1
|
Chen J, Li W, Yu L, Zhang B, Li Z, Zou P, Ding B, Dai X, Wang Q. Combined Effects of Ketogenic Diet and Aerobic Exercise on Skeletal Muscle Fiber Remodeling and Metabolic Adaptation in Simulated Microgravity Mice. Metabolites 2025; 15:270. [PMID: 40278399 PMCID: PMC12029359 DOI: 10.3390/metabo15040270] [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: 03/13/2025] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Objective: Prolonged microgravity environments impair skeletal muscle homeostasis by triggering fiber-type transitions and metabolic dysregulation. Although exercise and nutritional interventions may alleviate disuse atrophy, their synergistic effects under microgravity conditions remain poorly characterized. This study investigated the effects of an 8-week ketogenic diet combined with aerobic exercise in hindlimb-unloaded mice on muscle fiber remodeling and metabolic adaptation. Methods: Seven-week-old male C57BL/6J mice were randomly divided into six groups: normal diet control (NC), normal diet with hindlimb unloading (NH), normal diet with hindlimb unloading and exercise (NHE), ketogenic diet control (KC), ketogenic diet with hindlimb unloading (KH), and ketogenic diet with hindlimb unloading and exercise (KHE). During the last two weeks of intervention, hindlimb unloading was applied to simulate microgravity. Aerobic exercise groups performed moderate-intensity treadmill running (12 m/min, 60 min/day, and 6 days/week) for 8 weeks. Body weight, blood ketone, and glucose levels were measured weekly. Post-intervention assessments included the respiratory exchange ratio (RER), exhaustive exercise performance tests, and biochemical analyses of blood metabolic parameters. The skeletal muscle fiber-type composition was evaluated via immunofluorescence staining, lipid deposition was assessed using Oil Red O staining, glycogen content was analyzed by Periodic Acid-Schiff (PAS) staining, and gene expression was quantified using quantitative real-time PCR (RT-qPCR). Results: Hindlimb unloading significantly decreased body weight, induced muscle atrophy, and reduced exercise endurance in mice. However, the combination of KD and aerobic exercise significantly attenuated these adverse effects, as evidenced by increased proportions of oxidative muscle fibers (MyHC-I) and decreased proportions of glycolytic fibers (MyHC-IIb). Additionally, this combined intervention upregulated the expression of lipid metabolism-associated genes, including CPT-1b, HADH, PGC-1α, and FGF21, enhancing lipid metabolism and ketone utilization. These metabolic adaptations corresponded with improved exercise performance, demonstrated by the increased time to exhaustion in the KHE group compared to other hindlimb unloading groups. Conclusions: The combination of a ketogenic diet and aerobic exercise effectively ameliorates simulated microgravity-induced skeletal muscle atrophy and endurance impairment, primarily by promoting a fiber-type transition from MyHC-IIb to MyHC-I and enhancing lipid metabolism gene expression (CPT-1b, HADH, and PGC-1α). These findings underscore the potential therapeutic value of combined dietary and exercise interventions for mitigating muscle atrophy under simulated microgravity conditions.
Collapse
Affiliation(s)
- Jun Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China;
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
| | - Wenjiong Li
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Liang Yu
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China; (L.Y.); (B.Z.)
| | - Bowei Zhang
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China; (L.Y.); (B.Z.)
| | - Zhili Li
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Peng Zou
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Bai Ding
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Xiaoqian Dai
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Qirong Wang
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
| |
Collapse
|
2
|
Rodríguez-Rodríguez R, Baena M, Zagmutt S, Paraiso WK, Reguera AC, Fadó R, Casals N. International Union of Basic and Clinical Pharmacology: Fundamental insights and clinical relevance regarding the carnitine palmitoyltransferase family of enzymes. Pharmacol Rev 2025; 77:100051. [PMID: 40106976 DOI: 10.1016/j.pharmr.2025.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
The carnitine palmitoyltransferases (CPTs) play a key role in controlling the oxidation of long-chain fatty acids and are potential therapeutic targets for diseases with a strong metabolic component, such as obesity, diabetes, and cancer. Four distinct proteins are CPT1A, CPT1B, CPT1C, and CPT2, differing in tissue expression and catalytic activity. CPT1s are finely regulated by malonyl-CoA, a metabolite whose intracellular levels reflect the cell's nutritional state. Although CPT1C does not exhibit significant catalytic activity, it is capable of modulating the functioning of other neuronal proteins. Structurally, all CPTs share a Y-shaped catalytic tunnel that allows the entry of 2 substrates and accommodation of the acyl group in a hydrophobic pocket. Several molecules targeting these enzymes have been described, some showing potential in normalizing blood glucose levels in diabetic patients, and others that, through a central mechanism, are anorexigenic and enhance energy expenditure. However, given the critical roles that CPTs play in certain tissues, such as the heart, liver, and brain, it is essential to fully understand the differences between the various isoforms. We analyze in detail the structure of these proteins, their cellular and physiological functions, and their potential as therapeutic targets in diseases such as obesity, diabetes, and cancer. We also describe drugs identified to date as having inhibitory or activating capabilities for these proteins. This knowledge will support the design of new drugs specific to each isoform, and the development of nanomedicines that can selectively target particular tissues or cells. SIGNIFICANCE STATEMENT: Carnitine palmitoyltransferase (CPT) proteins, as gatekeepers of fatty acid oxidation, have great potential as pharmacological targets to treat metabolic diseases like obesity, diabetes, and cancer. In recent years, significant progress has been made in understanding the 3-dimensional structure of CPTs and their pathophysiological functions. A deeper understanding of the differences between the various CPT family members will enable the design of selective drugs and therapeutic approaches with fewer side effects.
Collapse
Affiliation(s)
- Rosalía Rodríguez-Rodríguez
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Miguel Baena
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Sebastián Zagmutt
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - West Kristian Paraiso
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Ana Cristina Reguera
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Rut Fadó
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Núria Casals
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Choi J, Smith DM, Scafidi S, Riddle RC, Wolfgang MJ. Carnitine palmitoyltransferase 1 facilitates fatty acid oxidation in a non-cell-autonomous manner. Cell Rep 2024; 43:115006. [PMID: 39671290 PMCID: PMC11726389 DOI: 10.1016/j.celrep.2024.115006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/18/2024] [Accepted: 11/06/2024] [Indexed: 12/15/2024] Open
Abstract
Mitochondrial fatty acid oxidation is facilitated by the combined activities of carnitine palmitoyltransferase 1 (Cpt1) and Cpt2, which generate and utilize acylcarnitines, respectively. We compare the response of mice with liver-specific deficiencies in the liver-enriched Cpt1a or the ubiquitously expressed Cpt2 and discover that they display unique metabolic, physiological, and molecular phenotypes. The loss of Cpt1a or Cpt2 results in the induction of the muscle-enriched isoenzyme Cpt1b in hepatocytes in a Pparα-dependent manner. However, hepatic Cpt1b does not contribute substantively to hepatic fatty acid oxidation when Cpt1a is absent. Liver-specific double knockout of Cpt1a and Cpt1b or Cpt2 eliminates the mitochondrial oxidation of non-esterified fatty acids. However, Cpt1a/Cpt1b double knockout mice retain fatty acid oxidation by utilizing extracellular long-chain acylcarnitines that are dependent on Cpt2. These data demonstrate the non-cell-autonomous intercellular metabolism of fatty acids in hepatocytes.
Collapse
Affiliation(s)
- Joseph Choi
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Danielle M Smith
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ryan C Riddle
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA; Research and Development Service, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Michael J Wolfgang
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Cheng X, Ju J, Huang W, Duan Z, Han Y. cpt1b Regulates Cardiomyocyte Proliferation Through Modulation of Glutamine Synthetase in Zebrafish. J Cardiovasc Dev Dis 2024; 11:344. [PMID: 39590187 PMCID: PMC11594654 DOI: 10.3390/jcdd11110344] [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: 08/13/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 11/28/2024] Open
Abstract
Carnitine palmitoyltransferase 1b (Cpt1b) is a crucial rate-limiting enzyme in fatty acid metabolism, but its role and mechanism in early cardiac development remains unclear. Here, we show that cpt1b regulates cardiomyocyte proliferation during zebrafish development. Knocking out entire cpt1b coding sequences leads to impaired cardiomyocyte proliferation, while cardiomyocyte-specific overexpression of cpt1b promotes cardiomyocyte proliferation. RNA sequencing analysis and pharmacological studies identified glutamine synthetase as a key downstream effector of cpt1b in regulating cardiomyocyte proliferation. Our study elucidates a novel mechanism whereby cpt1b promotes zebrafish cardiomyocyte proliferation through glutamine synthetase, which provides new perspectives on the significance of fatty acid metabolism in heart development and the interplay between fatty acid and amino acid metabolic pathways.
Collapse
Affiliation(s)
| | | | | | | | - Yanchao Han
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou 215000, China
| |
Collapse
|
5
|
Zhao Y, Lv H, Yu C, Liang J, Yu H, Du Z, Zhang R. Systemic inhibition of mitochondrial fatty acid β-oxidation impedes zebrafish ventricle regeneration. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167442. [PMID: 39059593 DOI: 10.1016/j.bbadis.2024.167442] [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: 11/29/2023] [Revised: 07/07/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Unlike humans and other mammals, zebrafish demonstrate a remarkable capacity to regenerate their injured hearts throughout life. Mitochondrial fatty acid β-oxidation (FAO) contributes to major energy demands of the adult hearts under physiological conditions; however, its functions in regulating cardiac regeneration and the underlying mechanisms are not completely understood. Different strategies targeting FAO have yield mixed outcomes. Here, we demonstrated that pharmacological inhibition of mitochondrial FAO with mildronate (MD) caused lipid accumulation in zebrafish larvae and suppressed ventricle regeneration. MD treatment impeded cardiogenic factor reactivation and cardiomyocyte (CM) proliferation, and impaired ventricle regeneration could be rescued by exogenous l-carnitine supplementation. Moreover, compared with the ablated hearts of wild-type fish, ventricle regeneration, cardiogenic factor reactivation and CM proliferation were significantly blocked in the ablated hearts of carnitine palmitoyltransferase-1b (cpt1b) knockout zebrafish. Further experiments suggested that NF-κB signaling and increased inflammation may be involved in the impediment of ventricle regeneration caused by systemic mitochondrial FAO inhibition. Overall, our study demonstrates the essential roles of mitochondrial FAO in zebrafish ventricle regeneration and reaffirms the sophisticated and multifaceted roles of FAO in heart regeneration with regard to different injury models and means of FAO inhibition.
Collapse
Affiliation(s)
- Yan Zhao
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Hongbo Lv
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Chunxiao Yu
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Jieling Liang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Hong Yu
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Zhenyu Du
- School of Life Sciences, East China Normal University, Shanghai, China.
| | - Ruilin Zhang
- TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| |
Collapse
|
6
|
Babcock SJ, Houten SM, Gillingham MB. A review of fatty acid oxidation disorder mouse models. Mol Genet Metab 2024; 142:108351. [PMID: 38430613 PMCID: PMC11073919 DOI: 10.1016/j.ymgme.2024.108351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024]
Abstract
Fatty acid oxidation disorders (FAODs) are a family of rare, genetic disorders that affect any part of the fatty acid oxidation pathway. Patients present with severe phenotypes, such as hypoketotic hypoglycemia, cardiomyopathy, and rhabdomyolysis, and currently manage these symptoms by the avoidance of fasting and maintaining a low-fat, high-carbohydrate diet. Because knowledge about FAODs is limited due to the small number of patients, rodent models have been crucial in learning more about these disorders, particularly in studying the molecular mechanisms involved in different phenotypes and in evaluating treatments for patients. The purpose of this review is to present the different FAOD mouse models and highlight the benefits and limitations of using these models. Specifically, we discuss the phenotypes of the available FAOD mouse models, the potential molecular causes of prominent FAOD phenotypes that have been studied using FAOD mouse models, and how FAOD mouse models have been used to evaluate treatments for patients.
Collapse
Affiliation(s)
- Shannon J Babcock
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
| | - Sander M Houten
- Deparment of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
7
|
Mishra A, Tavasoli M, Sokolenko S, McMaster CR, Pasumarthi KB. Atrial natriuretic peptide signaling co-regulates lipid metabolism and ventricular conduction system gene expression in the embryonic heart. iScience 2024; 27:108748. [PMID: 38235330 PMCID: PMC10792247 DOI: 10.1016/j.isci.2023.108748] [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: 05/15/2023] [Revised: 09/15/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024] Open
Abstract
It has been shown that atrial natriuretic peptide (ANP) and its high affinity receptor (NPRA) are involved in the formation of ventricular conduction system (VCS). Inherited genetic variants in fatty acid oxidation (FAO) genes are known to cause conduction abnormalities in newborn children. Although the effect of ANP on energy metabolism in noncardiac cell types is well documented, the role of lipid metabolism in VCS cell differentiation via ANP/NPRA signaling is not known. In this study, histological sections and primary cultures obtained from E11.5 mouse ventricles were analyzed to determine the role of metabolic adaptations in VCS cell fate determination and maturation. Exogenous treatment of E11.5 ventricular cells with ANP revealed a significant increase in lipid droplet accumulation, FAO and higher expression of VCS marker Cx40. Using specific inhibitors, we further identified PPARγ and FAO as critical downstream regulators of ANP-mediated regulation of metabolism and VCS formation.
Collapse
Affiliation(s)
- Abhishek Mishra
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Mahtab Tavasoli
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Stanislav Sokolenko
- Department of Process Engineering and Applied Science, Dalhousie University, Halifax, NS, Canada
| | | | | |
Collapse
|
8
|
Feng C, Bai H, Chang X, Wu Z, Dong W, Ma Q, Yang J. Aflatoxin B1-induced early developmental hepatotoxicity in larvae zebrafish. CHEMOSPHERE 2023; 340:139940. [PMID: 37634582 DOI: 10.1016/j.chemosphere.2023.139940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/31/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Aflatoxin B1 (AFB1) is a ubiquitous mycotoxin that causes oxidative damage in various organs. At present, the research studies on AFB1 are primarily focused on its effects on the terrestrial environment and animals. However, its toxicity mechanism in aquatic environments and aquatic animals has not been largely explored. Thus, in this study, zebrafish was used as a model to study the toxicity mechanism of AFB1 on the liver of developing larvae. The results showed that AFB1 exposure inhibited liver development and promoted fat accumulation in the liver. Transcriptome sequencing analysis showed that AFB1 affected liver redox metabolism and oxidoreductase activity. KEGG analysis showed that AFB1 inhibited the expression of gsto1, gpx4a, mgst3a, and idh1 in the glutathione metabolizing enzyme gene pathway, resulting in hepatic oxidative stress. At the same time, AFB1 also inhibited the expression of acox1, acsl1b, pparα, fabp2, and cpt1 genes in peroxidase and PPAR metabolic pathways, inducing hepatic steatosis and lipid droplet accumulation. Antioxidant N-Acetyl-l-cysteine (NAC) preconditioning up-regulated gsto1, gpx4a and idh1 genes, and improved the AFB1-induced lipid droplet accumulation in the liver. In summary, AFB1 induced hepatic oxidative stress and steatosis, resulting in abnormal liver fat metabolism and accumulation of cellular lipid droplets. NAC could be used as a potential preventative drug to improve AFB1-induced fat accumulation.
Collapse
Affiliation(s)
- Chi Feng
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology,Tongliao,Inner Mongolia, 028000, China; Department of Chemistry and Chemical Engineering, Beijing Forestry University, Beijing, 100083, China
| | - Hongxia Bai
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology,Tongliao,Inner Mongolia, 028000, China; Inner Mongolia Minzu Univ, Coll Anim Sci & Technol, Tongliao,Inner Mongolia, 028000, China
| | - Xu Chang
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology,Tongliao,Inner Mongolia, 028000, China; Inner Mongolia Minzu Univ, Coll Anim Sci & Technol, Tongliao,Inner Mongolia, 028000, China
| | - Zhixuan Wu
- Inner Mongolia Minzu Univ, Coll Anim Sci & Technol, Tongliao,Inner Mongolia, 028000, China
| | - Wu Dong
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology,Tongliao,Inner Mongolia, 028000, China; Inner Mongolia Minzu Univ, Coll Anim Sci & Technol, Tongliao,Inner Mongolia, 028000, China
| | - Qianqian Ma
- Inner Mongolia Minzu Univ, Inst Pharmaceut Chem & Pharmacol, Tongliao, Inner Mongolia, 028000, China
| | - Jingfeng Yang
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology,Tongliao,Inner Mongolia, 028000, China; Inner Mongolia Minzu Univ, Coll Anim Sci & Technol, Tongliao,Inner Mongolia, 028000, China.
| |
Collapse
|
9
|
Li X, Wu F, Günther S, Looso M, Kuenne C, Zhang T, Wiesnet M, Klatt S, Zukunft S, Fleming I, Poschet G, Wietelmann A, Atzberger A, Potente M, Yuan X, Braun T. Inhibition of fatty acid oxidation enables heart regeneration in adult mice. Nature 2023; 622:619-626. [PMID: 37758950 PMCID: PMC10584682 DOI: 10.1038/s41586-023-06585-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
Postnatal maturation of cardiomyocytes is characterized by a metabolic switch from glycolysis to fatty acid oxidation, chromatin reconfiguration and exit from the cell cycle, instating a barrier for adult heart regeneration1,2. Here, to explore whether metabolic reprogramming can overcome this barrier and enable heart regeneration, we abrogate fatty acid oxidation in cardiomyocytes by inactivation of Cpt1b. We find that disablement of fatty acid oxidation in cardiomyocytes improves resistance to hypoxia and stimulates cardiomyocyte proliferation, allowing heart regeneration after ischaemia-reperfusion injury. Metabolic studies reveal profound changes in energy metabolism and accumulation of α-ketoglutarate in Cpt1b-mutant cardiomyocytes, leading to activation of the α-ketoglutarate-dependent lysine demethylase KDM5 (ref. 3). Activated KDM5 demethylates broad H3K4me3 domains in genes that drive cardiomyocyte maturation, lowering their transcription levels and shifting cardiomyocytes into a less mature state, thereby promoting proliferation. We conclude that metabolic maturation shapes the epigenetic landscape of cardiomyocytes, creating a roadblock for further cell divisions. Reversal of this process allows repair of damaged hearts.
Collapse
Affiliation(s)
- Xiang Li
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Fan Wu
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Günther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mario Looso
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Carsten Kuenne
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ting Zhang
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Marion Wiesnet
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stephan Klatt
- Institute for Vascular Signaling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Sven Zukunft
- Institute for Vascular Signaling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signaling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Astrid Wietelmann
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ann Atzberger
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Michael Potente
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Max Delbrück Center for Molecular Medicine, Helmholtz Association of German Research Centres, Berlin, Germany
- Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Xuejun Yuan
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Instituto de Investigacion en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Instituto de Investigacion en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Chang YC, Lee HL, Yang W, Hsieh ML, Liu CC, Lee TY, Huang JY, Nong JY, Li FA, Chuang HL, Ding ZZ, Su WL, Chueh LY, Tsai YT, Chen CH, Mochly-Rosen D, Chuang LM. A common East-Asian ALDH2 mutation causes metabolic disorders and the therapeutic effect of ALDH2 activators. Nat Commun 2023; 14:5971. [PMID: 37749090 PMCID: PMC10520061 DOI: 10.1038/s41467-023-41570-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 09/11/2023] [Indexed: 09/27/2023] Open
Abstract
Obesity and type 2 diabetes have reached pandemic proportion. ALDH2 (acetaldehyde dehydrogenase 2, mitochondrial) is the key metabolizing enzyme of acetaldehyde and other toxic aldehydes, such as 4-hydroxynonenal. A missense Glu504Lys mutation of the ALDH2 gene is prevalent in 560 million East Asians, resulting in reduced ALDH2 enzymatic activity. We find that male Aldh2 knock-in mice mimicking human Glu504Lys mutation were prone to develop diet-induced obesity, glucose intolerance, insulin resistance, and fatty liver due to reduced adaptive thermogenesis and energy expenditure. We find reduced activity of ALDH2 of the brown adipose tissue from the male Aldh2 homozygous knock-in mice. Proteomic analyses of the brown adipose tissue from the male Aldh2 knock-in mice identifies increased 4-hydroxynonenal-adducted proteins involved in mitochondrial fatty acid oxidation and electron transport chain, leading to markedly decreased fatty acid oxidation rate and mitochondrial respiration of brown adipose tissue, which is essential for adaptive thermogenesis and energy expenditure. AD-9308 is a water-soluble, potent, and highly selective ALDH2 activator. AD-9308 treatment ameliorates diet-induced obesity and fatty liver, and improves glucose homeostasis in both male Aldh2 wild-type and knock-in mice. Our data highlight the therapeutic potential of reducing toxic aldehyde levels by activating ALDH2 for metabolic diseases.
Collapse
Affiliation(s)
- Yi-Cheng Chang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsiao-Lin Lee
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wenjin Yang
- Foresee Pharmaceuticals, Co.Ltd, Taipei, Taiwan
| | - Meng-Lun Hsieh
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Cai-Cin Liu
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
| | - Tung-Yuan Lee
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
| | - Jing-Yong Huang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jiun-Yi Nong
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Fu-An Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | - Zhi-Zhong Ding
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
| | - Wei-Lun Su
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
| | - Li-Yun Chueh
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
| | - Yi-Ting Tsai
- Laboratory Animal Center, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Lee-Ming Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Graduate Institute of Molecular Medicine, National Taiwan University, Taipei, Taiwan.
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
11
|
Pizzato HA, Wang Y, Wolfgang MJ, Finck BN, Patti GJ, Bhattacharya D. Mitochondrial pyruvate metabolism and glutaminolysis toggle steady-state and emergency myelopoiesis. J Exp Med 2023; 220:e20221373. [PMID: 37249600 PMCID: PMC10227646 DOI: 10.1084/jem.20221373] [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: 08/10/2022] [Revised: 03/23/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
To define the metabolic requirements of hematopoiesis, we examined blood lineages in mice conditionally deficient in genes required for long-chain fatty acid oxidation (Cpt2), glutaminolysis (Gls), or mitochondrial pyruvate import (Mpc2). Genetic ablation of Cpt2 or Gls minimally impacted most blood lineages. In contrast, deletion of Mpc2 led to a sharp decline in mature myeloid cells and a slower reduction in T cells, whereas other hematopoietic lineages were unaffected. Yet MPC2-deficient monocytes and neutrophils rapidly recovered due to a transient and specific increase in myeloid progenitor proliferation. Competitive bone marrow chimera and stable isotope tracing experiments demonstrated that this proliferative burst was progenitor intrinsic and accompanied by a metabolic switch to glutaminolysis. Myeloid recovery after loss of MPC2 or cyclophosphamide treatment was delayed in the absence of GLS. Reciprocally, MPC2 was not required for myeloid recovery after cyclophosphamide treatment. Thus, mitochondrial pyruvate metabolism maintains myelopoiesis under steady-state conditions, while glutaminolysis in progenitors promotes emergency myelopoiesis.
Collapse
Affiliation(s)
- Hannah A. Pizzato
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Yahui Wang
- Department of Chemistry, Washington University, Saint Louis, MO, USA
| | - Michael J. Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian N. Finck
- Division of Geriatrics and Nutritional Sciences, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Gary J. Patti
- Department of Chemistry, Washington University, Saint Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- Siteman Cancer Center, Washington University, Saint Louis, MO, USA
| | - Deepta Bhattacharya
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
12
|
Vianey-Saban C, Guffon N, Fouilhoux A, Acquaviva C. Fifty years of research on mitochondrial fatty acid oxidation disorders: The remaining challenges. J Inherit Metab Dis 2023; 46:848-873. [PMID: 37530674 DOI: 10.1002/jimd.12664] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023]
Abstract
Since the identification of the first disorder of mitochondrial fatty acid oxidation defects (FAOD) in 1973, more than 20 defects have been identified. Although there are some differences, most FAOD have similar clinical signs, which are mainly due to energy depletion and toxicity of accumulated metabolites. However, some of them have an unusual clinical phenotype or specific clinical signs. This manuscript focuses on what we have learnt so far on the pathophysiology of these disorders, which present with clinical signs that are not typical of categorical FAOD. It also highlights that some disorders have not yet been identified and tries to make assumptions to explain why. It also deals with new treatments under consideration in FAOD, including triheptanoin and similar anaplerotic substrates, ketone body treatments, RNA and gene therapy approaches. Finally, it suggests challenges for the diagnosis of FAOD in the coming years, both for symptomatic patients and for those diagnosed through newborn screening. The ultimate goal would be to identify all the patients born with FAOD and ensure for them the best possible quality of life.
Collapse
Affiliation(s)
- Christine Vianey-Saban
- Biochemical and Molecular Biology Laboratory, Metabolic Inborn Errors of Metabolism Unit, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Nathalie Guffon
- National Reference Centre for Hereditary Metabolic Diseases, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Alain Fouilhoux
- National Reference Centre for Hereditary Metabolic Diseases, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Cécile Acquaviva
- Biochemical and Molecular Biology Laboratory, Metabolic Inborn Errors of Metabolism Unit, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| |
Collapse
|
13
|
Gaston G, Babcock S, Ryals R, Elizondo G, DeVine T, Wafai D, Packwood W, Holden S, Raber J, Lindner JR, Pennesi ME, Harding CO, Gillingham MB. A G1528C Hadha knock-in mouse model recapitulates aspects of human clinical phenotypes for long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency. Commun Biol 2023; 6:890. [PMID: 37644104 PMCID: PMC10465608 DOI: 10.1038/s42003-023-05268-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Long chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) is a fatty acid oxidation disorder (FAOD) caused by a pathogenic variant, c.1528 G > C, in HADHA encoding the alpha subunit of trifunctional protein (TFPα). Individuals with LCHADD develop chorioretinopathy and peripheral neuropathy not observed in other FAODs in addition to the more ubiquitous symptoms of hypoketotic hypoglycemia, rhabdomyolysis and cardiomyopathy. We report a CRISPR/Cas9 generated knock-in murine model of G1528C in Hadha that recapitulates aspects of the human LCHADD phenotype. Homozygous pups are less numerous than expected from Mendelian probability, but survivors exhibit similar viability with wildtype (WT) littermates. Tissues of LCHADD homozygotes express TFPα protein, but LCHADD mice oxidize less fat and accumulate plasma 3-hydroxyacylcarnitines compared to WT mice. LCHADD mice exhibit lower ketones with fasting, exhaust earlier during treadmill exercise and develop a dilated cardiomyopathy compared to WT mice. In addition, LCHADD mice exhibit decreased visual performance, decreased cone function, and disruption of retinal pigment epithelium. Neurological function is affected, with impaired motor function during wire hang test and reduced open field activity. The G1528C knock-in mouse exhibits a phenotype similar to that observed in human patients; this model will be useful to explore pathophysiology and treatments for LCHADD in the future.
Collapse
Affiliation(s)
- Garen Gaston
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Shannon Babcock
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Renee Ryals
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - Gabriela Elizondo
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Tiffany DeVine
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Dahlia Wafai
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Sarah Holden
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
| | - Jacob Raber
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
- Departments of Neurology and Radiation Medicine, Oregon Health and Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center (ONPRC), Oregon Health and Science University, Portland, OR, USA
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Cardiovascular Division, University of Virginia Medical Center, Charlottesville, VA, USA
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - Cary O Harding
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
14
|
Zhang C, Lu LF, Li ZC, Han KJ, Wang XL, Chen DD, Xiong F, Li XY, Zhou L, Ge F, Li S. Zebrafish MAP2K7 Simultaneously Enhances Host IRF7 Stability and Degrades Spring Viremia of Carp Virus P Protein via Ubiquitination Pathway. J Virol 2023; 97:e0053223. [PMID: 37367226 PMCID: PMC10373533 DOI: 10.1128/jvi.00532-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/11/2023] [Indexed: 06/28/2023] Open
Abstract
During viral infection, host defensive proteins either enhance the host immune response or antagonize viral components directly. In this study, we report on the following two mechanisms employed by zebrafish mitogen-activated protein kinase kinase 7 (MAP2K7) to protect the host during spring viremia of carp virus (SVCV) infection: stabilization of host IRF7 and degradation of SVCV P protein. In vivo, map2k7+/- (map2k7-/- is a lethal mutation) zebrafish showed a higher lethality, more pronounced tissue damage, and more viral proteins in major immune organs than the controls. At the cellular level, overexpression of map2k7 significantly enhanced host cell antiviral capacity, and viral replication and proliferation were significantly suppressed. Additionally, MAP2K7 interacted with the C terminus of IRF7 and stabilized IRF7 by increasing K63-linked polyubiquitination. On the other hand, during MAP2K7 overexpression, SVCV P proteins were significantly decreased. Further analysis demonstrated that SVCV P protein was degraded by the ubiquitin-proteasome pathway, as the attenuation of K63-linked polyubiquitination was mediated by MAP2K7. Furthermore, the deubiquitinase USP7 was indispensable in P protein degradation. These results confirm the dual functions of MAP2K7 during viral infection. IMPORTANCE Normally, during viral infection, host antiviral factors individually modulate the host immune response or antagonize viral components to defense infection. In the present study, we report that zebrafish MAP2K7 plays a crucial positive role in the host antiviral process. According to the weaker antiviral capacity of map2k7+/- zebrafish than that of the control, we find that MAP2K7 reduces host lethality through two pathways, as follows: enhancing K63-linked polyubiquitination to promote host IRF7 stability and attenuating K63-mediated polyubiquitination to degrade the SVCV P protein. These two mechanisms of MAP2K7 reveal a special antiviral response in lower vertebrates.
Collapse
Affiliation(s)
- Can Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Long-Feng Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhuo-Cong Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ke-Jia Han
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Xue-Li Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Dan-Dan Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Feng Xiong
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xi-Yin Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Li Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Feng Ge
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shun Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
15
|
Berkay EG, Şoroğlu CV, Kalaycı T, Uyguner ZO, Akçapınar GB, Başaran S. A new enrichment approach for candidate gene detection in unexplained recurrent pregnancy loss and implantation failure. Mol Genet Genomics 2023; 298:253-272. [PMID: 36385415 DOI: 10.1007/s00438-022-01972-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022]
Abstract
Recurrent pregnancy loss (RPL) and implantation failure (RIF) are obstacles to livebirth and multifactorial conditions in which nearly half of the cases remain unexplained, and we aimed to identify maternal candidate gene variants and pathways for RPL and RIF by analyzing whole-exome sequencing (WES) data via a new detailed bioinformatics approach. A retrospective cohort study was applied to 35 women with normal chromosomal configuration diagnosed with unexplained RPL and/or RIF. WES and comprehensive bioinformatics analyses were performed. Published gene expression datasets (n = 46) were investigated for candidate genes. Variant effects on protein structure were analyzed for 12 proteins, and BUB1B was visualized in silico. WES and bioinformatics analyses are effective and applicable for studying URPL and RIF to detect mutations, as we suggest new candidates to explain the etiology. Forty-three variants in 39 genes were detected in 29 women, 7 of them contributing to oligogenic inheritance. These genes were related to implantation, placentation, coagulation, metabolism, immune system, embryological development, cell cycle-associated processes, and ovarian functions. WES, genomic variant analyses, expression data, and protein configuration studies offer new and promising ways to investigate the etiology of URPL and RIF. Discovering etiology-identifying genetic factors can help manage couples' needs and develop personalized therapies and new pharmaceutical products in the future. The classical approach with chromosomal analysis and targeted gene panel testing is insufficient in these cases; the exome data provide a promising way to detect and understand the possible clinical effects of the variant and its alteration on protein structure.
Collapse
Affiliation(s)
- Ezgi Gizem Berkay
- Department of Medical Genetics, Istanbul Medical Faculty, Istanbul University, 34093, Istanbul, Turkey. .,Department of Basic Sciences, Dentistry Faculty, Istanbul Kent University, 34433, Istanbul, Turkey.
| | - Can Veysel Şoroğlu
- Department of Medical Biotechnology, Institute of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, 34684, Istanbul, Turkey
| | - Tuğba Kalaycı
- Division of Medical Genetics, Department of Internal Medicine, Istanbul Medical Faculty, Istanbul University, 34093, Istanbul, Turkey
| | - Zehra Oya Uyguner
- Department of Medical Genetics, Istanbul Medical Faculty, Istanbul University, 34093, Istanbul, Turkey
| | - Günseli Bayram Akçapınar
- Department of Medical Biotechnology, Institute of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, 34684, Istanbul, Turkey
| | - Seher Başaran
- Department of Medical Genetics, Istanbul Medical Faculty, Istanbul University, 34093, Istanbul, Turkey
| |
Collapse
|
16
|
Wu Z, Rao S, Li J, Ding N, Chen J, Feng L, Ma S, Hu C, Dai H, Wen L, Jiang Q, Deng J, Deng M, Tan C. Dietary adenosine 5’-monophosphate supplementation increases food intake and remodels energy expenditure in mice. Food Nutr Res 2022; 66:7680. [PMID: 35844957 PMCID: PMC9250134 DOI: 10.29219/fnr.v66.7680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/30/2021] [Accepted: 10/04/2021] [Indexed: 11/20/2022] Open
Abstract
Background Methods Results Conclusions
Collapse
Affiliation(s)
- Zifang Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Sujuan Rao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jiaying Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ning Ding
- Guangzhou Customs Technology Center, 510623, China
| | - Jianzhao Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Li Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shuo Ma
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Chengjun Hu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Haonan Dai
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Lijun Wen
- Guangdong Hinabiotech Co., Ltd., Guangzhou, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Ming Deng,
| | - Ming Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Ming Deng,
| | - Chengquan Tan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
- Chengquan Tan, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
17
|
Yoshida Y, Shimizu I, Hsiao YT, Suda M, Katsuumi G, Seki M, Suzuki Y, Okuda S, Soga T, Minamino T. Differing impact of phosphoglycerate mutase 1-deficiency on brown and white adipose tissue. iScience 2022; 25:104268. [PMID: 35521515 PMCID: PMC9065309 DOI: 10.1016/j.isci.2022.104268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/03/2022] [Accepted: 04/13/2022] [Indexed: 11/27/2022] Open
Abstract
Brown adipose tissue (BAT) is a metabolically active organ that contributes to the thermogenic response to cold exposure. In addition, other thermogenic cells termed beige adipocytes are generated in white adipose tissue (WAT) by cold exposure. Although activation of brown/beige adipose tissue is associated with mobilization of both glucose and lipids, few studies have focused on the role of glycolytic enzymes in regulating adipose tissue function. We generated mouse models with specific deletion of the glycolytic enzyme phosphoglycerate mutase 1 (PGAM1) from adipose tissue. Deletion of Pgam1 from both BAT and WAT promoted whitening of BAT with beiging of visceral WAT, whereas deletion of Pgam1 from BAT alone led to whitening of BAT without beiging of WAT. Our results demonstrate a potential role of glycolytic enzymes in beiging of visceral WAT and suggest that PGAM1 would be a novel therapeutic target in obesity and diabetes. Pgam1 deletion leads to whitening of brown adipose tissue Pgam1 deletion promotes beiging of visceral white adipose tissue (WAT) Pgam1 deletion-induced beiging is associated with increased levels of amino acids
Collapse
Affiliation(s)
- Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.,Department of Advanced Senotherapeutics, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yung-Ting Hsiao
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata 997-0052, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| |
Collapse
|
18
|
Kappen C, Kruger C, Jones S, Salbaum JM. Nutrient Transporter Gene Expression in the Early Conceptus-Implications From Two Mouse Models of Diabetic Pregnancy. Front Cell Dev Biol 2022; 10:777844. [PMID: 35478964 PMCID: PMC9035823 DOI: 10.3389/fcell.2022.777844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/28/2022] [Indexed: 11/29/2022] Open
Abstract
Maternal diabetes in early pregnancy increases the risk for birth defects in the offspring, particularly heart, and neural tube defects. While elevated glucose levels are characteristic for diabetic pregnancies, these are also accompanied by hyperlipidemia, indicating altered nutrient availability. We therefore investigated whether changes in the expression of nutrient transporters at the conception site or in the early post-implantation embryo could account for increased birth defect incidence at later developmental stages. Focusing on glucose and fatty acid transporters, we measured their expression by RT-PCR in the spontaneously diabetic non-obese mouse strain NOD, and in pregnant FVB/N mouse strain dams with Streptozotocin-induced diabetes. Sites of expression in the deciduum, extra-embryonic, and embryonic tissues were determined by RNAscope in situ hybridization. While maternal diabetes had no apparent effects on levels or cellular profiles of expression, we detected striking cell-type specificity of particular nutrient transporters. For examples, Slc2a2/Glut2 expression was restricted to the endodermal cells of the visceral yolk sac, while Slc2a1/Glut1 expression was limited to the mesodermal compartment; Slc27a4/Fatp4 and Slc27a3/Fatp3 also exhibited reciprocally exclusive expression in the endodermal and mesodermal compartments of the yolk sac, respectively. These findings not only highlight the significance of nutrient transporters in the intrauterine environment, but also raise important implications for the etiology of birth defects in diabetic pregnancies, and for strategies aimed at reducing birth defects risk by nutrient supplementation.
Collapse
Affiliation(s)
- Claudia Kappen
- Department of Developmental Biology, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Claudia Kruger
- Department of Developmental Biology, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Sydney Jones
- Regulation of Gene Expression, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - J. Michael Salbaum
- Regulation of Gene Expression, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| |
Collapse
|
19
|
Ehrlich KC, Deng HW, Ehrlich M. Epigenetics of Mitochondria-Associated Genes in Striated Muscle. EPIGENOMES 2021; 6:1. [PMID: 35076500 PMCID: PMC8788487 DOI: 10.3390/epigenomes6010001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/04/2021] [Accepted: 12/16/2021] [Indexed: 11/16/2022] Open
Abstract
Striated muscle has especially large energy demands. We identified 97 genes preferentially expressed in skeletal muscle and heart, but not in aorta, and found significant enrichment for mitochondrial associations among them. We compared the epigenomic and transcriptomic profiles of the 27 genes associated with striated muscle and mitochondria. Many showed strong correlations between their tissue-specific transcription levels, and their tissue-specific promoter, enhancer, or open chromatin as well as their DNA hypomethylation. Their striated muscle-specific enhancer chromatin was inside, upstream, or downstream of the gene, throughout much of the gene as a super-enhancer (CKMT2, SLC25A4, and ACO2), or even overlapping a neighboring gene (COX6A2, COX7A1, and COQ10A). Surprisingly, the 3' end of the 1.38 Mb PRKN (PARK2) gene (involved in mitophagy and linked to juvenile Parkinson's disease) displayed skeletal muscle/myoblast-specific enhancer chromatin, a myoblast-specific antisense RNA, as well as brain-specific enhancer chromatin. We also found novel tissue-specific RNAs in brain and embryonic stem cells within PPARGC1A (PGC-1α), which encodes a master transcriptional coregulator for mitochondrial formation and metabolism. The tissue specificity of this gene's four alternative promoters, including a muscle-associated promoter, correlated with nearby enhancer chromatin and open chromatin. Our in-depth epigenetic examination of these genes revealed previously undescribed tissue-specific enhancer chromatin, intragenic promoters, regions of DNA hypomethylation, and intragenic noncoding RNAs that give new insights into transcription control for this medically important set of genes.
Collapse
Affiliation(s)
- Kenneth C. Ehrlich
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (K.C.E.); (H.-W.D.)
| | - Hong-Wen Deng
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (K.C.E.); (H.-W.D.)
| | - Melanie Ehrlich
- Center for Bioinformatics and Genomics, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; (K.C.E.); (H.-W.D.)
- Tulane Cancer Center and Hayward Genetics Center, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
20
|
Wang M, Wang K, Liao X, Hu H, Chen L, Meng L, Gao W, Li Q. Carnitine Palmitoyltransferase System: A New Target for Anti-Inflammatory and Anticancer Therapy? Front Pharmacol 2021; 12:760581. [PMID: 34764874 PMCID: PMC8576433 DOI: 10.3389/fphar.2021.760581] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/17/2021] [Indexed: 11/20/2022] Open
Abstract
Lipid metabolism involves multiple biological processes. As one of the most important lipid metabolic pathways, fatty acid oxidation (FAO) and its key rate-limiting enzyme, the carnitine palmitoyltransferase (CPT) system, regulate host immune responses and thus are of great clinical significance. The effect of the CPT system on different tissues or organs is complex: the deficiency or over-activation of CPT disrupts the immune homeostasis by causing energy metabolism disorder and inflammatory oxidative damage and therefore contributes to the development of various acute and chronic inflammatory disorders and cancer. Accordingly, agonists or antagonists targeting the CPT system may become novel approaches for the treatment of diseases. In this review, we first briefly describe the structure, distribution, and physiological action of the CPT system. We then summarize the pathophysiological role of the CPT system in chronic obstructive pulmonary disease, bronchial asthma, acute lung injury, chronic granulomatous disease, nonalcoholic fatty liver disease, hepatic ischemia–reperfusion injury, kidney fibrosis, acute kidney injury, cardiovascular disorders, and cancer. We are also concerned with the current knowledge in either preclinical or clinical studies of various CPT activators/inhibitors for the management of diseases. These compounds range from traditional Chinese medicines to novel nanodevices. Although great efforts have been made in studying the different kinds of CPT agonists/antagonists, only a few pharmaceuticals have been applied for clinical uses. Nevertheless, research on CPT activation or inhibition highlights the pharmacological modulation of CPT-dependent FAO, especially on different CPT isoforms, as a promising anti-inflammatory/antitumor therapeutic strategy for numerous disorders.
Collapse
Affiliation(s)
- Muyun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ximing Liao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiyang Hu
- Department of Vascular Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Liangzhi Chen
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linlin Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Yang Y, Xu X, Wu H, Yang J, Chen J, Morisseau C, Hammock BD, Bettaieb A, Zhao L. Differential Effects of 17,18-EEQ and 19,20-EDP Combined with Soluble Epoxide Hydrolase Inhibitor t-TUCB on Diet-Induced Obesity in Mice. Int J Mol Sci 2021; 22:ijms22158267. [PMID: 34361032 PMCID: PMC8347952 DOI: 10.3390/ijms22158267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022] Open
Abstract
17,18-Epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP) are bioactive epoxides produced from n-3 polyunsaturated fatty acid eicosapentaenoic acid and docosahexaenoic acid, respectively. However, these epoxides are quickly metabolized into less active diols by soluble epoxide hydrolase (sEH). We have previously demonstrated that an sEH inhibitor, t-TUCB, decreased serum triglycerides (TG) and increased lipid metabolic protein expression in the brown adipose tissue (BAT) of diet-induced obese mice. This study investigates the preventive effects of t-TUCB (T) alone or combined with 19,20-EDP (T + EDP) or 17,18-EEQ (T + EEQ) on BAT activation in the development of diet-induced obesity and metabolic disorders via osmotic minipump delivery in mice. Both T + EDP and T + EEQ groups showed significant improvement in fasting glucose, serum triglycerides, and higher core body temperature, whereas heat production was only significantly increased in the T + EEQ group. Moreover, both the T + EDP and T + EEQ groups showed less lipid accumulation in the BAT. Although UCP1 expression was not changed, PGC1α expression was increased in all three treated groups. In contrast, the expression of CPT1A and CPT1B, which are responsible for the rate-limiting step for fatty acid oxidation, was only increased in the T + EDP and T + EEQ groups. Interestingly, as a fatty acid transporter, CD36 expression was only increased in the T + EEQ group. Furthermore, both the T + EDP and T + EEQ groups showed decreased inflammatory NFκB signaling in the BAT. Our results suggest that 17,18-EEQ or 19,20-EDP combined with t-TUCB may prevent high-fat diet-induced metabolic disorders, in part through increased thermogenesis, upregulating lipid metabolic protein expression, and decreasing inflammation in the BAT.
Collapse
Affiliation(s)
- Yang Yang
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA; (Y.Y.); (X.X.); (H.W.); (A.B.)
| | - Xinyun Xu
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA; (Y.Y.); (X.X.); (H.W.); (A.B.)
| | - Haoying Wu
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA; (Y.Y.); (X.X.); (H.W.); (A.B.)
| | - Jun Yang
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA; (J.Y.); (C.M.); (B.D.H.)
| | - Jiangang Chen
- Department of Public Health, University of Tennessee, Knoxville, TN 37996, USA;
| | - Christophe Morisseau
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA; (J.Y.); (C.M.); (B.D.H.)
| | - Bruce D. Hammock
- Department of Entomology and Nematology, and Comprehensive Cancer Center, University of California, Davis, CA 95616, USA; (J.Y.); (C.M.); (B.D.H.)
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA; (Y.Y.); (X.X.); (H.W.); (A.B.)
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN 37996, USA
| | - Ling Zhao
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA; (Y.Y.); (X.X.); (H.W.); (A.B.)
- Correspondence: ; Tel.: +1-865-974-1833
| |
Collapse
|
22
|
Wang J, Xiang H, Lu Y, Wu T, Ji G. The role and therapeutic implication of CPTs in fatty acid oxidation and cancers progression. Am J Cancer Res 2021; 11:2477-2494. [PMID: 34249411 PMCID: PMC8263643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/10/2021] [Indexed: 06/13/2023] Open
Abstract
Cancer cells must maintain metabolic homeostasis under a wide range of conditions and meet their own energy needs in order to survive and reproduce. In addition to glycolysis, cancer cells can also perform various metabolic strategies, such as fatty acid oxidation (FAO). It has been found that the proliferation, survival, drug resistance and metastasis of cancer cells depend on FAO. The carnitine palmitoyltransferase (CPT), including CPT1 and CPT2, located on the mitochondrial membrane, are important mediators of FAO. In recent years, many researchers have found that CPT has a close relationship with the metabolic development of tumor cells, not only provides energy for cancer cells development and metastasis by promoting FAO but also affects the occurrence and invasion through other signal pathways or cytokines or microRNA. This review summarized the role of CPTs in several kinds of tumors and the developed targeted inhibitors of CPTs, as well as the potential gene therapy and immunotherapy of CPTs, hoping to better explore the mechanism and role of CPTs in the future and providing useful ideas for clinical treatment.
Collapse
Affiliation(s)
- Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Yifei Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| |
Collapse
|
23
|
Avram VF, Chamkha I, Åsander-Frostner E, Ehinger JK, Timar RZ, Hansson MJ, Muntean DM, Elmér E. Cell-Permeable Succinate Rescues Mitochondrial Respiration in Cellular Models of Statin Toxicity. Int J Mol Sci 2021; 22:E424. [PMID: 33401621 PMCID: PMC7796258 DOI: 10.3390/ijms22010424] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Statins are the cornerstone of lipid-lowering therapy. Although generally well tolerated, statin-associated muscle symptoms (SAMS) represent the main reason for treatment discontinuation. Mitochondrial dysfunction of complex I has been implicated in the pathophysiology of SAMS. The present study proposed to assess the concentration-dependent ex vivo effects of three statins on mitochondrial respiration in viable human platelets and to investigate whether a cell-permeable prodrug of succinate (complex II substrate) can compensate for statin-induced mitochondrial dysfunction. Mitochondrial respiration was assessed by high-resolution respirometry in human platelets, acutely exposed to statins in the presence/absence of the prodrug NV118. Statins concentration-dependently inhibited mitochondrial respiration in both intact and permeabilized cells. Further, statins caused an increase in non-ATP generating oxygen consumption (uncoupling), severely limiting the OXPHOS coupling efficiency, a measure of the ATP generating capacity. Cerivastatin (commercially withdrawn due to muscle toxicity) displayed a similar inhibitory capacity compared with the widely prescribed and tolerable atorvastatin, but did not elicit direct complex I inhibition. NV118 increased succinate-supported mitochondrial oxygen consumption in atorvastatin/cerivastatin-exposed platelets leading to normalization of coupled (ATP generating) respiration. The results acquired in isolated human platelets were validated in a limited set of experiments using atorvastatin in HepG2 cells, reinforcing the generalizability of the findings.
Collapse
Affiliation(s)
- Vlad F. Avram
- Department of Internal Medicine-Diabetes, Nutrition and Metabolic Diseases, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Romania, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania; (V.F.A.); (R.Z.T.)
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Romania, Spl. Tudor Vladimirescu No. 14, 300173 Timișoara, Romania
| | - Imen Chamkha
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, BMC A13, 221 84 Lund, Sweden; (I.C.); (E.Å.-F.); (J.K.E.); (M.J.H.)
- Abliva AB, Medicon Village, 223 81 Lund, Sweden
| | - Eleonor Åsander-Frostner
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, BMC A13, 221 84 Lund, Sweden; (I.C.); (E.Å.-F.); (J.K.E.); (M.J.H.)
- Abliva AB, Medicon Village, 223 81 Lund, Sweden
| | - Johannes K. Ehinger
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, BMC A13, 221 84 Lund, Sweden; (I.C.); (E.Å.-F.); (J.K.E.); (M.J.H.)
- Abliva AB, Medicon Village, 223 81 Lund, Sweden
| | - Romulus Z. Timar
- Department of Internal Medicine-Diabetes, Nutrition and Metabolic Diseases, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Romania, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania; (V.F.A.); (R.Z.T.)
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Romania, Spl. Tudor Vladimirescu No. 14, 300173 Timișoara, Romania
| | - Magnus J. Hansson
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, BMC A13, 221 84 Lund, Sweden; (I.C.); (E.Å.-F.); (J.K.E.); (M.J.H.)
- Abliva AB, Medicon Village, 223 81 Lund, Sweden
| | - Danina M. Muntean
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Romania, Spl. Tudor Vladimirescu No. 14, 300173 Timișoara, Romania
- Department of Functional Sciences-Pathophysiology, 2Center for Translational Research and Systems Medi-cine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Romania, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Eskil Elmér
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, BMC A13, 221 84 Lund, Sweden; (I.C.); (E.Å.-F.); (J.K.E.); (M.J.H.)
- Abliva AB, Medicon Village, 223 81 Lund, Sweden
| |
Collapse
|
24
|
Chen CP, Huang JP, Chern SR, Wu PS, Chen SW, Wu FT, Chen WL, Lee MS, Wang W. Prenatal diagnosis and molecular cytogenetic characterization of de novo distal 5p deletion and distal 22q duplication. Taiwan J Obstet Gynecol 2020; 59:140-145. [PMID: 32039783 DOI: 10.1016/j.tjog.2019.11.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2019] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE We present prenatal diagnosis and molecular cytogenetic characterization of de novo distal 5p deletion and distal 22q duplication. CASE REPORT A 34-year-old woman was underwent amniocentesis at 17 weeks of gestation because of advanced maternal age. Amniocentesis revealed a derivative chromosome 5 [der(5)] with an abnormal distal 5p segment of unknown origin. The parental karyotypes were normal. Array comparative genomic hybridization (aCGH) analysis was performed on the cultured amniocytes, and the result was arr 5p15.33p13.3 (22,149-29,760,922) × 1.0, arr 22q13.2q13.33 (42, 192, 065-51,178,264) × 3.0 [GRCh37 (hg19)] with a 29.739-Mb deletion of 5p15.33-p13.3 encompassing 55 [Online Mendelian Inheritance in Man (OMIM)] genes including TPPP, TERT, SRD5A1, SEMA5A and CTNND2, and an 8.986-Mb duplication of 22q13.2-q13.33 encompassing 82 OMIM genes including TRMU, SCO2, TYMP, CPT1B and SHANK3. The fetal karyotype was 46,XY,der(5)t(5; 22)(p13.3; q13.2)dn. The pregnancy was subsequently terminated, and a malformed fetus was delivered with facial dysmorphism. Postnatal polymorphic DNA marker analysis confirmed a maternal origin of the aberrant chromosome 5. CONCLUSION aCGH and polymorphic DNA marker analyses can determine the nature and parental origin of the de novo chromosome aberration, and the information acquired is useful for genetic counseling.
Collapse
Affiliation(s)
- Chih-Ping Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Institute of Clinical and Community Health Nursing, National Yang-Ming University, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Jian-Pei Huang
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan; MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Schu-Rern Chern
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | | | - Shin-Wen Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Fang-Tzu Wu
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Wen-Lin Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Meng-Shan Lee
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Wayseen Wang
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; Department of Bioengineering, Tatung University, Taipei, Taiwan
| |
Collapse
|
25
|
Patel BV, Yao F, Howenstine A, Takenaka R, Hyatt JA, Sears KE, Shewchuk BM. Emergent Coordination of the CHKB and CPT1B Genes in Eutherian Mammals: Implications for the Origin of Brown Adipose Tissue. J Mol Biol 2020; 432:6127-6145. [PMID: 33058877 DOI: 10.1016/j.jmb.2020.09.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 11/17/2022]
Abstract
Mitochondrial fatty acid oxidation (FAO) contributes to the proton motive force that drives ATP synthesis in many mammalian tissues. In eutherian (placental) mammals, brown adipose tissue (BAT) can also dissipate this proton gradient through uncoupling protein 1 (UCP1) to generate heat, but the evolutionary events underlying the emergence of BAT are unknown. An essential step in FAO is the transport of cytoplasmic long chain acyl-coenzyme A (acyl-CoA) into the mitochondrial matrix, which requires the action of carnitine palmitoyltransferase 1B (CPT1B) in striated muscle and BAT. In eutherians, the CPT1B gene is closely linked to the choline kinase beta (CHKB) gene, which is transcribed from the same DNA strand and terminates just upstream of CPT1B. CHKB is a rate-limiting enzyme in the synthesis of phosphatidylcholine (PC), a predominant mitochondrial membrane phospholipid, suggesting that the coordinated expression of CHKB and CPT1B may cooperatively enhance mitochondrial FAO. The present findings show that transcription of the eutherian CHKB and CPT1B genes is linked within a unitary epigenetic domain targeted to the CHKB gene, and that that this regulatory linkage appears to have resulted from an intergenic deletion in eutherians that significantly altered the distribution of CHKB and CPT1B expression. Informed by the timing of this event relative to the emergence of BAT, the phylogeny of CHKB-CPT1B synteny, and the insufficiency of UCP1 to account for eutherian BAT, these data support a mechanism for the emergence of BAT based on the acquisition of a novel capacity for adipocyte FAO in a background of extant UCP1.
Collapse
Affiliation(s)
- Bhavin V Patel
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Fanrong Yao
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Aidan Howenstine
- Department of Ecology & Evolutionary Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Risa Takenaka
- Department of Ecology & Evolutionary Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Jacob A Hyatt
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Karen E Sears
- Department of Ecology & Evolutionary Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Brian M Shewchuk
- Department of Biochemistry & Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States.
| |
Collapse
|
26
|
Kim HS, Ren G, Kim T, Bhatnagar S, Yang Q, Bahk YY, Kim JA. Metformin reduces saturated fatty acid-induced lipid accumulation and inflammatory response by restoration of autophagic flux in endothelial cells. Sci Rep 2020; 10:13523. [PMID: 32782332 PMCID: PMC7419289 DOI: 10.1038/s41598-020-70347-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy, an integral part of the waste recycling process, plays an important role in cellular physiology and pathophysiology. Impaired autophagic flux causes ectopic lipid deposition, which is defined as the accumulation of lipids in non-adipose tissue. Ectopic lipid accumulation is observed in patients with cardiometabolic syndrome, including obesity, diabetes, insulin resistance, and cardiovascular complications. Metformin is the first line of treatment for type 2 diabetes, and one of the underlying mechanisms for the anti-diabetic effect of metformin is mediated by the stimulation of AMP-activated protein kinase (AMPK). Because the activation of AMPK is crucial for the initiation of autophagy, we hypothesize that metformin reduces the accumulation of lipid droplets by increasing autophagic flux in vascular endothelial cells. Incubation of vascular endothelial cells with saturated fatty acid (SFA) increased the accumulation of lipid droplets and impaired autophagic flux. We observed that the accumulation of lipid droplets was reduced, and the autophagic flux was enhanced by treatment with metformin. The knock-down of AMPKα by using siRNA blunted the effect of metformin. Furthermore, treatment with SFA or inhibition of autophagy increased leukocyte adhesion, whereas treatment with metformin decreased the SFA-induced leukocyte adhesion. The results suggest a novel mechanism by which metformin protects vascular endothelium from SFA-induced ectopic lipid accumulation and pro-inflammatory responses. In conclusion, improving autophagic flux may be a therapeutic strategy to protect endothelial function from dyslipidemia and diabetic complications.
Collapse
Affiliation(s)
- Hae-Suk Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Guang Ren
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Teayoun Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Sushant Bhatnagar
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA
| | - Qinglin Yang
- Department of Nutrition, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Young Yil Bahk
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, 27478, Republic of Korea
| | - Jeong-A Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL, 35294, USA.
| |
Collapse
|
27
|
Liu D, Pan Y, Li K, Li D, Li P, Gao Z. Proteomics Reveals the Mechanism Underlying the Inhibition of Phytophthora sojae by Propyl Gallate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:8151-8162. [PMID: 32633954 DOI: 10.1021/acs.jafc.0c02371] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Phytophthora sojae is a serious soil-borne pathogen, and the major control measures undertaken include the induction of soybean-resistance genes, fungicides, and scientific and reasonable planting management. Owing to the safety and resistance of fungicides, it is of great importance to screen new control alternatives. In a preliminary study, we observed that propyl gallate (PG) exerts a considerable inhibitory effect on P. sojae and can effectively prevent and cure soybean diseases, although the underlying mechanism remains unclear. To explore the inhibitory mechanism of PG on P. sojae, we analyzed the differences in the protein profile of P. sojae before and after treatment with PG using tandem mass tag (TMT) proteomics. Proteomic analysis revealed that the number of differentially expressed proteins (DEPs) was 285, of which 75 were upregulated and 210 were downregulated, and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways primarily comprised glycolysis, tricarboxylic acid cycle, fatty acid metabolism, secondary metabolite generation, and other pathways. Among the DEPs involved in PG inhibition of P. sojae are two closely related uncharacterized proteins encoded by PHYSODRAFT_522340 and PHYSODRAFT_344464, denoted PsFACL and PsCPT herein. The CRISPR/Cas9 knockout technique revealed that PsFACL and PsCPT were involved in the growth rate and pathogenicity. In addition, the results of gas chromatography-mass spectrometry (GC-MS) showed that there were differences in fatty acid levels between wild-type (WT) and CRISPR/Cas9 knockout transformants. Knocking out PsFACL and PsCPT resulted in the restriction of the synthesis and β-oxidation of long-chain fatty acids, respectively. These suggest that PsFACL and PsCPT were also involved in the regulation of the fatty acid metabolism. Our results aid in understanding the mechanism underlying the inhibition of P. sojae growth by PG.
Collapse
Affiliation(s)
- Dong Liu
- College of Plant Protection, Anhui Agricultural University, 130 West of Changjiang Road, Hefei 230036, Anhui, China
- Department of Horticulture and Landscape, Anqing Vocational and Technical College, 99 North of Tianzhushan Road, Anqing 246003, Anhui, China
| | - Yuemin Pan
- College of Plant Protection, Anhui Agricultural University, 130 West of Changjiang Road, Hefei 230036, Anhui, China
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, Anhui Agricultural University, Hefei 230036, Anhui, China
- Key Laboratory of Biology and Sustainable Management of Plant Diseases and Pests of Anhui Higher Education Institutes, Anhui Agricultural University, Anqing 246003, Anhui, China
| | - Kunyuan Li
- College of Plant Protection, Anhui Agricultural University, 130 West of Changjiang Road, Hefei 230036, Anhui, China
| | - Dandan Li
- College of Plant Protection, Anhui Agricultural University, 130 West of Changjiang Road, Hefei 230036, Anhui, China
| | - Ping Li
- Department of Horticulture and Landscape, Anqing Vocational and Technical College, 99 North of Tianzhushan Road, Anqing 246003, Anhui, China
| | - Zhimou Gao
- College of Plant Protection, Anhui Agricultural University, 130 West of Changjiang Road, Hefei 230036, Anhui, China
| |
Collapse
|
28
|
Nassar ZD, Mah CY, Dehairs J, Burvenich IJG, Irani S, Centenera MM, Helm M, Shrestha RK, Moldovan M, Don AS, Holst J, Scott AM, Horvath LG, Lynn DJ, Selth LA, Hoy AJ, Swinnen JV, Butler LM. Human DECR1 is an androgen-repressed survival factor that regulates PUFA oxidation to protect prostate tumor cells from ferroptosis. eLife 2020; 9:e54166. [PMID: 32686647 PMCID: PMC7386908 DOI: 10.7554/elife.54166] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 07/16/2020] [Indexed: 12/27/2022] Open
Abstract
Fatty acid β-oxidation (FAO) is the main bioenergetic pathway in human prostate cancer (PCa) and a promising novel therapeutic vulnerability. Here we demonstrate therapeutic efficacy of targeting FAO in clinical prostate tumors cultured ex vivo, and identify DECR1, encoding the rate-limiting enzyme for oxidation of polyunsaturated fatty acids (PUFAs), as robustly overexpressed in PCa tissues and associated with shorter relapse-free survival. DECR1 is a negatively-regulated androgen receptor (AR) target gene and, therefore, may promote PCa cell survival and resistance to AR targeting therapeutics. DECR1 knockdown selectively inhibited β-oxidation of PUFAs, inhibited proliferation and migration of PCa cells, including treatment resistant lines, and suppressed tumor cell proliferation and metastasis in mouse xenograft models. Mechanistically, targeting of DECR1 caused cellular accumulation of PUFAs, enhanced mitochondrial oxidative stress and lipid peroxidation, and induced ferroptosis. These findings implicate PUFA oxidation via DECR1 as an unexplored facet of FAO that promotes survival of PCa cells.
Collapse
Grants
- Early Career Fellowship,1138648 National Health and Medical Research Council
- Project Grants C16/15/073 and C32/17/052 KU Leuven
- Future Fellowship,FT130101004 Australian Research Council
- Beat Cancer Fellowship,PRF1117 Cancer Council South Australia
- Revolutionary Team Award,MRTA3 Movember Foundation
- Project Grant,1121057 National Health and Medical Research Council
- Project Grant,1100626 National Health and Medical Research Council
- Fellowship,1084178 National Health and Medical Research Council
- Young Investigator Award,YI 1417 Prostate Cancer Foundation of Australia
- Project Grant,1164798 Cure Cancer Australia Foundation
- Group Leader Award EMBL Australia
- Robinson Fellowship University of Sydney
- Project Grants G.0841.15 and G.0C22.19N Fonds Wetenschappelijk Onderzoek
- 1138648 National Health and Medical Research Council
- 1121057 National Health and Medical Research Council
- 1100626 National Health and Medical Research Council
- 1084178 National Health and Medical Research Council
- YI 1417 Prostate Cancer Foundation of Australia
- 1164798 Cure Cancer Australia Foundation
- FT130101004 Australian Research Council
- PRF1117 Cancer Council South Australia
- MRTA3 Movember Foundation
- Freemasons Foundation Centre for Men's Health, University of Adelaide
Collapse
Affiliation(s)
- Zeyad D Nassar
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Chui Yan Mah
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Jonas Dehairs
- KU Leuven- University of Leuven, LKI- Leuven Cancer Institute, Department of Oncology, Laboratory of Lipid Metabolism and CancerLeuvenBelgium
| | - Ingrid JG Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe UniversityMelbourneAustralia
| | - Swati Irani
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Margaret M Centenera
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Madison Helm
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Raj K Shrestha
- Dame Roma Mitchell Cancer Research Laboratories, University of AdelaideAdelaideAustralia
| | - Max Moldovan
- South Australian Health and Medical Research InstituteAdelaideAustralia
| | - Anthony S Don
- NHMRC Clinical Trials Centre, and Centenary Institute, The University of SydneyCamperdownAustralia
| | - Jeff Holst
- Translational Cancer Metabolism Laboratory, School of Medical Sciences and Prince of Wales Clinical School, UNSW SydneySydneyAustralia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe UniversityMelbourneAustralia
| | - Lisa G Horvath
- Garvan Institute of Medical Research, NSW 2010; University of Sydney, NSW 2006; and University of New South WalesDarlinghurstAustralia
| | - David J Lynn
- South Australian Health and Medical Research InstituteAdelaideAustralia
- College of Medicine and Public Health, Flinders UniversityBedford ParkAustralia
| | - Luke A Selth
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- Dame Roma Mitchell Cancer Research Laboratories, University of AdelaideAdelaideAustralia
- College of Medicine and Public Health, Flinders UniversityBedford ParkAustralia
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of SydneyCamperdownAustralia
| | - Johannes V Swinnen
- KU Leuven- University of Leuven, LKI- Leuven Cancer Institute, Department of Oncology, Laboratory of Lipid Metabolism and CancerLeuvenBelgium
| | - Lisa M Butler
- University of Adelaide Medical School and Freemasons Foundation Centre for Men’s Health, University of AdelaideAdelaideAustralia
- South Australian Health and Medical Research InstituteAdelaideAustralia
| |
Collapse
|
29
|
Yu L, Gong L, Wang C, Hu N, Tang Y, Zheng L, Dai X, Li Y. Radix Polygoni Multiflori and Its Main Component Emodin Attenuate Non-Alcoholic Fatty Liver Disease in Zebrafish by Regulation of AMPK Signaling Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1493-1506. [PMID: 32346285 PMCID: PMC7167271 DOI: 10.2147/dddt.s243893] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/05/2020] [Indexed: 12/19/2022]
Abstract
Purpose Nonalcoholic fatty liver disease (NAFLD) has become a predictor of death in many diseases. This study was carried out to investigate the therapeutic effect of Radix Polygoni Multiflori Preparata (RPMP) and its main component emodin on egg yolk powder-induced NAFLD in zebrafish. Further investigation was performed to explore whether emodin was the main component of RPMP for the treatment of NAFLD as well as the underlying therapeutic mechanism of RPMP and emodin. Methods Zebrafish were divided into control group, egg yolk powder group, RPMP group and emodin group. The obesity of zebrafish was evaluated by body weight, body length and BMI. The content of lipid was detected by triglyceride (TG), total cholesterol (TC) reagent kit and the fatty acid was detected by nonesterified free fatty acids (NEFA) reagent kit. HE staining was used to detect the histological structure of liver. Whole-mount Oil red O staining and Frozen oil red O staining were carried out to investigate the lipid accumulation in liver. KEGG and STRING databases were performed to analyze the potential role of AMPK between insulin resistance (IR) and fatty acid oxidation. Western blot and RT-qPCR were carried out for mechanism research. Results RPMP and emodin significantly reduced zebrafish weight, body length and BMI. Both RPMP and emodin treatment could reduce the lipid deposition in zebrafish liver. RPMP significantly reduced the content of TG. However, emodin significantly reduced the contents of TG, TC and NEFA in zebrafish with NAFLD. The protein interaction network indicated that AMPK participated in both IR and fatty acid oxidation. Further investigation indicated that RPMP and emodin reduced hepatic lipogenesis via up-regulating the expressions of phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT2), amp-activated protein kinase alpha (AMPKα), proliferator-activated receptor alpha (PPARα), carnitine palmitoyl transferase 1a (CPT-1a) and acyl-coenzyme A oxidase 1 (ACOX1). Conclusion These findings suggest that emodin is the main component of RPMP for the treatment of NAFLD, which is closely related to the regulation of AMPK signaling pathway which increases IR and fatty acid oxidation.
Collapse
Affiliation(s)
- Linyuan Yu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Lihong Gong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Cheng Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Naihua Hu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Yunqiu Tang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Li Zheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Xuyang Dai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Yunxia Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| |
Collapse
|
30
|
McGlaughon JL, Pasquali M, Wallace K, Ross J, Senol-Cosar O, Shen W, Weaver MA, Feigenbaum A, Lyon E, Enns GM, Mao R, Baudet HG. Assessing the strength of evidence for genes implicated in fatty acid oxidation disorders using the ClinGen clinical validity framework. Mol Genet Metab 2019; 128:122-128. [PMID: 31399326 DOI: 10.1016/j.ymgme.2019.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 07/16/2019] [Indexed: 12/30/2022]
Abstract
Newborn screening is an incredibly useful tool for the early identification of many metabolic disorders, including fatty acid oxidation (FAO) disorders. In many cases, molecular tests are necessary to reach a final diagnosis, highlighting the need for a thorough evaluation of genes implicated in FAO disorders. Using the ClinGen (Clinical Genome Resource) clinical validity framework, thirty genes were analyzed for the strength of evidence supporting their association with FAO disorders. Evidence was gathered from the literature by biocurators and presented to disease experts for review in order to assign a clinical validity classification of Definitive, Strong, Moderate, Limited, Disputed, Refuted, or No Reported Evidence. Of the gene-disease relationships evaluated, 22/30 were classified as Definitive, three as Moderate, one as Limited, three as No Reported Evidence and one as Disputed. Gene-disease relationships with a Limited, Disputed, and No Reported Evidence were found on two, six, and up to four panels out of 30 FAO disorder-specific panels, respectively, in the National Institute of Health Genetic Testing Registry, while over 70% of the genes on panels are definitively associated with an FAO disorder. These results highlight the need to systematically assess the clinical relevance of genes implicated in fatty acid oxidation disorders in order to improve the interpretation of genetic testing results and diagnosis of patients with these disorders.
Collapse
Affiliation(s)
- Jennifer L McGlaughon
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Marzia Pasquali
- University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Kathleen Wallace
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Justyne Ross
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Ozlem Senol-Cosar
- Laboratory for Molecular Medicine, Partners Healthcare Personalized Medicine, Cambridge, MA, USA; Department of Pathology, Harvard Medical School/Brigham and Women's Hospital, Boston, MA, USA
| | - Wei Shen
- University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Meredith A Weaver
- American College of Medical Genetics and Genomics, Bethesda, MD, USA
| | - Annette Feigenbaum
- Department of Pediatrics, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Elaine Lyon
- University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Gregory M Enns
- Department of Pediatrics, Division of Medical Genetics, Stanford University, Stanford, CA, USA
| | - Rong Mao
- University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Heather G Baudet
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|
31
|
Circadian lipid synthesis in brown fat maintains murine body temperature during chronic cold. Proc Natl Acad Sci U S A 2019; 116:18691-18699. [PMID: 31451658 DOI: 10.1073/pnas.1909883116] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ambient temperature influences the molecular clock and lipid metabolism, but the impact of chronic cold exposure on circadian lipid metabolism in thermogenic brown adipose tissue (BAT) has not been studied. Here we show that during chronic cold exposure (1 wk at 4 °C), genes controlling de novo lipogenesis (DNL) including Srebp1, the master transcriptional regulator of DNL, acquired high-amplitude circadian rhythms in thermogenic BAT. These conditions activated mechanistic target of rapamycin 1 (mTORC1), an inducer of Srebp1 expression, and engaged circadian transcriptional repressors REV-ERBα and β as rhythmic regulators of Srebp1 in BAT. SREBP was required in BAT for the thermogenic response to norepinephrine, and depletion of SREBP prevented maintenance of body temperature both during circadian cycles as well as during fasting of chronically cold mice. By contrast, deletion of REV-ERBα and β in BAT allowed mice to maintain their body temperature in chronic cold. Thus, the environmental challenge of prolonged noncircadian exposure to cold temperature induces circadian induction of SREBP1 that drives fuel synthesis in BAT and is necessary to maintain circadian body temperature during chronic cold exposure. The requirement for BAT fatty acid synthesis has broad implications for adaptation to cold.
Collapse
|
32
|
Intrinsic expression of viperin regulates thermogenesis in adipose tissues. Proc Natl Acad Sci U S A 2019; 116:17419-17428. [PMID: 31341090 DOI: 10.1073/pnas.1904480116] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Viperin is an interferon (IFN)-inducible multifunctional protein. Recent evidence from high-throughput analyses indicates that most IFN-inducible proteins, including viperin, are intrinsically expressed in specific tissues; however, the respective intrinsic functions are unknown. Here we show that the intrinsic expression of viperin regulates adipose tissue thermogenesis, which is known to counter metabolic disease and contribute to the febrile response to pathogen invasion. Viperin knockout mice exhibit increased heat production, resulting in a reduction of fat mass, improvement of high-fat diet (HFD)-induced glucose tolerance, and enhancement of cold tolerance. These thermogenic phenotypes are attributed to an adipocyte-autonomous mechanism that regulates fatty acid β-oxidation. Under an HFD, viperin expression is increased, and its function is enhanced. Our findings reveal the intrinsic function of viperin as a novel mechanism regulating thermogenesis in adipose tissues, suggesting that viperin represents a molecular target for thermoregulation in clinical contexts.
Collapse
|
33
|
Mukhopadhyay A, Thomas T, Bosch RJ, Dwarkanath P, Thomas A, Duggan CP, Kurpad AV. Fetal sex modifies the effect of maternal macronutrient intake on the incidence of small-for-gestational-age births: a prospective observational cohort study. Am J Clin Nutr 2018; 108:814-820. [PMID: 30239558 PMCID: PMC6927877 DOI: 10.1093/ajcn/nqy161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 06/21/2018] [Indexed: 12/22/2022] Open
Abstract
Background Maternal macronutrient intake is likely to play a pivotal role in fetoplacental growth. Male fetuses grow faster and their growth is more responsive to maternal size. Objective We assessed the role of fetal sex in modifying the effect of maternal macronutrient intake on the risk of small-for-gestational-age (SGA) birth. Design This was a prospective, observational cohort study of 2035 births from an urban South Asian Indian population. Maternal intakes of total energy and macronutrients were recorded by validated food-frequency questionnaires. The interaction of trimester 1 macronutrient intake with fetal sex was tested on the outcome of SGA births. Results The prevalence of SGA was 28%. Trimester 1 macronutrient composition was high in carbohydrate and low in fat (means ± SDs-carbohydrate: 64.6% ± 5.1%; protein: 11.5% ± 1.1%; and fat: 23.9% ± 4.4% of energy). Higher carbohydrate and lower fat consumption were each associated with an increased risk of SGA [adjusted OR (AOR) per 5% of energy (95% CI): carbohydrate: 1.15 (1.01, 1.32); fat: 0.83 (0.71, 0.97)] specifically among male births (males: n = 1047; females: n = 988). Dietary intake of >70% of energy from carbohydrate was also associated with increased risk (AOR: 1.67; 95% CI: 1.00, 2.78), whereas >25% of energy from fat intake was associated with decreased risk (AOR: 0.61; 95% CI: 0.41, 0.90) of SGA in male births. Conclusions Higher carbohydrate and lower fat intakes early in pregnancy were associated with increased risk of male SGA births. Therefore, we speculate that fetal sex acts as a modifier of the role of maternal periconceptional nutrition in optimal fetoplacental growth.
Collapse
Affiliation(s)
- A Mukhopadhyay
- Divisions of Nutrition, St. John's National Academy of Health Sciences, Bangalore, India,Address correspondence to AM (e-mail: )
| | - T Thomas
- Epidemiology and Biostatistics, St. John's National Academy of Health Sciences
, Bangalore, India
| | - R J Bosch
- Departments of Biostatistics
, Boston, MA
| | - P Dwarkanath
- Divisions of Nutrition, St. John's National Academy of Health Sciences, Bangalore, India
| | - A Thomas
- Department of Obstetrics and Gynecology, St John's Medical College, St. John's National Academy of Health Sciences
, Bangalore, India
| | - C P Duggan
- Nutrition, Harvard TH Chan School of Public Health
, Boston, MA
| | - A V Kurpad
- Divisions of Nutrition, St. John's National Academy of Health Sciences, Bangalore, India
| |
Collapse
|
34
|
Hankir MK, Klingenspor M. Brown adipocyte glucose metabolism: a heated subject. EMBO Rep 2018; 19:embr.201846404. [PMID: 30135070 DOI: 10.15252/embr.201846404] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/22/2018] [Accepted: 07/20/2018] [Indexed: 11/09/2022] Open
Abstract
The energy expending and glucose sink properties of brown adipose tissue (BAT) make it an attractive target for new obesity and diabetes treatments. Despite decades of research, only recently have mechanistic studies started to provide a more complete and consistent picture of how activated brown adipocytes handle glucose. Here, we discuss the importance of intracellular glycolysis, lactate production, lipogenesis, lipolysis, and beta-oxidation for BAT thermogenesis in response to natural (temperature) and artificial (pharmacological and optogenetic) forms of sympathetic nervous system stimulation. It is now clear that together, these metabolic processes in series and in parallel flexibly power ATP-dependent and independent futile cycles in brown adipocytes to impact on whole-body thermal, energy, and glucose balance.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Department of Experimental Surgery, University Hospital Wuerzburg, Wuerzburg, Germany .,German Research Foundation Collaborative Research Center in Obesity Mechanisms 1052, University of Leipzig, Leipzig, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany .,EKFZ - Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| |
Collapse
|
35
|
Larsson M, Allan CM, Heizer PJ, Tu Y, Sandoval NP, Jung RS, Walzem RL, Beigneux AP, Young SG, Fong LG. Impaired thermogenesis and sharp increases in plasma triglyceride levels in GPIHBP1-deficient mice during cold exposure. J Lipid Res 2018; 59:706-713. [PMID: 29449313 PMCID: PMC5880501 DOI: 10.1194/jlr.m083832] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Indexed: 01/11/2023] Open
Abstract
Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), an endothelial cell protein, binds LPL in the subendothelial spaces and transports it to the capillary lumen. In Gpihbp1-/- mice, LPL remains stranded in the subendothelial spaces, causing hypertriglyceridemia, but how Gpihbp1-/- mice respond to metabolic stress (e.g., cold exposure) has never been studied. In wild-type mice, cold exposure increases LPL-mediated processing of triglyceride-rich lipoproteins (TRLs) in brown adipose tissue (BAT), providing fuel for thermogenesis and leading to lower plasma triglyceride levels. We suspected that defective TRL processing in Gpihbp1-/- mice might impair thermogenesis and blunt the fall in plasma triglyceride levels. Indeed, Gpihbp1-/- mice exhibited cold intolerance, but the effects on plasma triglyceride levels were paradoxical. Rather than falling, the plasma triglyceride levels increased sharply (from ∼4,000 to ∼15,000 mg/dl), likely because fatty acid release by peripheral tissues drives hepatic production of TRLs that cannot be processed. We predicted that the sharp increase in plasma triglyceride levels would not occur in Gpihbp1-/-Angptl4-/- mice, where LPL activity is higher and baseline plasma triglyceride levels are lower. Indeed, the plasma triglyceride levels in Gpihbp1-/-Angptl4-/- mice fell during cold exposure. Metabolic studies revealed increased levels of TRL processing in the BAT of Gpihbp1-/-Angptl4-/- mice.
Collapse
Affiliation(s)
- Mikael Larsson
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095.
| | - Christopher M Allan
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095
| | - Patrick J Heizer
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095
| | - Yiping Tu
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095
| | - Norma P Sandoval
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095
| | - Rachel S Jung
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095
| | - Rosemary L Walzem
- Department of Poultry Science and Faculty of Nutrition, Texas A&M University, College Station, TX 77843
| | - Anne P Beigneux
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095
| | - Stephen G Young
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095; Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095.
| | - Loren G Fong
- Departments of Medicine University of California Los Angeles, Los Angeles, CA 90095.
| |
Collapse
|
36
|
Winther S, Isidor MS, Basse AL, Skjoldborg N, Cheung A, Quistorff B, Hansen JB. Restricting glycolysis impairs brown adipocyte glucose and oxygen consumption. Am J Physiol Endocrinol Metab 2018; 314:E214-E223. [PMID: 29118013 DOI: 10.1152/ajpendo.00218.2017] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During thermogenic activation, brown adipocytes take up large amounts of glucose. In addition, cold stimulation leads to an upregulation of glycolytic enzymes. Here we have investigated the importance of glycolysis for brown adipocyte glucose consumption and thermogenesis. Using siRNA-mediated knockdown in mature adipocytes, we explored the effect of glucose transporters and glycolytic enzymes on brown adipocyte functions such as consumption of glucose and oxygen. Basal oxygen consumption in brown adipocytes was equally dependent on glucose and fatty acid oxidation, whereas isoproterenol (ISO)-stimulated respiration was fueled mainly by fatty acids, with a significant contribution from glucose oxidation. Knockdown of glucose transporters in brown adipocytes not only impaired ISO-stimulated glycolytic flux but also oxygen consumption. Diminishing glycolytic flux by knockdown of the first and final enzyme of glycolysis, i.e., hexokinase 2 (HK2) and pyruvate kinase M (PKM), respectively, decreased glucose uptake and ISO-stimulated oxygen consumption. HK2 knockdown had a more severe effect, which, in contrast to PKM knockdown, could not be rescued by supplementation with pyruvate. Hence, brown adipocytes rely on glucose consumption and glycolytic flux to achieve maximum thermogenic output, with glycolysis likely supporting thermogenesis not only by pyruvate formation but also by supplying intermediates for efferent metabolic pathways.
Collapse
Affiliation(s)
- Sally Winther
- Department of Biology, University of Copenhagen , Copenhagen , Denmark
| | - Marie S Isidor
- Department of Biology, University of Copenhagen , Copenhagen , Denmark
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Astrid L Basse
- Department of Biology, University of Copenhagen , Copenhagen , Denmark
| | - Nina Skjoldborg
- Department of Biology, University of Copenhagen , Copenhagen , Denmark
| | - Amanda Cheung
- Department of Biology, University of Copenhagen , Copenhagen , Denmark
| | - Bjørn Quistorff
- Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jacob B Hansen
- Department of Biology, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
37
|
Lee MS, Shin Y, Jung S, Kim Y. Effects of epigallocatechin-3-gallate on thermogenesis and mitochondrial biogenesis in brown adipose tissues of diet-induced obese mice. Food Nutr Res 2017; 61:1325307. [PMID: 28659734 PMCID: PMC5475335 DOI: 10.1080/16546628.2017.1325307] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/27/2017] [Indexed: 11/15/2022] Open
Abstract
Background: Epigallocatechin-3-gallate (EGCG) is the major polyphenol in green tea and has been considered a natural agent that can help to reduce the risk of obesity. Objective: The aim of this study was to investigate the effects of EGCG on thermogenesis and mitochondrial biogenesis in brown adipose tissue (BAT) of diet-induced obese mice. Methods: Male C57BL/6J mice were provided a high-fat diet for 8 weeks to induce obesity, following which they were divided into two groups: one on a high-fat control diet and the other on a 0.2% EGCG (w/w)-supplemented high-fat diet for another 8 weeks. Results: The EGCG-supplemented group showed decreased body weight gain, and plasma and liver lipids. EGCG-fed mice exhibited higher body temperature and mitochondrial DNA (mtDNA) content in BAT. The messenger RNA levels of genes related to thermogenesis and mitochondrial biogenesis in BAT were increased by EGCG. Moreover, adenosine monophosphate-activated protein kinase (AMPK) activity in BAT was stimulated by EGCG. Conclusions: The results suggest that EGCG may have anti-obesity properties through BAT thermogenesis and mitochondria biogenesis, which are partially associated with the regulation of genes related to thermogenesis and mitochondria biogenesis, and the increase in mtDNA replication and AMPK activation in BAT of diet-induced obese mice.
Collapse
Affiliation(s)
- Mak-Soon Lee
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Yoonjin Shin
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Sunyoon Jung
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Yangha Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
38
|
Kolwicz SC. Lipid partitioning during cardiac stress. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1861:1472-80. [PMID: 27040509 DOI: 10.1016/j.bbalip.2016.03.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/18/2016] [Accepted: 03/18/2016] [Indexed: 01/11/2023]
Abstract
It is well documented that fatty acids serve as the primary fuel substrate for the contracting myocardium. However, extensive research has identified significant changes in the myocardial oxidation of fatty acids during acute or chronic cardiac stress. As a result, the redistribution or partitioning of fatty acids due to metabolic derangements could have biological implications. Fatty acids can be stored as triacylglycerols, serve as critical components for biosynthesis of phospholipid membranes, and form the potent signaling molecules, diacylglycerol and ceramides. Therefore, the contribution of lipid metabolism to health and disease is more intricate than a balance of uptake and oxidation. In this review, the available data regarding alterations that occur in endogenous cardiac lipid pathways during the pathological stressors of ischemia-reperfusion and pathological hypertrophy/heart failure are highlighted. In addition, changes in endogenous lipids observed in exercise training models are presented for comparison. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.
Collapse
Affiliation(s)
- Stephen C Kolwicz
- Mitochondria and Metabolism Center, University of Washington, School of Medicine, 850 Republican St., Seattle, WA 98109, United States.
| |
Collapse
|
39
|
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
|
40
|
Requirement for the Mitochondrial Pyruvate Carrier in Mammalian Development Revealed by a Hypomorphic Allelic Series. Mol Cell Biol 2016; 36:2089-104. [PMID: 27215380 DOI: 10.1128/mcb.00166-16] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/13/2016] [Indexed: 11/20/2022] Open
Abstract
Glucose and oxygen are two of the most important molecules transferred from mother to fetus during eutherian pregnancy, and the metabolic fates of these nutrients converge at the transport and metabolism of pyruvate in mitochondria. Pyruvate enters the mitochondrial matrix through the mitochondrial pyruvate carrier (MPC), a complex in the inner mitochondrial membrane that consists of two essential components, MPC1 and MPC2. Here, we define the requirement for mitochondrial pyruvate metabolism during development with a progressive allelic series of Mpc1 deficiency in mouse. Mpc1 deletion was homozygous lethal in midgestation, but Mpc1 hypomorphs and tissue-specific deletion of Mpc1 presented as early perinatal lethality. The allelic series demonstrated that graded suppression of MPC resulted in dose-dependent metabolic and transcriptional changes. Steady-state metabolomics analysis of brain and liver from Mpc1 hypomorphic embryos identified compensatory changes in amino acid and lipid metabolism. Flux assays in Mpc1-deficient embryonic fibroblasts also reflected these changes, including a dramatic increase in mitochondrial alanine utilization. The mitochondrial alanine transaminase GPT2 was found to be necessary and sufficient for increased alanine flux upon MPC inhibition. These data show that impaired mitochondrial pyruvate transport results in biosynthetic deficiencies that can be mitigated in part by alternative anaplerotic substrates in utero.
Collapse
|
41
|
Lee J, Choi J, Scafidi S, Wolfgang MJ. Hepatic Fatty Acid Oxidation Restrains Systemic Catabolism during Starvation. Cell Rep 2016; 16:201-212. [PMID: 27320917 DOI: 10.1016/j.celrep.2016.05.062] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/14/2016] [Accepted: 05/16/2016] [Indexed: 12/30/2022] Open
Abstract
The liver is critical for maintaining systemic energy balance during starvation. To understand the role of hepatic fatty acid β-oxidation on this process, we generated mice with a liver-specific knockout of carnitine palmitoyltransferase 2 (Cpt2(L-/-)), an obligate step in mitochondrial long-chain fatty acid β-oxidation. Fasting induced hepatic steatosis and serum dyslipidemia with an absence of circulating ketones, while blood glucose remained normal. Systemic energy homeostasis was largely maintained in fasting Cpt2(L-/-) mice by adaptations in hepatic and systemic oxidative gene expression mediated in part by Pparα target genes including procatabolic hepatokines Fgf21, Gdf15, and Igfbp1. Feeding a ketogenic diet to Cpt2(L-/-) mice resulted in severe hepatomegaly, liver damage, and death with a complete absence of adipose triglyceride stores. These data show that hepatic fatty acid oxidation is not required for survival during acute food deprivation but essential for constraining adipocyte lipolysis and regulating systemic catabolism when glucose is limiting.
Collapse
Affiliation(s)
- Jieun Lee
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joseph Choi
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
42
|
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
|
43
|
Sekine S, Yao A, Hattori K, Sugawara S, Naguro I, Koike M, Uchiyama Y, Takeda K, Ichijo H. The Ablation of Mitochondrial Protein Phosphatase Pgam5 Confers Resistance Against Metabolic Stress. EBioMedicine 2016; 5:82-92. [PMID: 27077115 PMCID: PMC4816851 DOI: 10.1016/j.ebiom.2016.01.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/24/2016] [Accepted: 01/26/2016] [Indexed: 01/08/2023] Open
Abstract
Phosphoglycerate mutase family member 5 (PGAM5) is a mitochondrial protein phosphatase that has been reported to be involved in various stress responses from mitochondrial quality control to cell death. However, its roles in vivo are largely unknown. Here, we show that Pgam5-deficient mice are resistant to several metabolic insults. Under cold stress combined with fasting, Pgam5-deficient mice better maintained body temperature than wild-type mice and showed an extended survival rate. Serum triglycerides and lipid content in brown adipose tissue (BAT), a center of adaptive thermogenesis, were severely reduced in Pgam5-deficient mice. Moreover, although Pgam5 deficiency failed to maintain proper mitochondrial integrity in BAT, it reciprocally resulted in the dramatic induction of fibroblast growth factor 21 (FGF21) that activates various functions of BAT including thermogenesis. Thus, the enhancement of lipid metabolism and FGF21 may contribute to the cold resistance of Pgam5-deficient mice under fasting condition. Finally, we also found that Pgam5-deficient mice are resistant to high-fat-diet-induced obesity. Our study uncovered that PGAM5 is involved in the whole-body metabolism in response to stresses that impose metabolic challenges on mitochondria. The ablation of Pgam5, a mitochondria-resident protein phosphatase, protected mice from some metabolic stresses. Pgam5-deficient mice were resistant to a cold plus fasting stress and a high fat diet-induced obesity. Our study revealed that PGAM5 acts as a metabolic regulator in vivo.
Here, we revealed that the ablation of Pgam5, a mitochondria-resident protein phosphatase, protects mice from some metabolic stresses. When fasted mice were exposed to cold condition, Pgam5 deficiency promoted the lipid metabolism and the expression of metabolic hormone FGF21 in brown adipose tissue, a center of heat production, suggesting that these metabolic changes might ultimately contribute to their resistance. In addition, we found that Pgam5-deficient mice were dramatically resistant to high-fat diet-induced obesity. Our study not only provides the evidence that PGAM5 acts as a metabolic regulator in vivo but also raises the potential therapeutic target for metabolic diseases.
Collapse
Affiliation(s)
- Shiori Sekine
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Akari Yao
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuki Hattori
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Sho Sugawara
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Isao Naguro
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasuo Uchiyama
- Departments of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kohsuke Takeda
- Division of Cell Regulation, Graduate School of Briomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Corresponding author.
| |
Collapse
|
44
|
Frye RE, Cox D, Slattery J, Tippett M, Kahler S, Granpeesheh D, Damle S, Legido A, Goldenthal MJ. Mitochondrial Dysfunction may explain symptom variation in Phelan-McDermid Syndrome. Sci Rep 2016; 6:19544. [PMID: 26822410 PMCID: PMC4731780 DOI: 10.1038/srep19544] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 10/09/2015] [Indexed: 12/02/2022] Open
Abstract
Phelan-McDermid Syndrome (PMS), which is defined by a deletion within 22q13, demonstrates significant phenotypic variation. Given that six mitochondrial genes are located within 22q13, including complex I and IV genes, we hypothesize that mitochondrial complex activity abnormalities may explain phenotypic variation in PMS symptoms. Complex I, II, II + III and IV activity was measured in 51 PMS participants. Caretakers completed questionnaires and provided genetic information through the PMS foundation registry. Complex activity was abnormal in 59% of PMS participants. Abnormalities were found in complex I and IV but not complex II + III and II activity, consistent with disruption of genes within the 22q13 region. However, complex activity abnormalities were not related to specific gene deletions suggesting a "neighboring effect" of regional deletions on adjacent gene expression. A specific combination of symptoms (autism spectrum disorder, developmental regression, failure-to-thrive, exercise intolerance/fatigue) was associated with complex activity abnormalities. 64% of 106 individuals in the PMS foundation registry who did not have complex activity measured also endorsed this pattern of symptoms. These data suggest that mitochondrial abnormalities, specifically abnormalities in complex I and IV activity, may explain some phenotypic variation in PMS individuals. These results point to novel pathophysiology mechanisms and treatment targets for PMS patients.
Collapse
Affiliation(s)
- Richard E. Frye
- University of Arkansas for Medical Sciences, Department of Pediatrics, Arkansas Children’s Hospital Research Institute, Little Rock, Arkansas, AR 72202, USA
| | - Devin Cox
- Kansas University Medical Center, Kansas City, Kansas, KS, USA
| | - John Slattery
- University of Arkansas for Medical Sciences, Department of Pediatrics, Arkansas Children’s Hospital Research Institute, Little Rock, Arkansas, AR 72202, USA
| | - Marie Tippett
- University of Arkansas for Medical Sciences, Department of Pediatrics, Arkansas Children’s Hospital Research Institute, Little Rock, Arkansas, AR 72202, USA
| | - Stephen Kahler
- University of Arkansas for Medical Sciences, Department of Pediatrics, Arkansas Children’s Hospital Research Institute, Little Rock, Arkansas, AR 72202, USA
| | - Doreen Granpeesheh
- Center for Autism and Related Disorders, Inc., Woodland Hills, California, CA, USA
| | - Shirish Damle
- Drexel University College of Medicine, Department of Pediatrics, Neurology Section, St. Christopher’s Hospital for Children, Philadelphia, PA 19134, USA
| | - Agustin Legido
- Drexel University College of Medicine, Department of Pediatrics, Neurology Section, St. Christopher’s Hospital for Children, Philadelphia, PA 19134, USA
| | - Michael J. Goldenthal
- Drexel University College of Medicine, Department of Pediatrics, Neurology Section, St. Christopher’s Hospital for Children, Philadelphia, PA 19134, USA
| |
Collapse
|
45
|
Maurer SF, Fromme T, Grossman LI, Hüttemann M, Klingenspor M. The brown and brite adipocyte marker Cox7a1 is not required for non-shivering thermogenesis in mice. Sci Rep 2015; 5:17704. [PMID: 26635001 PMCID: PMC4669493 DOI: 10.1038/srep17704] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022] Open
Abstract
The cytochrome c oxidase subunit isoform Cox7a1 is highly abundant in skeletal muscle and heart and influences enzyme activity in these tissues characterised by high oxidative capacity. We identified Cox7a1, well-known as brown adipocyte marker gene, as a cold-responsive protein of brown adipose tissue. We hypothesised a mechanistic relationship between cytochrome c oxidase activity and Cox7a1 protein levels affecting the oxidative capacity of brown adipose tissue and thus non-shivering thermogenesis. We subjected wildtype and Cox7a1 knockout mice to different temperature regimens and tested characteristics of brown adipose tissue activation. Cytochrome c oxidase activity, uncoupling protein 1 expression and maximal norepinephrine-induced heat production were gradually increased during cold-acclimation, but unaffected by Cox7a1 knockout. Moreover, the abundance of uncoupling protein 1 competent brite cells in white adipose tissue was not influenced by presence or absence of Cox7a1. Skin temperature in the interscapular region of neonates was lower in uncoupling protein 1 knockout pups employed as a positive control, but not in Cox7a1 knockout pups. Body mass gain and glucose tolerance did not differ between wildtype and Cox7a1 knockout mice fed with high fat or control diet. We conclude that brown adipose tissue function in mice does not require the presence of Cox7a1.
Collapse
Affiliation(s)
- Stefanie F Maurer
- Chair of Molecular Nutritional Medicine, Technische Universität München, Else Kröner-Fresenius Center for Nutritional Medicine &ZIEL-Institute for Food and Health, 85350 Freising-Weihenstephan, Germany
| | - Tobias Fromme
- Chair of Molecular Nutritional Medicine, Technische Universität München, Else Kröner-Fresenius Center for Nutritional Medicine &ZIEL-Institute for Food and Health, 85350 Freising-Weihenstephan, Germany
| | - Lawrence I Grossman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technische Universität München, Else Kröner-Fresenius Center for Nutritional Medicine &ZIEL-Institute for Food and Health, 85350 Freising-Weihenstephan, Germany
| |
Collapse
|
46
|
Mitochondrial dysfunction in fatty acid oxidation disorders: insights from human and animal studies. Biosci Rep 2015; 36:e00281. [PMID: 26589966 PMCID: PMC4718505 DOI: 10.1042/bsr20150240] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/20/2015] [Indexed: 12/17/2022] Open
Abstract
Patients affected by FAOD commonly present with hepatopathy, cardiomyopathy, skeletal myopathy and encephalopathy. Human and animal evidences indicate that mitochondrial functions are disrupted by fatty acids and derivatives accumulating in these disorders, suggesting that lipotoxicity may contribute to their pathogenesis. Mitochondrial fatty acid oxidation (FAO) plays a pivotal role in maintaining body energy homoeostasis mainly during catabolic states. Oxidation of fatty acids requires approximately 25 proteins. Inherited defects of FAO have been identified in the majority of these proteins and constitute an important group of inborn errors of metabolism. Affected patients usually present with severe hepatopathy, cardiomyopathy and skeletal myopathy, whereas some patients may suffer acute and/or progressive encephalopathy whose pathogenesis is poorly known. In recent years growing evidence has emerged indicating that energy deficiency/disruption of mitochondrial homoeostasis is involved in the pathophysiology of some fatty acid oxidation defects (FAOD), although the exact underlying mechanisms are not yet established. Characteristic fatty acids and carnitine derivatives are found at high concentrations in these patients and more markedly during episodes of metabolic decompensation that are associated with worsening of clinical symptoms. Therefore, it is conceivable that these compounds may be toxic. We will briefly summarize the current knowledge obtained from patients and genetic mouse models with these disorders indicating that disruption of mitochondrial energy, redox and calcium homoeostasis is involved in the pathophysiology of the tissue damage in the more common FAOD, including medium-chain acyl-CoA dehydrogenase (MCAD), long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) and very long-chain acyl-CoA dehydrogenase (VLCAD) deficiencies. We will also provide evidence that the fatty acids and derivatives that accumulate in these diseases disrupt mitochondrial homoeostasis. The elucidation of the toxic mechanisms of these compounds may offer new perspectives for potential novel adjuvant therapeutic strategies in selected disorders of this group.
Collapse
|
47
|
Houten SM, Violante S, Ventura FV, Wanders RJA. The Biochemistry and Physiology of Mitochondrial Fatty Acid β-Oxidation and Its Genetic Disorders. Annu Rev Physiol 2015; 78:23-44. [PMID: 26474213 DOI: 10.1146/annurev-physiol-021115-105045] [Citation(s) in RCA: 568] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondrial fatty acid β-oxidation (FAO) is the major pathway for the degradation of fatty acids and is essential for maintaining energy homeostasis in the human body. Fatty acids are a crucial energy source in the postabsorptive and fasted states when glucose supply is limiting. But even when glucose is abundantly available, FAO is a main energy source for the heart, skeletal muscle, and kidney. A series of enzymes, transporters, and other facilitating proteins are involved in FAO. Recessively inherited defects are known for most of the genes encoding these proteins. The clinical presentation of these disorders may include hypoketotic hypoglycemia, (cardio)myopathy, arrhythmia, and rhabdomyolysis and illustrates the importance of FAO during fasting and in hepatic and (cardio)muscular function. In this review, we present the current state of knowledge on the biochemistry and physiological functions of FAO and discuss the pathophysiological processes associated with FAO disorders.
Collapse
Affiliation(s)
- Sander M Houten
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; ,
| | - Sara Violante
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; ,
| | - Fatima V Ventura
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences, iMed.ULisboa, 1649-003 Lisboa, Portugal; .,Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisboa, Portugal
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, University of Amsterdam, 1100 DE Amsterdam, The Netherlands; .,Department of Pediatrics, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
48
|
Rinaldi C, Schmidt T, Situ AJ, Johnson JO, Lee PR, Chen KL, Bott LC, Fadó R, Harmison GH, Parodi S, Grunseich C, Renvoisé B, Biesecker LG, De Michele G, Santorelli FM, Filla A, Stevanin G, Dürr A, Brice A, Casals N, Traynor BJ, Blackstone C, Ulmer TS, Fischbeck KH. Mutation in CPT1C Associated With Pure Autosomal Dominant Spastic Paraplegia. JAMA Neurol 2015; 72:561-70. [PMID: 25751282 PMCID: PMC5612424 DOI: 10.1001/jamaneurol.2014.4769] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
IMPORTANCE The family of genes implicated in hereditary spastic paraplegias (HSPs) is quickly expanding, mostly owing to the widespread availability of next-generation DNA sequencing methods. Nevertheless, a genetic diagnosis remains unavailable for many patients. OBJECTIVE To identify the genetic cause for a novel form of pure autosomal dominant HSP. DESIGN, SETTING, AND PARTICIPANTS We examined and followed up with a family presenting to a tertiary referral center for evaluation of HSP for a decade until August 2014. Whole-exome sequencing was performed in 4 patients from the same family and was integrated with linkage analysis. Sanger sequencing was used to confirm the presence of the candidate variant in the remaining affected and unaffected members of the family and screen the additional patients with HSP. Five affected and 6 unaffected participants from a 3-generation family with pure adult-onset autosomal dominant HSP of unknown genetic origin were included. Additionally, 163 unrelated participants with pure HSP of unknown genetic cause were screened. MAIN OUTCOME AND MEASURE Mutation in the neuronal isoform of carnitine palmitoyl-transferase (CPT1C) gene. RESULTS We identified the nucleotide substitution c.109C>T in exon 3 of CPT1C, which determined the base substitution of an evolutionarily conserved Cys residue for an Arg in the gene product. This variant strictly cosegregated with the disease phenotype and was absent in online single-nucleotide polymorphism databases and in 712 additional exomes of control participants. We showed that CPT1C, which localizes to the endoplasmic reticulum, is expressed in motor neurons and interacts with atlastin-1, an endoplasmic reticulum protein encoded by the ATL1 gene known to be mutated in pure HSPs. The mutation, as indicated by nuclear magnetic resonance spectroscopy studies, alters the protein conformation and reduces the mean (SD) number (213.0 [46.99] vs 81.9 [14.2]; P < .01) and size (0.29 [0.01] vs 0.26 [0.01]; P < .05) of lipid droplets on overexpression in cells. We also observed a reduction of mean (SD) lipid droplets in primary cortical neurons isolated from Cpt1c-/- mice as compared with wild-type mice (1.0 [0.12] vs 0.44 [0.05]; P < .001), suggesting a dominant negative mechanism for the mutation. CONCLUSIONS AND RELEVANCE This study expands the genetics of autosomal dominant HSP and is the first, to our knowledge, to link mutation in CPT1C with a human disease. The association of the CPT1C mutation with changes in lipid droplet biogenesis supports a role for altered lipid-mediated signal transduction in HSP pathogenesis.
Collapse
Affiliation(s)
- Carlo Rinaldi
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Thomas Schmidt
- Department of Biochemistry and Molecular Biology, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles
| | - Alan J Situ
- Department of Biochemistry and Molecular Biology, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles
| | - Janel O Johnson
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Philip R Lee
- Section on Nervous System Development and Plasticity, The Eunice Kennedy Shriver National Institute of Child and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Ke-Lian Chen
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Laura C Bott
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland5Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rut Fadó
- Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya, and CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Sant Cugat del Vallés, Spain
| | - George H Harmison
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Sara Parodi
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland7Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Benoît Renvoisé
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Leslie G Biesecker
- Genetic Disease Research Branch, National Human Genome Research Institute, and the National Institutes of Health Intramural Sequencing Center, National Institutes of Health, Bethesda, Maryland
| | - Giuseppe De Michele
- Department of Neurosciences, Reproductive Sciences, and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Filippo M Santorelli
- Neurogenetics Istituto di Ricovero e Cura a Carattere Scientifico, Stella Maris, Pisa, Italy
| | - Alessandro Filla
- Department of Neurosciences, Reproductive Sciences, and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Giovanni Stevanin
- Institut du Cerveau et de la Moelle Épinière, Paris, France12Laboratoire de Neurogénétique, École Pratique des Hautes Études-héSam Université, Institut du Cerveau et de la Moelle Épinière, Groupe Hospitalier Pitié-Salpêtrière, Paris, France13Sorbonne Univ
| | - Alexandra Dürr
- Institut du Cerveau et de la Moelle Épinière, Paris, France13Sorbonne Universités, Université Pierre et Marie Curie, Institut du Cerveau et de la Moelle Épinière, Paris, France14Department of Genetics, Assistance Publique Hopitaux de Paris, Groupe Hospita
| | - Alexis Brice
- Institut du Cerveau et de la Moelle Épinière, Paris, France13Sorbonne Universités, Université Pierre et Marie Curie, Institut du Cerveau et de la Moelle Épinière, Paris, France14Department of Genetics, Assistance Publique Hopitaux de Paris, Groupe Hospita
| | - Núria Casals
- Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya, and CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Sant Cugat del Vallés, Spain
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Craig Blackstone
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Tobias S Ulmer
- Department of Biochemistry and Molecular Biology, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles
| | - Kenneth H Fischbeck
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
49
|
Abdurrachim D, Luiken JJFP, Nicolay K, Glatz JFC, Prompers JJ, Nabben M. Good and bad consequences of altered fatty acid metabolism in heart failure: evidence from mouse models. Cardiovasc Res 2015; 106:194-205. [PMID: 25765936 DOI: 10.1093/cvr/cvv105] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 02/18/2015] [Indexed: 12/25/2022] Open
Abstract
The shift in substrate preference away from fatty acid oxidation (FAO) towards increased glucose utilization in heart failure has long been interpreted as an oxygen-sparing mechanism. Inhibition of FAO has therefore evolved as an accepted approach to treat heart failure. However, recent data indicate that increased reliance on glucose might be detrimental rather than beneficial for the failing heart. This review discusses new insights into metabolic adaptations in heart failure. A particular focus lies on data obtained from mouse models with modulations of cardiac FA metabolism at different levels of the FA metabolic pathway and how these differently affect cardiac function. Based on studies in which these mouse models were exposed to ischaemic and non-ischaemic heart failure, we discuss whether and when modulations in FA metabolism are protective against heart failure.
Collapse
Affiliation(s)
- Desiree Abdurrachim
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, High Tech Campus 11, 5656 AE, PO BOX 513, Eindhoven 5600 MB, The Netherlands
| | - Joost J F P Luiken
- Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Klaas Nicolay
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, High Tech Campus 11, 5656 AE, PO BOX 513, Eindhoven 5600 MB, The Netherlands
| | - Jan F C Glatz
- Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Jeanine J Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, High Tech Campus 11, 5656 AE, PO BOX 513, Eindhoven 5600 MB, The Netherlands
| | - Miranda Nabben
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, High Tech Campus 11, 5656 AE, PO BOX 513, Eindhoven 5600 MB, The Netherlands Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
50
|
Lee J, Ellis JM, Wolfgang MJ. Adipose fatty acid oxidation is required for thermogenesis and potentiates oxidative stress-induced inflammation. Cell Rep 2015; 10:266-79. [PMID: 25578732 PMCID: PMC4359063 DOI: 10.1016/j.celrep.2014.12.023] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 11/04/2014] [Accepted: 12/11/2014] [Indexed: 12/30/2022] Open
Abstract
To understand the contribution of adipose tissue fatty acid oxidation to whole-body metabolism, we generated mice with an adipose-specific knockout of carnitine palmitoyltransferase 2 (CPT2(A-/-)), an obligate step in mitochondrial long-chain fatty acid oxidation. CPT2(A-/-) mice became hypothermic after an acute cold challenge, and CPT2(A-/-) brown adipose tissue (BAT) failed to upregulate thermogenic genes in response to agonist-induced stimulation. The adipose-specific loss of CPT2 resulted in diet-dependent changes in adiposity but did not result in changes in body weight on low- or high-fat diets. Additionally, CPT2(A-/-) mice had suppressed high-fat diet-induced oxidative stress and inflammation in visceral white adipose tissue (WAT); however, high-fat diet-induced glucose intolerance was not improved. These data show that fatty acid oxidation is required for cold-induced thermogenesis in BAT and high-fat diet-induced oxidative stress and inflammation in WAT.
Collapse
Affiliation(s)
- Jieun Lee
- Department of Biological Chemistry, Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jessica M Ellis
- Department of Biological Chemistry, Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|