1
|
Güzel Y, Atakan MM, Turnagöl HH, Koşar ŞN. Effects of 10 weeks of walking-based exercise training on resting substrate oxidation in postmenopausal women with obesity. Eur J Clin Nutr 2025; 79:311-319. [PMID: 39578536 DOI: 10.1038/s41430-024-01546-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND AND AIMS Accumulating evidence supports the effectiveness of moderate-intensity aerobic training on metabolic health, with limited studies investigating change in resting substrate oxidation. The aim of this study was to explore whether 10 weeks of walking-based aerobic training would alter substrate oxidation in postmenopausal women with obesity. METHODS AND RESULTS Twenty-four postmenopausal women with obesity who were assigned into the control (n = 12) or exercise groups (n = 12) undertook a 10-week aerobic training program (3 d·week-1) that involved walking exercises at 50-70% of heart rate reserve on a treadmill, with exercise volume increased from 25 to 40 min·day-1. Resting metabolic rate (RMR) and body composition were measured pre- and post-training. Whole-body substrate oxidation was calculated using respiratory data collected during RMR measurement via indirect calorimetry. No significant change was noted (p > 0.05) in resting fat oxidation and carbohydrate oxidation in the exercise group. Resting respiratory exchange ratio and RMR did not alter in response to the training program (p > 0.05). CONCLUSION Our results show that a 10-week of moderate-intensity aerobic training does not modify substrate oxidation in postmenopausal women with obesity.
Collapse
Affiliation(s)
- Yasemin Güzel
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye.
| | - Muhammed Mustafa Atakan
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| | - Hüseyin Hüsrev Turnagöl
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| | - Şükran Nazan Koşar
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
2
|
Vieira-Lara MA, Bakker BM. The paradox of fatty-acid β-oxidation in muscle insulin resistance: Metabolic control and muscle heterogeneity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167172. [PMID: 38631409 DOI: 10.1016/j.bbadis.2024.167172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
The skeletal muscle is a metabolically heterogeneous tissue that plays a key role in maintaining whole-body glucose homeostasis. It is well known that muscle insulin resistance (IR) precedes the development of type 2 diabetes. There is a consensus that the accumulation of specific lipid species in the tissue can drive IR. However, the role of the mitochondrial fatty-acid β-oxidation in IR and, consequently, in the control of glucose uptake remains paradoxical: interventions that either inhibit or activate fatty-acid β-oxidation have been shown to prevent IR. We here discuss the current theories and evidence for the interplay between β-oxidation and glucose uptake in IR. To address the underlying intricacies, we (1) dive into the control of glucose uptake fluxes into muscle tissues using the framework of Metabolic Control Analysis, and (2) disentangle concepts of flux and catalytic capacities taking into account skeletal muscle heterogeneity. Finally, we speculate about hitherto unexplored mechanisms that could bring contrasting evidence together. Elucidating how β-oxidation is connected to muscle IR and the underlying role of muscle heterogeneity enhances disease understanding and paves the way for new treatments for type 2 diabetes.
Collapse
Affiliation(s)
- Marcel A Vieira-Lara
- Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Barbara M Bakker
- Laboratory of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
3
|
Sausa M, Fucarino A, Paladino L, Zummo FP, Fabbrizio A, Di Felice V, Rappa F, Barone R, Marino Gammazza A, Macaluso F. Probiotics as Potential Therapeutic Agents: Safeguarding Skeletal Muscle against Alcohol-Induced Damage through the Gut-Liver-Muscle Axis. Biomedicines 2024; 12:382. [PMID: 38397983 PMCID: PMC10886686 DOI: 10.3390/biomedicines12020382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Probiotics have shown the potential to counteract the loss of muscle mass, reduce physical fatigue, and mitigate inflammatory response following intense exercise, although the mechanisms by which they work are not very clear. The objective of this review is to describe the main harmful effects of alcohol on skeletal muscle and to provide important strategies based on the use of probiotics. The excessive consumption of alcohol is a worldwide problem and has been shown to be crucial in the progression of alcoholic liver disease (ALD), for which, to date, the only therapy available is lifestyle modification, including cessation of drinking. In ALD, alcohol contributes significantly to the loss of skeletal muscle, and also to changes in the intestinal microbiota, which are the basis for a series of problems related to the onset of sarcopenia. Some of the main effects of alcohol on the skeletal muscle are described in this review, with particular emphasis on the "gut-liver-muscle axis", which seems to be the primary cause of a series of muscle dysfunctions related to the onset of ALD. The modulation of the intestinal microbiota through probiotics utilization has appeared to be crucial in mitigating the muscle damage induced by the high amounts of alcohol consumed.
Collapse
Affiliation(s)
- Martina Sausa
- Department of Theoretical and Applied Sciences, eCampus University, 22060 Novedrate, Italy; (M.S.); (A.F.); (A.F.)
| | - Alberto Fucarino
- Department of Theoretical and Applied Sciences, eCampus University, 22060 Novedrate, Italy; (M.S.); (A.F.); (A.F.)
| | - Letizia Paladino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.P.); (F.P.Z.); (V.D.F.); (F.R.); (R.B.); (A.M.G.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Francesco Paolo Zummo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.P.); (F.P.Z.); (V.D.F.); (F.R.); (R.B.); (A.M.G.)
| | - Antonio Fabbrizio
- Department of Theoretical and Applied Sciences, eCampus University, 22060 Novedrate, Italy; (M.S.); (A.F.); (A.F.)
| | - Valentina Di Felice
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.P.); (F.P.Z.); (V.D.F.); (F.R.); (R.B.); (A.M.G.)
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.P.); (F.P.Z.); (V.D.F.); (F.R.); (R.B.); (A.M.G.)
| | - Rosario Barone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.P.); (F.P.Z.); (V.D.F.); (F.R.); (R.B.); (A.M.G.)
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.P.); (F.P.Z.); (V.D.F.); (F.R.); (R.B.); (A.M.G.)
| | - Filippo Macaluso
- Department of Theoretical and Applied Sciences, eCampus University, 22060 Novedrate, Italy; (M.S.); (A.F.); (A.F.)
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (L.P.); (F.P.Z.); (V.D.F.); (F.R.); (R.B.); (A.M.G.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| |
Collapse
|
4
|
Diaz-Vegas A, Madsen S, Cooke KC, Carroll L, Khor JXY, Turner N, Lim XY, Astore MA, Morris JC, Don AS, Garfield A, Zarini S, Zemski Berry KA, Ryan AP, Bergman BC, Brozinick JT, James DE, Burchfield JG. Mitochondrial electron transport chain, ceramide, and coenzyme Q are linked in a pathway that drives insulin resistance in skeletal muscle. eLife 2023; 12:RP87340. [PMID: 38149844 PMCID: PMC10752590 DOI: 10.7554/elife.87340] [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] [Indexed: 12/28/2023] Open
Abstract
Insulin resistance (IR) is a complex metabolic disorder that underlies several human diseases, including type 2 diabetes and cardiovascular disease. Despite extensive research, the precise mechanisms underlying IR development remain poorly understood. Previously we showed that deficiency of coenzyme Q (CoQ) is necessary and sufficient for IR in adipocytes and skeletal muscle (Fazakerley et al., 2018). Here, we provide new insights into the mechanistic connections between cellular alterations associated with IR, including increased ceramides, CoQ deficiency, mitochondrial dysfunction, and oxidative stress. We demonstrate that elevated levels of ceramide in the mitochondria of skeletal muscle cells result in CoQ depletion and loss of mitochondrial respiratory chain components, leading to mitochondrial dysfunction and IR. Further, decreasing mitochondrial ceramide levels in vitro and in animal models (mice, C57BL/6J) (under chow and high-fat diet) increased CoQ levels and was protective against IR. CoQ supplementation also rescued ceramide-associated IR. Examination of the mitochondrial proteome from human muscle biopsies revealed a strong correlation between the respirasome system and mitochondrial ceramide as key determinants of insulin sensitivity. Our findings highlight the mitochondrial ceramide-CoQ-respiratory chain nexus as a potential foundation of an IR pathway that may also play a critical role in other conditions associated with ceramide accumulation and mitochondrial dysfunction, such as heart failure, cancer, and aging. These insights may have important clinical implications for the development of novel therapeutic strategies for the treatment of IR and related metabolic disorders.
Collapse
Affiliation(s)
- Alexis Diaz-Vegas
- Charles Perkins Centre, School of life and Environmental Sciences, University of SydneySydneyAustralia
| | - Søren Madsen
- Charles Perkins Centre, School of life and Environmental Sciences, University of SydneySydneyAustralia
| | - Kristen C Cooke
- Charles Perkins Centre, School of life and Environmental Sciences, University of SydneySydneyAustralia
| | - Luke Carroll
- Charles Perkins Centre, School of life and Environmental Sciences, University of SydneySydneyAustralia
| | - Jasmine XY Khor
- Charles Perkins Centre, School of life and Environmental Sciences, University of SydneySydneyAustralia
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of SydneySydneyAustralia
| | - Nigel Turner
- Cellular Bioenergetics Laboratory, Victor Chang Cardiac Research InstituteSydneyAustralia
| | - Xin Y Lim
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of SydneySydneyAustralia
| | - Miro A Astore
- Center for Computational Biology and Center for Computational Mathematics, Flatiron InstituteNew YorkUnited States
| | | | - Anthony S Don
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of SydneySydneyAustralia
| | - Amanda Garfield
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Simona Zarini
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Karin A Zemski Berry
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Andrew P Ryan
- Lilly Research Laboratories, Division of Eli Lilly and CompanyIndianapolisUnited States
| | - Bryan C Bergman
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Joseph T Brozinick
- Lilly Research Laboratories, Division of Eli Lilly and CompanyIndianapolisUnited States
| | - David E James
- Charles Perkins Centre, School of life and Environmental Sciences, University of SydneySydneyAustralia
- Charles Perkins Centre and School of Medical Sciences, Faculty of Medicine and Health, University of SydneySydneyAustralia
| | - James G Burchfield
- Charles Perkins Centre, School of life and Environmental Sciences, University of SydneySydneyAustralia
| |
Collapse
|
5
|
Soler-Vázquez MC, Romero MDM, Todorcevic M, Delgado K, Calatayud C, Benitez-Amaro A, La Chica Lhoëst MT, Mera P, Zagmutt S, Bastías-Pérez M, Ibeas K, Casals N, Escolà-Gil JC, Llorente-Cortés V, Consiglio A, Serra D, Herrero L. Implantation of CPT1AM-expressing adipocytes reduces obesity and glucose intolerance in mice. Metab Eng 2023; 77:256-272. [PMID: 37088334 DOI: 10.1016/j.ymben.2023.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/14/2023] [Accepted: 04/16/2023] [Indexed: 04/25/2023]
Abstract
Obesity and its associated metabolic comorbidities are a rising global health and social issue, with novel therapeutic approaches urgently needed. Adipose tissue plays a key role in the regulation of energy balance and adipose tissue-derived mesenchymal stem cells (AT-MSCs) have gained great interest in cell therapy. Carnitine palmitoyltransferase 1A (CPT1A) is the gatekeeper enzyme for mitochondrial fatty acid oxidation. Here, we aimed to generate adipocytes expressing a constitutively active CPT1A form (CPT1AM) that can improve the obese phenotype in mice after their implantation. AT-MSCs were differentiated into mature adipocytes, subjected to lentivirus-mediated expression of CPT1AM or the GFP control, and subcutaneously implanted into mice fed a high-fat diet (HFD). CPT1AM-implanted mice showed lower body weight, hepatic steatosis and serum insulin and cholesterol levels alongside improved glucose tolerance. HFD-induced increases in adipose tissue hypertrophy, fibrosis, inflammation, endoplasmic reticulum stress and apoptosis were reduced in CPT1AM-implanted mice. In addition, the expression of mitochondrial respiratory chain complexes was enhanced in the adipose tissue of CPT1AM-implanted mice. Our results demonstrate that implantation of CPT1AM-expressing AT-MSC-derived adipocytes into HFD-fed mice improves the obese metabolic phenotype, supporting the future clinical use of this ex vivo gene therapy approach.
Collapse
Affiliation(s)
- M Carmen Soler-Vázquez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain
| | - María Del Mar Romero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Marijana Todorcevic
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain
| | - Katia Delgado
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain
| | - Carles Calatayud
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital- IDIBELL, E-08908, Hospitalet de Llobregat, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
| | - Aleyda Benitez-Amaro
- Lipids and Cardiovascular Pathology, Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), 08041, Barcelona, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041, Barcelona, Spain
| | - Maria Teresa La Chica Lhoëst
- Lipids and Cardiovascular Pathology, Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), 08041, Barcelona, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041, Barcelona, Spain; Universitat Autònoma de Barcelona, Spain
| | - Paula Mera
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Sebastián Zagmutt
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain
| | - Marianela Bastías-Pérez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain
| | - Kevin Ibeas
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Núria Casals
- Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain; Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), E-08195, Sant Cugat del Vallés, Barcelona, Spain
| | - Joan Carles Escolà-Gil
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029, Madrid, Spain
| | - Vicenta Llorente-Cortés
- Lipids and Cardiovascular Pathology, Institut d'Investigacions Biomèdiques de Barcelona (IIBB-CSIC), 08041, Barcelona, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041, Barcelona, Spain; CIBER of Cardiovascular (CIBERCV), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Antonella Consiglio
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital- IDIBELL, E-08908, Hospitalet de Llobregat, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain; Department of Molecular and Translational Medicine, University of Brescia, Piazza del Mercato, 15, 25121, Brescia, BS, Italy
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), E-08028, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain.
| |
Collapse
|
6
|
Den Hartogh DJ, Vlavcheski F, Tsiani E. Muscle Cell Insulin Resistance Is Attenuated by Rosmarinic Acid: Elucidating the Mechanisms Involved. Int J Mol Sci 2023; 24:ijms24065094. [PMID: 36982168 PMCID: PMC10049470 DOI: 10.3390/ijms24065094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/16/2023] [Accepted: 01/26/2023] [Indexed: 03/30/2023] Open
Abstract
Obesity and elevated blood free fatty acid (FFA) levels lead to impaired insulin action causing insulin resistance in skeletal muscle, and contributing to the development of type 2 diabetes mellitus (T2DM). Mechanistically, insulin resistance is associated with increased serine phosphorylation of the insulin receptor substrate (IRS) mediated by serine/threonine kinases including mTOR and p70S6K. Evidence demonstrated that activation of the energy sensor AMP-activated protein kinase (AMPK) may be an attractive target to counteract insulin resistance. We reported previously that rosemary extract (RE) and the RE polyphenol carnosic acid (CA) activated AMPK and counteracted the FFA-induced insulin resistance in muscle cells. The effect of rosmarinic acid (RA), another polyphenolic constituent of RE, on FFA-induced muscle insulin resistance has never been examined and is the focus of the current study. Muscle cell (L6) exposure to FFA palmitate resulted in increased serine phosphorylation of IRS-1 and reduced insulin-mediated (i) Akt activation, (ii) GLUT4 glucose transporter translocation, and (iii) glucose uptake. Notably, RA treatment abolished these effects, and restored the insulin-stimulated glucose uptake. Palmitate treatment increased the phosphorylation/activation of mTOR and p70S6K, kinases known to be involved in insulin resistance and RA significantly reduced these effects. RA increased the phosphorylation of AMPK, even in the presence of palmitate. Our data indicate that RA has the potential to counteract the palmitate-induced insulin resistance in muscle cells, and further studies are required to explore its antidiabetic properties.
Collapse
Affiliation(s)
- Danja J Den Hartogh
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Filip Vlavcheski
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
7
|
Luo Q, Das A, Oldoni F, Wu P, Wang J, Luo F, Fang Z. Role of ACSL5 in fatty acid metabolism. Heliyon 2023; 9:e13316. [PMID: 36816310 PMCID: PMC9932481 DOI: 10.1016/j.heliyon.2023.e13316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 12/07/2022] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Free fatty acids (FFAs) are essential energy sources for most body tissues. A fatty acid must be converted to fatty acyl-CoA to oxidize or be incorporated into new lipids. Acyl-CoA synthetase long-chain family member 5 (ACSL5) is localized in the endoplasmic reticulum and mitochondrial outer membrane, where it catalyzes the formation of fatty acyl-CoAs from long-chain fatty acids (C16-C20). Fatty acyl-CoAs are then used in lipid synthesis or β-oxidation mediated pathways. ACSL5 plays a pleiotropic role in lipid metabolism depending on substrate preferences, subcellular localization and tissue specificity. Here, we review the role of ACSL5 in fatty acid metabolism in multiple metabolic tissues, including the liver, small intestine, adipose tissue, and skeletal muscle. Given the increasing number of studies suggesting the role of ACSL5 in glucose and lipid metabolism, we also summarized the effects of ACSL5 on circulating lipids and insulin resistance.
Collapse
Affiliation(s)
- Qin Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Hunan, 410011, China
| | - Avash Das
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Federico Oldoni
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Panyun Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Hunan, 410011, China
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jiangang Wang
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Fei Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Hunan, 410011, China
| | - Zhenfei Fang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Hunan, 410011, China
| |
Collapse
|
8
|
Abascal-Saiz A, Duque-Alcorta M, Fioravantti V, Antolín E, Fuente-Luelmo E, Haro M, Ramos-Álvarez MP, Perdomo G, Bartha JL. The Relationship between Angiogenic Factors and Energy Metabolism in Preeclampsia. Nutrients 2022; 14:2172. [PMID: 35631313 PMCID: PMC9145768 DOI: 10.3390/nu14102172] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/15/2022] [Accepted: 05/19/2022] [Indexed: 11/18/2022] Open
Abstract
Antiangiogenic factors are currently used for the prediction of preeclampsia. The present study aimed to evaluate the relationship between antiangiogenic factors and lipid and carbohydrate metabolism in maternal plasma and placenta. We analyzed 56 pregnant women, 30 healthy and 26 with preeclampsia (including early and late onset). We compared antiangiogenic factors soluble Fms-like Tyrosine Kinase-1 (sfLt-1), placental growth factor (PlGF), and soluble endoglin (sEng)), lipid and carbohydrate metabolism in maternal plasma, and lipid metabolism in the placenta from assays of fatty acid oxidation, fatty acid esterification, and triglyceride levels in all groups. Antiangiogenic factors sFlt-1, sFlt-1/PlGF ratio, and sEng showed a positive correlation with triglyceride, free fatty acid, and C-peptide maternal serum levels. However, there was no relationship between angiogenic factors and placental lipid metabolism parameters. Free fatty acids were predictive of elevated sFlt-1 and sEng, while C-peptide was predictive of an elevated sFlt1/PlGF ratio. The findings in this study generate a model to predict elevated antiangiogenic factor values and the relationship between them with different products of lipid and carbohydrate metabolism in maternal serum and placenta in preeclampsia.
Collapse
Affiliation(s)
- Alejandra Abascal-Saiz
- Department of Obstetrics and Gynecology, Division of Maternal and Fetal Medicine, La Paz University Hospital, Paseo de la Castellana 261, 28046 Madrid, Spain; (A.A.-S.); (E.A.)
| | - Marta Duque-Alcorta
- Department of Clinical Chemistry, La Paz University Hospital, 28046 Madrid, Spain;
| | - Victoria Fioravantti
- Department of Pediatric Hematology and Oncology, Hospital Infantil Universitario Niño Jesus, 28009 Madrid, Spain;
| | - Eugenia Antolín
- Department of Obstetrics and Gynecology, Division of Maternal and Fetal Medicine, La Paz University Hospital, Paseo de la Castellana 261, 28046 Madrid, Spain; (A.A.-S.); (E.A.)
| | - Eva Fuente-Luelmo
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, 28668 Madrid, Spain; (E.F.-L.); (M.H.); (M.P.R.-Á.)
| | - María Haro
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, 28668 Madrid, Spain; (E.F.-L.); (M.H.); (M.P.R.-Á.)
| | - María P. Ramos-Álvarez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, 28668 Madrid, Spain; (E.F.-L.); (M.H.); (M.P.R.-Á.)
| | - Germán Perdomo
- Unidad de Excelencia Instituto de Biología y Genética Molecular, University of Valladolid-CSIC, 47003 Valladolid, Spain;
| | - José L. Bartha
- Department of Obstetrics and Gynecology, Division of Maternal and Fetal Medicine, La Paz University Hospital, Paseo de la Castellana 261, 28046 Madrid, Spain; (A.A.-S.); (E.A.)
| |
Collapse
|
9
|
Serine Palmitoyltransferase Gene Silencing Prevents Ceramide Accumulation and Insulin Resistance in Muscles in Mice Fed a High-Fat Diet. Cells 2022; 11:cells11071123. [PMID: 35406688 PMCID: PMC8997855 DOI: 10.3390/cells11071123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/15/2022] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
Skeletal muscles account for ~80% of insulin-stimulated glucose uptake and play a key role in lipid metabolism. Consumption of a high-fat diet (HFD) contributes to metabolic changes in muscles, including the development of insulin resistance. The studies carried out to date indicate that the accumulation of biologically active lipids, such as long-chain acyl-CoA, diacylglycerols and ceramides, play an important role in the development of insulin resistance in skeletal muscles. Unfortunately, it has not yet been clarified which of these lipid groups plays the dominant role in inducing these disorders. In order to explore this topic further, we locally silenced the gene encoding serine palmitoyltransferase (SPT) in the gastrocnemius muscle of animals with HFD-induced insulin resistance. This enzyme is primarily responsible for the first step of de novo ceramide biosynthesis. The obtained results confirm that the HFD induces the development of whole-body insulin resistance, which results in inhibition of the insulin pathway. This is associated with an increased level of biologically active lipids in the muscles. Our results also demonstrate that silencing the SPT gene with the shRNA plasmid reduces the accumulation of ceramides in gastrocnemius muscle, which, in turn, boosts the activity of the insulin signaling pathway. Furthermore, inhibition of ceramide synthesis does not significantly affect the content of other lipids, which suggests the leading role of ceramide in the lipid-related induction of skeletal muscle insulin resistance.
Collapse
|
10
|
Carnosic Acid Attenuates the Free Fatty Acid-Induced Insulin Resistance in Muscle Cells and Adipocytes. Cells 2022; 11:cells11010167. [PMID: 35011728 PMCID: PMC8750606 DOI: 10.3390/cells11010167] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/10/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
Elevated blood free fatty acids (FFAs), as seen in obesity, impair insulin action leading to insulin resistance and Type 2 diabetes mellitus. Several serine/threonine kinases including JNK, mTOR, and p70 S6K cause serine phosphorylation of the insulin receptor substrate (IRS) and have been implicated in insulin resistance. Activation of AMP-activated protein kinase (AMPK) increases glucose uptake, and in recent years, AMPK has been viewed as an important target to counteract insulin resistance. We reported previously that carnosic acid (CA) found in rosemary extract (RE) and RE increased glucose uptake and activated AMPK in muscle cells. In the present study, we examined the effects of CA on palmitate-induced insulin-resistant L6 myotubes and 3T3L1 adipocytes. Exposure of cells to palmitate reduced the insulin-stimulated glucose uptake, GLUT4 transporter levels on the plasma membrane, and Akt activation. Importantly, CA attenuated the deleterious effect of palmitate and restored the insulin-stimulated glucose uptake, the activation of Akt, and GLUT4 levels. Additionally, CA markedly attenuated the palmitate-induced phosphorylation/activation of JNK, mTOR, and p70S6K and activated AMPK. Our data indicate that CA has the potential to counteract the palmitate-induced muscle and fat cell insulin resistance.
Collapse
|
11
|
Lund J, Krapf SA, Sistek M, Bakke HG, Bartesaghi S, Peng XR, Rustan AC, Thoresen GH, Kase ET. SENP2 is vital for optimal insulin signaling and insulin-stimulated glycogen synthesis in human skeletal muscle cells. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100061. [PMID: 34909683 PMCID: PMC8663970 DOI: 10.1016/j.crphar.2021.100061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 11/17/2022] Open
Abstract
Sentrin-specific protease (SENP) 2 has been suggested as a possible novel drug target for the treatment of obesity and type 2 diabetes mellitus after observations of a palmitate-induced increase in SENP2 that lead to increased fatty acid oxidation and improved insulin sensitivity in skeletal muscle cells from mice. However, no precedent research has examined the role of SENP2 in human skeletal muscle cells. In the present work, we have investigated the impact of SENP2 on fatty acid and glucose metabolism as well as insulin sensitivity in human skeletal muscle using cultured primary human myotubes. Acute (4 h) oleic acid oxidation was reduced in SENP2-knockdown (SENP2-KD) cells compared to control cells, with no difference in uptake. After prelabeling (24 h) with oleic acid, total lipid content and incorporation into triacylglycerol was decreased, while incorporation into other lipids, as well as complete oxidation and β-oxidation was increased in SENP2-KD cells. Basal glucose uptake (i.e., not under insulin-stimulated conditions) was higher in SENP2-KD cells, whereas oxidation was similar to control myotubes. Further, basal glycogen synthesis was not different in SENP2-KD myotubes, but both insulin-stimulated glycogen synthesis and AktSer473 phosphorylation was completely blunted in SENP2-KD cells. In conclusion, SENP2 plays an important role in fatty acid and glucose metabolism in human myotubes. Interestingly, it also appears to have a pivotal role in regulating myotube insulin sensitivity. Future studies should examine the role of SENP2 in regulation of insulin sensitivity in other tissues and in vivo, defining the potential for SENP2 as a drug target.
Collapse
Affiliation(s)
- Jenny Lund
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
- Corresponding author. Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway.
| | - Solveig A. Krapf
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Medina Sistek
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Hege G. Bakke
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Stefano Bartesaghi
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM) BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM) BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Arild C. Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - G. Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Norway
| | - Eili T. Kase
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| |
Collapse
|
12
|
Hou X, Wang L, Zhao F, Liu X, Gao H, Shi L, Yan H, Wang L, Zhang L. Genome-Wide Expression Profiling of mRNAs, lncRNAs and circRNAs in Skeletal Muscle of Two Different Pig Breeds. Animals (Basel) 2021; 11:ani11113169. [PMID: 34827901 PMCID: PMC8614396 DOI: 10.3390/ani11113169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Variation exists in muscle-related traits, such as muscle growth and meat quality, between obese and lean pigs. In this study, the transcriptome profiles of skeletal muscle between Beijing Blackand Yorkshire pigs were characterized to explore the molecular mechanism underlying skeletal muscle-relatedtraits. Gene Ontology (GO) and KEGG pathway enrichment analyses showed that differentially expressed mRNAs, lncRNAs, and circRNAs involved in skeletal muscle development and fatty acid metabolism played a key role in the determination of muscle-related traits between different pig breeds. These results provide candidate genes responsible for muscle phenotypic variation and are valuable for pig breeding. Abstract RNA-Seq technology is widely used to analyze global changes in the transcriptome and investigate the influence on relevant phenotypic traits. Beijing Black pigs show differences in growth rate and meat quality compared to western pig breeds. However, the molecular mechanisms responsible for such phenotypic differences remain unknown. In this study, longissimus dorsi muscles from Beijing Black and Yorkshire pigs were used to construct RNA libraries and perform RNA-seq. Significantly different expressions were observed in 1051 mRNAs, 322 lncRNAs, and 82 circRNAs. GO and KEGG pathway annotation showed that differentially expressed mRNAs participated in skeletal muscle development and fatty acid metabolism, which determined the muscle-related traits. To explore the regulatory role of lncRNAs, the cis and trans-target genes were predicted and these lncRNAswere involved in the biological processes related to skeletal muscle development and fatty acid metabolismvia their target genes. CircRNAs play a ceRNA role by binding to miRNAs. Therefore, the potential miRNAs of differentially expressed circRNAs were predicted and interaction networks among circRNAs, miRNAs, and key regulatory mRNAs were constructed to illustrate the function of circRNAs underlying skeletal muscle development and fatty acid metabolism. This study provides new clues for elucidating muscle phenotypic variation in pigs.
Collapse
|
13
|
Cui W, Sathyanarayan A, Lopresti M, Aghajan M, Chen C, Mashek DG. Lipophagy-derived fatty acids undergo extracellular efflux via lysosomal exocytosis. Autophagy 2021; 17:690-705. [PMID: 32070194 PMCID: PMC8032247 DOI: 10.1080/15548627.2020.1728097] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 01/31/2020] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
The autophagic degradation of lipid droplets (LDs), termed lipophagy, is a major mechanism that contributes to lipid turnover in numerous cell types. While numerous factors, including nutrient deprivation or overexpression of PNPLA2/ATGL (patatin-like phospholipase domain containing 2) drive lipophagy, the trafficking of fatty acids (FAs) produced from this pathway is largely unknown. Herein, we show that PNPLA2 and nutrient deprivation promoted the extracellular efflux of FAs. Inhibition of autophagy or lysosomal lipid degradation attenuated FA efflux highlighting a critical role for lipophagy in this process. Rather than direct transport of FAs across the lysosomal membrane, lipophagy-derived FA efflux requires lysosomal fusion to the plasma membrane. The lysosomal Ca2+ channel protein MCOLN1/TRPML1 (mucolipin 1) regulates lysosomal-plasma membrane fusion and its overexpression increased, while inhibition blocked FA efflux. In addition, inhibition of autophagy/lipophagy or MCOLN1, or sequestration of extracellular FAs with BSA attenuated the oxidation and re-esterification of lipophagy-derived FAs. Overall, these studies show that the well-established pathway of lysosomal fusion to the plasma membrane is the primary route for the disposal of FAs derived from lipophagy. Moreover, the efflux of FAs and their reuptake or subsequent extracellular trafficking to adjacent cells may play an important role in cell-to-cell lipid exchange and signaling.Abbreviations: ACTB: beta actin; ADRA1A: adrenergic receptor alpha, 1a; ALB: albumin; ATG5: autophagy related 5; ATG7: autophagy related 7; BafA1: bafilomycin A1; BECN1: beclin 1; BHBA: beta-hydroxybutyrate; BSA: bovine serum albumin; CDH1: e-cadherin; CQ: chloroquine; CTSB: cathepsin B; DGAT: diacylglycerol O-acyltransferase; FA: fatty acid; HFD: high-fat diet; LAMP1: lysosomal-associated membrane protein 1; LD: lipid droplet; LIPA/LAL: lysosomal acid lipase A; LLME: Leu-Leu methyl ester hydrobromide; MAP1LC3B/LC3: microtubule associated protein 1 light chain 3 beta; MCOLN1/TRPML1: mucolipin 1; MEF: mouse embryo fibroblast; PBS: phosphate-buffered saline; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PLIN: perilipin; PNPLA2/ATGL patatin-like phospholipase domain containing 2; RUBCN (rubicon autophagy regulator); SM: sphingomyelin; TAG: triacylglycerol; TMEM192: transmembrane protein 192; VLDL: very low density lipoprotein.
Collapse
Affiliation(s)
- Wenqi Cui
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Aishwarya Sathyanarayan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Michael Lopresti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | | | - Chi Chen
- Department of Food Science and Nutrition, University of Minnesota, Minneapolis, MN, USA
| | - Douglas G. Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
14
|
Dang TTH, Yun JW. BMP10 positively regulates myogenic differentiation in C2C12 myoblasts via the Smad 1/5/8 signaling pathway. Mol Cell Biochem 2021; 476:2085-2097. [PMID: 33517521 DOI: 10.1007/s11010-021-04064-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Abstract
BMP10 plays an essential role in regulating cardiac growth, chamber maturation, and maintaining normal expressions of several key cardiogenic factors; however, other functional roles of BMP10 in muscle remain unexplored. This study therefore undertook to investigate the roles of BMP10 in muscle physiology, using mouse-derived C2C12 myoblasts. Bmp10 silencing prevented a number of biological processes such as myogenic differentiation, glucose uptake, and lipid catabolism, whereas exogenous induction of BMP10 in C2C12 cells significantly stimulated the expression of proteins and genes involved in these processes, as well as mitochondrial biogenesis and thermogenesis, resulting in reduced lipid accumulation. A mechanistic study revealed that BMP10 stimulates myogenesis mainly via the Smad 1/5/8 signaling pathway. In conclusion, our data unveiled a previously unknown mechanism in the regulation of lipid metabolisms by BMP10 in muscle cells and identified its significant roles in systemic metabolic homeostasis, shedding light on BMP10 as a pharmacotherapeutic target to treat metabolic disorders.
Collapse
Affiliation(s)
- Trang Thi Huyen Dang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
15
|
Katsnelson G, Ceddia RB. Docosahexaenoic and eicosapentaenoic fatty acids differentially regulate glucose and fatty acid metabolism in L6 rat skeletal muscle cells. Am J Physiol Cell Physiol 2020; 319:C1120-C1129. [PMID: 32966124 DOI: 10.1152/ajpcell.00304.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The objective of this study was to investigate whether the n-3 polyunsaturated fatty acids (PUFAs) docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) can directly regulate glucose and fat metabolism in skeletal muscle besides exerting anti-inflammatory effects. To accomplish this, L6 skeletal muscle cells were treated with 50 µM of either DHA or EPA for 1, 3, and 5 days. Here, we report that basal and insulin-stimulated rates of glucose uptake, glycogen synthesis, protein kinase B (AKT), and glycogen synthase kinase 3 (GSK3) phosphorylation were not affected by DHA or EPA. However, glucose and palmitate oxidation were consistently elevated by DHA treatment, whereas EPA only increased this variable transiently. Similarly, only DHA caused significant and sustained increases in AMP-activated protein kinase (AMPK) phosphorylation and protein levels of carnitine-palmitoyl transferase-1b (CPT1b) and peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) in skeletal muscle cells. DHA also caused a larger anti-inflammatory effect than EPA in these cells. In conclusion, besides exerting anti-inflammatory effects, DHA and EPA directly regulated glucose and fat metabolism in skeletal muscle cells, although DHA was more effective in doing so than EPA. Thus, by directly enhancing glucose and fat oxidation, DHA may increase glucose disposal and reduce intramyocellular lipid accumulation.
Collapse
Affiliation(s)
- Glen Katsnelson
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Rolando B Ceddia
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Abascal-Saiz A, Fuente-Luelmo E, Haro M, de la Calle M, Ramos-Álvarez MP, Perdomo G, Bartha JL. Placental Compartmentalization of Lipid Metabolism: Implications for Singleton and Twin Pregnancies. Reprod Sci 2020; 28:1150-1160. [PMID: 33171514 DOI: 10.1007/s43032-020-00385-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/01/2020] [Indexed: 11/27/2022]
Abstract
The study of placental lipid metabolism in uncomplicated pregnancies has not been developed in the literature to date. Its importance lies in expanding the knowledge of placental function to enable comparison with pathological pregnancies in future research. The aim of the present study was to compare the lipid metabolic activity and storage of the maternal and fetal sides of the placenta in healthy pregnancies. Moreover, we compare singleton vs. twin pregnancies to determine if placental metabolic needs differ. We analyzed placental explants from uncomplicated pregnancies, 20 from singleton and 8 from bichorial-biamniotic twin pregnancies (n = 28). Six cotyledon fragments were collected from each placenta at different distances from the umbilical cord, three close to the chorionic plate (hereinafter, we will refer to them as "fetal side") and another three close to the anchoring villi into the decidua basalis (referred to as "maternal side"). The samples were analyzed for quantitative assay placental fatty acid oxidation (FAO) and esterification (FAE) activities and triglyceride levels. The location of lipid storage in the chorionic villi was assessed by Oil red-O staining. Placental fatty acid oxidation did not show differences when comparing the maternal and fetal sides of the placenta or between single and twin pregnancies. When comparing placental sides, FAE was increased twofold in the maternal side compared to the fetal side of the placenta (P = 0.013). The tendency for lipogenesis in the placenta was exemplified by the FAE/FAO ratio, which was a 37.1% higher on the maternal side (P = 0.019). Despite this, triglyceride levels were five times higher in the fetal side than in the maternal one (P = 0.024). When analyzing singleton vs. twins, FAE was superior in the fetal side in multiple pregnancies (× 2.6, P = 0.007) and the FAE/FAO ratio was significantly higher in twins than in singleton pregnancies, on both sides of the placenta. Despite this finding, triglyceride levels were similar in twin and singleton pregnancies. Comparing the placentas of twins in the same pregnancy, there were no differences in lipid metabolism (FAO or FAE) or placental triglyceride levels between the two co-twins. Using Oil red-O staining, lipid storage in chorionic villi was found to be located on the syncytiotrophoblast cells and not in the connecting axis. The maternal side of the placenta is more active in the esterification of fatty acids, while the storage of neutral lipids concentrates on the fetal side. Moreover, multiple gestations have increased esterification without changes in the concentration of placental triglycerides, probably due to a higher transfer to the fetal circulation in response to the greater energy demand from twin fetuses.
Collapse
Affiliation(s)
- Alejandra Abascal-Saiz
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Eva Fuente-Luelmo
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - María Haro
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - María de la Calle
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - María P Ramos-Álvarez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, CEU-San Pablo University, Madrid, Spain
| | - Germán Perdomo
- Department of Health Sciences, University of Burgos, Burgos, Spain.,Institute of Molecular Biology and Genetic (IMBG), CSIC - University of Valladolid, Valladolid, Spain
| | - José L Bartha
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain.
| |
Collapse
|
17
|
Halama A, Suleiman NN, Kulinski M, Bettahi I, Hassoun S, Alkasem M, Abdalhakam I, Iskandarani A, Samra TA, Atkin SL, Suhre K, Abou-Samra AB. The metabolic footprint of compromised insulin sensitivity under fasting and hyperinsulinemic-euglycemic clamp conditions in an Arab population. Sci Rep 2020; 10:17164. [PMID: 33051490 PMCID: PMC7555540 DOI: 10.1038/s41598-020-73723-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/15/2020] [Indexed: 11/29/2022] Open
Abstract
Metabolic pathways that are corrupted at early stages of insulin resistance (IR) remain elusive. This study investigates changes in body metabolism in clinically healthy and otherwise asymptomatic subjects that may become apparent already under compromised insulin sensitivity (IS) and prior to IR. 47 clinically healthy Arab male subjects with a broad range of IS, determined by hyperinsulinemic-euglycemic clamp (HIEC), were investigated. Untargeted metabolomics and complex lipidomics were conducted on serum samples collected under fasting and HIEC conditions. Linear models were used to identify associations between metabolites concentrations and IS levels. Among 1896 identified metabolites, 551 showed significant differences between fasting and HIEC, reflecting the metabolic switch in energy utilization. At fasting, 336 metabolites, predominantly di- and tri-acylglycerols, showed significant differences between subjects with low and high levels of IS. Changes in amino acid, carbohydrate and fatty acid metabolism in response to insulin were impaired in subjects with low IS. Association of altered mannose and amino acids with IS was also replicated in an independent cohort of T2D patients. We identified metabolic phenotypes that characterize clinically healthy Arab subjects with low levels of IS at their fasting state. Our study is providing further insights into the metabolic pathways that precede IR.
Collapse
Affiliation(s)
- Anna Halama
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar.
| | - Noor N Suleiman
- Department of Internal Medicine, Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar
| | - Michal Kulinski
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ilham Bettahi
- Department of Internal Medicine, Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shaimaa Hassoun
- Department of Internal Medicine, Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar
| | - Meis Alkasem
- Department of Internal Medicine, Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ibrahem Abdalhakam
- Department of Internal Medicine, Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar
| | - Ahmad Iskandarani
- Department of Internal Medicine, Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Tareq A Samra
- Department of Internal Medicine, Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Stephen L Atkin
- Weill Cornell Medicine-Qatar, Doha, Qatar.,Royal College of Surgeons in Ireland, Busaiteen, Bahrain
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar.
| | - Abdul Badi Abou-Samra
- Department of Internal Medicine, Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar. .,Weill Cornell Medicine-Qatar, Doha, Qatar.
| |
Collapse
|
18
|
Ferreira MDR, Oliva ME, Aiassa V, D'Alessandro ME. Salvia hispanica L. (chia) seed improves skeletal muscle lipotoxicity and insulin sensitivity in rats fed a sucrose-rich diet by modulating intramuscular lipid metabolism. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
19
|
Zhang W, Bai Y, Chen Z, Li X, Fu S, Huang L, Lin S, Du H. Comprehensive analysis of long non-coding RNAs and mRNAs in skeletal muscle of diabetic Goto-Kakizaki rats during the early stage of type 2 diabetes. PeerJ 2020; 8:e8548. [PMID: 32095365 PMCID: PMC7023842 DOI: 10.7717/peerj.8548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/12/2020] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle long non-coding RNAs (lncRNAs) were reported to be involved in the development of type 2 diabetes (T2D). However, little is known about the mechanism of skeletal muscle lncRNAs on hyperglycemia of diabetic Goto-Kakizaki (GK) rats at the age of 3 and 4 weeks. To elucidate this, we used RNA-sequencing to profile the skeletal muscle transcriptomes including lncRNAs and mRNAs, in diabetic GK and control Wistar rats at the age of 3 and 4 weeks. In total, there were 438 differentially expressed mRNAs (DEGs) and 401 differentially expressed lncRNAs (DELs) in skeletal muscle of 3-week-old GK rats compared with age-matched Wistar rats, and 1000 DEGs and 726 DELs between GK rats and Wistar rats at 4 weeks of age. The protein-protein interaction analysis of overlapping DEGs between 3 and 4 weeks, the correlation analysis of DELs and DEGs, as well as the prediction of target DEGs of DELs showed that these DEGs (Pdk4, Stc2, Il15, Fbxw7 and Ucp3) might play key roles in hyperglycemia, glucose intolerance, and increased fatty acid oxidation. Considering the corresponding co-expressed DELs with high correlation coefficients or targeted DELs of these DEGs, our study indicated that these dysregulated lncRNA-mRNA pairs (NONRATG017315.2-Pdk4, NONRATG003318.2-Stc2, NONRATG011882.2-Il15, NONRATG013497.2-Fbxw7, MSTRG.1662-Ucp3) might be related to above biological processes in GK rats at the age of 3 and 4 weeks. Our study could provide more comprehensive knowledge of mRNAs and lncRNAs in skeletal muscle of GK rats at 3 and 4 weeks of age. And our study may provide deeper understanding of the underlying mechanism in T2D of GK rats at the age of 3 and 4 weeks.
Collapse
Affiliation(s)
- Wenlu Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yunmeng Bai
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Zixi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Xingsong Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Shuying Fu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Lizhen Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Shudai Lin
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
20
|
Transcriptome Changes of Skeletal Muscle RNA-Seq Speculates the Mechanism of Postprandial Hyperglycemia in Diabetic Goto-Kakizaki Rats During the Early Stage of T2D. Genes (Basel) 2019; 10:genes10060406. [PMID: 31141985 PMCID: PMC6627578 DOI: 10.3390/genes10060406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022] Open
Abstract
To address how skeletal muscle contributes to postprandial hyperglycemia, we performed skeletal muscle transcriptome analysis of diabetic Goto-Kakizaki (GK) and control Wistar rats by RNA sequencing (RNA-Seq). We obtained 600 and 1785 differentially expressed genes in GK rats compared to those Wistar rats at three and four weeks of age, respectively. Specifically, Tbc1d4, involved in glucose uptake, was significantly downregulated in the skeletal muscle of GK aged both three and four weeks compared to those of age-matched Wistar rats. Pdk4, related to glucose uptake and oxidation, was significantly upregulated in the skeletal muscle of GK aged both three and four weeks compared to that of age-matched Wistar rats. Genes (Acadl, Acsl1 and Fabp4) implicated in fatty acid oxidation were significantly upregulated in the skeletal muscle of GK aged four weeks compared to those of age-matched Wistar rats. The overexpression or knockout of Tbc1d4, Pdk4, Acadl, Acsl1 and Fabp4 has been reported to change glucose uptake and fatty acid oxidation directly in rodents. By taking the results of previous studies into consideration, we speculated that dysregulation of key dysregulated genes (Tbc1d4, Pdk4, Acadl, Acsl1 and Fabp4) may lead to a decrease in glucose uptake and oxidation, and an increase in fatty acid oxidation in GK skeletal muscle at three and four weeks, which may, in turn, contribute to postprandial hyperglycemia. Our research revealed transcriptome changes in GK skeletal muscle at three and four weeks. Tbc1d4, Acadl, Acsl1 and Fabp4 were found to be associated with early diabetes in GK rats for the first time, which may provide a new scope for pathogenesis of postprandial hyperglycemia.
Collapse
|
21
|
Attenuation of Free Fatty Acid-Induced Muscle Insulin Resistance by Rosemary Extract. Nutrients 2018; 10:nu10111623. [PMID: 30400151 PMCID: PMC6267446 DOI: 10.3390/nu10111623] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 12/13/2022] Open
Abstract
Elevated blood free fatty acids (FFAs), as seen in obesity, impair muscle insulin action leading to insulin resistance and Type 2 diabetes mellitus. Serine phosphorylation of the insulin receptor substrate (IRS) is linked to insulin resistance and a number of serine/threonine kinases including JNK, mTOR and p70 S6K have been implicated in this process. Activation of the energy sensor AMP-activated protein kinase (AMPK) increases muscle glucose uptake, and in recent years AMPK has been viewed as an important target to counteract insulin resistance. We reported recently that rosemary extract (RE) increased muscle cell glucose uptake and activated AMPK. However, the effect of RE on FFA-induced muscle insulin resistance has never been examined. In the current study, we investigated the effect of RE in palmitate-induced insulin resistant L6 myotubes. Exposure of myotubes to palmitate reduced the insulin-stimulated glucose uptake, increased serine phosphorylation of IRS-1, and decreased the insulin-stimulated phosphorylation of Akt. Importantly, exposure to RE abolished these effects and the insulin-stimulated glucose uptake was restored. Treatment with palmitate increased the phosphorylation/activation of JNK, mTOR and p70 S6K whereas RE completely abolished these effects. RE increased the phosphorylation of AMPK even in the presence of palmitate. Our data indicate that rosemary extract has the potential to counteract the palmitate-induced muscle cell insulin resistance and further studies are required to explore its antidiabetic properties.
Collapse
|
22
|
Excess Hydrocortisone Hampers Placental Nutrient Uptake Disrupting Cellular Metabolism. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5106174. [PMID: 30402483 PMCID: PMC6198558 DOI: 10.1155/2018/5106174] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/31/2018] [Accepted: 09/20/2018] [Indexed: 11/17/2022]
Abstract
Low birth weight increases neonatal morbidity and mortality, and surviving infants have increased risk of metabolic and cardiovascular disturbances later in life, as well as other neurological, psychiatric, and immune complications. A gestational excess of glucocorticoids (GCs) is a well-known cause for fetal growth retardation, but the biological basis for this association remains elusive. Placental growth is closely related to fetal growth. The placenta is the main regulator of nutrient transport to the fetus, resulting from the difference between placental nutrient uptake and the placenta's own metabolism. The aim of this study was to analyze how excess hydrocortisone affects placental glucose and lipid metabolism. Human placenta explants from term physiological pregnancies were cultured for 18 hours under different hydrocortisone concentrations (2.75, 5.5, and 55 mM; 1, 2, and 20 mg/ml). Placental glucose and lipid uptake and the metabolic partitioning of fatty acids were quantified by isotopic techniques, and expression of specific glucose transporter GLUT1 was quantified by western blot. Cell viability was assessed by MTT, immunohistochemistry and caspase activity. We found that excess hydrocortisone impairs glucose uptake and lipoprotein lipase (LPL) activity, coincident with a GC-dose dependent inhibition of fatty acid oxidation and esterification. None of the experimental conditions showed an increased cell death. In conclusion, our results show that GC overexposure exerts a dysfunctional effect on lipid transport and metabolism and glucose uptake in human placental explants. These findings could well be directly related to a reduced placental growth and possibly to a reduced supply of nutrients to the fetus and the consequent fetal growth retardation and metabolic programming.
Collapse
|
23
|
Tardelli LP, Breda L, Marques LF, Gomes Carvalho Lima NC, Furtado de Camargo T, Scherer BR, Moreira NF, Dias JF, Dalia RA, Thomazini BF, Corezolla do Amaral ME, Alves AA. High lipid and low carbohydrate content diet, immediately after weaning, causes hepatic injury, systemic oxidative stress and diminishment of lipids in white adipose tissue. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
24
|
Bhatt BA, Dedousis N, Sipula IJ, O'Doherty RM. Elevated metabolic rate and skeletal muscle oxidative metabolism contribute to the reduced susceptibility of NF-κB p50 null mice to obesity. Physiol Rep 2018; 6:e13836. [PMID: 30251338 PMCID: PMC6153426 DOI: 10.14814/phy2.13836] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/03/2018] [Accepted: 07/04/2018] [Indexed: 01/01/2023] Open
Abstract
Mice with a deletion of the p50 subunit of the proinflammatory nuclear factor kappa B pathway (NF-κB p50) have reduced weight compared to wild-type control mice. However, the physiological underpinning of this phenotype remains unknown. This study addressed this issue. Compared to littermate controls, lean male p50 null mice (p50-/- ) had an increased metabolic rate (~20%) that was associated with increased skeletal muscle (SkM, ~35%), but not liver, oxidative metabolism. These metabolic alterations were accompanied by decreases in adiposity, and tissue and plasma triglyceride levels (all ~30%). Notably, there was a marked decrease in skeletal muscle, but not liver, DGAT2 gene expression (~70%), but a surprising reduction in muscle PPARα and CPT1 (both ~20%) gene expression. Exposure to a high-fat diet accentuated the diminished adiposity of p50-/- mice despite elevated caloric intake, whereas plasma triglycerides and free fatty acids (both ~30%), and liver (~40%) and SkM (~50%) triglyceride accumulation were again reduced compared to WT. Although SkM cytokine expression (IL-6 and TNFα, each ~100%) were increased in p50-/- mice, neither cytokine acutely increased SkM oxidative metabolism. We conclude that the reduced susceptibility to diet-induced obesity and dyslipidemia in p50-/- mice results from an increase in metabolic rate, which is associated with elevated skeletal muscle oxidative metabolism and decreased DGAT2 expression.
Collapse
Affiliation(s)
- Bankim A. Bhatt
- Department of MedicineDivision of Endocrinology and MetabolismUniversity of PittsburghPittsburghPennsylvania
| | - Nikolaos Dedousis
- Department of MedicineDivision of Endocrinology and MetabolismUniversity of PittsburghPittsburghPennsylvania
| | - Ian J. Sipula
- Department of MedicineDivision of Endocrinology and MetabolismUniversity of PittsburghPittsburghPennsylvania
| | - Robert M. O'Doherty
- Department of MedicineDivision of Endocrinology and MetabolismUniversity of PittsburghPittsburghPennsylvania
- Department of Microbiology and Molecular GeneticsUniversity of PittsburghPittsburghPennsylvania
| |
Collapse
|
25
|
Mora C, Pintado C, Rubio B, Mazuecos L, López V, Fernández A, Salamanca A, Bárcena B, Fernández-Agulló T, Arribas C, Gallardo N, Andrés A. Central leptin regulates heart lipid content by selectively increasing PPAR β/δ expression. J Endocrinol 2018; 236:43-56. [PMID: 29109080 DOI: 10.1530/joe-17-0554] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/06/2017] [Indexed: 01/03/2023]
Abstract
The role of central leptin in regulating the heart from lipid accumulation in lean leptin-sensitive animals has not been fully elucidated. Herein, we investigated the effects of central leptin infusion on the expression of genes involved in cardiac metabolism and its role in the control of myocardial triacylglyceride (TAG) accumulation in adult Wistar rats. Intracerebroventricular (icv) leptin infusion (0.2 µg/day) for 7 days markedly decreased TAG levels in cardiac tissue. Remarkably, the cardiac anti-steatotic effects of central leptin were associated with the selective upregulation of gene and protein expression of peroxisome proliferator-activated receptor β/δ (PPARβ/δ, encoded by Pparb/d) and their target genes, adipose triglyceride lipase (encoded by Pnpla2, herefater referred to as Atgl), hormone sensitive lipase (encoded by Lipe, herefater referred to as Hsl), pyruvate dehydrogenase kinase 4 (Pdk4) and acyl CoA oxidase 1 (Acox1), involved in myocardial intracellular lipolysis and mitochondrial/peroxisomal fatty acid utilization. Besides, central leptin decreased the expression of stearoyl-CoA deaturase 1 (Scd1) and diacylglycerol acyltransferase 1 (Dgat1) involved in TAG synthesis and increased the CPT-1 independent palmitate oxidation, as an index of peroxisomal β-oxidation. Finally, the pharmacological inhibition of PPARβ/δ decreased the effects on gene expression and cardiac TAG content induced by leptin. These results indicate that leptin, acting at central level, regulates selectively the cardiac expression of PPARβ/δ, contributing in this way to regulate the cardiac TAG accumulation in rats, independently of its effects on body weight.
Collapse
Affiliation(s)
- Cristina Mora
- BiochemistryFaculty of Science and Technological Chemistry, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), Ciudad Real, Spain
| | - Cristina Pintado
- BiochemistryFaculty of Environmental Sciences and and CRIB, UCLM, Toledo, Spain
| | - Blanca Rubio
- BiochemistryFaculty of Science and Technological Chemistry, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), Ciudad Real, Spain
| | - Lorena Mazuecos
- BiochemistryFaculty of Science and Technological Chemistry, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), Ciudad Real, Spain
| | - Virginia López
- BiochemistryFaculty of Science and Technological Chemistry, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), Ciudad Real, Spain
| | - Alejandro Fernández
- BiochemistryFaculty of Science and Technological Chemistry, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), Ciudad Real, Spain
| | - Aurora Salamanca
- BiochemistryFaculty of Science and Technological Chemistry, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), Ciudad Real, Spain
| | - Brenda Bárcena
- BiochemistryFaculty of Science and Technological Chemistry, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), Ciudad Real, Spain
| | | | - Carmen Arribas
- BiochemistryFaculty of Environmental Sciences and and CRIB, UCLM, Toledo, Spain
| | - Nilda Gallardo
- BiochemistryFaculty of Science and Technological Chemistry, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), Ciudad Real, Spain
| | - Antonio Andrés
- BiochemistryFaculty of Science and Technological Chemistry, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha (UCLM), Ciudad Real, Spain
| |
Collapse
|
26
|
Dębski B, Nowicki T, Zalewski W, Bartoszewicz A, Twardoń J. Effect of Pregnancy and Stage of Lactation on Energy Processes in Isolated Blood Cells of Dairy Cows. J Vet Res 2017; 61:211-215. [PMID: 29978075 PMCID: PMC5894394 DOI: 10.1515/jvetres-2017-0027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/10/2017] [Indexed: 11/15/2022] Open
Abstract
Introduction The transition period is the most challenging time for dairy cattle, which is characterised not only by negative energy balance but also by fatty tissue mobilisation. Material and Methods The efficiency of energy pathways, β-oxidation in WBC and glycolysis in RBC (based on deoxyglucose transmembrane transport) were estimated. Insulin in blood plasma was determined using ELISA. Results After calving and up to one month after delivery, a significant drop in blood plasma level was noticed, simultaneously with a rise in β-oxidation from 18.93 ±3.64 to 30.32 ±5.28 pmol/min/mg protein in WBC. A strong negative correlation between these two indices (r = -0.68) was found. During the period of transition to lactation an increase in glucose cross-membrane transportation from 41.44 ±4.92 to 50.49 ±6.41 μmol/h/g Hb was observed. A strong positive correlation between glucose transportation in RBC and β-oxidation in WBC (r = 0.71) was noticed. These data are in agreement with results of studies on dairy cows using liver slices from dairy cows in late pregnancy and different stages of lactation, in which changes in gene expression were analysed. Conclusion It seems that measuring fatty acids oxidation and glycolysis using isolated blood cells may be an adequate and relatively simple method for energy state analysis to estimate the state of dairy cow metabolism and animal health.
Collapse
Affiliation(s)
- Bogdan Dębski
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, 02-766 Warsaw, Poland
| | | | - Wojciech Zalewski
- Department of Reproduction and Clinic of Farm Animals, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Agnieszka Bartoszewicz
- Department of Reproduction and Clinic of Farm Animals, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Jan Twardoń
- Department of Reproduction and Clinic of Farm Animals, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| |
Collapse
|
27
|
Somatic growth, aging, and longevity. NPJ Aging Mech Dis 2017; 3:14. [PMID: 28970944 PMCID: PMC5622030 DOI: 10.1038/s41514-017-0014-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 02/01/2023] Open
Abstract
Although larger species of animals typically live longer than smaller species, the relationship of body size to longevity within a species is generally opposite. The longevity advantage of smaller individuals can be considerable and is best documented in laboratory mice and in domestic dogs. Importantly, it appears to apply broadly, including humans. It is not known whether theses associations represent causal links between various developmental and physiological mechanisms affecting growth and/or aging. However, variations in growth hormone (GH) signaling are likely involved because GH is a key stimulator of somatic growth, and apparently also exerts various “pro-aging” effects. Mechanisms linking GH, somatic growth, adult body size, aging, and lifespan likely involve target of rapamycin (TOR), particularly one of its signaling complexes, mTORC1, as well as various adjustments in mitochondrial function, energy metabolism, thermogenesis, inflammation, and insulin signaling. Somatic growth, aging, and longevity are also influenced by a variety of hormonal and nutritional signals, and much work will be needed to answer the question of why smaller individuals may be likely to live longer.
Collapse
|
28
|
Qiu F, Xie L, Ma JE, Luo W, Zhang L, Chao Z, Chen S, Nie Q, Lin Z, Zhang X. Lower Expression of SLC27A1 Enhances Intramuscular Fat Deposition in Chicken via Down-Regulated Fatty Acid Oxidation Mediated by CPT1A. Front Physiol 2017; 8:449. [PMID: 28706492 PMCID: PMC5489693 DOI: 10.3389/fphys.2017.00449] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/14/2017] [Indexed: 01/11/2023] Open
Abstract
Intramuscular fat (IMF) is recognized as the predominant factor affecting meat quality due to its positive correlation with tenderness, juiciness, and flavor. Chicken IMF deposition depends on the balance among lipid synthesis, transport, uptake, and subsequent metabolism, involving a lot of genes and pathways, however, its precise molecular mechanisms remain poorly understood. In the present study, the breast muscle tissue of female Wenchang chickens (WC) (higher IMF content, 1.24 in D120 and 1.62 in D180) and female White Recessive Rock chickens (WRR; lower IMF content, 0.53 in D120 and 0.90 in D180) were subjected to RNA-sequencing (RNA-seq) analysis. Results showed that many genes related to lipid catabolism, such as SLC27A1, LPL, ABCA1, and CPT1A were down-regulated in WC chickens, and these genes were involved in the PPAR signaling pathway and formed an IPA® network related to lipid metabolism. Furthermore, SLC27A1 was more down-regulated in WRR.D180.B than in WRR.D120.B. Decreased cellular triglyceride (TG) and up-regulated CPT1A were observed in the SLC27A1 overexpression QM-7 cells, and increased cellular triglyceride (TG) and down-regulated CPT1A were observed in the SLC27A1 knockdown QM-7 cells. These results suggest that lower lipid catabolism exists in WC chickens but not in WRR chickens, and lower expression of SLC27A1 facilitate IMF deposition in chicken via down-regulated fatty acid oxidation mediated by CPT1A. These findings indicate that reduced lipid catabolism, rather than increased lipid anabolism, contributes to chicken IMF deposition.
Collapse
Affiliation(s)
- Fengfang Qiu
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China.,School of Chemistry, Biology and Material Science, East China University of TechnologyNanchang, China
| | - Liang Xie
- Department of Poultry Science, Institute of Animal Science and Veterinary, Hainan Academy of Agricultural SciencesHaikou, China
| | - Jing-E Ma
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Wen Luo
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Li Zhang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Zhe Chao
- Department of Poultry Science, Institute of Animal Science and Veterinary, Hainan Academy of Agricultural SciencesHaikou, China
| | - Shaohao Chen
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Qinghua Nie
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| | - Zhemin Lin
- Department of Poultry Science, Institute of Animal Science and Veterinary, Hainan Academy of Agricultural SciencesHaikou, China
| | - Xiquan Zhang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural UniversityGuangzhou, China
| |
Collapse
|
29
|
Chen G, Li H, Zhao Y, Zhu H, Cai E, Gao Y, Liu S, Yang H, Zhang L. Saponins from stems and leaves of Panax ginseng prevent obesity via regulating thermogenesis, lipogenesis and lipolysis in high-fat diet-induced obese C57BL/6 mice. Food Chem Toxicol 2017; 106:393-403. [PMID: 28599882 DOI: 10.1016/j.fct.2017.06.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 06/02/2017] [Accepted: 06/05/2017] [Indexed: 02/06/2023]
Abstract
In this study, high-fat diet (HFD)-induced obesity in mouse model was used to evaluate the dietary effect of saponins from stems and leaves of Panax ginseng (SLG), and to explore its mechanism of action in producing anti-obesity effects. The results indicate that SLG showed significant anti-obesity effects in diet-induced obese mice, represented by decreased serum levels of free fatty acids (FFA), total cholesterol (TC), triglycerides (TG), low-density lipoprotein (LDL)-cholesterol, glucose, leptin and insulin, as well as a reduction in overall body and liver weight, epididymal adipose tissue weight, and food efficiency, and inhibition of abnormal increases in acyl carnitine levels normally caused by an HFD. Additionally, the down-regulated expression of PPARγ, FAS, CD36, FATP2 and up-regulated expression of CPT-1, UCP-2, PPARα, HSL, and ATGL in liver tissue was induced by SLG. In addition, the SLG groups showed decreased PPARγ, aP2 and leptin mRNA levels and increased expression of PPARα, PGC-1α, UCP-1 and UCP-3 genes in adipose tissues, compared with the HFD group. In short, SLG may play a key role in producing anti-obesity effects in mice fed an HFD, and its mechanism may be related to regulation of thermogenesis, lipogenesis and lipolysis.
Collapse
Affiliation(s)
- Guilin Chen
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - Haijun Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China.
| | - Hongyan Zhu
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - Enbo Cai
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - Yugang Gao
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China.
| | - Shuangli Liu
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - He Yang
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | - Lianxue Zhang
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| |
Collapse
|
30
|
Fucho R, Casals N, Serra D, Herrero L. Ceramides and mitochondrial fatty acid oxidation in obesity. FASEB J 2016; 31:1263-1272. [PMID: 28003342 DOI: 10.1096/fj.201601156r] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/06/2016] [Indexed: 12/12/2022]
Abstract
Obesity is an epidemic, complex disease that is characterized by increased glucose, lipids, and low-grade inflammation in the circulation, among other factors. It creates the perfect scenario for the production of ceramide, the building block of the sphingolipid family of lipids, which is involved in metabolic disorders such as obesity, diabetes, and cardiovascular disease. In addition, obesity causes a decrease in fatty acid oxidation (FAO), which contributes to lipid accumulation within the cells, conferring more susceptibility to cell dysfunction. C16:0 ceramide, a specific ceramide species, has been identified recently as the principal mediator of obesity-derived insulin resistance, impaired fatty acid oxidation, and hepatic steatosis. In this review, we have sought to cover the importance of the ceramide species and their metabolism, the main ceramide signaling pathways in obesity, and the link between C16:0 ceramide, FAO, and obesity.-Fucho, R., Casals, N., Serra, D., Herrero, L. Ceramides and mitochondrial fatty acid oxidation in obesity.
Collapse
Affiliation(s)
- Raquel Fucho
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | - Núria Casals
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain; and.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain; .,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina, Universitat de Barcelona, Barcelona, Spain; .,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
31
|
Capel F, Cheraiti N, Acquaviva C, Hénique C, Bertrand-Michel J, Vianey-Saban C, Prip-Buus C, Morio B. Oleate dose-dependently regulates palmitate metabolism and insulin signaling in C2C12 myotubes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2000-2010. [PMID: 27725263 DOI: 10.1016/j.bbalip.2016.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/10/2016] [Accepted: 10/05/2016] [Indexed: 01/22/2023]
Abstract
Because the protective effect of oleate against palmitate-induced insulin resistance may be lessened in skeletal muscle once cell metabolism is overloaded by fatty acids (FAs), we examined the impact of varying amounts of oleate on palmitate metabolic channeling and insulin signaling in C2C12 myotubes. Cells were exposed to 0.5mM of palmitate and to increasing doses of oleate (0.05, 0.25 and 0.5mM). Impacts of FA treatments on radio-labelled FA fluxes, on cellular content in diacylglycerols (DAG), triacylglycerols (TAG), ceramides, acylcarnitines, on PKCθ, MAPKs (ERK1/2, p38) and NF-ΚB activation, and on insulin-dependent Akt phosphorylation were examined. Low dose of oleate (0.05mM) was sufficient to improve palmitate complete oxidation to CO2 (+29%, P<0.05) and to alter the cellular acylcarnitine profile. Insulin-induced Akt phosphorylation was 48% higher in that condition vs. palmitate alone (p<0.01). Although DAG and ceramide contents were significantly decreased with 0.05mM of oleate vs. palmitate alone (-47 and -28%, respectively, p<0.01), 0.25mM of oleate was required to decrease p38 MAPK and PKCθ phosphorylation, thus further improving the insulin signaling (+32%, p<0.05). By contrast, increasing oleate concentration from 0.25 to 0.5mM, thus increasing total amount of FA from 0.75 to 1mM, deteriorated the insulin signaling pathway (-30%, p<0.01). This was observed despite low contents in DAG and ceramides, and enhanced palmitate incorporation into TAG (+27%, p<0.05). This was associated with increased incomplete FA β-oxidation and impairment of acylcarnitine profile. In conclusion, these combined data place mitochondrial β-oxidation at the center of the regulation of muscle insulin sensitivity, besides p38 MAPK and PKCθ.
Collapse
Affiliation(s)
- Frédéric Capel
- INRA UMR1019 Nutrition Humaine, Laboratoire de Nutrition Humaine, Université d'Auvergne, CRNH, 58 rue Montalembert BP321, 63009 Clermont Ferrand CEDEX 1, France.
| | - Naoufel Cheraiti
- INRA UMR1019 Nutrition Humaine, Laboratoire de Nutrition Humaine, Université d'Auvergne, CRNH, 58 rue Montalembert BP321, 63009 Clermont Ferrand CEDEX 1, France.
| | - Cécile Acquaviva
- Service Maladies Héréditaires du Métabolisme, Centre de Biologie et Pathologie Est, CHU de Lyon, France.
| | - Carole Hénique
- Institut Cochin, Département d'Endocrinologie, Métabolisme and Diabète, INSERM U1016/CNRS UMR8104/UMR-S8104, Bâtiment Faculté, 24 rue du faubourg Saint Jacques, 75014 Paris, France.
| | - Justine Bertrand-Michel
- MetaToul-Lipidomic, MetaboHUB, INSERM UMR 1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France University of Toulouse, UMR1048, Paul Sabatier University, France.
| | - Christine Vianey-Saban
- Service Maladies Héréditaires du Métabolisme, Centre de Biologie et Pathologie Est, CHU de Lyon, France.
| | - Carina Prip-Buus
- Institut Cochin, Département d'Endocrinologie, Métabolisme and Diabète, INSERM U1016/CNRS UMR8104/UMR-S8104, Bâtiment Faculté, 24 rue du faubourg Saint Jacques, 75014 Paris, France.
| | - Béatrice Morio
- INRA UMR1019 Nutrition Humaine, Laboratoire de Nutrition Humaine, Université d'Auvergne, CRNH, 58 rue Montalembert BP321, 63009 Clermont Ferrand CEDEX 1, France; INRA UMR1397, Laboratoire CarMeN, Inserm UMR1060, Université Lyon 1, INSA de Lyon, Faculté de Médecine Lyon Sud, BP 12, 165 Chemin du Grand Revoyet, 69921 Oullins Cedex, France.
| |
Collapse
|
32
|
Vavrova E, Lenoir V, Alves-Guerra MC, Denis RG, Castel J, Esnous C, Dyck JRB, Luquet S, Metzger D, Bouillaud F, Prip-Buus C. Muscle expression of a malonyl-CoA-insensitive carnitine palmitoyltransferase-1 protects mice against high-fat/high-sucrose diet-induced insulin resistance. Am J Physiol Endocrinol Metab 2016; 311:E649-60. [PMID: 27507552 DOI: 10.1152/ajpendo.00020.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 08/09/2016] [Indexed: 12/17/2022]
Abstract
Impaired skeletal muscle mitochondrial fatty acid oxidation (mFAO) has been implicated in the etiology of insulin resistance. Carnitine palmitoyltransferase-1 (CPT1) is a key regulatory enzyme of mFAO whose activity is inhibited by malonyl-CoA, a lipogenic intermediate. Whereas increasing CPT1 activity in vitro has been shown to exert a protective effect against lipid-induced insulin resistance in skeletal muscle cells, only a few studies have addressed this issue in vivo. We thus examined whether a direct modulation of muscle CPT1/malonyl-CoA partnership is detrimental or beneficial for insulin sensitivity in the context of diet-induced obesity. By using a Cre-LoxP recombination approach, we generated mice with skeletal muscle-specific and inducible expression of a mutated CPT1 form (CPT1mt) that is active but insensitive to malonyl-CoA inhibition. When fed control chow, homozygous CPT1mt transgenic (dbTg) mice exhibited decreased CPT1 sensitivity to malonyl-CoA inhibition in isolated muscle mitochondria, which was sufficient to substantially increase ex vivo muscle mFAO capacity and whole body fatty acid utilization in vivo. Moreover, dbTg mice were less prone to high-fat/high-sucrose (HFHS) diet-induced insulin resistance and muscle lipotoxicity despite similar body weight gain, adiposity, and muscle malonyl-CoA content. Interestingly, these CPT1mt-protective effects in dbTg-HFHS mice were associated with preserved muscle insulin signaling, increased muscle glycogen content, and upregulation of key genes involved in muscle glucose metabolism. These beneficial effects of muscle CPT1mt expression suggest that a direct modulation of the malonyl-CoA/CPT1 partnership in skeletal muscle could represent a potential strategy to prevent obesity-induced insulin resistance.
Collapse
Affiliation(s)
- Eliska Vavrova
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France; Centre National de la Recherche Scientifique, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Paris Diderot, Paris, France
| | - Véronique Lenoir
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France; Centre National de la Recherche Scientifique, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie-Clotilde Alves-Guerra
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France; Centre National de la Recherche Scientifique, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Raphaël G Denis
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, Centre National de la Recherche Scientifique UMR8251, Paris, France
| | - Julien Castel
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, Centre National de la Recherche Scientifique UMR8251, Paris, France
| | - Catherine Esnous
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France; Centre National de la Recherche Scientifique, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Serge Luquet
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, Centre National de la Recherche Scientifique UMR8251, Paris, France
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale, U964, Centre National de la Recherche Scientifique, UMR7104, Université de Strasbourg, Illkirch, France
| | - Frédéric Bouillaud
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France; Centre National de la Recherche Scientifique, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Carina Prip-Buus
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France; Centre National de la Recherche Scientifique, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France;
| |
Collapse
|
33
|
Calderon-Dominguez M, Sebastián D, Fucho R, Weber M, Mir JF, García-Casarrubios E, Obregón MJ, Zorzano A, Valverde ÁM, Serra D, Herrero L. Carnitine Palmitoyltransferase 1 Increases Lipolysis, UCP1 Protein Expression and Mitochondrial Activity in Brown Adipocytes. PLoS One 2016; 11:e0159399. [PMID: 27438137 PMCID: PMC4954705 DOI: 10.1371/journal.pone.0159399] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/03/2016] [Indexed: 02/07/2023] Open
Abstract
The discovery of active brown adipose tissue (BAT) in adult humans and the fact that it is reduced in obese and diabetic patients have put a spotlight on this tissue as a key player in obesity-induced metabolic disorders. BAT regulates energy expenditure through thermogenesis; therefore, harnessing its thermogenic fat-burning power is an attractive therapeutic approach. We aimed to enhance BAT thermogenesis by increasing its fatty acid oxidation (FAO) rate. Thus, we expressed carnitine palmitoyltransferase 1AM (CPT1AM), a permanently active mutant form of CPT1A (the rate-limiting enzyme in FAO), in a rat brown adipocyte (rBA) cell line through adenoviral infection. We found that CPT1AM-expressing rBA have increased FAO, lipolysis, UCP1 protein levels and mitochondrial activity. Additionally, enhanced FAO reduced the palmitate-induced increase in triglyceride content and the expression of obese and inflammatory markers. Thus, CPT1AM-expressing rBA had enhanced fat-burning capacity and improved lipid-induced derangements. This indicates that CPT1AM-mediated increase in brown adipocytes FAO may be a new approach to the treatment of obesity-induced disorders.
Collapse
Affiliation(s)
- María Calderon-Dominguez
- Department of Biochemistry and Physiology, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - David Sebastián
- Institute for Research in Biomedicine (IRB Barcelona) and Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Raquel Fucho
- Department of Biochemistry and Physiology, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Minéia Weber
- Department of Biochemistry and Physiology, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Joan F. Mir
- Department of Biochemistry and Physiology, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Ester García-Casarrubios
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM) and Instituto de Investigación Sanitaria La Paz, 28029, Madrid, Spain
| | - María Jesús Obregón
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM) and Instituto de Investigación Sanitaria La Paz, 28029, Madrid, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona) and Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Ángela M. Valverde
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, E-28029, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM) and Instituto de Investigación Sanitaria La Paz, 28029, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
- * E-mail:
| |
Collapse
|
34
|
Zhang L, Tian F, Gao X, Wang X, Wu C, Li N, Li J. N-3 Polyunsaturated Fatty Acids Improve Liver Lipid Oxidation-Related Enzyme Levels and Increased the Peroxisome Proliferator-Activated Receptor α Expression Level in Mice Subjected to Hemorrhagic Shock/Resuscitation. Nutrients 2016; 8:237. [PMID: 27110821 PMCID: PMC4848705 DOI: 10.3390/nu8040237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/09/2016] [Accepted: 04/15/2016] [Indexed: 01/02/2023] Open
Abstract
Appropriate metabolic interventions after hemorrhagic shock/resuscitation injury have not yet been identified. We aimed to examine the effects of fish oil on lipid metabolic intervention after hemorrhagic shock/resuscitation. Firstly, 48 C57BL/6 mice were assigned to six groups (n = 8 per group). The sham group did not undergo surgery, while mice in the remaining groups were sacrificed 1–5 days after hemorrhagic shock/resuscitation. In the second part, mice were treated with saline or fish oil (n = 8 per group) five days after injury. We determined serum triglyceride levels and liver tissues were collected and prepared for qRT-PCR or Western blot analysis. We found that triglyceride levels were increased five days after hemorrhagic shock/resuscitation, but decreased after addition of fish oil. After injury, the protein and gene expression of carnitine palmitoyltransferase 1A, fatty acid transport protein 1, and peroxisome proliferator-activated receptor-α decreased significantly in liver tissue. In contrast, after treatment with fish oil, the expression levels of these targets increased compared with those in the saline group. The present results suggest n-3 polyunsaturated fatty acids could improve lipid oxidation-related enzymes in liver subjected to hemorrhagic shock/resuscitation. This function is possibly accomplished through activating the peroxisome proliferator-activated receptor-α pathway.
Collapse
Affiliation(s)
- Li Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Feng Tian
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Clinical College of Southern Medical University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Chao Wu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Ning Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Jieshou Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| |
Collapse
|
35
|
Calderon-Dominguez M, Mir JF, Fucho R, Weber M, Serra D, Herrero L. Fatty acid metabolism and the basis of brown adipose tissue function. Adipocyte 2016; 5:98-118. [PMID: 27386151 PMCID: PMC4916887 DOI: 10.1080/21623945.2015.1122857] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/13/2015] [Accepted: 11/12/2015] [Indexed: 12/21/2022] Open
Abstract
Obesity has reached epidemic proportions, leading to severe associated pathologies such as insulin resistance, cardiovascular disease, cancer and type 2 diabetes. Adipose tissue has become crucial due to its involvement in the pathogenesis of obesity-induced insulin resistance, and traditionally white adipose tissue has captured the most attention. However in the last decade the presence and activity of heat-generating brown adipose tissue (BAT) in adult humans has been rediscovered. BAT decreases with age and in obese and diabetic patients. It has thus attracted strong scientific interest, and any strategy to increase its mass or activity might lead to new therapeutic approaches to obesity and associated metabolic diseases. In this review we highlight the mechanisms of fatty acid uptake, trafficking and oxidation in brown fat thermogenesis. We focus on BAT's morphological and functional characteristics and fatty acid synthesis, storage, oxidation and use as a source of energy.
Collapse
Affiliation(s)
- María Calderon-Dominguez
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan F. Mir
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Raquel Fucho
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Minéia Weber
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
36
|
Han L, Li J, Chen Y, Wang W, Zhang D, Liu G. Effects of Ghrelin on Triglyceride Accumulation and Glucose Uptake in Primary Cultured Rat Myoblasts under Palmitic Acid-Induced High Fat Conditions. Int J Endocrinol 2015; 2015:635863. [PMID: 26713091 DOI: 10.1155/2015/635863] [cited] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 09/11/2015] [Accepted: 09/20/2015] [Indexed: 01/26/2025] Open
Abstract
This study aimed to study the effects of acylated ghrelin on glucose and triglyceride metabolism in rat myoblasts under palmitic acid- (PA-) induced high fat conditions. Rat myoblasts were treated with 0, 10(-11), 10(-9), or 10(-7) M acylated ghrelin and 0.3 mM PA for 12 h. Triglyceride accumulation was determined by Oil-Red-O staining and the glycerol phosphate dehydrogenase-peroxidase enzymatic method, and glucose uptake was determined by isotope tracer. The glucose transporter 4 (GLUT4), AMP-activated protein kinase (AMPK), acetyl-CoA carboxylase (ACC), and uncoupling protein 3 (UCP3) were assessed by RT-PCR and western blot. Compared to 0.3 mM PA, ghrelin at 10(-9) and 10(-7) M reduced triglyceride content (5.855 ± 0.352 versus 5.030 ± 0.129 and 4.158 ± 0.254 mM, P < 0.05) and prevented PA-induced reduction of glucose uptake (1.717 ± 0.264 versus 2.233 ± 0.333 and 2.333 ± 0.273 10(-2) pmol/g/min, P < 0.05). The relative protein expression of p-AMPKα/AMPKα, UCP3, and p-ACC under 0.3 mM PA was significantly reduced compared to controls (all P < 0.05), but those in the 10(-9) and 10(-7) M ghrelin groups were significantly protected from 0.3 mM PA (all P < 0.05). In conclusion, acylated ghrelin reduced PA-induced triglyceride accumulation and prevented the PA-induced decrease in glucose uptake in rat myoblasts. These effects may involve fatty acid oxidation.
Collapse
Affiliation(s)
- Lingling Han
- Department of Endocrinology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jia Li
- Department of Endocrinology, The Fourth Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Ying Chen
- Shenyang 93303 Military Hospital, Shenyang, Liaoning 110011, China
| | - Wei Wang
- Department of Endocrinology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Dan Zhang
- Department of Endocrinology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Guoliang Liu
- Department of Endocrinology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| |
Collapse
|
37
|
Van Hoeck V, Rizos D, Gutierrez-Adan A, Pintelon I, Jorssen E, Dufort I, Sirard MA, Verlaet A, Hermans N, Bols PEJ, Leroy JLMR. Interaction between differential gene expression profile and phenotype in bovine blastocysts originating from oocytes exposed to elevated non-esterified fatty acid concentrations. Reprod Fertil Dev 2015; 27:372-84. [PMID: 24360349 DOI: 10.1071/rd13263] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/28/2013] [Indexed: 02/03/2023] Open
Abstract
Maternal metabolic disorders linked to lipolysis are major risk factors for reproductive failure. A notable feature of such disorders is increased non-esterified fatty acid (NEFA) concentrations in the blood, which are reflected in the ovarian follicular fluid. Elevated NEFA concentrations impact on the maturing oocyte and even alter subsequent embryo physiology. The aetiological mechanisms have not been fully elucidated. Therefore, in the present study, bovine in vitro maturing cumulus-oocyte complexes were exposed (24 h) to three different maturation treatments containing: (1) physiological (72 µM) NEFA concentrations (=control); (2) elevated (75 µM) stearic acid (SA) concentrations (=HIGH SA); and (3) elevated (425 µM) NEFA concentrations (=HIGH COMBI). Zygotes were fertilised and cultured following standard procedures. Transcriptomic analyses in resulting Day 7.5 blastocysts revealed that the major pathways affected are related to lipid and carbohydrate metabolism in HIGH COMBI embryos and to lipid metabolism and cell death in HIGH SA embryos. Furthermore, lower glutathione content and a reduced number of lipid droplets per cell were observed in HIGH SA-exposed oocytes and resulting morulae, respectively, compared with their HIGH COMBI-exposed counterparts. Vitrified embryos originating from HIGH SA-exposed oocytes tended to exhibit lower survival rates compared with controls. These data suggest possible mechanisms explaining why females across species suffering lipolytic disorders experience difficulties in conceiving.
Collapse
Affiliation(s)
- V Van Hoeck
- Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - D Rizos
- Departamento de Reproducción Animal y Conservación de Recursos Zoogenéticos, INIA, Ctra. de la Coruna Km 5.9, 28040 Madrid, Spain
| | - A Gutierrez-Adan
- Departamento de Reproducción Animal y Conservación de Recursos Zoogenéticos, INIA, Ctra. de la Coruna Km 5.9, 28040 Madrid, Spain
| | - I Pintelon
- Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - E Jorssen
- Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - I Dufort
- Département des Sciences Animales Pavillon des services, INAF, Université Laval, G1V 0A6 Québec, Canada
| | - M A Sirard
- Département des Sciences Animales Pavillon des services, INAF, Université Laval, G1V 0A6 Québec, Canada
| | - A Verlaet
- Departement Pharmaceutical Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - N Hermans
- Departement Pharmaceutical Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - P E J Bols
- Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - J L M R Leroy
- Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
38
|
Tumova J, Malisova L, Andel M, Trnka J. Protective Effect of Unsaturated Fatty Acids on Palmitic Acid-Induced Toxicity in Skeletal Muscle Cells is not Mediated by PPARδ Activation. Lipids 2015; 50:955-64. [DOI: 10.1007/s11745-015-4058-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/22/2015] [Indexed: 11/28/2022]
|
39
|
Koo YD, Choi JW, Kim M, Chae S, Ahn BY, Kim M, Oh BC, Hwang D, Seol JH, Kim YB, Park YJ, Chung SS, Park KS. SUMO-Specific Protease 2 (SENP2) Is an Important Regulator of Fatty Acid Metabolism in Skeletal Muscle. Diabetes 2015; 64:2420-31. [PMID: 25784542 PMCID: PMC4477359 DOI: 10.2337/db15-0115] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 02/27/2015] [Indexed: 12/27/2022]
Abstract
Small ubiquitin-like modifier (SUMO)-specific proteases (SENPs) that reverse protein modification by SUMO are involved in the control of numerous cellular processes, including transcription, cell division, and cancer development. However, the physiological function of SENPs in energy metabolism remains unclear. Here, we investigated the role of SENP2 in fatty acid metabolism in C2C12 myotubes and in vivo. In C2C12 myotubes, treatment with saturated fatty acids, like palmitate, led to nuclear factor-κB-mediated increase in the expression of SENP2. This increase promoted the recruitment of peroxisome proliferator-activated receptor (PPAR)δ and PPARγ, through desumoylation of PPARs, to the promoters of the genes involved in fatty acid oxidation (FAO), such as carnitine-palmitoyl transferase-1 (CPT1b) and long-chain acyl-CoA synthetase 1 (ACSL1). In addition, SENP2 overexpression substantially increased FAO in C2C12 myotubes. Consistent with the cell culture system, muscle-specific SENP2 overexpression led to a marked increase in the mRNA levels of CPT1b and ACSL1 and thereby in FAO in the skeletal muscle, which ultimately alleviated high-fat diet-induced obesity and insulin resistance. Collectively, these data identify SENP2 as an important regulator of fatty acid metabolism in skeletal muscle and further implicate that muscle SENP2 could be a novel therapeutic target for the treatment of obesity-linked metabolic disorders.
Collapse
Affiliation(s)
- Young Do Koo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Woo Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Myungjin Kim
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Sehyun Chae
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Korea
| | - Byung Yong Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Byung Chul Oh
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Daehee Hwang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Korea
| | - Jae Hong Seol
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine, Seoul National University, Seoul, Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Soo Chung
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
40
|
Shikonin protects against obesity through the modulation of adipogenesis, lipogenesis, and β-oxidation in vivo. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.04.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
41
|
Malandrino MI, Fucho R, Weber M, Calderon-Dominguez M, Mir JF, Valcarcel L, Escoté X, Gómez-Serrano M, Peral B, Salvadó L, Fernández-Veledo S, Casals N, Vázquez-Carrera M, Villarroya F, Vendrell JJ, Serra D, Herrero L. Enhanced fatty acid oxidation in adipocytes and macrophages reduces lipid-induced triglyceride accumulation and inflammation. Am J Physiol Endocrinol Metab 2015; 308:E756-69. [PMID: 25714670 DOI: 10.1152/ajpendo.00362.2014] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 02/19/2015] [Indexed: 12/31/2022]
Abstract
Lipid overload in obesity and type 2 diabetes is associated with adipocyte dysfunction, inflammation, macrophage infiltration, and decreased fatty acid oxidation (FAO). Here, we report that the expression of carnitine palmitoyltransferase 1A (CPT1A), the rate-limiting enzyme in mitochondrial FAO, is higher in human adipose tissue macrophages than in adipocytes and that it is differentially expressed in visceral vs. subcutaneous adipose tissue in both an obese and a type 2 diabetes cohort. These observations led us to further investigate the potential role of CPT1A in adipocytes and macrophages. We expressed CPT1AM, a permanently active mutant form of CPT1A, in 3T3-L1 CARΔ1 adipocytes and RAW 264.7 macrophages through adenoviral infection. Enhanced FAO in palmitate-incubated adipocytes and macrophages reduced triglyceride content and inflammation, improved insulin sensitivity in adipocytes, and reduced endoplasmic reticulum stress and ROS damage in macrophages. We conclude that increasing FAO in adipocytes and macrophages improves palmitate-induced derangements. This indicates that enhancing FAO in metabolically relevant cells such as adipocytes and macrophages may be a promising strategy for the treatment of chronic inflammatory pathologies such as obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Maria Ida Malandrino
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Raquel Fucho
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Minéia Weber
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María Calderon-Dominguez
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Francesc Mir
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Lorea Valcarcel
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Escoté
- Endocrinology and Diabetes Unit, Joan XXIII University Hospital, Institut d'Investigació Sanitària Pere i Virgili, Universitat Rovira i Virgili, Tarragona, Spain; Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - María Gómez-Serrano
- Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Belén Peral
- Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Laia Salvadó
- Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain; Pharmacology Unit, Department of Pharmacology and Therapeutic Chemistry and Institut de Biomedicina de la Universitat de Barcelona, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain; and
| | - Sonia Fernández-Veledo
- Endocrinology and Diabetes Unit, Joan XXIII University Hospital, Institut d'Investigació Sanitària Pere i Virgili, Universitat Rovira i Virgili, Tarragona, Spain; Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Núria Casals
- Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain; Pharmacology Unit, Department of Pharmacology and Therapeutic Chemistry and Institut de Biomedicina de la Universitat de Barcelona, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain; and
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Joan J Vendrell
- Endocrinology and Diabetes Unit, Joan XXIII University Hospital, Institut d'Investigació Sanitària Pere i Virgili, Universitat Rovira i Virgili, Tarragona, Spain; Institut de Biomedicina de la Universitat de Barcelona de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Molecular Biology, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Madrid, Spain;
| |
Collapse
|
42
|
Hepatocyte growth factor is elevated in amniotic fluid from obese women and regulates placental glucose and fatty acid metabolism. Placenta 2015; 36:381-8. [PMID: 25690371 DOI: 10.1016/j.placenta.2015.01.199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/26/2015] [Accepted: 01/28/2015] [Indexed: 12/24/2022]
Abstract
INTRODUCTION To evaluate the impact of the pro-inflammatory cytokine hepatocyte growth factor (HGF) on the regulation of glucose and lipid placental metabolism. METHODS HGF levels were quantified in amniotic fluid and placenta from control and obese women. 2-deoxy-glucose (2-DOG) uptake, glycolysis, fatty acid oxidation (FAO), fatty acid esterification, de novo fatty acid synthesis, triglyceride levels and carnitine palmitoyltransferase activities (CPT) were measured in placental explants upon addition of pathophysiological HGF levels. RESULTS In obese women, total- and -activated-HGF levels in amniotic fluid were elevated ∼24%, and placental HGF levels were ∼3-fold higher than in control women. At a similar dose to that present in amniotic fluid of obese women, HGF (30 ng/mL) increased Glut-1 levels and 2-DOG uptake by ∼25-30% in placental explants. HGF-mediated effect on 2-DOG uptake was dependent on the activation of phosphatidylinositol 3-kinase signaling pathway. In addition, HGF decreased ∼20% FAO, whereas esterification and de novo fatty acid synthesis increased ∼15% and ∼25% respectively, leading to 2-fold triglyceride accumulation in placental explants. In parallel, HGF reduced CPT-I activity ∼70%. DISCUSSION HGF is a cytokine elevated in amniotic fluid and placental tissue of obese women, which through its ability to stimulate 2-DOG uptake and metabolism impairs FAO and enhances esterification and de novo fatty acid synthesis, leading to accumulation of placental triglycerides.
Collapse
|
43
|
Beneficial role of bitter melon supplementation in obesity and related complications in metabolic syndrome. J Lipids 2015; 2015:496169. [PMID: 25650336 PMCID: PMC4306384 DOI: 10.1155/2015/496169] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 12/05/2014] [Indexed: 02/06/2023] Open
Abstract
Diabetes, obesity, and metabolic syndrome are becoming epidemic both in developed and developing countries in recent years. Complementary and alternative medicines have been used since ancient era for the treatment of diabetes and cardiovascular diseases. Bitter melon is widely used as vegetables in daily food in Bangladesh and several other countries in Asia. The fruits extract of bitter melon showed strong antioxidant and hypoglycemic activities in experimental condition both in vivo and in vitro. Recent scientific evaluation of this plant extracts also showed potential therapeutic benefit in diabetes and obesity related metabolic dysfunction in experimental animals and clinical studies. These beneficial effects are mediated probably by inducing lipid and fat metabolizing gene expression and increasing the function of AMPK and PPARs, and so forth. This review will thus focus on the recent findings on beneficial effect of Momordica charantia extracts on metabolic syndrome and discuss its potential mechanism of actions.
Collapse
|
44
|
Alkhalidy H, Moore W, Zhang Y, McMillan R, Wang A, Ali M, Suh KS, Zhen W, Cheng Z, Jia Z, Hulver M, Liu D. Small Molecule Kaempferol Promotes Insulin Sensitivity and Preserved Pancreatic β -Cell Mass in Middle-Aged Obese Diabetic Mice. J Diabetes Res 2015; 2015:532984. [PMID: 26064984 PMCID: PMC4439495 DOI: 10.1155/2015/532984] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 04/09/2015] [Accepted: 04/20/2015] [Indexed: 12/20/2022] Open
Abstract
Insulin resistance and a progressive decline in functional β-cell mass are hallmarks of developing type 2 diabetes (T2D). Thus, searching for natural, low-cost compounds to target these two defects could be a promising strategy to prevent the pathogenesis of T2D. Here, we show that dietary intake of flavonol kaempferol (0.05% in the diet) significantly ameliorated hyperglycemia, hyperinsulinemia, and circulating lipid profile, which were associated with the improved peripheral insulin sensitivity in middle-aged obese mice fed a high-fat (HF) diet. Kaempferol treatment reversed HF diet impaired glucose transport-4 (Glut4) and AMP-dependent protein kinase (AMPK) expression in both muscle and adipose tissues from obese mice. In vitro, kaempferol increased lipolysis and prevented high fatty acid-impaired glucose uptake, glycogen synthesis, AMPK activity, and Glut4 expression in skeletal muscle cells. Using another mouse model of T2D generated by HF diet feeding and low doses of streptozotocin injection, we found that kaempferol treatment significantly improved hyperglycemia, glucose tolerance, and blood insulin levels in obese diabetic mice, which are associated with the improved islet β-cell mass. These results demonstrate that kaempferol may be a naturally occurring anti-diabetic agent by improving peripheral insulin sensitivity and protecting against pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Hana Alkhalidy
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - William Moore
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Yanling Zhang
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Ryan McMillan
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
- The Metabolic Phenotyping Core, Virginia Tech, Blacksburg, VA 24061, USA
| | - Aihua Wang
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Mostafa Ali
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Kyung-Shin Suh
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Wei Zhen
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Zhiyong Cheng
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Zhenquan Jia
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, NC 27412, USA
| | - Matthew Hulver
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Dongmin Liu
- Department of Human Nutrition, Foods & Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
- *Dongmin Liu:
| |
Collapse
|
45
|
Han L, Li J, Chen Y, Wang W, Zhang D, Liu G. Effects of Ghrelin on Triglyceride Accumulation and Glucose Uptake in Primary Cultured Rat Myoblasts under Palmitic Acid-Induced High Fat Conditions. Int J Endocrinol 2015; 2015:635863. [PMID: 26713091 PMCID: PMC4680104 DOI: 10.1155/2015/635863] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 09/11/2015] [Accepted: 09/20/2015] [Indexed: 02/08/2023] Open
Abstract
This study aimed to study the effects of acylated ghrelin on glucose and triglyceride metabolism in rat myoblasts under palmitic acid- (PA-) induced high fat conditions. Rat myoblasts were treated with 0, 10(-11), 10(-9), or 10(-7) M acylated ghrelin and 0.3 mM PA for 12 h. Triglyceride accumulation was determined by Oil-Red-O staining and the glycerol phosphate dehydrogenase-peroxidase enzymatic method, and glucose uptake was determined by isotope tracer. The glucose transporter 4 (GLUT4), AMP-activated protein kinase (AMPK), acetyl-CoA carboxylase (ACC), and uncoupling protein 3 (UCP3) were assessed by RT-PCR and western blot. Compared to 0.3 mM PA, ghrelin at 10(-9) and 10(-7) M reduced triglyceride content (5.855 ± 0.352 versus 5.030 ± 0.129 and 4.158 ± 0.254 mM, P < 0.05) and prevented PA-induced reduction of glucose uptake (1.717 ± 0.264 versus 2.233 ± 0.333 and 2.333 ± 0.273 10(-2) pmol/g/min, P < 0.05). The relative protein expression of p-AMPKα/AMPKα, UCP3, and p-ACC under 0.3 mM PA was significantly reduced compared to controls (all P < 0.05), but those in the 10(-9) and 10(-7) M ghrelin groups were significantly protected from 0.3 mM PA (all P < 0.05). In conclusion, acylated ghrelin reduced PA-induced triglyceride accumulation and prevented the PA-induced decrease in glucose uptake in rat myoblasts. These effects may involve fatty acid oxidation.
Collapse
Affiliation(s)
- Lingling Han
- Department of Endocrinology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jia Li
- Department of Endocrinology, The Fourth Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Ying Chen
- Shenyang 93303 Military Hospital, Shenyang, Liaoning 110011, China
| | - Wei Wang
- Department of Endocrinology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Dan Zhang
- Department of Endocrinology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Guoliang Liu
- Department of Endocrinology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
- *Guoliang Liu:
| |
Collapse
|
46
|
Rodriguez S, Ellis JM, Wolfgang MJ. Chemical-genetic induction of Malonyl-CoA decarboxylase in skeletal muscle. BMC BIOCHEMISTRY 2014; 15:20. [PMID: 25152047 PMCID: PMC4236586 DOI: 10.1186/1471-2091-15-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 08/13/2014] [Indexed: 01/02/2023]
Abstract
Background Defects in skeletal muscle fatty acid oxidation have been implicated in the etiology of insulin resistance. Malonyl-CoA decarboxylase (MCD) has been a target of investigation because it reduces the concentration of malonyl-CoA, a metabolite that inhibits fatty acid oxidation. The in vivo role of muscle MCD expression in the development of insulin resistance remains unclear. Results To determine the role of MCD in skeletal muscle of diet induced obese and insulin resistant mouse models we generated mice expressing a muscle specific transgene for MCD (Tg-fMCDSkel) stabilized posttranslationally by the small molecule, Shield-1. Tg-fMCDSkel and control mice were placed on either a high fat or low fat diet for 3.5 months. Obese and glucose intolerant as well as lean control Tg-fMCDSkel and nontransgenic control mice were treated with Shield-1 and changes in their body weight and insulin sensitivity were determined upon induction of MCD. Inducing MCD activity >5-fold in skeletal muscle over two weeks did not alter body weight or glucose intolerance of obese mice. MCD induction further potentiated the defects in insulin signaling of obese mice. In addition, key enzymes in fatty acid oxidation were suppressed following MCD induction. Conclusion Acute induction of MCD in the skeletal muscle of obese and glucose intolerant mice did not improve body weight and decreased insulin sensitivity compared to obese nontransgenic controls. Induction of MCD in skeletal muscle resulted in a suppression of mitochondrial oxidative genes suggesting a redundant and metabolite driven regulation of gene expression.
Collapse
Affiliation(s)
| | | | - Michael J Wolfgang
- Department of Biological Chemistry, Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, 725 N, Wolfe St,, 475 Rangos Building, Baltimore, Maryland 21205, USA.
| |
Collapse
|
47
|
Verma MK, Yateesh AN, Smitha R, Neelima K, Pallavi PM, Reddy M, Poornima J, Oommen AM, Jagannath MR, Somesh BP. Integrated analysis of chronic lipotoxicity on muscle metabolism and stress and its reversal by antioxidants. SPRINGERPLUS 2014; 3:251. [PMID: 24936385 PMCID: PMC4048371 DOI: 10.1186/2193-1801-3-251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 05/12/2014] [Indexed: 12/21/2022]
Abstract
Apart from elevated glucose, triglyceride and cholesterol, elevated levels of serum free-fatty acid (FFA) are observed in diabetic patients. Increased FFA load can cause multiple dysregulation which are collectively known as lipotoxicity. Impacts of FFA induced lipotoxicity were evaluated on various cellular responses of metabolism and stress in skeletal muscle myotubes. Under lipotoxicity, oxidative capacity of C2C12 myotubes was reduced and decreased levels ATP and NAD were observed. Lipotoxicity augmented non-oxidative disposal of metabolites in terms of lactate release, IMTG and ceramide synthesis. Concomitantly, insulin resistance was also observed. These impacts were in conjunction with increased cellular stress, inflammation, proteolysis and apoptosis. Quenching of lipotoxicity mediated oxidative stress by antioxidant reverted its deleterious impacts and restored insulin stimulated glucose uptake. In conclusion, the in vitro lipotoxicity makes a system which resembles in vivo pathology of muscle as seen in diabetic patients and represents an integrated perspective of lipotoxicity on various parameters of metabolism and stress.
Collapse
Affiliation(s)
- Mahesh Kumar Verma
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| | - Aggunda Nagaraju Yateesh
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| | - Rachapalli Smitha
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| | - Korrapati Neelima
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| | - Puttrevana M Pallavi
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| | - Madhusudhan Reddy
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| | - Jayaram Poornima
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| | - Anup M Oommen
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| | - Madanahalli R Jagannath
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| | - Baggavalli P Somesh
- Connexios Life Sciences Private Ltd, No. 49, First Main road, 3rd phase, JP Nagar, Bangalore, 560 078 India
| |
Collapse
|
48
|
Maternal protein restriction impairs the transcriptional metabolic flexibility of skeletal muscle in adult rat offspring. Br J Nutr 2014; 112:328-37. [DOI: 10.1017/s0007114514000865] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Skeletal muscle exhibits a remarkable flexibility in the usage of fuel in response to the nutrient intake and energy demands of the organism. In fact, increased physical activity and fasting trigger a transcriptional programme in skeletal muscle cells leading to a switch from carbohydrate to lipid oxidation. Impaired metabolic flexibility has been reported to be associated with obesity and type 2 diabetes, but it is not known whether the disability to adapt to metabolic demands is a cause or a consequence of these pathological conditions. Inasmuch as a poor nutritional environment during early life is a predisposing factor for the development of metabolic diseases in adulthood, in the present study, we aimed to determine the long-term effects of maternal malnutrition on the metabolic flexibility of offspring skeletal muscle. To this end, the transcriptional responses of the soleus and extensor digitorum longus muscles to fasting were evaluated in adult rats born to dams fed a control (17 % protein) or a low-protein (8 % protein, protein restricted (PR)) diet throughout pregnancy and lactation. With the exception of reduced body weight and reduced plasma concentrations of TAG, PR rats exhibited a metabolic profile that was the same as that of the control rats. In the fed state, PR rats exhibited an enhanced expression of key regulatory genes of fatty acid oxidation including CPT1a, PGC-1α, UCP3 and PPARα and an impaired expression of genes that increase the capacity for fat oxidation in response to fasting. These results suggest that impaired metabolic inflexibility precedes and may contribute to the development of metabolic disorders associated with early malnutrition.
Collapse
|
49
|
Fukumori R, Sugino T, Shingu H, Moriya N, Kobayashi H, Hasegawa Y, Kojima M, Kangawa K, Obitsu T, Kushibiki S, Taniguchi K. Ingestion of medium chain fatty acids by lactating dairy cows increases concentrations of plasma ghrelin. Domest Anim Endocrinol 2013; 45:216-23. [PMID: 24209506 DOI: 10.1016/j.domaniend.2013.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/12/2013] [Accepted: 09/12/2013] [Indexed: 10/26/2022]
Abstract
The purpose of this study was to elucidate the effects of medium-chain fatty acids (MCFAs) on plasma ghrelin concentration in lactating dairy cows. Five early-lactating Holstein cows were randomly assigned to 2 dietary treatments in a crossover design with 2-wk periods. Treatments consisted of diets supplemented or not (control) with calcium salts of MCFAs (MCFA-Ca; 1.5% dry matter). Plasma hormone and metabolite concentrations in blood samples taken from the jugular vein were measured on the morning of feeding on day 14 of each period. Dry matter intake, milk protein, and lactose content of cows fed the MCFA-Ca diet were decreased compared with controls, but with no change in milk yield. Plasma ghrelin concentrations were higher in cows fed the MCFA-Ca diet; however, no significant effect was found on glucagon-like peptide-1 concentrations in plasma. Plasma insulin concentrations decreased, but plasma glucagon concentrations remained unchanged in cows fed the MCFA-Ca diet. The concentrations of nonesterified FAs, total cholesterol, and β-hydroxybutyrate in plasma increased in these cows. In conclusion, dietary MCFAs increase the plasma ghrelin concentrations in lactating dairy cows.
Collapse
Affiliation(s)
- R Fukumori
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8528, Japan; National Institute of Livestock and Grassland Science, Tsukuba 305-0901, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
ASTORINO TODDA, SCHUBERT MATTHEWM, PALUMBO ELYSE, STIRLING DOUGLAS, MCMILLAN DAVIDW. Effect of Two Doses of Interval Training on Maximal Fat Oxidation in Sedentary Women. Med Sci Sports Exerc 2013; 45:1878-86. [DOI: 10.1249/mss.0b013e3182936261] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|