1
|
Gao C, Gong N, Chen F, Hu S, Zhou Q, Gao X. The Effects of Astaxanthin on Metabolic Syndrome: A Comprehensive Review. Mar Drugs 2024; 23:9. [PMID: 39852511 PMCID: PMC11766962 DOI: 10.3390/md23010009] [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: 11/07/2024] [Revised: 12/21/2024] [Accepted: 12/25/2024] [Indexed: 01/26/2025] Open
Abstract
Metabolic syndrome (MS) represents a complex cluster of metabolic disorders primarily characterized by obesity, insulin resistance, hyperglycemia, dyslipidemia, hypertension, and hyperuricemia. Diet and functional ingredients play a pivotal role in seeking non-pharmacological strategies to prevent and ameliorate MS. Astaxanthin (AST), a carotenoid found in various marine organisms, exhibits exceptional antioxidant properties and holds great promise as a natural compound that improves MS. This article introduces the basic properties of AST, including its absorptance and metabolic pathways, along with various isomers. Most importantly, we comprehensively review the effects and mechanisms of AST on improving the primary components of MS. These mechanisms primarily involve regulating signal transduction, transport, or metabolic pathways within the body, as well as influencing intestinal microbiota and metabolites, thereby exerting positive effects on metabolism and inhibiting the occurrence of MS. This review emphasizes the potential efficacy of AST in managing MS. However, more studies are needed to confirm the clinical effect of AST on MS and reveal potential molecular mechanisms.
Collapse
Affiliation(s)
- Chunhao Gao
- College of Life Sciences, Qingdao University, Qingdao 266071, China; (C.G.); (N.G.); (S.H.)
| | - Nengyun Gong
- College of Life Sciences, Qingdao University, Qingdao 266071, China; (C.G.); (N.G.); (S.H.)
| | - Fangtian Chen
- Department of Marine Technology, Rizhao Polytechnic, Shandong Engineering and Technology Research Center for Marine Crustacean Resources Comprehensive Utilization, Shandong Engineering Research Center for Efficient Utilization Technology of Marine Food Resources, Rizhao Key Laboratory of Efficient Utilization of Marine Food Resources, Rizhao 276826, China;
| | - Shiran Hu
- College of Life Sciences, Qingdao University, Qingdao 266071, China; (C.G.); (N.G.); (S.H.)
| | - Qingxin Zhou
- Department of Marine Technology, Rizhao Polytechnic, Shandong Engineering and Technology Research Center for Marine Crustacean Resources Comprehensive Utilization, Shandong Engineering Research Center for Efficient Utilization Technology of Marine Food Resources, Rizhao Key Laboratory of Efficient Utilization of Marine Food Resources, Rizhao 276826, China;
| | - Xiang Gao
- College of Life Sciences, Qingdao University, Qingdao 266071, China; (C.G.); (N.G.); (S.H.)
| |
Collapse
|
2
|
Mambrini SP, Grillo A, Colosimo S, Zarpellon F, Pozzi G, Furlan D, Amodeo G, Bertoli S. Diet and physical exercise as key players to tackle MASLD through improvement of insulin resistance and metabolic flexibility. Front Nutr 2024; 11:1426551. [PMID: 39229589 PMCID: PMC11370663 DOI: 10.3389/fnut.2024.1426551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) has emerged as a prevalent health concern, encompassing a wide spectrum of liver-related disorders. Insulin resistance, a key pathophysiological feature of MASLD, can be effectively ameliorated through dietary interventions. The Mediterranean diet, rich in whole grains, fruits, vegetables, legumes, and healthy fats, has shown promising results in improving insulin sensitivity. Several components of the Mediterranean diet, such as monounsaturated fats and polyphenols, exert anti-inflammatory and antioxidant effects, thereby reducing hepatic steatosis and inflammation. Furthermore, this dietary pattern has been associated with a higher likelihood of achieving MASLD remission. In addition to dietary modifications, physical exercise, particularly resistance exercise, plays a crucial role in enhancing metabolic flexibility. Resistance exercise training promotes the utilization of fatty acids as an energy source. It enhances muscle glucose uptake and glycogen storage, thus reducing the burden on the liver to uptake excess blood glucose. Furthermore, resistance exercise stimulates muscle protein synthesis, contributing to an improved muscle-to-fat ratio and overall metabolic health. When implemented synergistically, the Mediterranean diet and resistance exercise can elicit complementary effects in combating MASLD. Combined interventions have demonstrated additive benefits, including greater improvements in insulin resistance, increased metabolic flexibility, and enhanced potential for MASLD remission. This underscores the importance of adopting a multifaceted approach encompassing dietary modifications and regular physical exercise to effectively manage MASLD. This narrative review explores the biological mechanisms of diet and physical exercise in addressing MASLD by targeting insulin resistance and decreased metabolic flexibility.
Collapse
Affiliation(s)
- Sara Paola Mambrini
- Nutrition Science Research Lab, Ospedale S. Giuseppe, Istituto Auxologico Italiano IRCCS, Piancavallo, Italy
| | | | - Santo Colosimo
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
- PhD School of Nutrition Science, University of Milan, Milan, Italy
| | - Francesco Zarpellon
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Giorgia Pozzi
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Davide Furlan
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | | | - Simona Bertoli
- Nutrition Science Research Lab, Ospedale S. Giuseppe, Istituto Auxologico Italiano IRCCS, Piancavallo, Italy
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| |
Collapse
|
3
|
Chen FX, Wan Q, Fang J, Peng L, Li QL, Hu J. The Src1-PGC1α-AP1 complex-dependent secretion of substance P induces inflammation and apoptosis in encephalomyocarditis virus-infected mice. Cytokine 2023; 165:156186. [PMID: 36989655 DOI: 10.1016/j.cyto.2023.156186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Substance P (SP), a neuropeptide consisting of 11 amino acid residues, is involved in the pathogenesis of encephalomyocarditis virus (EMCV)-induced myocarditis by stimulating the production of proinflammatory cytokines. However, the underlying mechanism that regulates SP production is still unknown. In this study, we report the transcriptional regulation of the Tachykinin Precursor 1 (TAC1) gene that encodes SP by a transcriptional complex composed of Steroid Receptor Coactivator 1 (Src1), Peroxisome proliferator-activated receptor-gamma coactivator 1 (PGC1α), and Activator Protein 1 (AP1) transcription factor. Infection of mice with EMCV induced the accumulation of PGC1α and increased TAC1 expression, thereby promoting the secretion of SP, initiating apoptosis, and elevating proinflammatory cytokine levels. In vitro overexpression of the Src1-PGC1α-AP1 members also induced TAC1 expression, increased the SP concentration, initiated apoptosis, and elevated proinflammatory cytokine concentrations. Depletion or inhibition of the Src1-PGC1α-AP1 complex reversed these effects. The administration of gossypol, an Src1 inhibitor, or SR1892, a PGC1α inhibitor, to EMCV-infected mice attenuated myocarditis. Taken together, our results reveal that the upregulation of TAC1 and the secretion of SP in EMCV-induced myocarditis are dependent on the Src1-PGC1α-AP1 complex. Targeting the Src1-PGC1α-AP1 complex may represent a new therapeutic strategy for myocarditis.
Collapse
Affiliation(s)
- Fa-Xiu Chen
- Department of Geriatrics, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, Jiangxi, China; Department of Geriatrics, Jiangxi Provincial People's Hospital, Nanchang 330006, Jiangxi, China
| | - Qin Wan
- Department of Geriatrics, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, Jiangxi, China; Department of Geriatrics, Jiangxi Provincial People's Hospital, Nanchang 330006, Jiangxi, China
| | - Jing Fang
- Department of Geriatrics, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, Jiangxi, China; Department of Geriatrics, Jiangxi Provincial People's Hospital, Nanchang 330006, Jiangxi, China
| | - Le Peng
- Department of Geriatrics, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, Jiangxi, China; Department of Geriatrics, Jiangxi Provincial People's Hospital, Nanchang 330006, Jiangxi, China
| | - Qing-Ling Li
- Department of Geriatrics, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, Jiangxi, China; Department of Geriatrics, Jiangxi Provincial People's Hospital, Nanchang 330006, Jiangxi, China.
| | - Jian Hu
- Department of Geriatrics, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, Jiangxi, China; Department of Geriatrics, Jiangxi Provincial People's Hospital, Nanchang 330006, Jiangxi, China.
| |
Collapse
|
4
|
Coulter AA, Greenway FL, Zhang D, Ghosh S, Coulter CR, James SL, He Y, Cusimano LA, Rebello CJ. Naringenin and β-carotene convert human white adipocytes to a beige phenotype and elevate hormone- stimulated lipolysis. Front Endocrinol (Lausanne) 2023; 14:1148954. [PMID: 37143734 PMCID: PMC10153092 DOI: 10.3389/fendo.2023.1148954] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/20/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction Naringenin, a peroxisome proliferator-activated receptor (PPAR) activator found in citrus fruits, upregulates markers of thermogenesis and insulin sensitivity in human adipose tissue. Our pharmacokinetics clinical trial demonstrated that naringenin is safe and bioavailable, and our case report showed that naringenin causes weight loss and improves insulin sensitivity. PPARs form heterodimers with retinoic-X-receptors (RXRs) at promoter elements of target genes. Retinoic acid is an RXR ligand metabolized from dietary carotenoids. The carotenoid β-carotene reduces adiposity and insulin resistance in clinical trials. Our goal was to examine if carotenoids strengthen the beneficial effects of naringenin on human adipocyte metabolism. Methods Human preadipocytes from donors with obesity were differentiated in culture and treated with 8µM naringenin + 2µM β-carotene (NRBC) for seven days. Candidate genes involved in thermogenesis and glucose metabolism were measured as well as hormone-stimulated lipolysis. Results We found that β-carotene acts synergistically with naringenin to boost UCP1 and glucose metabolism genes including GLUT4 and adiponectin, compared to naringenin alone. Protein levels of PPARα, PPARγ and PPARγ-coactivator-1α, key modulators of thermogenesis and insulin sensitivity, were also upregulated after treatment with NRBC. Transcriptome sequencing was conducted and the bioinformatics analyses of the data revealed that NRBC induced enzymes for several non-UCP1 pathways for energy expenditure including triglyceride cycling, creatine kinases, and Peptidase M20 Domain Containing 1 (PM20D1). A comprehensive analysis of changes in receptor expression showed that NRBC upregulated eight receptors that have been linked to lipolysis or thermogenesis including the β1-adrenergic receptor and the parathyroid hormone receptor. NRBC increased levels of triglyceride lipases and agonist-stimulated lipolysis in adipocytes. We observed that expression of RXRγ, an isoform of unknown function, was induced ten-fold after treatment with NRBC. We show that RXRγ is a coactivator bound to the immunoprecipitated PPARγ protein complex from white and beige human adipocytes. Discussion There is a need for obesity treatments that can be administered long-term without side effects. NRBC increases the abundance and lipolytic response of multiple receptors for hormones released after exercise and cold exposure. Lipolysis provides the fuel for thermogenesis, and these observations suggest that NRBC has therapeutic potential.
Collapse
Affiliation(s)
- Ann A. Coulter
- Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Frank L. Greenway
- Clinical Trials, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Dachuan Zhang
- Biostatistics, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Sujoy Ghosh
- Adjunct Faculty, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Cathryn R. Coulter
- Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Sarah L. James
- Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Yanlin He
- Brain Glycemic and Metabolism Control, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Luke A. Cusimano
- Cusimano Plastic and Reconstructive Surgery, Baton Rouge, LA, United States
| | - Candida J. Rebello
- Nutrition and Chronic Disease, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| |
Collapse
|
5
|
Ishaq A, Tchkonia T, Kirkland JL, Siervo M, Saretzki G. Palmitate induces DNA damage and senescence in human adipocytes in vitro that can be alleviated by oleic acid but not inorganic nitrate. Exp Gerontol 2022; 163:111798. [PMID: 35390489 PMCID: PMC9214712 DOI: 10.1016/j.exger.2022.111798] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022]
Abstract
Hypertrophy in white adipose tissue (WAT) can result in sustained systemic inflammation, hyperlipidaemia, insulin resistance, and onset of senescence in adipocytes. Inflammation and hypertrophy can be induced in vitro using palmitic acid (PA). WAT adipocytes have innately low β-oxidation capacity, while inorganic nitrate can promote a beiging phenotype, with promotion of β-oxidation when cells are exposed to nitrate during differentiation. We hypothesized that treatment of human adipocytes with PA in vitro can induce senescence, which might be attenuated by nitrate treatment through stimulation of β-oxidation to remove accumulated lipids. Differentiated subcutaneous and omental adipocytes were treated with PA and nitrate and senescence markers were analyzed. PA induced DNA damage and increased p16INK4a levels in both human subcutaneous and omental adipocytes in vitro. However, lipid accumulation and lipid droplet size increased after PA treatment only in subcutaneous adipocytes. Thus, hypertrophy and senescence seem not to be causally associated. Contrary to our expectations, subsequent treatment of PA-induced adipocytes with nitrate did not attenuate PA-induced lipid accumulation or senescence. Instead, we found a significantly beneficial effect of oleic acid (OA) on human subcutaneous adipocytes when applied together with PA, which reduced the DNA damage caused by PA treatment.
Collapse
Affiliation(s)
- Abbas Ishaq
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Tamara Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, United States of America
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, United States of America
| | - Mario Siervo
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK; School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Gabriele Saretzki
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle upon Tyne, UK.
| |
Collapse
|
6
|
Wang M, Xu W, Yu J, Liu Y, Ma H, Ji C, Zhang C, Xue J, Li R, Cui H. Astaxanthin From Haematococcus pluvialis Prevents High-Fat Diet-Induced Hepatic Steatosis and Oxidative Stress in Mice by Gut-Liver Axis Modulating Properties. Front Nutr 2022; 9:840648. [PMID: 35495929 PMCID: PMC9039660 DOI: 10.3389/fnut.2022.840648] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Scope Evidence is mounting that astaxanthin (ATX), a xanthophyll carotenoid, used as a nutritional supplement to prevent chronic metabolic diseases. The present study aims to identify the potential function of ATX supplementation in preventing steatohepatitis and hepatic oxidative stress in diet-induced obese mice. Methods and Results In this study, ATX as dose of 0.25, 0.5, and 0.75% have orally administered to mice along with a high-fat diet (HFD) to investigate the role of ATX in regulating liver lipid metabolism and gut microbiota. The study showed that ATX dose-dependently reduces body weight, lipid droplet formation, hepatic triglycerides and ameliorated hepatic steatosis and oxidative stress. 0.75% ATX altered the levels of 34 lipid metabolites related to hepatic cholesterol and fatty acid metabolism which might be associated with downregulation of lipogenesis-related genes and upregulation of bile acid biosynthesis-related genes. The result also revealed that ATX alleviates HFD-induced gut microbiota dysbiosis by significantly inhibiting the growth of obesity-related Parabacteroides and Desulfovibrio while promoting the growth of Allobaculum and Akkermansia. Conclusion The study results suggested that dietary ATX may prevent the development of hepatic steatosis and oxidative stress with the risk of metabolic disease by gut-liver axis modulating properties.
Collapse
Affiliation(s)
- Meng Wang
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Wenxin Xu
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Jie Yu
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Yingying Liu
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Haotian Ma
- Health Science Center, College of Forensic Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Chunli Ji
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Chunhui Zhang
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Jinai Xue
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Runzhi Li
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China.,State Key Laboratory of Integrative Sustainable Dryland Agriculture, Shanxi Agricultural University, Taiyuan, China
| | - Hongli Cui
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China.,State Key Laboratory of Integrative Sustainable Dryland Agriculture, Shanxi Agricultural University, Taiyuan, China
| |
Collapse
|
7
|
Christoffersen BØ, Sanchez‐Delgado G, John LM, Ryan DH, Raun K, Ravussin E. Beyond appetite regulation: Targeting energy expenditure, fat oxidation, and lean mass preservation for sustainable weight loss. Obesity (Silver Spring) 2022; 30:841-857. [PMID: 35333444 PMCID: PMC9310705 DOI: 10.1002/oby.23374] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/09/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022]
Abstract
New appetite-regulating antiobesity treatments such as semaglutide and agents under investigation such as tirzepatide show promise in achieving weight loss of 15% or more. Energy expenditure, fat oxidation, and lean mass preservation are important determinants of weight loss and weight-loss maintenance beyond appetite regulation. This review discusses prior failures in clinical development of weight-loss drugs targeting energy expenditure and explores novel strategies for targeting energy expenditure: mitochondrial proton leak, uncoupling, dynamics, and biogenesis; futile calcium and substrate cycling; leptin for weight maintenance; increased sympathetic nervous system activity; and browning of white fat. Relevant targets for preserving lean mass are also reviewed: growth hormone, activin type II receptor inhibition, and urocortin 2 and 3. We endorse moderate modulation of energy expenditure and preservation of lean mass in combination with efficient appetite reduction as a means of obtaining a significant, safe, and long-lasting weight loss. Furthermore, we suggest that the regulatory guidelines should be revisited to focus more on the quality of weight loss and its maintenance rather than the absolute weight loss. Commitment to this research focus both from a scientific and from a regulatory point of view could signal the beginning of the next era in obesity therapies.
Collapse
Affiliation(s)
| | | | - Linu Mary John
- Global Obesity and Liver Disease ResearchGlobal Drug DiscoveryNovo Nordisk A/SMåløvDenmark
| | - Donna H. Ryan
- Pennington Biomedical Research CenterLouisiana State UniversityBaton RougeLouisianaUSA
| | - Kirsten Raun
- Global Obesity and Liver Disease ResearchGlobal Drug DiscoveryNovo Nordisk A/SMåløvDenmark
| | - Eric Ravussin
- Pennington Biomedical Research CenterLouisiana State UniversityBaton RougeLouisianaUSA
| |
Collapse
|
8
|
Los posibles mecanismos de pardeamiento del tejido adiposo blanco: una diana novedosa para el tratamiento de la obesidad. NUTR HOSP 2022; 39:411-424. [PMID: 35001637 DOI: 10.20960/nh.03852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
The increase of the obesity pandemic worldwide over the last several decades has generated a constant need for the scientific world to develop new possibilities to combat obesity. Since the discovery that brown adipose tissue (BAT) exists in adult humans, and BAT activation contributes to a negative energy balance, much more attention has been focused on the understanding of the molecular switches and their different regulatory mechanisms turning on energy expenditure. Recent insights have revealed that a range of stimuli including cold exposure, physical activity and diet, and critical transcription molecules such as PPARγ, PRDM16, PGC-1α and UCP1, aiming at the induction of BAT activation, could cause the browning of white adipose tissue, thereby dissipating energy and increasing heat production. An increasing number of studies that point to the white adipose tissue (WAT) browning strategies aiming at diet-induced and/or genetically determined obesity have been tested in mouse models as well as in human studies. Findings suggested that browning stimulating drugs have been currently or previously assayed as a therapy against obesity. As PPARα agonists, fibrate drugs effectively reduced plasma triglyceride, increased high-density lipoproteins, and improved glycemic control and heat production in brown adipose tissue, which has been used in the treatment of metabolic disorders. Many kinds of natural products promote white adipose tissue browning, such as alkaloids, flavonoids, terpenoids, and long-chain fatty acids, which can also ameliorate metabolic disorders including obesity, insulin resistance and diabetes. The aim of this review is to summarize the transcriptional regulators as well as the various mediators that have been regarded as potential therapeutic targets in the process of WAT browning.
Collapse
|
9
|
PPARs-Orchestrated Metabolic Homeostasis in the Adipose Tissue. Int J Mol Sci 2021; 22:ijms22168974. [PMID: 34445679 PMCID: PMC8396609 DOI: 10.3390/ijms22168974] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 01/12/2023] Open
Abstract
It has been more than three decades since peroxisome proliferator-activated receptors (PPARs) were first discovered. Many investigations have revealed the central regulators of PPARs in lipid and glucose homeostasis in response to different nutrient conditions. PPARs have attracted much attention due to their ability to improve metabolic syndromes, and they have also been proposed as classical drug targets for the treatment of hyperlipidemia and type 2 diabetes (T2D) mellitus. In parallel, adipose tissue is known to play a unique role in the pathogenesis of insulin resistance and metabolic syndromes due to its ability to “safely” store lipids and secrete cytokines that regulate whole-body metabolism. Adipose tissue relies on a complex and subtle network of transcription factors to maintain its normal physiological function, by coordinating various molecular events, among which PPARs play distinctive and indispensable roles in adipocyte differentiation, lipid metabolism, adipokine secretion, and insulin sensitivity. In this review, we discuss the characteristics of PPARs with special emphasis on the roles of the different isotypes in adipocyte biology.
Collapse
|
10
|
Morigny P, Boucher J, Arner P, Langin D. Lipid and glucose metabolism in white adipocytes: pathways, dysfunction and therapeutics. Nat Rev Endocrinol 2021; 17:276-295. [PMID: 33627836 DOI: 10.1038/s41574-021-00471-8] [Citation(s) in RCA: 290] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 12/14/2022]
Abstract
In mammals, the white adipocyte is a cell type that is specialized for storage of energy (in the form of triacylglycerols) and for energy mobilization (as fatty acids). White adipocyte metabolism confers an essential role to adipose tissue in whole-body homeostasis. Dysfunction in white adipocyte metabolism is a cardinal event in the development of insulin resistance and associated disorders. This Review focuses on our current understanding of lipid and glucose metabolic pathways in the white adipocyte. We survey recent advances in humans on the importance of adipocyte hypertrophy and on the in vivo turnover of adipocytes and stored lipids. At the molecular level, the identification of novel regulators and of the interplay between metabolic pathways explains the fine-tuning between the anabolic and catabolic fates of fatty acids and glucose in different physiological states. We also examine the metabolic alterations involved in the genesis of obesity-associated metabolic disorders, lipodystrophic states, cancers and cancer-associated cachexia. New challenges include defining the heterogeneity of white adipocytes in different anatomical locations throughout the lifespan and investigating the importance of rhythmic processes. Targeting white fat metabolism offers opportunities for improved patient stratification and a wide, yet unexploited, range of therapeutic opportunities.
Collapse
Affiliation(s)
- Pauline Morigny
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1297, Toulouse, France
- University of Toulouse, Paul Sabatier University, I2MC, UMR1297, Toulouse, France
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jeremie Boucher
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Peter Arner
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Dominique Langin
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1297, Toulouse, France.
- University of Toulouse, Paul Sabatier University, I2MC, UMR1297, Toulouse, France.
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague and Paul Sabatier University, Toulouse, France.
- Toulouse University Hospitals, Laboratory of Clinical Biochemistry, Toulouse, France.
| |
Collapse
|
11
|
Brown Adipose Tissue and Its Role in Insulin and Glucose Homeostasis. Int J Mol Sci 2021; 22:ijms22041530. [PMID: 33546400 PMCID: PMC7913527 DOI: 10.3390/ijms22041530] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/21/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
The increased worldwide prevalence of obesity, insulin resistance, and their related metabolic complications have prompted the scientific world to search for new possibilities to combat obesity. Brown adipose tissue (BAT), due to its unique protein uncoupling protein 1 (UPC1) in the inner membrane of the mitochondria, has been acknowledged as a promising approach to increase energy expenditure. Activated brown adipocytes dissipate energy, resulting in heat production. In other words, BAT burns fat and increases the metabolic rate, promoting a negative energy balance. Moreover, BAT alleviates metabolic complications like dyslipidemia, impaired insulin secretion, and insulin resistance in type 2 diabetes. The aim of this review is to explore the role of BAT in total energy expenditure, as well as lipid and glucose homeostasis, and to discuss new possible activators of brown adipose tissue in humans to treat obesity and metabolic disorders.
Collapse
|
12
|
Shan Y, Zhang S, Gao B, Liang S, Zhang H, Yu X, Zhao J, Ye L, Yang Q, Shang W. Adipose Tissue SIRT1 Regulates Insulin Sensitizing and Anti-Inflammatory Effects of Berberine. Front Pharmacol 2020; 11:591227. [PMID: 33390968 PMCID: PMC7774030 DOI: 10.3389/fphar.2020.591227] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
Berberine (BBR), which is an active component of Coptis chinensis Franch, has been reported to improve glucose metabolism and insulin resistance in animal and human studies, predominantly via activation of the 5′-adenosine monophosphate kinase (AMPK) pathway and suppression of the inflammation response. However, the mechanisms underlying the effects of BBR on AMPK and inflammation remain unclear. In this present study, we found that BBR upregulated SIRT1 expression in 3T3L-1 adipocytes and adipose tissue. Inhibition of SIRT1 blunted the BBR-induced increase in glucose consumption and uptake in adipocytes. The BBR-induced activation of the AMPK pathway and AKT phosphorylation in adipocytes and adipose tissue were also attenuated by inhibition or knockout of Sirt1. The BBR-induced improvement of systemic insulin sensitivity was impaired by Sirt1 knockout in HFD-induced obese mice. The suppressing effects of BBR on systemic and local inflammatory responses, such as serum concentrations and expression of inflammatory cytokines, phosphorylation of c-Jun N-terminal kinase (JNK) and IKKβ, and the accumulation of F4/80-positive macrophages in adipose tissue were also attenuated in Sirt1 knockout mice. The BBR-induced decrease in PGC-1α acetylation was reversed by inhibition or knockout of Sirt1 in adipocytes and adipose tissue. Together, these results indicate that adipose tissue SIRT1 is a key regulator of the insulin sensitizing and anti-inflammatory effects of BBR, which contributes to the improvement of metabolic dysregulation.
Collapse
Affiliation(s)
- Yun Shan
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuchen Zhang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bin Gao
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shu Liang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Zhang
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xizhong Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Juan Zhao
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lifang Ye
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qin Yang
- Department of Medicine and Physiology, UC Irvine Diabetes Center, Center for Epigenetics and Metabolism, University of California at Irvine, Irvine, CA, United States
| | - Wenbin Shang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
13
|
Wu H, Li X, Shen C. Peroxisome proliferator-activated receptor gamma in white and brown adipocyte regulation and differentiation. Physiol Res 2020; 69:759-773. [PMID: 32901494 DOI: 10.33549/physiolres.934411] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In as early as 1997, the World Health Organization officially recognized obesity as a chronic disease. The current epidemic of obesity and overweightness has aroused great interest in the study of adipose tissue formation. The transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma) binds to the target gene promoter regulatory sequences, acting as a key factor in regulating the differentiation of preadipocytes in the adipose tissue, and plays an important role in regulating the adipocyte metabolism. A further understanding of the structure and expression characteristics of PPARgamma, in addition to its mechanisms of action in adipocyte differentiation, may be applied to control obesity and prevent obesity-related diseases. In this article, recent studies investigating the effect of regulating PPARgamma on adipocyte differentiation are reviewed. In particular, the structural characteristics, expression patterns, and molecular mechanisms of PPARgamma function in adipocyte differentiation are considered.
Collapse
Affiliation(s)
- H Wu
- Nutritional Department, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Gaoqiao Town, Pudong New Area, Shanghai, China.
| | | | | |
Collapse
|
14
|
Yu P, Liu M, Zhang B, Yu Y, Su E, Xie S, Zhang L, Yang X, Jiang H, Chen R, Zou Y, Ge J. Cardiomyocyte-restricted high-mobility group box 1 (HMGB1) deletion leads to small heart and glycolipid metabolic disorder through GR/PGC-1α signalling. Cell Death Discov 2020; 6:106. [PMID: 33101708 PMCID: PMC7575537 DOI: 10.1038/s41420-020-00340-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/13/2020] [Accepted: 09/24/2020] [Indexed: 11/22/2022] Open
Abstract
Cardiac growth and remodelling are key biological processes influencing the physiological performance of the heart, and a previous study showed a critical role for intracellular HMGB1 in vitro. However, the in vivo study, which used conditional Hmgb1 ablation, did not show a significant effect on cellular or organic function. We have demonstrated the extracellular effect of HMGB1 as a pro-inflammatory molecule on cardiac remodelling. In this study, we found that HMGB1 deletion by cTnT-Cre in mouse hearts altered glucocorticoid receptor (GR) function and glycolipid metabolism, eventually leading to growth retardation, small heart and heart failure. The subcellular morphology did not show a significant change caused by HMGB1 knockout. The heart showed significant elevation of glycolysis, free fatty acid deposition and related enzyme changes. Transcriptomic analysis revealed a list of differentially expressed genes that coincide with glucocorticoid receptor function in neonatal mice and a significant increase in inflammatory genes in adult mice. Cardiac HMGB1 knockout led to a series of changes in PGC-1α, UCP3 and GyK, which were the cause of metabolic changes and further impacted cardiac function. Ckmm-Cre Hmgb1fl/fl mice did not show a specific phenotype, which was consistent with the reported negative result of cardiomyocyte-specific Hmgb1 deletion via MHC-Cre. We concluded that HMGB1 plays essential roles in maintaining normal cardiac growth, and different phenotype from cardiac-specific HMGB1-deficient mice may be caused by the cross with mice of different Cre strains.
Collapse
Affiliation(s)
- Peng Yu
- Department of Endocrinology and Metabolism, Fudan Institute of Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming Liu
- Department of General Practice, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Baoli Zhang
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Ying Yu
- Department of General Practice, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Enyong Su
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Shiyao Xie
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lei Zhang
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xue Yang
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Hong Jiang
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Ruizhen Chen
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
15
|
Gerbec ZJ, Hashemi E, Nanbakhsh A, Holzhauer S, Yang C, Mei A, Tsaih SW, Lemke A, Flister MJ, Riese MJ, Thakar MS, Malarkannan S. Conditional Deletion of PGC-1α Results in Energetic and Functional Defects in NK Cells. iScience 2020; 23:101454. [PMID: 32858341 PMCID: PMC7474003 DOI: 10.1016/j.isci.2020.101454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/30/2019] [Accepted: 08/10/2020] [Indexed: 01/07/2023] Open
Abstract
During an immune response, natural killer (NK) cells activate specific metabolic pathways to meet the increased energetic and biosynthetic demands associated with effector functions. Here, we found in vivo activation of NK cells during Listeria monocytogenes infection-augmented transcription of genes encoding mitochondria-associated proteins in a manner dependent on the transcriptional coactivator PGC-1α. Using an Ncr1Cre-based conditional knockout mouse, we found that PGC-1α was crucial for optimal NK cell effector functions and bioenergetics, as the deletion of PGC-1α was associated with decreased cytotoxic potential and cytokine production along with altered ADP/ATP ratios. Lack of PGC-1α also significantly impaired the ability of NK cells to control B16F10 tumor growth in vivo, and subsequent gene expression analysis showed that PGC-1α mediates transcription required to maintain mitochondrial activity within the tumor microenvironment. Together, these data suggest that PGC-1α-dependent transcription of specific target genes is required for optimal NK cell function during the response to infection or tumor growth.
Collapse
Affiliation(s)
- Zachary J. Gerbec
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Arash Nanbakhsh
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Sandra Holzhauer
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
| | - Chao Yang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ao Mei
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shirng-Wern Tsaih
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Angela Lemke
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael J. Flister
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew J. Riese
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Laboratory of Lymphocyte Signaling, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Monica S. Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
16
|
Hassan QN, Queen NJ, Cao L. Regulation of aging and cancer by enhanced environmental activation of a hypothalamic-sympathoneural-adipocyte axis. Transl Cancer Res 2020; 9:5687-5699. [PMID: 33134111 PMCID: PMC7595574 DOI: 10.21037/tcr.2020.02.39] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/11/2020] [Indexed: 12/20/2022]
Abstract
Social and environmental factors impact cancer and energy balance profoundly. Years ago, our lab established the existence of a novel brain-fat interaction we termed the "hypothalamic-sympathoneural-adipocyte (HSA) axis", through which complex environmental stimuli provided by an enriched environment regulate body composition, energy balance, and development of cancer. We have spent a significant portion of the past decade to further characterize the broad health benefits of an enriched environment (for example, leanness, enhanced immune function, and cancer resistance), and to identify mediators in the brain and periphery along the HSA axis. This review summarizes our recent work regarding the interface between endocrinology, immunology, cancer biology, aging, and neuroscience. We will discuss the interplay between these systemic phenomena and how the HSA axis can be targeted for regulation of cancer and aging.
Collapse
Affiliation(s)
- Quais N. Hassan
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Medical Scientist Training Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
17
|
Anti-obesity effect of cocoa proteins (Theobroma cacao L.) variety “Criollo” and the expression of genes related to the dysfunction of white adipose tissue in high-fat diet-induced obese rats. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
18
|
Peroxisome Proliferator Activated Receptor Gamma Controls Mature Brown Adipocyte Inducibility through Glycerol Kinase. Cell Rep 2019; 22:760-773. [PMID: 29346772 DOI: 10.1016/j.celrep.2017.12.067] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/03/2017] [Accepted: 12/20/2017] [Indexed: 01/08/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been suggested as the master regulators of adipose tissue formation. However, their role in regulating brown fat functionality has not been resolved. To address this question, we generated mice with inducible brown fat-specific deletions of PPARα, β/δ, and γ, respectively. We found that both PPARα and β/δδ are dispensable for brown fat function. In contrast, we could show that ablation of PPARγ in vitro and in vivo led to a reduced thermogenic capacity accompanied by a loss of inducibility by β-adrenergic signaling, as well as a shift from oxidative fatty acid metabolism to glucose utilization. We identified glycerol kinase (Gyk) as a partial mediator of PPARγ function and could show that Gyk expression correlates with brown fat thermogenic capacity in human brown fat biopsies. Thus, Gyk might constitute the link between PPARγ-mediated regulation of brown fat function and activation by β-adrenergic signaling.
Collapse
|
19
|
Larson CJ. Translational Pharmacology and Physiology of Brown Adipose Tissue in Human Disease and Treatment. Handb Exp Pharmacol 2019; 251:381-424. [PMID: 30689089 DOI: 10.1007/164_2018_184] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Human brown adipose tissue (BAT) is experimentally modeled to better understand the biology of this important metabolic tissue, and also to enable the potential discovery and development of novel therapeutics for obesity and sequelae resulting from the persistent positive energy balance. This chapter focuses on translation into humans of findings and hypotheses generated in nonhuman models of BAT pharmacology. Given the demonstrated challenges of sustainably reducing caloric intake in modern humans, potential solutions to obesity likely lie in increasing energy expenditure. The energy-transforming activities of a single cell in any given tissue can be conceptualized as a flow of chemical energy from energy-rich substrate molecules into energy-expending, endergonic biological work processes through oxidative degradation of organic molecules ingested as nutrients. Despite the relatively tight coupling between metabolic reactions and products, some expended energy is incidentally lost as heat, and in this manner a significant fraction of the energy originally captured from the environment nonproductively transforms into heat rather than into biological work. In human and other mammalian cells, some processes are even completely uncoupled, and therefore purely energy consuming. These molecular and cellular actions sum up at the physiological level to adaptive thermogenesis, the endogenous physiology in which energy is nonproductively released as heat through uncoupling of mitochondria in brown fat and potentially skeletal muscle. Adaptive thermogenesis in mammals occurs in three forms, mostly in skeletal muscle and brown fat: shivering thermogenesis in skeletal muscle, non-shivering thermogenesis in brown fat, and diet-induced thermogenesis in brown fat. At the cellular level, the greatest energy transformations in humans and other eukaryotes occur in the mitochondria, where creating energetic inefficiency by uncoupling the conversion of energy-rich substrate molecules into ATP usable by all three major forms of biological work occurs by two primary means. Basal uncoupling occurs as a passive, general, nonspecific leak down the proton concentration gradient across the membrane in all mitochondria in the human body, a gradient driving a key step in ATP synthesis. Inducible uncoupling, which is the active conduction of protons across gradients through processes catalyzed by proteins, occurs only in select cell types including BAT. Experiments in rodents revealed UCP1 as the primary mammalian molecule accounting for the regulated, inducible uncoupling of BAT, and responsive to both cold and pharmacological stimulation. Cold stimulation of BAT has convincingly translated into humans, and older clinical observations with nonselective 2,4-DNP validate that human BAT's participation in pharmacologically mediated, though nonselective, mitochondrial membrane decoupling can provide increased energy expenditure and corresponding body weight loss. In recent times, however, neither beta-adrenergic antagonism nor unselective sympathomimetic agonism by ephedrine and sibutramine provide convincing evidence that more BAT-selective mechanisms can impact energy balance and subsequently body weight. Although BAT activity correlates with leanness, hypothesis-driven selective β3-adrenergic agonism to activate BAT in humans has only provided robust proof of pharmacologic activation of β-adrenergic receptor signaling, limited proof of the mechanism of increased adaptive thermogenesis, and no convincing evidence that body weight loss through negative energy balance upon BAT activation can be accomplished outside of rodents. None of the five demonstrably β3 selective molecules with sufficient clinical experience to merit review provided significant weight loss in clinical trials (BRL 26830A, TAK 677, L-796568, CL 316,243, and BRL 35135). Broader conclusions regarding the human BAT therapeutic hypothesis are limited by the absence of data from most studies demonstrating specific activation of BAT thermogenesis in most studies. Additionally, more limited data sets with older or less selective β3 agonists also did not provide strong evidence of body weight effects. Encouragingly, β3-adrenergic agonists, catechins, capsinoids, and nutritional extracts, even without robust negative energy balance outcomes, all demonstrated increased total energy expenditure that in some cases could be associated with concomitant activation of BAT, though the absence of body weight loss indicates that in no cases did the magnitude of negative energy balance reach sufficient levels. Glucocorticoid receptor agonists, PPARg agonists, and thyroid hormone receptor agonists all possess defined molecular and cellular pharmacology that preclinical models predicted to be efficacious for negative energy balance and body weight loss, yet their effects on human BAT thermogenesis upon translation were inconsistent with predictions and disappointing. A few new mechanisms are nearing the stage of clinical trials and may yet provide a more quantitatively robust translation from preclinical to human experience with BAT. In conclusion, translation into humans has been demonstrated with BAT molecular pharmacology and cell biology, as well as with physiological response to cold. However, despite pharmacologically mediated, statistically significant elevation in total energy expenditure, translation into biologically meaningful negative energy balance was not achieved, as indicated by the absence of measurable loss of body weight over the duration of a clinical study.
Collapse
Affiliation(s)
- Christopher J Larson
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
20
|
Moody L, Xu GB, Chen H, Pan YX. Epigenetic regulation of carnitine palmitoyltransferase 1 (Cpt1a) by high fat diet. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:141-152. [PMID: 30605728 DOI: 10.1016/j.bbagrm.2018.12.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/22/2018] [Accepted: 12/27/2018] [Indexed: 12/15/2022]
Abstract
Carnitine palmitoyltransferase 1 (Cpt1a) is a rate-limiting enzyme that mediates the transport of fatty acids into the mitochondria for subsequent beta-oxidation. The objective of this study was to uncover how diet mediates the transcriptional regulation of Cpt1a. Pregnant Sprague Dawley rats were exposed to either a high-fat (HF) or low-fat control diet during gestation and lactation. At weaning, male offspring received either a HF or control diet, creating 4 groups: lifelong control diet (C/C; n = 12), perinatal HF diet (HF/C; n = 9), post-weaning HF diet (C/HF; n = 10), and lifelong HF diet (HF/HF; n = 10). Only HF/HF animals had higher hepatic Cpt1a mRNA expression than C/C. Epigenetic analysis revealed reduced DNA methylation (DNAMe) and increased histone 3 lysine 4 dimethylation (H3K4Me2) upstream and within the promoter of Cpt1a in the HF/HF group. This was accompanied by increased peroxisome proliferator activated receptor alpha (PPARα) and CCAAT/enhancer binding protein beta (C/EBPβ) binding directly downstream of the Cpt1a transcription start site within the first intron. Findings were confirmed in rat hepatoma H4IIEC3 cells treated with non-esterified fatty acid (NEFA). After 12 h of NEFA treatment, there was an enrichment of SWI/SNF related matrix associated actin dependent regulator of chromatin subfamily D member 1 (BAF60a or SMARCD1) in the first intron of Cpt1a. We conclude that dietary fat elevates hepatic Cpt1a expression via a highly coordinated transcriptional mechanism involving increased H3K4Me2, reduced DNAMe, and recruitment of C/EBPβ, PPARα, PGC1α, and BAF60a to the gene.
Collapse
Affiliation(s)
- Laura Moody
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America.
| | - Guanying Bianca Xu
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America.
| | - Hong Chen
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America.
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States of America; Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.
| |
Collapse
|
21
|
Madsen L, Myrmel LS, Fjære E, Øyen J, Kristiansen K. Dietary Proteins, Brown Fat, and Adiposity. Front Physiol 2018; 9:1792. [PMID: 30631281 PMCID: PMC6315128 DOI: 10.3389/fphys.2018.01792] [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: 09/03/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022] Open
Abstract
High protein diets have become popular for body weight maintenance and weight loss despite controversies regarding efficacy and safety. Although both weight gain and weight loss are determined by energy consumption and expenditure, data from rodent trials consistently demonstrate that the protein:carbohydrate ratio in high fat diets strongly influences body and fat mass gain per calorie eaten. Here, we review data from rodent trials examining how high protein diets may modulate energy metabolism and the mechanisms by which energy may be dissipated. We discuss the possible role of activating brown and so-called beige/BRITE adipocytes including non-canonical UCP1-independent thermogenesis and futile cycles, where two opposing metabolic pathways are operating simultaneously. We further review data on how the gut microbiota may affect energy expenditure. Results from human and rodent trials demonstrate that human trials are less consistent than rodent trials, where casein is used almost exclusively as the protein source. The lack of consistency in results from human trials may relate to the specific design of human trials, the possible distinct impact of different protein sources, and/or the differences in the efficiency of high protein diets to attenuate obesity development in lean subjects vs. promoting weight loss in obese subjects.
Collapse
Affiliation(s)
- Lise Madsen
- Institute of Marine Research, Bergen, Norway.,Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Even Fjære
- Institute of Marine Research, Bergen, Norway
| | | | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Løvsletten NG, Bakke SS, Kase ET, Ouwens DM, Thoresen GH, Rustan AC. Increased triacylglycerol - Fatty acid substrate cycling in human skeletal muscle cells exposed to eicosapentaenoic acid. PLoS One 2018; 13:e0208048. [PMID: 30496314 PMCID: PMC6264501 DOI: 10.1371/journal.pone.0208048] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/09/2018] [Indexed: 11/18/2022] Open
Abstract
It has previously been shown that pretreatment of differentiated human skeletal muscle cells (myotubes) with eicosapentaenoic acid (EPA) promoted increased uptake of fatty acids and increased triacylglycerol accumulation, compared to pretreatment with oleic acid (OA) and palmitic acid (PA). The aim of the present study was to examine whether EPA could affect substrate cycling in human skeletal muscle cells by altering lipolysis rate of intracellular TAG and re-esterification of fatty acids. Fatty acid metabolism was studied in human myotubes using a mixture of fatty acids, consisting of radiolabelled oleic acid as tracer (14C-OA) together with EPA or PA. Co-incubation of myotubes with EPA increased cell-accumulation and incomplete fatty acid oxidation of 14C-OA compared to co-incubation with PA. Lipid distribution showed higher incorporation of 14C-OA into all cellular lipids after co-incubation with EPA relative to PA, with most markedly increases (3 to 4-fold) for diacylglycerol and triacylglycerol. Further, the increases in cellular lipids after co-incubation with EPA were accompanied by higher lipolysis and fatty acid re-esterification rate. Correspondingly, basal respiration, proton leak and maximal respiration were significantly increased in cells exposed to EPA compared to PA. Microarray and Gene Ontology (GO) enrichment analysis showed that EPA, related to PA, significantly changed i.e. the GO terms "Neutral lipid metabolic process" and "Regulation of lipid storage". Finally, an inhibitor of diacylglycerol acyltransferase 1 decreased the effect of EPA to promote fatty acid accumulation. In conclusion, incubation of human myotubes with EPA, compared to PA, increased processes of fatty acid turnover and oxidation suggesting that EPA may activate futile substrate cycling of fatty acids in human myotubes. Increased TAG-FA cycling may be involved in the potentially favourable effects of long-chain polyunsaturated n-3 fatty acids on skeletal muscle and whole-body energy metabolism.
Collapse
Affiliation(s)
- Nils G. Løvsletten
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
- * E-mail:
| | - Siril S. Bakke
- Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Eili T. Kase
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - D. Margriet Ouwens
- German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - G. Hege Thoresen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Arild C. Rustan
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| |
Collapse
|
23
|
Worsch S, Heikenwalder M, Hauner H, Bader BL. Dietary n-3 long-chain polyunsaturated fatty acids upregulate energy dissipating metabolic pathways conveying anti-obesogenic effects in mice. Nutr Metab (Lond) 2018; 15:65. [PMID: 30275870 PMCID: PMC6158869 DOI: 10.1186/s12986-018-0291-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022] Open
Abstract
Background We previously reported on the anti-obesogenic and anti-inflammatory effects associated with n-3 long-chain polyunsaturated fatty acids (LCPUFA) in our diet-induced obesity (DIO) mouse model. Two isocaloric high-fat diets (HFDs; 48 kJ% fat), HFD (HF) and n-3 LCPUFA-enriched HFD (HF/n-3), and a control diet (C; 13 kJ% fat) were used. The underlying mechanisms however have largely remained unclear. Here, we assessed whether the reduced fat mass reflected n-3 LCPUFA-induced expression changes in lipid metabolism of the intestine, liver, and interscapular brown adipose tissue (iBAT), as well as increased iBAT thermogenic capacity. Methods For HF/n-3, saturated and monounsaturated fatty acids were partially substituted by n-3 LCPUFA eicosapentaenoic acid and docosahexaenoic acid to achieve a balanced n-6/n-3 PUFA ratio (0.84) compared to the unbalanced ratios of HF (13.5) and C (9.85). Intestine, liver and iBAT from male C57BL/6 J mice, fed defined soybean/palm oil-based diets for 12 weeks, were further analysed. Gene and protein expression analyses, immunohistochemistry and correlation analyses for metabolic interactions were performed. Results Compared to HF and C, our analyses suggest significantly diminished de novo lipogenesis (DNL) and/or increased hepatic and intestinal fatty acid oxidation (ω-oxidation and peroxisomal β-oxidation) in HF/n-3 mice. For iBAT, the thermogenic potential was enhanced upon HF/n-3 consistent with upregulated expression for uncoupling protein-1 and genes involved in mitochondrial biogenesis. In addition, a higher capacity for the supply and oxidation of fatty acids was observed and expression and correlation analyses indicated a coordinated regulation of energy metabolism and futile cycling of triacylglycerol (TAG). Moreover, HF/n-3 significantly increased the number of anti-inflammatory macrophages and eosinophils and significantly enhanced the levels of activated AMP-activated protein kinase α (AMPKα), peroxisome proliferator-activated receptor α (PPARα) and fibroblast growth factor 21 (FGF21). Conclusions Our data suggest that by targeting transcriptional regulatory pathways, AMPKα, and FGF21 as potential mediators, HF/n-3 activated less efficient pathways for energy production, such as peroxisomal β-oxidation, increased ATP consumption upon the induction of futile cycling of TAG, and additionally increased the thermogenic and oxidative potential of iBAT. Therefore, we consider n-3 LCPUFA as the potent inducer for upregulating energy dissipating metabolic pathways conveying anti-obesogenic effects in mice. Electronic supplementary material The online version of this article (10.1186/s12986-018-0291-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stefanie Worsch
- 1Else Kroener-Fresenius-Center for Nutritional Medicine, Chair of Nutritional Medicine, Technical University of Munich, Freising, Germany.,2ZIEL - Institute for Food and Health, Nutritional Medicine Unit, Technical University of Munich, Freising, Germany
| | - Mathias Heikenwalder
- 4Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans Hauner
- 1Else Kroener-Fresenius-Center for Nutritional Medicine, Chair of Nutritional Medicine, Technical University of Munich, Freising, Germany.,2ZIEL - Institute for Food and Health, Nutritional Medicine Unit, Technical University of Munich, Freising, Germany.,Else Kroener-Fresenius-Center for Nutritional Medicine, University Hospital Klinikum rechts der Isar, Uptown München-Campus D, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992 Munich, Germany
| | - Bernhard L Bader
- 1Else Kroener-Fresenius-Center for Nutritional Medicine, Chair of Nutritional Medicine, Technical University of Munich, Freising, Germany.,2ZIEL - Institute for Food and Health, Nutritional Medicine Unit, Technical University of Munich, Freising, Germany.,Else Kroener-Fresenius-Center for Nutritional Medicine, University Hospital Klinikum rechts der Isar, Uptown München-Campus D, Technical University of Munich, Georg-Brauchle-Ring 60/62, 80992 Munich, Germany
| |
Collapse
|
24
|
García MDC, Pazos P, Lima L, Diéguez C. Regulation of Energy Expenditure and Brown/Beige Thermogenic Activity by Interleukins: New Roles for Old Actors. Int J Mol Sci 2018; 19:E2569. [PMID: 30158466 PMCID: PMC6164446 DOI: 10.3390/ijms19092569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/25/2018] [Indexed: 12/16/2022] Open
Abstract
Obesity rates and the burden of metabolic associated diseases are escalating worldwide Energy burning brown and inducible beige adipocytes in human adipose tissues (ATs) have attracted considerable attention due to their therapeutic potential to counteract the deleterious metabolic effects of nutritional overload and overweight. Recent research has highlighted the relevance of resident and recruited ATs immune cell populations and their signalling mediators, cytokines, as modulators of the thermogenic activity of brown and beige ATs. In this review, we first provide an overview of the developmental, cellular and functional heterogeneity of the AT organ, as well as reported molecular switches of its heat-producing machinery. We also discuss the key contribution of various interleukins signalling pathways to energy and metabolic homeostasis and their roles in the biogenesis and function of brown and beige adipocytes. Besides local actions, attention is also drawn to their influence in the central nervous system (CNS) networks governing energy expenditure.
Collapse
Affiliation(s)
- María Del Carmen García
- Department of Physiology/Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain.
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III (ISCIII, Ministerio de Economía y Competitividad (MINECO)), C/Monforte de Lemos 3-5, Pabellón 11. Planta 0, 28029 Madrid, Spain.
| | - Patricia Pazos
- Department of Physiology/Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain.
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III (ISCIII, Ministerio de Economía y Competitividad (MINECO)), C/Monforte de Lemos 3-5, Pabellón 11. Planta 0, 28029 Madrid, Spain.
| | - Luis Lima
- Department of Physiology/Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain.
| | - Carlos Diéguez
- Department of Physiology/Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain.
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III (ISCIII, Ministerio de Economía y Competitividad (MINECO)), C/Monforte de Lemos 3-5, Pabellón 11. Planta 0, 28029 Madrid, Spain.
| |
Collapse
|
25
|
Régnier M, Polizzi A, Lippi Y, Fouché E, Michel G, Lukowicz C, Smati S, Marrot A, Lasserre F, Naylies C, Batut A, Viars F, Bertrand-Michel J, Postic C, Loiseau N, Wahli W, Guillou H, Montagner A. Insights into the role of hepatocyte PPARα activity in response to fasting. Mol Cell Endocrinol 2018; 471:75-88. [PMID: 28774777 DOI: 10.1016/j.mce.2017.07.035] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/04/2017] [Accepted: 07/28/2017] [Indexed: 12/28/2022]
Abstract
The liver plays a central role in the regulation of fatty acid metabolism. Hepatocytes are highly sensitive to nutrients and hormones that drive extensive transcriptional responses. Nuclear hormone receptors are key transcription factors involved in this process. Among these factors, PPARα is a critical regulator of hepatic lipid catabolism during fasting. This study aimed to analyse the wide array of hepatic PPARα-dependent transcriptional responses during fasting. We compared gene expression in male mice with a hepatocyte specific deletion of PPARα and their wild-type littermates in the fed (ad libitum) and 24-h fasted states. Liver samples were acquired, and transcriptome and lipidome analyses were performed. Our data extended and confirmed the critical role of hepatocyte PPARα as a central for regulator of gene expression during starvation. Interestingly, we identified novel PPARα-sensitive genes, including Cxcl-10, Rab30, and Krt23. We also found that liver phospholipid remodelling was a novel fasting-sensitive pathway regulated by PPARα. These results may contribute to investigations on transcriptional control in hepatic physiology and underscore the clinical relevance of drugs that target PPARα in liver pathologies, such as non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Marion Régnier
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Arnaud Polizzi
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Yannick Lippi
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Edwin Fouché
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Géraldine Michel
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Céline Lukowicz
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Sarra Smati
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France; Institut National de La Santé et de La Recherche Médicale (INSERM), UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Alain Marrot
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Frédéric Lasserre
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Claire Naylies
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Aurélie Batut
- Metatoul-Lipidomic Facility, MetaboHUB, Institut National de La Santé et de La Recherche Médicale (INSERM), UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Fanny Viars
- Metatoul-Lipidomic Facility, MetaboHUB, Institut National de La Santé et de La Recherche Médicale (INSERM), UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Justine Bertrand-Michel
- Metatoul-Lipidomic Facility, MetaboHUB, Institut National de La Santé et de La Recherche Médicale (INSERM), UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Catherine Postic
- Institut National de La Santé et de La Recherche Médicale (INSERM), U1016, Institut Cochin, Paris, France
| | - Nicolas Loiseau
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Walter Wahli
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France; Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, 308232, Singapore; Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| | - Hervé Guillou
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France.
| | - Alexandra Montagner
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France; Institut National de La Santé et de La Recherche Médicale (INSERM), UMR1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.
| |
Collapse
|
26
|
Araki M, Nakagawa Y, Oishi A, Han SI, Wang Y, Kumagai K, Ohno H, Mizunoe Y, Iwasaki H, Sekiya M, Matsuzaka T, Shimano H. The Peroxisome Proliferator-Activated Receptor α (PPARα) Agonist Pemafibrate Protects against Diet-Induced Obesity in Mice. Int J Mol Sci 2018; 19:ijms19072148. [PMID: 30041488 PMCID: PMC6073532 DOI: 10.3390/ijms19072148] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/13/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a therapeutic target for hyperlipidemia. Pemafibrate (K-877) is a new selective PPARα modulator activating PPARα transcriptional activity. To determine the effects of pemafibrate on diet-induced obesity, wild-type mice were fed a high-fat diet (HFD) containing pemafibrate for 12 weeks. Like fenofibrate, pemafibrate significantly suppressed HFD-induced body weight gain; decreased plasma glucose, insulin and triglyceride (TG) levels; and increased plasma fibroblast growth factor 21 (FGF21). However, compared to the dose of fenofibrate, a relatively low dose of pemafibrate showed these effects. Pemafibrate activated PPARα transcriptional activity in the liver, increasing both hepatic expression and plasma levels of FGF21. Additionally, pemafibrate increased the expression of genes involved in thermogenesis and fatty acid oxidation, including Ucp1, Cidea and Cpt1b in inguinal adipose tissue (iWAT) and the mitochondrial marker Elovl3 in brown adipose tissue (BAT). Therefore, pemafibrate activates thermogenesis in iWAT and BAT by increasing plasma levels of FGF21. Additionally, pemafibrate induced the expression of Atgl and Hsl in epididymal white adipose tissue, leading to the activation of lipolysis. Taken together, pemafibrate suppresses diet-induced obesity in mice and improves their obesity-related metabolic abnormalities. We propose that pemafibrate may be useful for the suppression and improvement of obesity-induced metabolic abnormalities.
Collapse
Affiliation(s)
- Masaya Araki
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Yoshimi Nakagawa
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Asayo Oishi
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Song-Iee Han
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Yunong Wang
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Kae Kumagai
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Hiroshi Ohno
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Yuhei Mizunoe
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Hitoshi Iwasaki
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Motohiro Sekiya
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
- Japan Agency for Medical Research and Development⁻Core Research for Evolutional Science and Technology (AMED-CREST), Chiyoda-ku, Tokyo 100-1004, Japan.
| |
Collapse
|
27
|
Kang M, Liu X, Fu Y, Timothy Garvey W. Improved systemic metabolism and adipocyte biology in miR-150 knockout mice. Metabolism 2018; 83:139-148. [PMID: 29352962 PMCID: PMC6142816 DOI: 10.1016/j.metabol.2017.12.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Short non-coding micro-RNAs (miRNAs) are post-transcriptional factors that directly regulate protein expression by degrading or inhibiting target mRNAs; however, the role of miRNAs in obesity and cardiometabolic disease remains unclarified. Based on our earlier study demonstrating that miR-150 influences lipid metabolism, we have studied effects of miR-150 on systemic metabolism and adipocyte biology. MATERIALS AND METHODS Metabolic phenotypes including body weight, food intake, body composition, glucose tolerance and insulin sensitivity were assessed in WT and global miR-150 KO male mice fed a high-fat diet. Molecular changes in epididymal adipose tissue were evaluated through qRT-PCR and Western blotting. RESULTS miR-150 KO mice displayed lower body weight characterized by a reduction in % fat mass while % lean mass was increased. Lower body weight was associated with reduced food consumption and an increase in circulating leptin concentrations, as well as enhanced insulin sensitivity and glucose tolerance compared with WT mice. Absence of miR-150 resulted in increased mTOR expression known to participate in increased leptin production leading to reduction of food intake. Expression of PGC-1α, another target gene of miR-150, was also increased together with upregulation of PPARα and glycerol kinase in adipose tissue as well as other genes participating in triglyceride degradation and lipid oxidation. CONCLUSION miR-150 KO mice showed metabolic benefits accompanied by reduced body weight, decreased energy intake, and enhanced lipid metabolism. miR-150 may represent both a biomarker and novel therapeutic target regarding obesity and insulin resistance.
Collapse
Affiliation(s)
- Minsung Kang
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Xiaobing Liu
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yuchang Fu
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - W Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA; Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
28
|
A map of the PGC-1α- and NT-PGC-1α-regulated transcriptional network in brown adipose tissue. Sci Rep 2018; 8:7876. [PMID: 29777200 PMCID: PMC5959870 DOI: 10.1038/s41598-018-26244-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/04/2018] [Indexed: 11/19/2022] Open
Abstract
Transcriptional coactivator PGC-1α and its splice variant NT-PGC-1α play crucial roles in regulating cold-induced thermogenesis in brown adipose tissue (BAT). PGC-1α and NT-PGC-1α are highly induced by cold in BAT and subsequently bind to and coactivate many transcription factors to regulate expression of genes involved in mitochondrial biogenesis, fatty acid oxidation, respiration and thermogenesis. To identify the complete repertoire of PGC-1α and NT-PGC-1α target genes in BAT, we analyzed genome-wide DNA-binding and gene expression profiles. We find that PGC-1α-/NT-PGC-1α binding broadly associates with cold-mediated transcriptional activation. In addition to their known target genes in mitochondrial biogenesis and oxidative metabolism, PGC-1α and NT-PGC-1α additionally target a broad spectrum of genes involved in diverse biological pathways including ubiquitin-dependent protein catabolism, ribonucleoprotein complex biosynthesis, phospholipid biosynthesis, angiogenesis, glycogen metabolism, phosphorylation, and autophagy. Our findings expand the number of genes and biological pathways that may be regulated by PGC-1α and NT-PGC-1α and provide further insight into the transcriptional regulatory network in which PGC-1α and NT-PGC-1α coordinate a comprehensive transcriptional response in BAT in response to cold.
Collapse
|
29
|
Abstract
Huntington's disease (HD) as an inherited neurodegenerative disorder leads to neuronal loss in striatum. Progressive motor dysfunction, cognitive decline, and psychiatric disturbance are the main clinical symptoms of the HD. This disease is caused by expansion of the CAG repeats in exon 1 of the huntingtin which encodes Huntingtin protein (Htt). Various cellular and molecular events play role in the pathology of HD. Mitochondria as important organelles play crucial roles in the most of neurodegenerative disorders like HD. Critical roles of the mitochondria in neurons are ATP generation, Ca2+ buffering, ROS generation, and antioxidant activity. Neurons as high-demand energy cells closely related to function, maintenance, and dynamic of mitochondria. In the most neurological disorders, mitochondrial activities and dynamic are disrupted which associate with high ROS level, low ATP generation, and apoptosis. Accumulation of mutant huntingtin (mHtt) during this disease may evoke mitochondrial dysfunction. Here, we review recent findings to support this hypothesis that mHtt could cause mitochondrial defects. In addition, by focusing normal huntingtin functions in neurons, we purpose mitochondria and Huntingtin association in normal condition. Moreover, mHtt affects various cellular signaling which ends up to mitochondrial biogenesis. So, it could be a potential candidate to decline ATP level in HD. We conclude how mitochondrial biogenesis plays a central role in the neuronal survival and activity and how mHtt affects mitochondrial trafficking, maintenance, integrity, function, dynamics, and hemostasis and makes neurons vulnerable to degeneration in HD.
Collapse
|
30
|
Goto T, Hirata M, Aoki Y, Iwase M, Takahashi H, Kim M, Li Y, Jheng HF, Nomura W, Takahashi N, Kim CS, Yu R, Seno S, Matsuda H, Aizawa-Abe M, Ebihara K, Itoh N, Kawada T. The hepatokine FGF21 is crucial for peroxisome proliferator-activated receptor-α agonist-induced amelioration of metabolic disorders in obese mice. J Biol Chem 2017; 292:9175-9190. [PMID: 28404815 DOI: 10.1074/jbc.m116.767590] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 04/12/2017] [Indexed: 01/05/2023] Open
Abstract
Obesity causes excess fat accumulation in white adipose tissues (WAT) and also in other insulin-responsive organs such as the skeletal muscle, increasing the risk for insulin resistance, which can lead to obesity-related metabolic disorders. Peroxisome proliferator-activated receptor-α (PPARα) is a master regulator of fatty acid oxidation whose activator is known to improve hyperlipidemia. However, the molecular mechanisms underlying PPARα activator-mediated reduction in adiposity and improvement of metabolic disorders are largely unknown. In this study we investigated the effects of PPARα agonist (fenofibrate) on glucose metabolism dysfunction in obese mice. Fenofibrate treatment reduced adiposity and attenuated obesity-induced dysfunctions of glucose metabolism in obese mice fed a high-fat diet. However, fenofibrate treatment did not improve glucose metabolism in lipodystrophic A-Zip/F1 mice, suggesting that adipose tissue is important for the fenofibrate-mediated amelioration of glucose metabolism, although skeletal muscle actions could not be completely excluded. Moreover, we investigated the role of the hepatokine fibroblast growth factor 21 (FGF21), which regulates energy metabolism in adipose tissue. In WAT of WT mice, but not of FGF21-deficient mice, fenofibrate enhanced the expression of genes related to brown adipocyte functions, such as Ucp1, Pgc1a, and Cpt1b Fenofibrate increased energy expenditure and attenuated obesity, whole body insulin resistance, and adipocyte dysfunctions in WAT in high-fat-diet-fed WT mice but not in FGF21-deficient mice. These findings indicate that FGF21 is crucial for the fenofibrate-mediated improvement of whole body glucose metabolism in obese mice via the amelioration of WAT dysfunctions.
Collapse
Affiliation(s)
- Tsuyoshi Goto
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan, .,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501 Japan
| | - Mariko Hirata
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Yumeko Aoki
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Mari Iwase
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Haruya Takahashi
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Minji Kim
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Yongjia Li
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Huei-Fen Jheng
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan
| | - Wataru Nomura
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501 Japan
| | - Nobuyuki Takahashi
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501 Japan
| | - Chu-Sook Kim
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 680-749, South Korea
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 680-749, South Korea
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita 565-0871, Japan
| | - Hideo Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita 565-0871, Japan
| | - Megumi Aizawa-Abe
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto 606-8507, Japan, and
| | - Ken Ebihara
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto 606-8507, Japan, and
| | - Nobuyuki Itoh
- Department of Genetic Biochemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto 606-8501, Japan
| | - Teruo Kawada
- From the Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji 611-0011, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto 606-8501 Japan
| |
Collapse
|
31
|
Barquissau V, Ghandour RA, Ailhaud G, Klingenspor M, Langin D, Amri EZ, Pisani DF. Control of adipogenesis by oxylipins, GPCRs and PPARs. Biochimie 2016; 136:3-11. [PMID: 28034718 DOI: 10.1016/j.biochi.2016.12.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/02/2016] [Accepted: 12/23/2016] [Indexed: 01/15/2023]
Abstract
Oxylipins are bioactive metabolites derived from the oxygenation of ω3 and ω6 polyunsaturated fatty acids, triggered essentially by cyclooxygenase and lipoxygenase activities. Oxylipins are involved in the development and function of adipose tissue and their productions are strictly related to diet quality and quantity. Oxylipins signal via cell surface membrane (G Protein-coupled receptors) and nuclear receptors (peroxisome proliferator-activated receptors), two pathways playing a pivotal role in adipocyte biology. In this review, we made an attempt to cover the available knowledge about synthesis and molecular function of oxylipins known to modulate adipogenesis, adipocyte function and phenotype conversion, with a focus on their interaction with peroxisome proliferator-activated nuclear receptor family.
Collapse
Affiliation(s)
- Valentin Barquissau
- Inserm, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, 31432, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, 31432, France
| | | | | | - Martin Klingenspor
- Technische Universität München, Chair of Molecular Nutritional Medicine, Else Kröner-Fresenius Center, 85350, Freising-Weihenstephan, Germany
| | - Dominique Langin
- Inserm, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, 31432, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, 31432, France; Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, 31059, France
| | | | | |
Collapse
|
32
|
Regulation of Brown and White Adipocyte Transcriptome by the Transcriptional Coactivator NT-PGC-1α. PLoS One 2016; 11:e0159990. [PMID: 27454177 PMCID: PMC4959749 DOI: 10.1371/journal.pone.0159990] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/28/2016] [Indexed: 02/07/2023] Open
Abstract
The β3-adrenergic receptor (AR) signaling pathway is a major component of adaptive thermogenesis in brown and white adipose tissue during cold acclimation. The β3-AR signaling highly induces the expression of transcriptional coactivator PGC-1α and its splice variant N-terminal (NT)-PGC-1α, which in turn activate the transcription program of adaptive thermogenesis by co-activating a number of transcription factors. We previously reported that NT-PGC-1α is able to increase mitochondrial number and activity in cultured brown adipocytes by promoting the expression of mitochondrial and thermogenic genes. In the present study, we performed genome-wide profiling of NT-PGC-1α-responsive genes in brown adipocytes to identify genes potentially regulated by NT-PGC-1α. Canonical pathway analysis revealed that a number of genes upregulated by NT-PGC-1α are highly enriched in mitochondrial pathways including fatty acid transport and β-oxidation, TCA cycle and electron transport system, thus reinforcing the crucial role of NT-PGC-1α in the enhancement of mitochondrial function. Moreover, canonical pathway analysis of NT-PGC-1α-responsive genes identified several metabolic pathways including glycolysis and fatty acid synthesis. In order to validate the identified genes in vivo, we utilized the FL-PGC-1α-/- mouse that is deficient in full-length PGC-1α (FL-PGC-1α) but expresses a slightly shorter and functionally equivalent form of NT-PGC-1α (NT-PGC-1α254). The β3-AR-induced increase of NT-PGC-1α254 in FL-PGC-1α-/- brown and white adipose tissue was closely associated with elevated expression of genes involved in thermogenesis, mitochondrial oxidative metabolism, glycolysis and fatty acid synthesis. Increased adipose tissue thermogenesis by β3-AR activation resulted in attenuation of adipose tissue expansion in FL-PGC-1α-/- adipose tissue under the high-fat diet condition. Together, the data strengthen our previous findings that NT-PGC-1α regulates mitochondrial genes involved in thermogenesis and oxidative metabolism in brown and white adipocytes and further suggest that NT-PGC-1α regulates a broad spectrum of genes to meet cellular needs for adaptive thermogenesis.
Collapse
|
33
|
Zhuang W, Zhang H, Pan J, Li Z, Wei T, Cui H, Liu Z, Guan Q, Dong H, Zhang Z. PEDF and PEDF-derived peptide 44mer inhibit oxygen-glucose deprivation-induced oxidative stress through upregulating PPARγ via PEDF-R in H9c2 cells. Biochem Biophys Res Commun 2016; 472:482-8. [PMID: 26966066 DOI: 10.1016/j.bbrc.2016.02.110] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 01/30/2023]
Abstract
Pigment epithelial-derived factor (PEDF) is a glycoprotein with broad biological activities including inhibiting oxygen-glucose deprivation(OGD)-induced cardiomyocytes apoptosis through its anti-oxidative properties. PEDF derived peptide-44mer shows similar cytoprotective effect to PEDF. However, the molecular mechanisms mediating cardiomyocytes apoptosis have not been fully established. Here we found that PEDF and 44mer decreased the content of ROS. This content was abolished by either PEDF-R small interfering RNA (siRNA) or PPARγ antagonist. The level of Lysophosphatidic acid (LPA) and phospholipase A2 (PLA2) was observed as drawn from the ELISA assays. PEDF and 44mer sequentially induced PPARγ expression was observed both in qPCR and Western blot assays. The level of LPA and PLA2 and PPARγ expression increased by PEDF and 44mer was significantly attenuated by PEDF-R siRNA. However, PEDF and 44mer inhibited the H9c2 cells and cultured neonatal rat myocardial cells apoptosis rate. On the other hand, TUNEL assay and cleavage of procaspase-3 showed that PEDF-R siRNA or PPARγ antagonist increased the apoptosis again. We conclude that under OGD condition, PEDF and 44mer reduce H9c2 cells apoptosis and inhibit OGD-induced oxidative stress via its receptor PEDF-R and the PPARγ signaling pathway.
Collapse
Affiliation(s)
- Wei Zhuang
- Research Facility Center for Morphology, 209 Tong shan Road, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, China
| | - Hao Zhang
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical College, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
| | - Jiajun Pan
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical College, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
| | - Zhimin Li
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical College, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
| | - Tengteng Wei
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical College, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
| | - Huazhu Cui
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical College, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
| | - Zhiwei Liu
- Research Facility Center for Morphology, 209 Tong shan Road, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, China
| | - Qiuhua Guan
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, China
| | - Hongyan Dong
- Research Facility Center for Morphology, 209 Tong shan Road, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, China.
| | - Zhongming Zhang
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical College, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
34
|
Barquissau V, Beuzelin D, Pisani DF, Beranger GE, Mairal A, Montagner A, Roussel B, Tavernier G, Marques MA, Moro C, Guillou H, Amri EZ, Langin D. White-to-brite conversion in human adipocytes promotes metabolic reprogramming towards fatty acid anabolic and catabolic pathways. Mol Metab 2016; 5:352-365. [PMID: 27110487 PMCID: PMC4837301 DOI: 10.1016/j.molmet.2016.03.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/13/2016] [Indexed: 12/29/2022] Open
Abstract
Objective Fat depots with thermogenic activity have been identified in humans. In mice, the appearance of thermogenic adipocytes within white adipose depots (so-called brown-in-white i.e., brite or beige adipocytes) protects from obesity and insulin resistance. Brite adipocytes may originate from direct conversion of white adipocytes. The purpose of this work was to characterize the metabolism of human brite adipocytes. Methods Human multipotent adipose-derived stem cells were differentiated into white adipocytes and then treated with peroxisome proliferator-activated receptor (PPAR)γ or PPARα agonists between day 14 and day 18. Gene expression profiling was determined using DNA microarrays and RT-qPCR. Variations of mRNA levels were confirmed in differentiated human preadipocytes from primary cultures. Fatty acid and glucose metabolism was investigated using radiolabelled tracers, Western blot analyses and assessment of oxygen consumption. Pyruvate dehydrogenase kinase 4 (PDK4) knockdown was achieved using siRNA. In vivo, wild type and PPARα-null mice were treated with a β3-adrenergic receptor agonist (CL316,243) to induce appearance of brite adipocytes in white fat depot. Determination of mRNA and protein levels was performed on inguinal white adipose tissue. Results PPAR agonists promote a conversion of white adipocytes into cells displaying a brite molecular pattern. This conversion is associated with transcriptional changes leading to major metabolic adaptations. Fatty acid anabolism i.e., fatty acid esterification into triglycerides, and catabolism i.e., lipolysis and fatty acid oxidation, are increased. Glucose utilization is redirected from oxidation towards glycerol-3-phophate production for triglyceride synthesis. This metabolic shift is dependent on the activation of PDK4 through inactivation of the pyruvate dehydrogenase complex. In vivo, PDK4 expression is markedly induced in wild-type mice in response to CL316,243, while this increase is blunted in PPARα-null mice displaying an impaired britening response. Conclusions Conversion of human white fat cells into brite adipocytes results in a major metabolic reprogramming inducing fatty acid anabolic and catabolic pathways. PDK4 redirects glucose from oxidation towards triglyceride synthesis and favors the use of fatty acids as energy source for uncoupling mitochondria. PPARγ and α agonists induce conversion of human white into brite adipocytes. Fatty acid anabolism and catabolism are activated in human brite adipocytes. Glucose use in brite adipocytes is redirected from oxidation to glyceroneogenesis. PDK4 induction is responsible for the shift from glucose to fatty acid oxidation.
Collapse
Affiliation(s)
- V Barquissau
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - D Beuzelin
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - D F Pisani
- University of Nice Sophia Antipolis, Nice, France; CNRS, iBV, UMR 7277, Nice, France; INSERM, iBV, U 1091, Nice, France
| | - G E Beranger
- University of Nice Sophia Antipolis, Nice, France; CNRS, iBV, UMR 7277, Nice, France; INSERM, iBV, U 1091, Nice, France
| | - A Mairal
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - A Montagner
- University of Toulouse, Paul Sabatier University, France; INRA, UMR 1331, TOXALIM, Toulouse, France
| | - B Roussel
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - G Tavernier
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - M-A Marques
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - C Moro
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France
| | - H Guillou
- University of Toulouse, Paul Sabatier University, France; INRA, UMR 1331, TOXALIM, Toulouse, France
| | - E-Z Amri
- University of Nice Sophia Antipolis, Nice, France; CNRS, iBV, UMR 7277, Nice, France; INSERM, iBV, U 1091, Nice, France
| | - D Langin
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, Paul Sabatier University, France; Toulouse University Hospitals, Laboratory of Clinical Biochemistry, Toulouse, France.
| |
Collapse
|
35
|
Roman S, Agil A, Peran M, Alvaro-Galue E, Ruiz-Ojeda FJ, Fernández-Vázquez G, Marchal JA. Brown adipose tissue and novel therapeutic approaches to treat metabolic disorders. Transl Res 2015; 165:464-79. [PMID: 25433289 DOI: 10.1016/j.trsl.2014.11.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/16/2014] [Accepted: 11/04/2014] [Indexed: 01/08/2023]
Abstract
In humans, 2 functionally different types of adipose tissue coexist: white adipose tissue (WAT) and brown adipose tissue (BAT). WAT is involved in energy storage, whereas BAT is involved in energy expenditure. Increased amounts of WAT may contribute to the development of metabolic disorders, such as obesity-associated type 2 diabetes mellitus and cardiovascular diseases. In contrast, the thermogenic function of BAT allows high consumption of fatty acids because of the activity of uncoupling protein 1 in the internal mitochondrial membrane. Interestingly, obesity reduction and insulin sensitization have been achieved by BAT activation-regeneration in animal models. This review describes the origin, function, and differentiation mechanisms of BAT to identify new therapeutic strategies for the treatment of metabolic disorders related to obesity. On the basis of the animal studies, novel approaches for BAT regeneration combining stem cells from the adipose tissue with active components, such as melatonin, may have potential for the treatment of metabolic disorders in humans.
Collapse
Affiliation(s)
- Sabiniano Roman
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada, Spain; Tissue Engineering Group, Kroto Research Institute, University of Sheffield, Sheffield, UK
| | - Ahmad Agil
- Department of Pharmacology and Neurosciences Institute, Faculty of Medicine, Biosanitary Institute of Granada (ibs.GRANADA), Hospitals Unversity/University of Granada, Granada, Spain
| | - Macarena Peran
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada, Spain; Department of Health Sciences, University of Jaén, Jaén, Spain
| | - Eduardo Alvaro-Galue
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada, Spain; Wake Forest Institute for Regenerative Medicine Wake Forest School of Medicine, Winston Salem, NC
| | - Francisco J Ruiz-Ojeda
- Institute of Nutrition and Food Technology, Centre for Biomedical Research, University of Granada, Granada, Spain
| | | | - Juan A Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada, Spain; Department of Human Anatomy and Embryology, Biosanitary Institute of Granada (ibs.GRANADA), Hospitals Unversity/University of Granada, Granada, Spain.
| |
Collapse
|
36
|
Yu J, Zhang S, Cui L, Wang W, Na H, Zhu X, Li L, Xu G, Yang F, Christian M, Liu P. Lipid droplet remodeling and interaction with mitochondria in mouse brown adipose tissue during cold treatment. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:918-28. [PMID: 25655664 DOI: 10.1016/j.bbamcr.2015.01.020] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/22/2014] [Accepted: 01/16/2015] [Indexed: 01/17/2023]
Abstract
Brown adipose tissue (BAT) maintains animal body temperature by non-shivering thermogenesis, which is through uncoupling protein 1 (UCP1) that uncouples oxidative phosphorylation and utilizes β-oxidation of fatty acids released from triacylglycerol (TAG) in lipid droplets (LDs). Increasing BAT activity and "browning" other tissues such as white adipose tissue (WAT) can enhance the expenditure of excess stored energy, and in turn reduce prevalence of metabolic diseases. Although many studies have characterized the biology of BAT and brown adipocytes, BAT LDs especially their activation induced by cold exposure remain to be explored. We have isolated LDs from mouse interscapular BAT and characterized the full proteome using mass spectrometry. Both morphological and biochemical experiments showed that the LDs could tightly associate with mitochondria. Under cold treatment mouse BAT started expressing LD structure protein PLIN-2/ADRP and increased expression of PLIN1. Both hormone sensitive lipase (HSL) and adipose TAG lipase (ATGL) were increased in LDs. In addition, isolated BAT LDs showed increased levels of the mitochondrial protein UCP1, and prolonged cold exposure could stimulate BAT mitochondrial cristae biogenesis. These changes were in agreement with the data from transcriptional analysis. Our results provide the BAT LD proteome for the first time and show that BAT LDs facilitate heat production by coupling increasing TAG hydrolysis through recruitment of ATGL and HSL to the organelle and expression of another LD resident protein PLIN2/ADRP, as well as by tightly associating with activated mitochondria. These findings will benefit the study of BAT activation and the interaction between LDs and mitochondria.
Collapse
Affiliation(s)
- Jinhai Yu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Liujuan Cui
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Weiyi Wang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Huimin Na
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaotong Zhu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linghai Li
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Guoheng Xu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Fuquan Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Mark Christian
- Division of Translational and Systems Medicine, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
37
|
Renes J, Rosenow A, Roumans N, Noben JP, Mariman EC. Calorie restriction-induced changes in the secretome of human adipocytes, comparison with resveratrol-induced secretome effects. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:1511-22. [DOI: 10.1016/j.bbapap.2014.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 12/17/2022]
|
38
|
Ameliorative Effects of Mulberry (Morus albaL.) Leaves on Hyperlipidemia in Rats Fed a High-Fat Diet: Induction of Fatty Acid Oxidation, Inhibition of Lipogenesis, and Suppression of Oxidative Stress. Biosci Biotechnol Biochem 2014; 74:2385-95. [DOI: 10.1271/bbb.100392] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Liu BH, Lin YY, Wang YC, Huang CW, Chen CC, Wu SC, Mersmann HJ, Cheng WTK, Ding ST. Porcine adiponectin receptor 1 transgene resists high-fat/sucrose diet-induced weight gain, hepatosteatosis and insulin resistance in mice. Exp Anim 2014; 62:347-60. [PMID: 24172199 PMCID: PMC4160961 DOI: 10.1538/expanim.62.347] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Adiponectin and its receptors have been demonstrated to play important roles in regulating glucose and lipid metabolism in mice. Obesity, type II diabetes and cardiovascular disease are highly correlated with down-regulated adiponectin signaling. In this study, we generated mice overexpressing the porcine Adipor1 transgene (pAdipor1) to study its beneficial effects in metabolic syndromes as expressed in diet-induced obesity, hepatosteatosis and insulin resistance. Wild-type (WT) and pAdipor1 transgenic mice were fed ad libitum with a standard chow diet (Chow) or a high-fat/sucrose diet (HFSD) for 24 weeks, beginning at 6 to 7 weeks of age. There were 12 mice per genetic/diet/sex group. When challenged with HFSD to induce obesity, the pAdipor1 transgenic mice resisted development of weight gain, hepatosteatosis and insulin resistance. These mice had lowered plasma adiponectin, triglyceride and glycerol concentrations compared to WT mice. Moreover, we found that (indicated by mRNA levels) fatty acid oxidation was enhanced in skeletal muscle and adipose tissue, and liver lipogenesis was inhibited. The pAdipor1 transgene also restored HFSD-reduced phosphoenolpyruvate carboxykinase 1 (Pck1) and glucose transporter 4 mRNA in the adipose tissues, implying that the increased Pck1 may promote glyceroneogenesis to reduce glucose intolerance and thus activate the flux of glyceride-glycerol to resist diet-induced weight gain in the adipose tissues. Taken together, we demonstrated that pAdipor1 can prevent diet-induced weight gain and insulin resistance. Our findings may provide potential therapeutic strategies for treating metabolic syndromes and obesity, such as treatment with an ADIPOR1 agonist or activation of Adipor1 downstream targets.
Collapse
Affiliation(s)
- Bing-Hsien Liu
- Department of Animal Science and Technology, National Taiwan University, No. 50, Ln. 155, Sec. 3, Keelung Rd., Da'an Dist., Taipei City 106, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kiskinis E, Chatzeli L, Curry E, Kaforou M, Frontini A, Cinti S, Montana G, Parker MG, Christian M. RIP140 represses the "brown-in-white" adipocyte program including a futile cycle of triacylglycerol breakdown and synthesis. Mol Endocrinol 2014; 28:344-356. [PMID: 24479876 PMCID: PMC4207910 DOI: 10.1210/me.2013-1254] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 01/22/2014] [Indexed: 11/30/2022] Open
Abstract
Receptor-interacting protein 140 (RIP140) is a corepressor of nuclear receptors that is highly expressed in adipose tissues. We investigated the role of RIP140 in conditionally immortal preadipocyte cell lines prepared from white or brown fat depots. In white adipocytes, a large set of brown fat-associated genes was up-regulated in the absence of RIP140. In contrast, a relatively minor role can be ascribed to RIP140 in the control of basal gene expression in differentiated brown adipocytes because significant changes were observed only in Ptgds and Fabp3. The minor role of RIP140 in brown adipocytes correlates with the similar histology and uncoupling protein 1 and CIDEA staining in knockout compared with wild-type brown adipose tissue (BAT). In contrast, RIP140 knockout sc white adipose tissue (WAT) shows increased numbers of multilocular adipocytes with elevated staining for uncoupling protein 1 and CIDEA. Furthermore in a white adipocyte cell line, the markers of BRITE adipocytes, Tbx1, CD137, Tmem26, Cited1, and Epsti1 were repressed in the presence of RIP140 as was Prdm16. Microarray analysis of wild-type and RIP140-knockout white fat revealed elevated expression of genes associated with cold-induced expression or high expression in BAT. A set of genes associated with a futile cycle of triacylglycerol breakdown and resynthesis and functional assays revealed that glycerol kinase and glycerol-3-phosphate dehydrogenase activity as well as [(3)H]glycerol incorporation were elevated in the absence of RIP140. Thus, RIP140 blocks the BRITE program in WAT, preventing the expression of brown fat genes and inhibiting a triacylglycerol futile cycle, with important implications for energy homeostasis.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/physiology
- Adipocytes, Brown/metabolism
- Adipose Tissue, Brown/cytology
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/cytology
- Adipose Tissue, White/metabolism
- Animals
- Cell Differentiation
- Cells, Cultured
- Female
- Gene Silencing
- Lipid Metabolism
- Male
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Nuclear Proteins/physiology
- Nuclear Receptor Interacting Protein 1
- Oligonucleotide Array Sequence Analysis
- Transcriptome
- Triglycerides/biosynthesis
- Triglycerides/metabolism
Collapse
Affiliation(s)
- Evangelos Kiskinis
- Department of Stem Cell and Regenerative Biology (E.K.), Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138; Institute of Reproductive and Developmental Biology (L.C., E.C., M.G.P.), Faculty of Medicine, Imperial College London, W12 0NN, United Kingdom; Department of Mathematics (M.K., G.M.), Statistics Section, Imperial College London, London SW7 2AZ, United Kingdom; Department of Experimental and Clinical Medicine (A.F., S.C.), University of Ancona, (Politecnica delle Marche), 60126 Ancona, Italy; Division of Metabolic and Vascular Health (M.C.), Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chen L, Song J, Cui J, Hou J, Zheng X, Li C, Liu L. microRNAs regulate adipocyte differentiation. Cell Biol Int 2013; 37:533-546. [PMID: 23504919 DOI: 10.1002/cbin.10063] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 01/22/2013] [Indexed: 02/05/2023]
Abstract
The number of adipocytes is relevant to the extent of differentiation from pluripotent stem cells into pre-adipocytes, whereas the size of adipocytes relates to the extent of differentiation from pre-adipocytes into mature fat cells and the accumulation of triglyceride. Investigation of the molecular regulatory mechanism of adipocyte differentiation is not only essential for understanding the physiological processes of adipogenesis, but it is also important for identifying new biomarkers and therapeutic targets for some metabolic diseases, such as obesity and diabetes. microRNAs (miRNAs) appear to play important roles in adipocyte differentiation. During adipogenesis, miRNAs can accelerate or inhibit adipocyte differentiation by acting on transcription factors, regulating signalling pathways related to adipogenesis, or blocking the mitotic clonal expansion stage, thus regulating adipocyte development. The regulatory role of some miRNAs varies in different species or different cells. In this review, the biological characteristics of miRNA and the adipocyte differentiation process are concisely discussed. Recent advances in our understanding of the role of miRNAs in adipocytes development or adipogenesis are discussed.
Collapse
Affiliation(s)
- Lin Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | | | | | | | | | | | | |
Collapse
|
42
|
Beranger GE, Karbiener M, Barquissau V, Pisani DF, Scheideler M, Langin D, Amri EZ. In vitro brown and “brite”/“beige” adipogenesis: Human cellular models and molecular aspects. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:905-14. [DOI: 10.1016/j.bbalip.2012.11.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Revised: 10/31/2012] [Accepted: 11/02/2012] [Indexed: 11/30/2022]
|
43
|
Flachs P, Rossmeisl M, Kuda O, Kopecky J. Stimulation of mitochondrial oxidative capacity in white fat independent of UCP1: A key to lean phenotype. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:986-1003. [DOI: 10.1016/j.bbalip.2013.02.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 02/06/2013] [Accepted: 02/09/2013] [Indexed: 02/06/2023]
|
44
|
Ahmadi N, Nabavi V, Hajsadeghi F, Zeb I, Flores F, Ebrahimi R, Budoff M. Aged garlic extract with supplement is associated with increase in brown adipose, decrease in white adipose tissue and predict lack of progression in coronary atherosclerosis. Int J Cardiol 2013; 168:2310-4. [PMID: 23453866 DOI: 10.1016/j.ijcard.2013.01.182] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 11/24/2012] [Accepted: 01/18/2013] [Indexed: 01/04/2023]
Abstract
BACKGROUND Aged garlic extract with supplement (AGE-S) significantly reduces coronary artery calcium (CAC). We evaluated the effects of AGE-S on change in white (wEAT) and brown (bEAT) epicardial adipose tissue, homocysteine and CAC. METHODS Sixty subjects, randomized to a daily capsule of placebo vs. AGE-S inclusive of aged garlic-extract (250 mg) plus vitamin-B12 (100 μg), folic-acid (300 μg), vitamin-B6 (12.5mg) and L-arginine (100mg) underwent CAC, wEAT and bEAT measurements at baseline and 12 months. The postcuff deflation temperature-rebound index of vascular function was assessed using a reactive-hyperemia procedure. Vascular dysfunction was defined according to the tertiles of temperature-rebound at 1 year of follow-up. CAC progression was defined as an annual-increase in CAC>15%. RESULTS From baseline to 12 months, there was a strong correlation between increase in wEAT and CAC (r(2)=0.54, p=0.0001). At 1 year, the risks of CAC progression and increased wEAT and homocysteine were significantly lower in AGE-S to placebo (p<0.05). Similarly, bEAT and temperature-rebound were significantly higher in AGE-S as compared to placebo (p<0.05). Strong association between increase in temperature-rebound and bEAT/wEAT ratio (r(2)=0.80, p=0.001) was noted, which was more robust in AGE-S. Maximum beneficial effect of AGE-S was noted with increase in bEAT/wEAT ratio, temperature-rebound, and lack of progression of homocysteine and CAC. CONCLUSIONS AGE-S is associated with increase in bEAT/wEAT ratio, reduction of homocysteine and lack of progression of CAC. Increases in bEAT/wEAT ratio correlated strongly with increases in vascular function measured by temperature-rebound and predicted a lack of CAC progression and plaque stabilization in response to AGE-S.
Collapse
Affiliation(s)
- Naser Ahmadi
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, USA; Greater Los Angeles Veterans Administration Medical Center, UCLA-School of Medicine, Los Angeles, CA, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Ducreux S, Gregory P, Schwaller B. Inverse regulation of the cytosolic Ca²⁺ buffer parvalbumin and mitochondrial volume in muscle cells via SIRT1/PGC-1α axis. PLoS One 2012; 7:e44837. [PMID: 23028640 PMCID: PMC3441610 DOI: 10.1371/journal.pone.0044837] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/09/2012] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscles show a high plasticity to cope with various physiological demands. Different muscle types can be distinguished by the force, endurance, contraction/relaxation kinetics (fast-twitch vs. slow-twitch muscles), oxidative/glycolytic capacity, and also with respect to Ca²⁺-signaling components. Changes in Ca²⁺ signaling and associated Ca²⁺-dependent processes are thought to underlie the high adaptive capacity of muscle fibers. Here we investigated the consequences and the involved mechanisms caused by the ectopic expression of the Ca²⁺-binding protein parvalbumin (PV) in C2C12 myotubes in vitro, and conversely, the effects caused by its absence in in fast-twitch muscles of parvalbumin null-mutant (PV⁻/⁻) mice in vivo. The absence of PV in fast-twitch muscle tibialis anterior (TA) resulted in an increase in the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and of its positive regulator, the deacetylase sirtuin 1 (SIRT1). TA muscles from PV⁻/⁻ mice also have an increased mitochondrial volume. Mild ionophore treatment of control (PV-devoid) C2C12 myotubes causing a moderate elevation in [Ca²⁺](c) resulted in an increase in mitochondrial volume, together with elevated PGC-1α and SIRT1 expression levels, whilst it increased PV expression levels in myotubes stably transfected with PV. In PV-expressing myotubes the mitochondrial volume, PGC-1α and SIRT1 were significantly lower than in control C2C12 myotubes already at basal conditions and application of ionophore had no effect on either one. SIRT1 activation causes a down-regulation of PV in transfected myotubes, whilst SIRT1 inhibition has the opposite effect. We conclude that PV expression and mitochondrial volume in muscle cells are inversely regulated via a SIRT1/PGC-1α signaling axis.
Collapse
Affiliation(s)
- Sylvie Ducreux
- Unit of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Patrick Gregory
- Unit of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Beat Schwaller
- Unit of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
46
|
Viguerie N, Picard F, Hul G, Roussel B, Barbe P, Iacovoni JS, Valle C, Langin D, Saris WHM. Multiple effects of a short-term dexamethasone treatment in human skeletal muscle and adipose tissue. Physiol Genomics 2011; 44:141-51. [PMID: 22108209 DOI: 10.1152/physiolgenomics.00032.2011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Glucocorticoids are frequently prescribed drugs with important side-effects such as glucose intolerance and tissue remodeling. The goal was to explore the molecular basis of the response of skeletal muscle and adipose tissue during a short-term dexamethasone treatment to better understand the induction of side-effects of glucocorticoids on these metabolic tissues. Fifteen healthy male subjects were assigned to a 4-day treatment with dexamethasone at 4 mg/day. The primary outcome measures were changes in gene expression profiling of subcutaneous skeletal muscle and adipose tissue. Urinary cortisol, plasma, and metabolic biochemistry were also assessed. In both tissues the prominent observation was a response to stress and increased inflammatory responses. An upregulation of the serum amyloid A was detected in skeletal muscle, adipose tissue, and plasma, whereas circulating levels of C reactive protein, another acute phase protein, decreased along with a worsened insulin sensitivity index. As tissue-specific features, tissue remodeling was shown in skeletal muscle while the adipose tissue exhibited a decreased energy metabolism. Several limitations might be raised due to the small number of subjects investigated: a possible cross talk with the mineralocorticoid receptor, and a single time point may not identify regulations occurring during longitudinal treatment. In line with the known physiological effect of glucocorticoids the early modulation of stress response genes was observed. An unexpected feature was the upregulation of the inflammatory and immune pathways. The identification of novel impact on two glucocorticoid target tissues provides a molecular basis for the design of more specific glucocorticoids devoid of adverse effects.
Collapse
Affiliation(s)
- Nathalie Viguerie
- Inserm, UMR1048, Obesity Research Laboratory, Institut des Maladies Métaboliques et Cardiovasculaires, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Castillero E, Nieto-Bona MP, Fernández-Galaz C, Martín AI, López-Menduiña M, Granado M, Villanúa MA, López-Calderón A. Fenofibrate, a PPAR{alpha} agonist, decreases atrogenes and myostatin expression and improves arthritis-induced skeletal muscle atrophy. Am J Physiol Endocrinol Metab 2011; 300:E790-9. [PMID: 21304067 DOI: 10.1152/ajpendo.00590.2010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Arthritis is a chronic inflammatory illness that induces cachexia, which has a direct impact on morbidity and mortality. Fenofibrate, a selective PPARα activator prescribed to treat human dyslipidemia, has been reported to decrease inflammation in rheumatoid arthritis patients. The aim of this study was to elucidate whether fenofibrate is able to ameliorate skeletal muscle wasting in adjuvant-induced arthritis, an experimental model of rheumatoid arthritis. On day 4 after adjuvant injection, control and arthritic rats were treated with 300 mg/kg fenofibrate until day 15, when all rats were euthanized. Fenofibrate decreased external signs of arthritis and liver TNFα and blocked arthritis-induced decreased in PPARα expression in the gastrocnemius muscle. Arthritis decreased gastrocnemius weight, which results from a decrease in cross-section area and myofiber size, whereas fenofibrate administration to arthritic rats attenuated the decrease in both gastrocnemius weight and fast myofiber size. Fenofibrate treatment prevented arthritis-induced increase in atrogin-1 and MuRF1 expression in the gastrocnemius. Neither arthritis nor fenofibrate administration modify Akt-FoxO3 signaling. Myostatin expression was not modified by arthritis, but fenofibrate decreased myostatin expression in the gastrocnemius of arthritic rats. Arthritis increased muscle expression of MyoD, PCNA, and myogenin in the rats treated with vehicle but not in those treated with fenofibrate. The results indicate that, in experimental arthritis, fenofibrate decreases skeletal muscle atrophy through inhibition of the ubiquitin-proteasome system and myostatin.
Collapse
Affiliation(s)
- Estíbaliz Castillero
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense, Madrid, Spain 28040.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Goto T, Lee JY, Teraminami A, Kim YI, Hirai S, Uemura T, Inoue H, Takahashi N, Kawada T. Activation of peroxisome proliferator-activated receptor-alpha stimulates both differentiation and fatty acid oxidation in adipocytes. J Lipid Res 2011; 52:873-84. [PMID: 21324916 PMCID: PMC3073464 DOI: 10.1194/jlr.m011320] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Peroxisome proliferator-activated receptor-α (PPARα) is a dietary lipid sensor, whose activation results in hypolipidemic effects. In this study, we investigated whether PPARα activation affects energy metabolism in white adipose tissue (WAT). Activation of PPARα by its agonist (bezafibrate) markedly reduced adiposity in KK mice fed a high-fat diet. In 3T3-L1 adipocytes, addition of GW7647, a highly specific PPARα agonist, during adipocyte differentiation enhanced glycerol-3-phosphate dehydrogenase activity, insulin-stimulated glucose uptake, and adipogenic gene expression. However, triglyceride accumulation was not increased by PPARα activation. PPARα activation induced expression of target genes involved in FA oxidation and stimulated FA oxidation. In WAT of KK mice treated with bezafibrate, both adipogenic and FA oxidation-related genes were significantly upregulated. These changes in mRNA expression were not observed in PPARα-deficient mice. Bezafibrate treatment enhanced FA oxidation in isolated adipocytes, suppressing adipocyte hypertrophy. Chromatin immunoprecipitation (ChIP) assay revealed that PPARα was recruited to promoter regions of both adipogenic and FA oxidation-related genes in the presence of GW7647 in 3T3-L1 adipocytes. These findings indicate that the activation of PPARα affects energy metabolism in adipocytes, and PPARα activation in WAT may contribute to the clinical effects of fibrate drugs.
Collapse
Affiliation(s)
- Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Aguilar V, Annicotte JS, Escote X, Vendrell J, Langin D, Fajas L. Cyclin G2 regulates adipogenesis through PPAR gamma coactivation. Endocrinology 2010; 151:5247-54. [PMID: 20844002 PMCID: PMC3000854 DOI: 10.1210/en.2010-0461] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cell cycle regulators such as cyclins, cyclin-dependent kinases, or retinoblastoma protein play important roles in the differentiation of adipocytes. In the present paper, we investigated the role of cyclin G2 as a positive regulator of adipogenesis. Cyclin G2 is an unconventional cyclin which expression is up-regulated during growth inhibition or apoptosis. Using the 3T3-F442A cell line, we observed an up-regulation of cyclin G2 expression at protein and mRNA levels throughout the process of cell differentiation, with a further induction of adipogenesis when the protein is transiently overexpressed. We show here that the positive regulatory effects of cyclin G2 in adipocyte differentiation are mediated by direct binding of cyclin G2 to peroxisome proliferator-activated receptor γ (PPARγ), the key regulator of adipocyte differentiation. The role of cyclin G2 as a novel PPARγ coactivator was further demonstrated by chromatin immunoprecipitation assays, which showed that the protein is present in the PPARγ-responsive element of the promoter of aP2, which is a PPARγ target gene. Luciferase reporter gene assays, showed that cyclin G2 positively regulates the transcriptional activity of PPARγ. The role of cyclin G2 in adipogenesis is further underscored by its increased expression in mice fed a high-fat diet. Taken together, our results demonstrate a novel role for cyclin G2 in the regulation of adipogenesis.
Collapse
Affiliation(s)
- Victor Aguilar
- IRCM, Institut de recherche en cancérologie de Montpellier
INSERM : U896Université Montpellier ICRLC Val d'Aurelle - Paul Lamarque F-34298 Montpellier,FR
| | - Jean-Sébastien Annicotte
- IRCM, Institut de recherche en cancérologie de Montpellier
INSERM : U896Université Montpellier ICRLC Val d'Aurelle - Paul Lamarque F-34298 Montpellier,FR
| | - Xavier Escote
- Endocrinology and Diabetes Unit
University Hospital of TarragonaResearch Department. Pere Virgili Institute, 43007 Tarragona,,ES
| | - Joan Vendrell
- Endocrinology and Diabetes Unit
University Hospital of TarragonaResearch Department. Pere Virgili Institute, 43007 Tarragona,,ES
| | - Dominique Langin
- I2MR, Institut de médecine moléculaire de Rangueil
INSERM : U858IFR31IFR150Université Paul Sabatier - Toulouse IIIInstitut Louis Bugnard 1, avenue Jean Poulhes BP 84225 31432 TOULOUSE CEDEX 4,FR
| | - Lluis Fajas
- IRCM, Institut de recherche en cancérologie de Montpellier
INSERM : U896Université Montpellier ICRLC Val d'Aurelle - Paul Lamarque F-34298 Montpellier,FR
- * Correspondence should be adressed to: Lluis Fajas
| |
Collapse
|
50
|
Kelly AK, Waters SM, McGee M, Fonseca RG, Carberry C, Kenny DA. mRNA expression of genes regulating oxidative phosphorylation in the muscle of beef cattle divergently ranked on residual feed intake. Physiol Genomics 2010; 43:12-23. [PMID: 20923863 DOI: 10.1152/physiolgenomics.00213.2009] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Our objective was to evaluate the effects of phenotypic ranking on residual feed intake (RFI) on the transcription of genes 1) involved in the respiratory chain complex and 2) coding for transcriptional factors regulating mitochondrial biogenesis, across two contrasting diet types. Beef heifers (n = 86) fed a diet comprising 70:30 concentrate-corn silage [low forage (LF)] over a 82-day period were ranked on RFI. The 10 highest (feed inefficient, high-RFI) and 10 lowest (feed efficient, low-RFI) ranking animals were selected for the current study. Biopsies of the M. longissimus dorsi were harvested following initial selection (LF diet) and again following a 6 wk period while the animals were offered a high-forage (HF) grass silage-only diet. Real-time PCR was used to quantify mRNA transcripts of 17 genes associated with cellular energetic efficiency. The mRNA expression of UCP3 tended to be upregulated (2.2-fold, P = 0.06) for the high-RFI compared with the low-RFI animals. mRNA transcripts coding for the transcription factor PGC-1α was 1.7-fold higher (P = 0.01) in low compared with high-RFI animals. A phenotype × diet interaction was evident for the abundance of ANT1 mRNA transcript, with greater (P = 0.04) expression levels detected in the low-RFI phenotype during the HF period, but no difference (P = 0.50) between phenotypes during the LF period. A phenotype × diet interaction was also evident for COX II with greater expression levels detected (P = 0.04) in the low compared with the high RFI phenotype while on LF but not the HF diet (P = 0.22). These data suggest an association between cellular energetic efficiency and RFI in cattle.
Collapse
Affiliation(s)
- Alan K Kelly
- UCD School of Agriculture, Food Science and Veterinary Medicine, College of Life Sciences, University College Dublin, Belfield, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|