, Hong SH, Park YJ, Sung JH, Suh W, Lee KW, Jung K, Lim C, Kim JH, Kim H, Park KS, Park SG. MD001, a Novel Peroxisome Proliferator-activated Receptor α/γ Agonist, Improves Glucose and Lipid Metabolism. Sci Rep 2019; 9:1656. [PMID: 30733541 PMCID: PMC6367362 DOI: 10.1038/s41598-018-38281-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 12/20/2018] [Indexed: 01/14/2023] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR)-α/γ dual agonists have been developed to treat metabolic diseases; however, most of them exhibit side effects such as body weight gain and oedema. Therefore, we developed a novel PPARα/γ dual agonist that modulates glucose and lipid metabolism without adverse effects. We synthesised novel compounds composed of coumarine and chalcone, determined their crystal structures, and then examined their binding affinity toward PPARα/γ. We investigated the expression of PPARα and PPARγ target genes by chemicals in HepG2, differentiated 3T3-L1, and C2C12 cells. We examined the effect of chemicals on glucose and lipid metabolism in db/db mice. Only MD001 functions as a PPARα/γ dual agonist in vitro. MD001 increased the transcriptional activity of PPARα and PPARγ, resulting in enhanced expression of genes related to β-oxidation and fatty acid and glucose uptake. MD001 significantly improved blood metabolic parameters, including triglycerides, free fatty acids, and glucose, in db/db mice. In addition, MD001 ameliorated hepatic steatosis by stimulating β-oxidation in vitro and in vivo. Our results demonstrated the beneficial effects of the novel compound MD001 on glucose and lipid metabolism as a PPARα/γ dual agonist. Consequently, MD001 may show potential as a novel drug candidate for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Seok-Ho Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, Korea
| | - Shin Hee Hong
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Korea
| | - Young-Joon Park
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Korea
| | - Jong-Hyuk Sung
- College of Pharmacy, Yonsei University, Songdo, Incheon, 405-750, Korea
| | - Wonhee Suh
- College of Pharmacy, Chung-Ang University, Seoul, 156-756, Korea
| | - Kyeong Won Lee
- Marine Biotechnology Research Center, Korea Institute of Ocean Science & Technology 787 Haeanlo, Ansan, Gyeonggi-do, 426-744, Korea
| | - Kiwon Jung
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, Korea
| | - Changjin Lim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, Korea
| | - Jin-Hee Kim
- College of Pharmacy, Yonsei University, Songdo, Incheon, 405-750, Korea
| | - Hyoungsu Kim
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Korea
| | - Kyong Soo Park
- The Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Sang Gyu Park
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Korea.
| |
Collapse
|
19
|
Rodriguez‐Cuenca S, Carobbio S, Barceló‐Coblijn G, Prieur X, Relat J, Amat R, Campbell M, Dias AR, Bahri M, Gray SL, Vidal‐Puig A. P465L-PPARγ mutation confers partial resistance to the hypolipidaemic action of fibrates. Diabetes Obes Metab 2018; 20:2339-2350. [PMID: 29790245 PMCID: PMC6589924 DOI: 10.1111/dom.13370] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/04/2018] [Accepted: 05/12/2018] [Indexed: 12/13/2022]
Abstract
AIMS Familial partial lipodystrophic syndrome 3 (FPLD3) is associated with mutations in the transcription factor PPARγ. One of these mutations, the P467L, confers a dominant negative effect. We and others have previously investigated the pathophysiology associated with this mutation using a humanized mouse model that recapitulates most of the clinical symptoms observed in patients who have been phenotyped under different experimental conditions. One of the key clinical manifestations observed, both in humans and mouse models, is the ectopic accumulation of fat in the liver. With this study we aim to dissect the molecular mechanisms that contribute to the excessive accumulation of lipids in the liver and characterize the negative effect of this PPARγ mutation on the activity of PPARα in vivo when activated by fibrates. MATERIAL AND METHODS P465L-PPAR mutant and wild-type mice were divided into 8 experimental groups, 4 different conditions per genotype. Briefly, mice were fed a chow diet or a high-fat diet (HFD 45% Kcal from fat) for a period of 28 days and treated with WY14643 or vehicle for five days before culling. At the end of the experiment, tissues and plasma were collected. We performed extensive gene expression, fatty acid composition and histological analysis in the livers. The serum collected was used to measure several metabolites and to perform basic lipoprotein profile. RESULTS P465L mice showed increased levels of insulin and free fatty acids (FFA) as well as increased liver steatosis. They also exhibit decreased levels of very low density lipoproteins (VLDL) when fed an HFD. We also provide evidence of impaired expression of a number of well-established PPARα target genes in the P465L mutant livers. CONCLUSION Our data demonstrate that P465L confers partial resistance to the hypolipidemic action of fibrates. These results show that the fatty liver phenotype observed in P465L mutant mice is not only the consequence of dysfunctional adipose tissue, but also involves defective liver metabolism. All in all, the deleterious effects of P465L-PPARγ mutation may be magnified by their collateral negative effect on PPARα function.
Collapse
Affiliation(s)
- Sergio Rodriguez‐Cuenca
- University of Cambridge Metabolic Research Laboratories, Level 4Wellcome Trust‐MRC Institute of Metabolic ScienceCambridgeUK
| | - Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Level 4Wellcome Trust‐MRC Institute of Metabolic ScienceCambridgeUK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome CampusHinxtonUK
| | - Gwendolyn Barceló‐Coblijn
- Institut d'Investigació Sanitària Illes Balears (IdISBa, Balearic Islands Health Research Institute)PalmaSpain
| | - Xavier Prieur
- Département des Sciences de la Vie, L'Institut du Thorax, INSERM, CNRSUniversité de NantesNantesFrance
| | - Joana Relat
- Department of Nutrition, Food Science and Gastronomy, School of Pharmacy and Food Science, Food and Nutrition Torribera Campus. University of Barcelona (UB), Santa Coloma de Gramenet (Spain); INSA‐UB, Nutrition and Food Safety Research InstituteUniversity of BarcelonaBarcelonaSpain
| | - Ramon Amat
- Cell Signaling Unit, Departament de Ciències Experimentals i de la SalutUniversitat Pompeu Fabra (UPF)BarcelonaSpain
| | - Mark Campbell
- University of Cambridge Metabolic Research Laboratories, Level 4Wellcome Trust‐MRC Institute of Metabolic ScienceCambridgeUK
| | - Ana Rita Dias
- University of Cambridge Metabolic Research Laboratories, Level 4Wellcome Trust‐MRC Institute of Metabolic ScienceCambridgeUK
| | - Myriam Bahri
- University of Cambridge Metabolic Research Laboratories, Level 4Wellcome Trust‐MRC Institute of Metabolic ScienceCambridgeUK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome CampusHinxtonUK
| | - Sarah L. Gray
- Northern Medical ProgramUniversity of Northern British ColumbiaPrince GeorgeCanada
| | - Antonio Vidal‐Puig
- University of Cambridge Metabolic Research Laboratories, Level 4Wellcome Trust‐MRC Institute of Metabolic ScienceCambridgeUK
| |
Collapse
|
20
|
Lee J, Kim E, Kim Y, Yoo SH. Leucrose, a Sucrose Isomer, Suppresses Hepatic Fat Accumulation by Regulating Hepatic Lipogenesis and Fat Oxidation in High-fat Diet-induced Obese Mice. J Cancer Prev 2018; 23:99-106. [PMID: 30003071 PMCID: PMC6037208 DOI: 10.15430/jcp.2018.23.2.99] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/21/2018] [Accepted: 06/24/2018] [Indexed: 11/15/2022] Open
Abstract
Obesity is currently one of the most serious public health problems and it can lead to numerous metabolic diseases. Leucrose, d-glucopyranosyl-α-(1-5)-d-fructopyranose, is an isoform of sucrose and it is naturally found in pollen and honey. The aim of this study was to investigate the effect of leucrose on metabolic changes induced by a high-fat diet (HFD) that lead to obesity. C57BL/6 mice were fed a 60% HFD or a HFD with 25% (L25) or 50% (L50) of its total sucrose content replaced with leucrose for 12 weeks. Leucrose supplementation improved fasting blood glucose levels and hepatic triglyceride content. In addition, leucrose supplementation reduced mRNA levels of lipogenesis-related genes, including peroxisome proliferator-activated receptor γ, sterol regulatory element binding protein 1C, and fatty acid synthase in HFD mice. Conversely, mRNA levels of β oxidation-related genes, such as carnitine palmitoyltransferase 1A and acyl CoA oxidase, returned to control levels with leucrose supplementation. Taken together, these results demonstrated the therapeutic potential of leucrose to prevent metabolic abnormalities by mediating regulation of plasma glucose level and hepatic triglyceride accumulation.
Collapse
Affiliation(s)
- Jihye Lee
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Eunju Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Yuri Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Sang-Ho Yoo
- Department of Food Science and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul, Korea
| |
Collapse
|
21
|
Zhao Z, Xu D, Wang Z, Wang L, Han R, Wang Z, Liao L, Chen Y. Hepatic PPARα function is controlled by polyubiquitination and proteasome-mediated degradation through the coordinated actions of PAQR3 and HUWE1. Hepatology 2018; 68:289-303. [PMID: 29331071 PMCID: PMC6055728 DOI: 10.1002/hep.29786] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 12/16/2017] [Accepted: 01/10/2018] [Indexed: 01/14/2023]
Abstract
UNLABELLED Peroxisome proliferator-activated receptor α (PPARα) is a key transcriptional factor that regulates hepatic lipid catabolism by stimulating fatty acid oxidation and ketogenesis in an adaptive response to nutrient starvation. However, how PPARα is regulated by posttranslational modification is poorly understood. In this study, we identified that progestin and adipoQ receptor 3 (PAQR3) promotes PPARα ubiquitination through the E3 ubiquitin ligase HUWE1, thereby negatively modulating PPARα functions both in vitro and in vivo. Adenovirus-mediated Paqr3 knockdown and liver-specific deletion of the Paqr3 gene reduced hepatic triglyceride levels while increasing fatty acid oxidation and ketogenesis upon fasting. PAQR3 deficiency enhanced the fasting-induced expression of PPARα target genes, including those involved in fatty acid oxidation and fibroblast growth factor 21, a key molecule that mediates the metabolism-modulating effects of PPARα. PAQR3 directly interacted with PPARα and increased the polyubiquitination and proteasome-mediated degradation of PPARα. Furthermore, the E3 ubiquitin ligase HUWE1 was identified to mediate PPARα polyubiquitination. Additionally, PAQR3 enhanced the interaction between HUWE1 and PPARα. CONCLUSION Ubiquitination modification through the coordinated action of PAQR3 with HUWE1 plays a crucial role in regulating the activity of PPARα in response to starvation. (Hepatology 2018;68:289-303).
Collapse
Affiliation(s)
- Zilong Zhao
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological SciencesUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Daqian Xu
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological SciencesUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Zheng Wang
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological SciencesUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Lin Wang
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological SciencesUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Ruomei Han
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological SciencesUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Zhenzhen Wang
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological SciencesUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Lujian Liao
- Shanghai Key Laboratory of Regulatory Biology, School of Life SciencesEast China Normal UniversityShanghaiChina
| | - Yan Chen
- CAS Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological SciencesUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
22
|
Jung HY, Kim B, Ryu HG, Ji Y, Park S, Choi SH, Lee D, Lee IK, Kim M, Lee YJ, Song W, Lee YH, Choi HJ, Hyun CK, Holzapfel WH, Kim KT. Amodiaquine improves insulin resistance and lipid metabolism in diabetic model mice. Diabetes Obes Metab 2018. [PMID: 29516607 DOI: 10.1111/dom.13284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIMS Although peroxisome proliferator-activated receptors (PPARs)α/γ dual agonists can be beneficial for treatment of dyslipidemia in patients with type 2 diabetes, their use is limited owing to various side effects, including body weight gain, edema, and heart failure. We aimed to demonstrate that amodiaquine, an antimalarial agent, has potential as a PPARα/γ dual agonist with low risk of adverse effects. METHODS We screened a Prestwick library (Prestwick Chemical; Illkirch, France) to identify novel PPARα/γ dual agonists and selected amodiaquine (4-[(7-chloroquinolin-4-yl)amino]-2-[(diethylamino)methyl]phenol), which activated both PPAR-α & -γ, for further investigation. We performed both in vitro, including glucose uptake assay and fatty acid oxidation assay, and in vivo studies to elucidate the anti-diabetic and anti-obesity effects of amodiaquine. RESULTS Amodiaquine selectively activated the transcriptional activities of PPARα/γ and enhanced both fatty acid oxidation and glucose uptake without altering insulin secretion in vitro. In high-fat diet-induced obese and genetically modified obese/diabetic mice, amodiaquine not only remarkably ameliorated insulin resistance, hyperlipidemia, and fatty liver but also decreased body weight gain. CONCLUSION Our findings suggest that amodiaquine exerts beneficial effects on glucose and lipid metabolism by concurrent activation of PPARα/γ. Furthermore, amodiaquine acts as an alternative insulin-sensitizing agent with a positive influence on lipid metabolism and has potential to prevent and treat type 2 diabetes while reducing the risk of lipid abnormalities.
Collapse
Affiliation(s)
- Hoe-Yune Jung
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- R&D Center, NovMetaPharma Co., Ltd., Pohang, Republic of Korea
| | - Bobae Kim
- Department of Advanced Green Energy and Environment, Handong Global University, Pohang, Republic of Korea
- School of Life Science, Handong Global University, Pohang, Republic of Korea
| | - Hye Guk Ryu
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Yosep Ji
- Department of Advanced Green Energy and Environment, Handong Global University, Pohang, Republic of Korea
| | - Soyoung Park
- Department of Advanced Green Energy and Environment, Handong Global University, Pohang, Republic of Korea
| | - Seung Hee Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Dohyun Lee
- R&D Center, NovMetaPharma Co., Ltd., Pohang, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Republic of Korea
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Munki Kim
- Bio Convergence Team, Advanced Bio Convergence Center, Pohang, Republic of Korea
| | - You Jeong Lee
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- Academy of Immunology and Microbiology, Institute for Basic Science (IBS), Pohang, Republic of Korea
| | - Woojin Song
- Functional Neuroanatomy of Metabolism Regulation Laboratory, Department of Anatomy, Division of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Hee Lee
- Functional Neuroanatomy of Metabolism Regulation Laboratory, Department of Anatomy, Division of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyung Jin Choi
- Functional Neuroanatomy of Metabolism Regulation Laboratory, Department of Anatomy, Division of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chang-Kee Hyun
- School of Life Science, Handong Global University, Pohang, Republic of Korea
| | - Wilhelm H Holzapfel
- Department of Advanced Green Energy and Environment, Handong Global University, Pohang, Republic of Korea
| | - Kyong-Tai Kim
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| |
Collapse
|
23
|
Li G, Brocker CN, Xie C, Yan T, Noguchi A, Krausz KW, Xiang R, Gonzalez FJ. Hepatic peroxisome proliferator-activated receptor alpha mediates the major metabolic effects of Wy-14643. J Gastroenterol Hepatol 2018; 33:1138-1145. [PMID: 29141109 PMCID: PMC6334298 DOI: 10.1111/jgh.14046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/13/2017] [Accepted: 11/06/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Peroxisome proliferator-activated receptor alpha (PPARα) is a molecular target of various fibrate drugs clinically used to lower serum lipids. However, the tissue-specific functions of PPARα remain to be elucidated. This study aimed to explore the tissue-specific functions of PPARα in response to Wy-14643. METHODS A hepatocyte-specific Ppara knockout mouse line was used to explore the impact of hepatic PPARα activity on the systemic response to treatment with the potent PPARα agonist Wy-14643. RESULTS Wy-14643 mainly activated hepatic PPARα and regulated the expression of PPARα target genes in liver. Hepatic Ppara disruption abolished the triglyceride lowering effects of Wy-14643, prevented agonist-induced hypophagia, and ablated PPARα target gene response in the liver. CONCLUSIONS These findings indicate that Wy-14643 treatment mainly activates hepatic PPARα, and the hypolipidemic and hypophagic effects of Wy-14643 are dependent on PPARα activation within hepatocytes.
Collapse
Affiliation(s)
- Guolin Li
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA,Laboratory of Aging Biochemistry, College of Life Sciences, Changsha, Hunan, China,The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Chad N Brocker
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Tingting Yan
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Audrey Noguchi
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Rong Xiang
- The State Key Laboratory of Medical Genetics and School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
24
|
Tewari N, Awad S, Duška F, Williams JP, Bennett A, Macdonald IA, Lobo DN. Postoperative inflammation and insulin resistance in relation to body composition, adiposity and carbohydrate treatment: A randomised controlled study. Clin Nutr 2018; 38:204-212. [PMID: 29454501 PMCID: PMC6380471 DOI: 10.1016/j.clnu.2018.01.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 01/25/2018] [Indexed: 12/17/2022]
Abstract
Background & aims The aims of this study were to identify whether differences in distribution of adipose tissue and skeletal muscle in obese and non-obese individuals contribute to the magnitude of the postoperative inflammatory response and insulin resistance, with and without preoperative treatment with carbohydrate drinks. Methods Thirty-two adults (16 obese/16 non-obese) undergoing elective major open abdominal surgery participated in this 2 × 2 factorial, randomised, double-blind, placebo-controlled study. Participants received Nutricia preOp® or placebo (800 ml on the night before surgery/400 ml 2–3 h preoperatively) after stratifying for obesity. Insulin sensitivity was measured using the hyperinsulinaemic-euglycaemic clamp preoperatively and on the 1st postoperative day. Vastus lateralis, omental and subcutaneous fat biopsies were taken pre- and postoperatively and analysed after RNA extraction. The primary endpoint was within subject differences in insulin sensitivity. Results Major abdominal surgery was associated with a 42% reduction in insulin sensitivity from mean(SD) M value of 37.3(11.8) μmol kg−1 fat free mass (FFM) to 21.7(7.4) μmol kg−1 FFM, but this was not influenced by obesity or preoperative carbohydrate treatment. Activation of the triggering receptor expressed on myeloid cells (TREM1) pathway was seen in response to surgery in omental fat samples. In postoperative muscle samples, gene expression differences indicated activation of the peroxisome proliferator-activated receptor (PPAR-α)/retinoid X-receptor (RXR-α) pathway in obese but not in non-obese participants. There were no significant changes in gene expression pathways associated with carbohydrate treatment. Conclusion The reduction in insulin sensitivity associated with major abdominal surgery was confirmed but there were no differences associated with preoperative carbohydrates or obesity.
Collapse
Affiliation(s)
- Nilanjana Tewari
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Sherif Awad
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK; The East-Midlands Bariatric and Metabolic Institute (EMBMI), Derby Teaching Hospitals NHS Foundation Trust, Royal Derby Hospital, Derby DE22 3NE, UK
| | - František Duška
- Department of Anaesthesia and Intensive Care, Kralovske Vinohrady University Hospital and The Third Faculty of Medicine, Prague, Czech Republic; Department of Critical Care, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Julian P Williams
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Andrew Bennett
- FRAME Laboratory, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK; School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Ian A Macdonald
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK; MRC/ARUK Centre for Musculoskeletal Ageing Research, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Dileep N Lobo
- Gastrointestinal Surgery, Nottingham Digestive Diseases Centre, National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK; MRC/ARUK Centre for Musculoskeletal Ageing Research, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| |
Collapse
|
25
|
Abstract
Obesity is a worldwide epidemic that predisposes individuals to cardiometabolic complications, such as type 2 diabetes mellitus (T2DM) and nonalcoholic fatty liver disease (NAFLD), which are all related to inappropriate ectopic lipid deposition. Identification of the pathogenic molecular mechanisms and effective therapeutic approaches are highly needed. The peroxisome proliferator-activated receptors (PPARs) modulate several biological processes that are perturbed in obesity, including inflammation, lipid and glucose metabolism and overall energy homeostasis. Here, we review how PPARs regulate the functions of adipose tissues, such as adipogenesis, lipid storage and adaptive thermogenesis, under healthy and pathological conditions. We also discuss the clinical use and mechanism of PPAR agonists in the treatment of obesity comorbidities such as dyslipidaemia, T2DM and NAFLD. First generation PPAR agonists, primarily those acting on PPARγ, are associated with adverse effects that outweigh their clinical benefits, which led to the discontinuation of their development. An improved understanding of the physiological roles of PPARs might, therefore, enable the development of safe, new PPAR agonists with improved therapeutic potential.
Collapse
Affiliation(s)
- Barbara Gross
- Université de Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Michal Pawlak
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, 02-109 Warsaw, Poland
| | - Philippe Lefebvre
- Université de Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Bart Staels
- Université de Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| |
Collapse
|
26
|
Yagi S, Kaido T, Iida T, Yoshizawa A, Okajima H, Uemoto S. New-onset diabetes mellitus after living-donor liver transplantation: association with graft synthetic function. Surg Today 2016; 47:733-742. [PMID: 27837276 DOI: 10.1007/s00595-016-1444-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 09/20/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND PURPOSE It is now known that post-transplant graft function after deceased-donor liver transplantation and living-donor liver transplantation (LDLT) differ; however, there is no report assessing the relationship between graft function and the development of new-onset diabetes mellitus after transplantation (NODAT). We conducted this study to identify the predictive risk factors for NODAT, including graft function after LDLT. METHODS The subjects of this study were 175 adult recipients who underwent LDLT at Kyoto University Hospital between 2006 and 2010, and survived for more than 3 months (median observation period, 1046 days). RESULTS The 1-, 2-, and 3-year incidences of NODAT after LDLT were 26.1, 32.0, and 33.4%, respectively. Pre-transplant diabetes was associated with poor survival (p = 0.0048), whereas NODAT was not associated with patient survival. In the multivariate analysis, recipient age ≥40, a tacrolimus trough level ≥8 ng/mL 3 months after LDLT, and cholinesterase (ChE) <185 IU/L 3 months after LDLT were the independent risk factors for NODAT. CONCLUSIONS Poor graft synthetic function 3 months after LDLT as well as older age of the recipient and a higher tacrolimus concentration were strongly associated with NODAT development after LDLT.
Collapse
Affiliation(s)
- Shintaro Yagi
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Toshimi Kaido
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taku Iida
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Atsushi Yoshizawa
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hideaki Okajima
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shinji Uemoto
- Department of Hepatobiliary, Pancreas and Transplant Surgery, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
27
|
Guo Y, Yu J, Deng J, Liu B, Xiao Y, Li K, Xiao F, Yuan F, Liu Y, Chen S, Guo F. A Novel Function of Hepatic FOG2 in Insulin Sensitivity and Lipid Metabolism Through PPARα. Diabetes 2016; 65:2151-63. [PMID: 27207553 DOI: 10.2337/db15-1565] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 04/21/2016] [Indexed: 11/13/2022]
Abstract
Friend of GATA 2 (FOG2) is a transcriptional cofactor involved mostly in cardiac function. The aim of this study was to investigate the role of hepatic FOG2 in insulin sensitivity and lipid accumulation. FOG2 overexpression by adenovirus-expressing FOG2 (Ad-FOG2) significantly attenuates insulin signaling in hepatocytes in vitro. Opposite effects were observed when FOG2 was knocked down through adenovirus-expressing small hairpin RNA for FOG2 (Ad-shFOG2). Furthermore, FOG2 knockdown by Ad-shFOG2 ameliorated insulin resistance in leptin receptor-mutated (db/db) mice, and FOG2 overexpression by Ad-FOG2 attenuated insulin sensitivity in C57BL/6J wild-type (WT) mice. In addition, Ad-FOG2 reduced, whereas Ad-shFOG2 promoted, hepatic triglyceride (TG) accumulation in WT mice under fed or fasted conditions, which was associated with increased or decreased hepatic peroxisome proliferator-activated receptor α (PPARα) expression, respectively. Moreover, the improved insulin sensitivity and increased hepatic TG accumulation by Ad-shFOG2 were largely reversed by adenovirus-expressing PPARα (Ad-PPARα) in WT mice. Finally, we generated FOG2 liver-specific knockout mice and found that they exhibit enhanced insulin sensitivity and elevated hepatic TG accumulation, which were also reversed by Ad-PPARα. Taken together, the results demonstrate a novel function of hepatic FOG2 in insulin sensitivity and lipid metabolism through PPARα.
Collapse
Affiliation(s)
- Yajie Guo
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Junjie Yu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Jiali Deng
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Bin Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Yuzhong Xiao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Kai Li
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Fei Xiao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Feixiang Yuan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Yong Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Shanghai Chen
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
28
|
Bae JC, Han JM, Kwon S, Jee JH, Yu TY, Lee MK, Kim JH. LDL-C/apoB and HDL-C/apoA-1 ratios predict incident chronic kidney disease in a large apparently healthy cohort. Atherosclerosis 2016; 251:170-176. [DOI: 10.1016/j.atherosclerosis.2016.06.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/25/2016] [Accepted: 06/15/2016] [Indexed: 11/27/2022]
|
29
|
Massaro M, Scoditti E, Pellegrino M, Carluccio MA, Calabriso N, Wabitsch M, Storelli C, Wright M, De Caterina R. Therapeutic potential of the dual peroxisome proliferator activated receptor (PPAR)α/γ agonist aleglitazar in attenuating TNF-α-mediated inflammation and insulin resistance in human adipocytes. Pharmacol Res 2016; 107:125-136. [PMID: 26976796 DOI: 10.1016/j.phrs.2016.02.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 12/13/2022]
Abstract
Adipose tissue inflammation is a mechanistic link between obesity and its related sequelae, including insulin resistance and type 2 diabetes. Dual ligands of peroxisome proliferator activated receptor (PPAR)α and γ, combining in a single molecule the metabolic and inflammatory-regulatory properties of α and γ agonists, have been proposed as a promising therapeutic strategy to antagonize adipose tissue inflammation. Here we investigated the effects of the dual PPARα/γ agonist aleglitazar on human adipocytes challenged with inflammatory stimuli. Human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes were treated with aleglitazar or - for comparison - the selective agonists for PPARα or γ fenofibrate or rosiglitazone, respectively, for 24h before stimulation with TNF-α. Aleglitazar, at concentrations as low as 10nmol/L, providing the half-maximal transcriptional activation of both PPARα and PPARγ, reduced the stimulated expression of several pro-inflammatory mediators including interleukin (IL)-6, the chemokine CXC-L10, and monocyte chemoattractant protein (MCP)-1. Correspondingly, media from adipocytes treated with aleglitazar reduced monocyte migration, consistent with suppression of MCP-1 secretion. Under the same conditions, aleglitazar also reversed the TNF-α-mediated suppression of insulin-stimulated ser473 Akt phosphorylation and decreased the TNF-α-induced ser312 IRS1 phosphorylation, two major switches in insulin-mediated metabolic activities, restoring glucose uptake in insulin-resistant adipocytes. Such effects were similar to those obtainable with a combination of single PPARα and γ agonists. In conclusion, aleglitazar reduces inflammatory activation and dysfunction in insulin signaling in activated adipocytes, properties that may benefit diabetic and obese patients. The effect of aleglitazar was consistent with dual PPARα and γ agonism, but with no evidence of synergism.
Collapse
Affiliation(s)
- Marika Massaro
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Egeria Scoditti
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Mariangela Pellegrino
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy; Department of Biological and Environmental Science and Technology (DISTEBA), University of Salento, Lecce, Italy
| | | | - Nadia Calabriso
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Martin Wabitsch
- Division of Pediatric Endocrinology, Diabetes and Obesity, Department of Pediatrics and Adolescent Medicine, University of Ulm, Germany
| | - Carlo Storelli
- Department of Biological and Environmental Science and Technology (DISTEBA), University of Salento, Lecce, Italy
| | | | - Raffaele De Caterina
- G. dAnnunzio University and Center of Excellence on Aging, Chieti, Italy; G. Monasterio Foundation for Clinical Research, Pisa, Italy.
| |
Collapse
|
30
|
Orime K, Shirakawa J, Togashi Y, Tajima K, Inoue H, Nagashima Y, Terauchi Y. Lipid-lowering agents inhibit hepatic steatosis in a non-alcoholic steatohepatitis-derived hepatocellular carcinoma mouse model. Eur J Pharmacol 2015; 772:22-32. [PMID: 26724391 DOI: 10.1016/j.ejphar.2015.12.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/22/2015] [Accepted: 12/23/2015] [Indexed: 01/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is associated with various metabolic disorders, and the therapeutic strategies for treating NAFLD and non-alcoholic steatohepatitis (NASH) have not been fully established. In the present study, we examined whether lipid-lowering agents inhibited the progression of NAFLD and tumorigenesis in a non-alcoholic steatohepatitis-derived hepatocellular carcinoma model mouse (STAM mice) generated by streptozotocin injection and a high-fat diet. Seven-week-old STAM mice were divided into groups fed a high-fat diet (Ctl) or a high-fat diet supplemented with ezetimibe (Ez), fenofibrate (Ff), rosuvastatin (Rs), ezetimibe plus fenofibrate (EF), or ezetimibe plus rosuvastatin (ER) for 4 weeks. At the end of the experiments, an oral glucose tolerance test, an insulin tolerance test, biochemical analyses using serum and liver, and a histological analysis of liver were performed in 11-week-old STAM mice. The lipid-lowering agents did not affect the body weight or the casual blood glucose levels in any of the groups. The serum triglyceride level was significantly decreased by Ff, Rs, and EF. Glucose tolerance was improved by Ez and Ff, but none of these agents improved insulin sensitivity. A histochemical analysis revealed that the lipid-lowering agents, with the exception of Rs, significantly inhibited the progression of hepatic steatosis. Nonetheless, no significant changes in the incidence of hepatic tumors were observed in any of the groups. Lipid-lowering agents inhibited the progression of hepatic steatosis without suppressing tumorigenesis in STAM mice. Our data has implications for the mechanism underlying steatosis-independent hepatic tumorigenesis in mice.
Collapse
Affiliation(s)
- Kazuki Orime
- Department of Endocrinology and Metabolism, Yokohama-City University, Yokohama, Japan
| | - Jun Shirakawa
- Department of Endocrinology and Metabolism, Yokohama-City University, Yokohama, Japan
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Yokohama-City University, Yokohama, Japan
| | - Kazuki Tajima
- Department of Endocrinology and Metabolism, Yokohama-City University, Yokohama, Japan
| | - Hideaki Inoue
- Department of Endocrinology and Metabolism, Yokohama-City University, Yokohama, Japan
| | - Yoji Nagashima
- Department of Molecular Pathology, Yokohama-City University, Yokohama, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Yokohama-City University, Yokohama, Japan.
| |
Collapse
|
31
|
Effect of dietary amylose/amylopectin ratio on growth performance, carcass traits, and meat quality in finishing pigs. Meat Sci 2015; 108:55-60. [DOI: 10.1016/j.meatsci.2015.05.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/10/2015] [Accepted: 05/26/2015] [Indexed: 01/10/2023]
|
32
|
Cuadros DF, Miller FD, Nagelkerke N, Abu-Raddad LJ. Association between HCV infection and diabetes type 2 in Egypt: is it time to split up? Ann Epidemiol 2015; 25:918-23. [PMID: 26499381 DOI: 10.1016/j.annepidem.2015.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/06/2015] [Accepted: 09/07/2015] [Indexed: 12/15/2022]
Abstract
PURPOSE There is a conflicting evidence about the association between hepatitis C virus (HCV) infection and diabetes mellitus. The objective of this study was to assess this association in Egypt, the country with the highest HCV prevalence in the world. METHODS The source of data was from the Egypt Demographic and Health Survey conducted in 2008. Using multivariable logistic regression analyses to account for known confounders, the association was investigated at two levels']: (1) HCV exposure (HCV antibody status) and diabetes mellitus and (2) diabetes mellitus and chronic HCV infection (HCV RNA status) among HCV-exposed individuals. RESULTS We found no evidence for an association between HCV antibody status and diabetes (adjusted odds ratio [OR] = 0.87; 95% confidence interval [CI], 0.63-1.19). However, among HCV-exposed individuals, we found an evidence for an association between diabetes and active HCV infection (adjusted OR = 2.44, 95% CI, 1.30-4.57). CONCLUSIONS Although it does not appear that HCV exposure and diabetes are linked, there might be an association between diabetes and chronic HCV infection. The HCV-diabetes relationship may be more complex than previously anticipated. Therefore, a call for an "amicable divorce" to the HCV-diabetes relationship could be premature.
Collapse
Affiliation(s)
- Diego F Cuadros
- Infectious Disease Epidemiology Group, Weill Cornell Medical College-Qatar, Cornell University, Qatar Foundation-Education City, Doha, Qatar; Department of Healthcare Policy and Research, Weill Cornell Medical College, Cornell University, New York, NY.
| | - F DeWolfe Miller
- Department of Tropical Medicine and Medical Microbiology and Pharmacology, University of Hawaii, Honolulu
| | - Nico Nagelkerke
- Department of Public Health, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Laith J Abu-Raddad
- Infectious Disease Epidemiology Group, Weill Cornell Medical College-Qatar, Cornell University, Qatar Foundation-Education City, Doha, Qatar; Department of Healthcare Policy and Research, Weill Cornell Medical College, Cornell University, New York, NY; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
33
|
Pirinixic acids: flexible fatty acid mimetics with various biological activities. Future Med Chem 2015; 7:1597-616. [DOI: 10.4155/fmc.15.87] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Pirinixic acid is a typical fatty acid mimetic and was developed as synthetic antihyperlipidemic agent. While its target remained unknown in the early development, it has later been characterized as dual PPARα/γ agonist. Based on this activity, pirinixic acid has served as a lead compound for several structure–activity relationship (SAR) studies addressing diverse targets for lipid mimetics. Many structural variants of pirinixic acid descendants have been developed and thereby potent agents on metabolic, inflammatory and neuroprotective targets were discovered of which some have proven in vivo efficacy. This article reviews pirinixic acid descendants along with their in vitro-pharmacological profiles, summarizes their in vivo data and finally gives a future perspective for this valuable class of fatty acid mimetics.
Collapse
|
34
|
Ku CS, Kim B, Pham TX, Yang Y, Weller CL, Carr TP, Park YK, Lee JY. Hypolipidemic Effect of a Blue-Green Alga (Nostoc commune) Is Attributed to Its Nonlipid Fraction by Decreasing Intestinal Cholesterol Absorption in C57BL/6J Mice. J Med Food 2015; 18:1214-22. [PMID: 26161942 DOI: 10.1089/jmf.2014.0121] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We previously demonstrated that Nostoc commune var. sphaeroids Kützing (NO), a blue-green alga (BGA), exerts a hypolipidemic effect in vivo and its lipid extract regulates the expression of genes involved in cholesterol and lipid metabolism in vitro. The objective of this study was to investigate whether the hypolipidemic effect of NO is attributed to an algal lipid or a delipidated fraction in vivo compared with Spirulina platensis (SP). Male C57BL/6J mice were fed an AIN-93M diet containing 2.5% or 5% of BGA (w/w) or a lipid extract equivalent to 5% of BGA for 4 weeks to measure plasma and liver lipids, hepatic gene expression, intestinal cholesterol absorption, and fecal sterol excretion. Plasma total cholesterol (TC) was significantly lower in 2.5% and 5% NO-fed groups, while plasma triglyceride (TG) levels were decreased in the 5% NO group compared with controls. However, neither NO organic extract (NOE) nor SP-fed groups altered plasma lipids. Hepatic mRNA levels of sterol regulatory element-binding protein 2, 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGR), carnitine palmitoyltransferase-1α, and acyl-CoA oxidase 1 were induced in 5% NO-fed mice, while there were no significant changes in hepatic lipogenic gene expression between groups. NO, but not NOE and SP groups, significantly decreased intestinal cholesterol absorption. When HepG2 cells and primary mouse hepatocytes were incubated with NOE and SP organic extract (SPE), there were marked decreases in protein levels of HMGR, low-density lipoprotein receptor, and fatty acid synthase. In conclusion, the nonlipid fraction of NO exerts TC and TG-lowering effects primarily by inhibiting intestinal cholesterol absorption and by increasing hepatic fatty acid oxidation, respectively.
Collapse
Affiliation(s)
- Chai Siah Ku
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut, USA
| | - Bohkyung Kim
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut, USA
| | - Tho X Pham
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut, USA
| | - Yue Yang
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut, USA
| | - Curtis L Weller
- 2 Department of Biological Systems Engineering, University of Nebraska , Lincoln, Nebraska, USA
| | - Timothy P Carr
- 3 Department of Nutrition and Health Sciences, University of Nebraska , Lincoln, Nebraska, USA
| | - Young-Ki Park
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut, USA
| | - Ji-Young Lee
- 1 Department of Nutritional Sciences, University of Connecticut , Storrs, Connecticut, USA
| |
Collapse
|
35
|
Urushima H, Sanada Y, Sakaue M, Matsuzawa Y, Ito T, Maeda K. Maltitol Prevents the Progression of Fatty Liver Degeneration in Mice Fed High-Fat Diets. J Med Food 2015; 18:1081-7. [PMID: 26061453 DOI: 10.1089/jmf.2014.3380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) progresses to nonalcoholic steatohepatitis, ultimately leading to cirrhosis and liver cancer. It is important to prevent this progression during the initial stages of hepatic fatty degeneration. Maltitol is a polyol produced by the hydrogenation of maltose. We investigated the efficacy of maltitol for treating hepatic fatty degeneration in C57BL/6 male mice using a high-fat diet model. Intake of 5.0% maltitol for 8 weeks significantly suppressed weight gain, hepatic fatty degeneration, hyperglycemia, and hypercholesterolemia. With maltitol intake, sterol regulatory element-binding protein 1c (SREBP1c) mRNA expression was significantly decreased, and farnesoid X receptor (FXR), peroxisome proliferator-activated receptor α (PPARα), and hydroxymethylglutaryl-Co reductase expressions were significantly higher in the liver. The increase in SREBP1c and suppression of FXR and PPARα expressions are correlated with NAFLD. Our results suggest that maltitol may prevent steatosis of NAFLD with a high-fat diet.
Collapse
Affiliation(s)
- Hayato Urushima
- 1 Department of Integrative Medicine, Graduate School of Medicine, Osaka University , Osaka, Japan
| | - Yasuaki Sanada
- 1 Department of Integrative Medicine, Graduate School of Medicine, Osaka University , Osaka, Japan
| | - Miki Sakaue
- 1 Department of Integrative Medicine, Graduate School of Medicine, Osaka University , Osaka, Japan
| | | | - Toshinori Ito
- 1 Department of Integrative Medicine, Graduate School of Medicine, Osaka University , Osaka, Japan
| | - Kazuhisa Maeda
- 1 Department of Integrative Medicine, Graduate School of Medicine, Osaka University , Osaka, Japan
| |
Collapse
|
36
|
Deregulated coenzyme A, loss of metabolic flexibility and diabetes. Biochem Soc Trans 2015; 42:1118-22. [PMID: 25110012 DOI: 10.1042/bst20140156] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
CoA (coenzyme A) is an essential cofactor that is emerging as a global regulator of energy metabolism. Tissue CoA levels are tightly regulated and vary in response to different conditions including nutritional state and diabetes. Recent studies reveal the ability of this cofactor to control the output of key metabolic pathways. CoA regulation is important for the maintenance of metabolic flexibility and glucose homoeostasis.
Collapse
|
37
|
Abstract
Microsomal triglyceride transfer protein (MTP) is one of the promising targets for the therapy of dyslipidemia and MTP inhibition can lead to robust plasma low-density lipoprotein cholesterol (LDL-C) reduction. Lomitapide, a small-molecule MTP inhibitor, was recently approved by the US FDA as an additional treatment for homozygous familial hypercholesterolemia (hoFH). However, liver-related side effects, including hepatic fat accumulation and transaminase elevations, are the main safety concerns associated with MTP inhibitors. Here, we review recent knowledge on the mechanisms underlying liver toxicity of MTP inhibitors. The contribution of altered levels of intracellular triglycerides, cholesteryl esters, and free cholesterols toward cellular dysfunction is specifically addressed. On this basis, therapies targeted to attenuate cellular lipid accumulation, to reduce risk factors for non-alcoholic fatty liver disease (NAFLD) (i.e., insulin resistance and oxidative stress) and to specifically inhibit intestinal MTP may be useful for ameliorating liver damage induced by MTP inhibitors. In particular, weight loss through lifestyle interventions is expected to be the most effective and safest way to minimize the undesirable side effects. Specific dietary supplementation might also have protective effects against hepatosteatosis. Despite that, to date, few clinical data support these therapeutic options in MTP inhibition-related liver damage, such proposed approaches may be further explored in the future for their use in preventing unwanted effects of MTP inhibitors.
Collapse
|
38
|
SteatoNet: the first integrated human metabolic model with multi-layered regulation to investigate liver-associated pathologies. PLoS Comput Biol 2014; 10:e1003993. [PMID: 25500563 PMCID: PMC4263370 DOI: 10.1371/journal.pcbi.1003993] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 10/15/2014] [Indexed: 12/15/2022] Open
Abstract
Current state-of-the-art mathematical models to investigate complex biological processes, in particular liver-associated pathologies, have limited expansiveness, flexibility, representation of integrated regulation and rely on the availability of detailed kinetic data. We generated the SteatoNet, a multi-pathway, multi-tissue model and in silico platform to investigate hepatic metabolism and its associated deregulations. SteatoNet is based on object-oriented modelling, an approach most commonly applied in automotive and process industries, whereby individual objects correspond to functional entities. Objects were compiled to feature two novel hepatic modelling aspects: the interaction of hepatic metabolic pathways with extra-hepatic tissues and the inclusion of transcriptional and post-transcriptional regulation. SteatoNet identification at normalised steady state circumvents the need for constraining kinetic parameters. Validation and identification of flux disturbances that have been proven experimentally in liver patients and animal models highlights the ability of SteatoNet to effectively describe biological behaviour. SteatoNet identifies crucial pathway branches (transport of glucose, lipids and ketone bodies) where changes in flux distribution drive the healthy liver towards hepatic steatosis, the primary stage of non-alcoholic fatty liver disease. Cholesterol metabolism and its transcription regulators are highlighted as novel steatosis factors. SteatoNet thus serves as an intuitive in silico platform to identify systemic changes associated with complex hepatic metabolic disorders.
Collapse
|
39
|
Ericsson A, Turner N, Hansson GI, Wallenius K, Oakes ND. Pharmacological PPARα activation markedly alters plasma turnover of the amino acids glycine, serine and arginine in the rat. PLoS One 2014; 9:e113328. [PMID: 25486018 PMCID: PMC4259322 DOI: 10.1371/journal.pone.0113328] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 10/27/2014] [Indexed: 11/18/2022] Open
Abstract
The current study extends previously reported PPARα agonist WY 14,643 (30 µmol/kg/day for 4 weeks) effects on circulating amino acid concentrations in rats fed a 48% saturated fat diet. Steady-state tracer experiments were used to examine in vivo kinetic mechanisms underlying altered plasma serine, glycine and arginine levels. Urinary urea and creatinine excretion were measured to assess whole-body amino acid catabolism. WY 14,643 treated animals demonstrated reduced efficiency to convert food consumed to body weight gain while liver weight was increased compared to controls. WY 14,643 raised total amino acid concentration (38%), largely explained by glycine, serine and threonine increases. 3H-glycine, 14C-serine and 14C-arginine tracer studies revealed elevated rates of appearance (Ra) for glycine (45.5 ± 5.8 versus 17.4 ± 2.7 µmol/kg/min) and serine (21.0 ± 1.4 versus 12.0 ± 1.0) in WY 14,643 versus control. Arginine was substantially decreased (-62%) in plasma with estimated Ra reduced from 3.1 ± 0.3 to 1.2 ± 0.2 µmol/kg/min in control versus WY 14,643. Nitrogen excretion over 24 hours was unaltered. Hepatic arginase activity was substantially decreased by WY 14,643 treatment. In conclusion, PPARα agonism potently alters metabolism of several specific amino acids in the rat. The changes in circulating levels of serine, glycine and arginine reflected altered fluxes into the plasma rather than changes in clearance or catabolism. This suggests that PPARα has an important role in modulating serine, glycine and arginine de novo synthesis.
Collapse
Affiliation(s)
- Anette Ericsson
- Department of Bioscience, AstraZeneca R&D Mölndal, Mölndal, Sweden
- * E-mail:
| | - Nigel Turner
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Pharmacology, University of New South Wales, Sydney, Australia
| | - Göran I. Hansson
- Department of Bioscience, AstraZeneca R&D Mölndal, Mölndal, Sweden
| | | | | |
Collapse
|
40
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a clinicopathological change characterized by the accumulation of triglycerides in hepatocytes and has frequently been associated with obesity, type 2 diabetes mellitus, hyperlipidemia, and insulin resistance. It is an increasingly recognized condition that has become the most common liver disorder in developed countries, affecting over one-third of the population and is associated with increased cardiovascular- and liver-related mortality. NAFLD is a spectrum of disorders, beginning as simple steatosis. In about 15% of all NAFLD cases, simple steatosis can evolve into non-alcoholic steatohepatitis, a medley of inflammation, hepatocellular injury, and fibrosis, often resulting in cirrhosis and even hepatocellular cancer. However, the molecular mechanism underlying NAFLD progression is not completely understood. Its pathogenesis has often been interpreted by the “double-hit” hypothesis. The primary insult or the “first hit” includes lipid accumulation in the liver, followed by a “second hit” in which proinflammatory mediators induce inflammation, hepatocellular injury, and fibrosis. Nowadays, a more complex model suggests that fatty acids (FAs) and their metabolites may be the true lipotoxic agents that contribute to NAFLD progression; a multiple parallel hits hypothesis has also been suggested. In NAFLD patients, insulin resistance leads to hepatic steatosis via multiple mechanisms. Despite the excess hepatic accumulation of FAs in NAFLD, it has been described that not only de novo FA synthesis is increased, but FAs are also taken up from the serum. Furthermore, a decrease in mitochondrial FA oxidation and secretion of very-low-density lipoproteins has been reported. This review discusses the molecular mechanisms that underlie the pathophysiological changes of hepatic lipid metabolism that contribute to NAFLD.
Collapse
Affiliation(s)
- Alba Berlanga
- Group GEMMAIR (AGAUR) and Applied Medicine Research Group, Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), IISPV, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Esther Guiu-Jurado
- Group GEMMAIR (AGAUR) and Applied Medicine Research Group, Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), IISPV, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - José Antonio Porras
- Group GEMMAIR (AGAUR) and Applied Medicine Research Group, Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), IISPV, Hospital Universitari Joan XXIII, Tarragona, Spain ; Department of Internal Medicine, Hospital Universitari Joan XXIII Tarragona, Tarragona, Spain
| | - Teresa Auguet
- Group GEMMAIR (AGAUR) and Applied Medicine Research Group, Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), IISPV, Hospital Universitari Joan XXIII, Tarragona, Spain ; Department of Internal Medicine, Hospital Universitari Joan XXIII Tarragona, Tarragona, Spain
| |
Collapse
|
41
|
Romagnolo DF, Zempleni J, Selmin OI. Nuclear receptors and epigenetic regulation: opportunities for nutritional targeting and disease prevention. Adv Nutr 2014; 5:373-85. [PMID: 25022987 PMCID: PMC4085186 DOI: 10.3945/an.114.005868] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Posttranslational modifications of histones, alterations in the recruitment and functions of non-histone proteins, DNA methylation, and changes in expression of noncoding RNAs contribute to current models of epigenetic regulation. Nuclear receptors (NRs) are a group of transcription factors that, through ligand-binding, act as sensors to changes in nutritional, environmental, developmental, pathophysiologic, and endocrine conditions and drive adaptive responses via gene regulation. One mechanism through which NRs direct gene expression is the assembly of transcription complexes with cofactors and coregulators that possess chromatin-modifying properties. Chromatin modifications can be transient or become part of the cellular "memory" and contribute to genomic imprinting. Because many food components bind to NRs, they can ultimately influence transcription of genes associated with biologic processes, such as inflammation, proliferation, apoptosis, and hormonal response, and alter the susceptibility to chronic diseases (e.g., cancer, diabetes, obesity). The objective of this review is to highlight how NRs influence epigenetic regulation and the relevance of dietary compound-NR interactions in human nutrition and for disease prevention and treatment. Identifying gene targets of unliganded and bound NRs may assist in the development of epigenetic maps for food components and dietary patterns. Progress in these areas may lead to the formulation of disease-prevention models based on epigenetic control by individual or associations of food ligands of NRs.
Collapse
Affiliation(s)
- Donato F Romagnolo
- Department of Nutritional Sciences and University of Arizona Cancer Center, University of Arizona, Tucson, AZ; and
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE
| | - Ornella I Selmin
- Department of Nutritional Sciences and University of Arizona Cancer Center, University of Arizona, Tucson, AZ; and
| |
Collapse
|
42
|
Bang CY, Choung SY. Enzogenol improves diabetes-related metabolic change in C57BL/KsJ-db/db mice, a model of type 2 diabetes mellitus. J Pharm Pharmacol 2014; 66:875-85. [DOI: 10.1111/jphp.12211] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 12/07/2013] [Indexed: 01/09/2023]
Abstract
Abstract
Objectives
Dietary use of pine bark extract has been associated with reduced risk of inflammation and diabetes. In this study, we investigated the antidiabetic effects of enzogenol, proanthocyanidins-rich bioflavonoid extract derived from the pine bark of New Zealand Pinus radiata trees, using C57BL/KsJ-db/db mice.
Methods
After 1-week acclimation period, the db/db mice were divided into vehicle-treated, Enzogenol-treated (12.5, 25 and 50 mg/kg; EZ) and positive control (tea polyphenol 50 mg/kg; TPP) groups.
Key findings
The administration of EZ improved the glucose tolerance and lowered the glycosylated haemoglobin (HbA1C), insulin and glucagon levels in blood. Interestingly, EZ and TPP treatments resulted in reduced hepatic free fatty acid, cholesterol and triglyceride levels in db/db mice. EZ and TPP treatments significantly elevated hepatic AMPK activity, and the expression of proteins related to glucose homeostasis and lipid metabolism, such as glucokinase, peroxisome proliferator-activated receptor (PPAR)α and long-chain acyl-CoA dehydrogenase protein level with a simultaneous reduction of glucose-6-phosphatase and phosphoenolpyruvate carboxykinase protein expression. In addition, the EZ administration groups had an increased hepatic glycogen synthase expression in db/db mice.
Conclusions
These results suggest that EZ may be beneficial in improving insulin resistance and hyperglycaemia in type 2 diabetic mice by enhancing the glucose and lipids metabolism.
Collapse
Affiliation(s)
- Chae-Young Bang
- Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Se-Young Choung
- Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Kubo M, Fukui M, Ito Y, Kitao T, Shirahase H, Hinoi E, Yoneda Y. Insulin Sensitization by a Novel Partial Peroxisome Proliferator-Activated Receptor γ Agonist With Protein Tyrosine Phosphatase 1B Inhibitory Activity in Experimental Osteoporotic Rats. J Pharmacol Sci 2014; 124:276-85. [DOI: 10.1254/jphs.13236fp] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
44
|
Nakamura MT, Yudell BE, Loor JJ. Regulation of energy metabolism by long-chain fatty acids. Prog Lipid Res 2013; 53:124-44. [PMID: 24362249 DOI: 10.1016/j.plipres.2013.12.001] [Citation(s) in RCA: 548] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 12/03/2013] [Accepted: 12/04/2013] [Indexed: 12/12/2022]
Abstract
In mammals, excess energy is stored primarily as triglycerides, which are mobilized when energy demands arise. This review mainly focuses on the role of long chain fatty acids (LCFAs) in regulating energy metabolism as ligands of peroxisome proliferator-activated receptors (PPARs). PPAR-alpha expressed primarily in liver is essential for metabolic adaptation to starvation by inducing genes for beta-oxidation and ketogenesis and by downregulating energy expenditure through fibroblast growth factor 21. PPAR-delta is highly expressed in skeletal muscle and induces genes for LCFA oxidation during fasting and endurance exercise. PPAR-delta also regulates glucose metabolism and mitochondrial biogenesis by inducing FOXO1 and PGC1-alpha. Genes targeted by PPAR-gamma in adipocytes suggest that PPAR-gamma senses incoming non-esterified LCFAs and induces the pathways to store LCFAs as triglycerides. Adiponectin, another important target of PPAR-gamma may act as a spacer between adipocytes to maintain their metabolic activity and insulin sensitivity. Another topic of this review is effects of skin LCFAs on energy metabolism. Specific LCFAs are required for the synthesis of skin lipids, which are essential for water barrier and thermal insulation functions of the skin. Disturbance of skin lipid metabolism often causes apparent resistance to developing obesity at the expense of normal skin function.
Collapse
Affiliation(s)
- Manabu T Nakamura
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana, IL 61801, USA.
| | - Barbara E Yudell
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana, IL 61801, USA
| | - Juan J Loor
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana, IL 61801, USA
| |
Collapse
|
45
|
Lu J, Huang G, Hu S, Wang Z, Guan S. 1,3-Dichloro-2-propanol induced hyperlipidemia in C57BL/6J mice via AMPK signaling pathway. Food Chem Toxicol 2013; 64:403-9. [PMID: 24333398 DOI: 10.1016/j.fct.2013.11.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/26/2013] [Accepted: 11/29/2013] [Indexed: 11/18/2022]
Abstract
1,3-Dichloro-2-propanol (1,3-DCP) is a well-known contaminant that has been detected in a wide range of foods. Dietary intake represents the greatest source of exposure to 1,3-DCP. In the study, we first found 1,3-DCP could induce hyperlipidemia in C57BL/6J mice below 1 mg/kg/day. We investigated serum lipid profile, liver total cholesterol (TC) and triglyceride (TG), histopathology of Liver and adipose tissue. The results showed 1,3-DCP dose dependently increased serum TG, TC and low-density lipoprotein cholesterol (LDL-C), decreased serum high-density lipoprotein cholesterol (HDL-C), increased relative liver weight, liver TG and TC, relative adipose tissue weight and enlarged the size of adipose cells. Because AMPK signal pathway is important in the process of lipid metabolism, we further investigated the effects of 1,3-DCP on AMPK signaling pathway in murine models. The results showed that 1,3-DCP (0.1-1 mg/kg/day) decreased p-AMPK/tAMPK ratio, p-ACC/tACC ratio, PPARα expression, but increased FAT, SREBP1, HMGCR and FAS expression. These observations indicated that 1,3-DCP induced hyperlipidemia in C57BL/6J mice at least partially through regulating AMPK signaling pathway.
Collapse
Affiliation(s)
- Jing Lu
- Department of Food Quality and Safety, Jilin University, Changchun, People's Republic of China
| | - Guoren Huang
- Department of Food Quality and Safety, Jilin University, Changchun, People's Republic of China
| | - Sizhuo Hu
- Department of Food Quality and Safety, Jilin University, Changchun, People's Republic of China
| | - Zhenning Wang
- Department of Food Quality and Safety, Jilin University, Changchun, People's Republic of China
| | - Shuang Guan
- Department of Food Quality and Safety, Jilin University, Changchun, People's Republic of China.
| |
Collapse
|
46
|
Fenofibrate, a peroxisome proliferator-activated receptor α ligand, prevents abnormal liver function induced by a fasting–refeeding process. Biochem Biophys Res Commun 2013; 442:22-7. [DOI: 10.1016/j.bbrc.2013.10.140] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 10/26/2013] [Indexed: 12/18/2022]
|
47
|
Abstract
Alcohol consumption is a major cause of liver disease. It also associates with increased cardiovascular risk and Type 2 diabetes. ALD (alcoholic liver disease) and NAFLD (non-alcoholic fatty liver disease) share pathological features, pathogenic mechanisms and pattern of disease progression. In NAFLD, steatosis, lipotoxicity and liver inflammation participate to hepatic insulin resistance. The aim of the present study was to verify the effect of alcohol on hepatic insulin sensitivity and to evaluate the role of alcohol-induced steatosis and inflammation on glucose homoeostasis. C57BL/6J mice were fed for 20 days a modified Lieber-DeCarli diet in which the alcohol concentration was gradually increased up to 35% of daily caloric intake. OH (alcohol liquid diet)-fed mice had liver steatosis and inflammatory infiltration. In addition, these mice developed insulin resistance in the liver, but not in muscles, as demonstrated by euglycaemic-hyperinsulinaemic clamp and analysis of the insulin signalling cascade. Treatment with the PPAR-α (peroxisome-proliferator-activated receptor-α) agonist Wy14,643 protected against OH-induced steatosis and KC (Kupffer cell) activation and almost abolished OH-induced insulin resistance. As KC activation may modulate insulin sensitivity, we repeated the clamp studies in mice depleted in KC to decipher the role of macrophages. Depletion of KC using liposomes-encapsuled clodronate in OH-fed mice failed both to improve hepatic steatosis and to restore insulin sensitivity as assessed by clamp. Our study shows that chronic alcohol consumption induces steatosis, KC activation and hepatic insulin resistance in mice. PPAR-α agonist treatment that prevents steatosis and dampens hepatic inflammation also prevents alcohol-induced hepatic insulin resistance. However, KC depletion has little impact on OH-induced metabolic disturbances.
Collapse
|
48
|
|
49
|
Lim S, Meigs JB. Ectopic fat and cardiometabolic and vascular risk. Int J Cardiol 2013; 169:166-76. [PMID: 24063931 DOI: 10.1016/j.ijcard.2013.08.077] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 06/16/2013] [Accepted: 08/28/2013] [Indexed: 12/16/2022]
Abstract
Given that the variation in how regional adipose tissue handles and stores excess dietary energy has substantial cardiometabolic implications, ectopic fat distribution might be an important predictor of cardiometabolic and vascular risk, in addition to overall obesity itself. Conceptually, ectopic fat depots may be divided into systemically acting fat depots and locally acting fat depots. Systemically acting fat depots include visceral fat, fat in the liver, muscle, or neck, and subcutaneous fat. Accumulation in the abdominal visceral area, compared with overall obesity, has an equally or more important role in the development of cardiometabolic risk. Fat depots in liver/muscle tissue cause adverse cardiometabolic effects by affecting energy metabolism. Fat depots in lower-body subcutaneous areas may be protective regarding cardiometabolic risk, by trapping remnant energy. Fat accumulation in the neck is a unique type of fat depot that may increase cardiovascular risk by increasing insulin resistance. Locally acting fat depots include pericardial fat, perivascular fat, and renal sinus fat. These fat depots have effects primarily on adjacent anatomic organs, directly via lipotoxicity and indirectly via cytokine secretion. Pericardial fat is associated with coronary atherosclerosis. Perivascular fat may play an independent role in adverse vascular biology, including arterial stiffness. Renal sinus fat is a unique fat depot that may confer additional cardiometabolic risk. Thus, ectopic fat depots may contribute to the understanding of the link between body composition and cardiometabolic risk. In this review, we focus on the role and clinical implications of ectopic fat depots in cardiometabolic and vascular risk.
Collapse
Affiliation(s)
- Soo Lim
- General Medicine Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seoul, South Korea.
| | | |
Collapse
|
50
|
Wang Y, Zhang Y, Qian H, Lu J, Zhang Z, Min X, Lang M, Yang H, Wang N, Zhang P. The g0/g1 switch gene 2 is an important regulator of hepatic triglyceride metabolism. PLoS One 2013; 8:e72315. [PMID: 23951308 PMCID: PMC3741160 DOI: 10.1371/journal.pone.0072315] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 07/10/2013] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease is associated with obesity and insulin resistance. Factors that regulate the disposal of hepatic triglycerides contribute to the development of hepatic steatosis. G0/G1 switch gene 2 (G0S2) is a target of peroxisome proliferator-activated receptors and plays an important role in regulating lipolysis in adipocytes. Therefore, we investigated whether G0S2 plays a role in hepatic lipid metabolism. Adenovirus-mediated expression of G0S2 (Ad-G0S2) potently induced fatty liver in mice. The liver mass of Ad-G0S2-infected mice was markedly increased with excess triglyceride content compared to the control mice. G0S2 did not change cellular cholesterol levels in hepatocytes. G0S2 was found to be co-localized with adipose triglyceride lipase at the surface of lipid droplets. Hepatic G0S2 overexpression resulted in an increase in plasma Low-density lipoprotein (LDL)/Very-Low-density (VLDL) lipoprotein cholesterol level. Plasma High-density lipoprotein (HDL) cholesterol and ketone body levels were slightly decreased in Ad-G0S2 injected mice. G0S2 also increased the accumulation of neutral lipids in cultured HepG2 and L02 cells. However, G0S2 overexpression in the liver significantly improved glucose tolerance in mice. Livers expressing G0S2 exhibited increased 6-(N-(7-nitrobenz-2-oxa-1-3-diazol-4-yl) amino)-6-deoxyglucose uptake compared with livers transfected with control adenovirus. Taken together, our results provide evidence supporting an important role for G0S2 as a regulator of triglyceride content in the liver and suggest that G0S2 may be a molecular target for the treatment of insulin resistance and other obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Yinfang Wang
- Cardiovascular Research Center, Hubei University of Medicine, Hubei, China
- Department of Physiology, Hubei University of Medicine, Hubei, China
| | - Yahui Zhang
- Department of Pathophysiology, Hubei University of Medicine, Hubei, China
| | - Hang Qian
- Cardiovascular Research Center, Hubei University of Medicine, Hubei, China
| | - Juan Lu
- Cardiovascular Research Center, Hubei University of Medicine, Hubei, China
- Department of Physiology, Hubei University of Medicine, Hubei, China
| | - Zhifeng Zhang
- Cardiovascular Research Center, Hubei University of Medicine, Hubei, China
- Department of Physiology, Hubei University of Medicine, Hubei, China
| | - Xinwen Min
- Cardiovascular Research Center, Hubei University of Medicine, Hubei, China
| | - Mingjian Lang
- Cardiovascular Research Center, Hubei University of Medicine, Hubei, China
| | - Handong Yang
- Cardiovascular Research Center, Hubei University of Medicine, Hubei, China
| | - Nanping Wang
- Cardiovascular Research Center, Xi’an Jiaotong University School of Medicine, Xi’an, China
| | - Peng Zhang
- Cardiovascular Research Center, Hubei University of Medicine, Hubei, China
- Department of Physiology, Hubei University of Medicine, Hubei, China
- * E-mail:
| |
Collapse
|