1
|
Engin AB. Mechanism of Obesity-Related Lipotoxicity and Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:131-166. [PMID: 39287851 DOI: 10.1007/978-3-031-63657-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The link between cellular exposure to fatty acid species and toxicity phenotypes remains poorly understood. However, structural characterization and functional profiling of human plasma free fatty acids (FFAs) analysis has revealed that FFAs are located either in the toxic cluster or in the cluster that is transcriptionally responsive to lipotoxic stress and creates genetic risk factors. Genome-wide short hairpin RNA screen has identified more than 350 genes modulating lipotoxicity. Hypertrophic adipocytes in obese adipose are both unable to expand further to store excess lipids in the diet and are resistant to the antilipolytic action of insulin. In addition to lipolysis, the inability of packaging the excess lipids into lipid droplets causes circulating fatty acids to reach toxic levels in non-adipose tissues. Deleterious effects of accumulated lipid in non-adipose tissues are known as lipotoxicity. Although triglycerides serve a storage function for long-chain non-esterified fatty acid and their products such as ceramide and diacylglycerols (DAGs), overloading of palmitic acid fraction of saturated fatty acids (SFAs) raises ceramide levels. The excess DAG and ceramide load create harmful effects on multiple organs and systems, inducing chronic inflammation in obesity. Thus, lipotoxic inflammation results in β cells death and pancreatic islets dysfunction. Endoplasmic reticulum stress stimuli induce lipolysis by activating cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) and extracellular signal-regulated kinase (Erk) 1/2 signaling in adipocytes. However, palmitic acid-induced endoplasmic reticulum stress-c-Jun N-terminal kinase (JNK)-autophagy axis in hypertrophic adipocytes is a pro-survival mechanism against endoplasmic reticulum stress and cell death induced by SFAs. Endoplasmic reticulum-localized acyl-coenzyme A (CoA): glycerol-3-phosphate acyltransferase (GPAT) enzymes are mediators of lipotoxicity, and inhibiting these enzymes has therapeutic potential for lipotoxicity. Lipotoxicity increases the number of autophagosomes, which engulf palmitic acid, and thus suppress the autophagic turnover. Fatty acid desaturation promotes palmitate detoxification and storages into triglycerides. As therapeutic targets of glucolipotoxicity, in addition to caloric restriction and exercise, there are four different pharmacological approaches, which consist of metformin, glucagon-like peptide 1 (GLP-1) receptor agonists, peroxisome proliferator-activated receptor-gamma (PPARγ) ligands thiazolidinediones, and chaperones are still used in clinical practice. Furthermore, induction of the brown fat-like phenotype with the mixture of eicosapentanoic acid and docosahexaenoic acid appears as a potential therapeutic application for treatment of lipotoxicity.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
2
|
Wang H, Liu X, Zhou Q, Liu L, Jia Z, Qi Y, Xu F, Zhang Y. Current status and emerging trends of cardiac metabolism from the past 20 years: A bibliometric study. Heliyon 2023; 9:e21952. [PMID: 38045208 PMCID: PMC10692779 DOI: 10.1016/j.heliyon.2023.e21952] [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: 04/21/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 12/05/2023] Open
Abstract
Background Abnormal cardiac metabolism is a key factor in the development of cardiovascular diseases. Consequently, there has been considerable emphasis on researching and developing drugs that regulate metabolism. This study employed bibliometric methods to comprehensively and objectively analyze the relevant literature, offering insights into the knowledge dynamics in this field. Methods The data source for this study was the Web of Science Core Collection (WoSCC), from which the collected data were imported into bibliometric software for analysis. Results The United States was the leading contributor, accounting for 38.33 % of publications. The University of Washington and Damian J. Tyler were the most active institution and author, respectively. The American Journal of Physiology-Heart and Circulatory Physiology, Journal of Molecular and Cellular Cardiology, Cardiovascular Research, Circulation Research, and American Journal of Physiology-Endocrinology and Metabolism were highly influential journals that published numerous high-quality articles on cardiac metabolism. Common keywords in this research area included heart failure, insulin resistance, skeletal muscle, mitochondria, as well as topic words such as cardiac metabolism, fatty acid oxidation, glucose metabolism, and myocardial metabolism. Co-citation analysis has shown that research on heart failure and in vitro modeling of cardiovascular disease has gained prominence in recent years and making it a research hotspot. Conclusion Research on cardiac metabolism is steadily growing, with a specific focus on heart failure and the interplay between mitochondrial dysfunction, insulin resistance, and cardiac metabolism. An emerging trend in this field involves the enhancement of maturation in human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) through the manipulation of cardiac metabolism.
Collapse
Affiliation(s)
- Hongqin Wang
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolin Liu
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingbing Zhou
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Liu
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Zijun Jia
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Yifei Qi
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Li X, Bi X. Integrated Control of Fatty Acid Metabolism in Heart Failure. Metabolites 2023; 13:615. [PMID: 37233656 PMCID: PMC10220550 DOI: 10.3390/metabo13050615] [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: 03/27/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Disrupted fatty acid metabolism is one of the most important metabolic features in heart failure. The heart obtains energy from fatty acids via oxidation. However, heart failure results in markedly decreased fatty acid oxidation and is accompanied by the accumulation of excess lipid moieties that lead to cardiac lipotoxicity. Herein, we summarized and discussed the current understanding of the integrated regulation of fatty acid metabolism (including fatty acid uptake, lipogenesis, lipolysis, and fatty acid oxidation) in the pathogenesis of heart failure. The functions of many enzymes and regulatory factors in fatty acid homeostasis were characterized. We reviewed their contributions to the development of heart failure and highlighted potential targets that may serve as promising new therapeutic strategies.
Collapse
Affiliation(s)
| | - Xukun Bi
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China;
| |
Collapse
|
4
|
Myocardial Viability – An Important Decision Making Factor in the Treatment Protocol for Patients with Ischemic Heart Disease. ACTA MEDICA BULGARICA 2022. [DOI: 10.2478/amb-2022-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Ischemic heart disease (IHD) affects > 110 million individuals worldwide and represents an important contributor to the rise in the prevalence of heart failure and the associated mortality and morbidity. Despite modern therapies, up to one-third of patients with acute myocardial infarction would develop heart failure. IHD is a pathologic condition of the myocardium resulting from the imbalance in a given moment between its oxygen demands and the actual perfusion. Acute and chronic forms of the disease may potentially lead to extensive and permanent damage of the cardiac muscle. From a clinical point of view, determination of the still viable extent of myocardium is crucial for the therapeutic protocol – since ischemia is the underlying cause, then revascularization should provide for a better prognosis. Different methods for evaluation of myocardial viability have been described – each one presenting some advantages over the others, being, in the same time, inferior in some respects. The review offers a relatively comprehensive overview of methods available for determining myocardial viability.
Collapse
|
5
|
Heart Failure and Drug Therapies: A Metabolic Review. Int J Mol Sci 2022; 23:ijms23062960. [PMID: 35328390 PMCID: PMC8950643 DOI: 10.3390/ijms23062960] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality globally with at least 26 million people worldwide living with heart failure (HF). Metabolism has been an active area of investigation in the setting of HF since the heart demands a high rate of ATP turnover to maintain homeostasis. With the advent of -omic technologies, specifically metabolomics and lipidomics, HF pathologies have been better characterized with unbiased and holistic approaches. These techniques have identified novel pathways in our understanding of progression of HF and potential points of intervention. Furthermore, sodium-glucose transport protein 2 inhibitors, a drug that has changed the dogma of HF treatment, has one of the strongest types of evidence for a potential metabolic mechanism of action. This review will highlight cardiac metabolism in both the healthy and failing heart and then discuss the metabolic effects of heart failure drugs.
Collapse
|
6
|
Lipke K, Kubis-Kubiak A, Piwowar A. Molecular Mechanism of Lipotoxicity as an Interesting Aspect in the Development of Pathological States-Current View of Knowledge. Cells 2022; 11:cells11050844. [PMID: 35269467 PMCID: PMC8909283 DOI: 10.3390/cells11050844] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Free fatty acids (FFAs) play numerous vital roles in the organism, such as contribution to energy generation and reserve, serving as an essential component of the cell membrane, or as ligands for nuclear receptors. However, the disturbance in fatty acid homeostasis, such as inefficient metabolism or intensified release from the site of storage, may result in increased serum FFA levels and eventually result in ectopic fat deposition, which is unfavorable for the organism. The cells are adjusted for the accumulation of FFA to a limited extent and so prolonged exposure to elevated FFA levels results in deleterious effects referred to as lipotoxicity. Lipotoxicity contributes to the development of diseases such as insulin resistance, diabetes, cardiovascular diseases, metabolic syndrome, and inflammation. The nonobvious organs recognized as the main lipotoxic goal of action are the pancreas, liver, skeletal muscles, cardiac muscle, and kidneys. However, lipotoxic effects to a significant extent are not organ-specific but affect fundamental cellular processes occurring in most cells. Therefore, the wider perception of cellular lipotoxic mechanisms and their interrelation may be beneficial for a better understanding of various diseases’ pathogenesis and seeking new pharmacological treatment approaches.
Collapse
|
7
|
Ketema EB, Lopaschuk GD. Post-translational Acetylation Control of Cardiac Energy Metabolism. Front Cardiovasc Med 2021; 8:723996. [PMID: 34409084 PMCID: PMC8365027 DOI: 10.3389/fcvm.2021.723996] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 06/30/2021] [Indexed: 12/17/2022] Open
Abstract
Perturbations in myocardial energy substrate metabolism are key contributors to the pathogenesis of heart diseases. However, the underlying causes of these metabolic alterations remain poorly understood. Recently, post-translational acetylation-mediated modification of metabolic enzymes has emerged as one of the important regulatory mechanisms for these metabolic changes. Nevertheless, despite the growing reports of a large number of acetylated cardiac mitochondrial proteins involved in energy metabolism, the functional consequences of these acetylation changes and how they correlate to metabolic alterations and myocardial dysfunction are not clearly defined. This review summarizes the evidence for a role of cardiac mitochondrial protein acetylation in altering the function of major metabolic enzymes and myocardial energy metabolism in various cardiovascular disease conditions.
Collapse
Affiliation(s)
- Ezra B Ketema
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| | - Gary D Lopaschuk
- Department of Pediatrics, Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
8
|
Pasqua T, Rocca C, Giglio A, Angelone T. Cardiometabolism as an Interlocking Puzzle between the Healthy and Diseased Heart: New Frontiers in Therapeutic Applications. J Clin Med 2021; 10:721. [PMID: 33673114 PMCID: PMC7918460 DOI: 10.3390/jcm10040721] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiac metabolism represents a crucial and essential connecting bridge between the healthy and diseased heart. The cardiac muscle, which may be considered an omnivore organ with regard to the energy substrate utilization, under physiological conditions mainly draws energy by fatty acids oxidation. Within cardiomyocytes and their mitochondria, through well-concerted enzymatic reactions, substrates converge on the production of ATP, the basic chemical energy that cardiac muscle converts into mechanical energy, i.e., contraction. When a perturbation of homeostasis occurs, such as an ischemic event, the heart is forced to switch its fatty acid-based metabolism to the carbohydrate utilization as a protective mechanism that allows the maintenance of its key role within the whole organism. Consequently, the flexibility of the cardiac metabolic networks deeply influences the ability of the heart to respond, by adapting to pathophysiological changes. The aim of the present review is to summarize the main metabolic changes detectable in the heart under acute and chronic cardiac pathologies, analyzing possible therapeutic targets to be used. On this basis, cardiometabolism can be described as a crucial mechanism in keeping the physiological structure and function of the heart; furthermore, it can be considered a promising goal for future pharmacological agents able to appropriately modulate the rate-limiting steps of heart metabolic pathways.
Collapse
Affiliation(s)
- Teresa Pasqua
- Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
| | - Anita Giglio
- Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy;
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
- National Institute of Cardiovascular Research (I.N.R.C.), 40126 Bologna, Italy
| |
Collapse
|
9
|
Allosteric, transcriptional and post-translational control of mitochondrial energy metabolism. Biochem J 2019; 476:1695-1712. [PMID: 31217327 DOI: 10.1042/bcj20180617] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/24/2022]
Abstract
The heart is the organ with highest energy turnover rate (per unit weight) in our body. The heart relies on its flexible and powerful catabolic capacity to continuously generate large amounts of ATP utilizing many energy substrates including fatty acids, carbohydrates (glucose and lactate), ketones and amino acids. The normal health mainly utilizes fatty acids (40-60%) and glucose (20-40%) for ATP production while ketones and amino acids have a minor contribution (10-15% and 1-2%, respectively). Mitochondrial oxidative phosphorylation is the major contributor to cardiac energy production (95%) while cytosolic glycolysis has a marginal contribution (5%). The heart can dramatically and swiftly switch between energy-producing pathways and/or alter the share from each of the energy substrates based on cardiac workload, availability of each energy substrate and neuronal and hormonal activity. The heart is equipped with a highly sophisticated and powerful mitochondrial machinery which synchronizes cardiac energy production from different substrates and orchestrates the rate of ATP production to accommodate its contractility demands. This review discusses mitochondrial cardiac energy metabolism and how it is regulated. This includes a discussion on the allosteric control of cardiac energy metabolism by short-chain coenzyme A esters, including malonyl CoA and its effect on cardiac metabolic preference. We also discuss the transcriptional level of energy regulation and its role in the maturation of cardiac metabolism after birth and cardiac adaptability for different metabolic conditions and energy demands. The role post-translational modifications, namely phosphorylation, acetylation, malonylation, succinylation and glutarylation, play in regulating mitochondrial energy metabolism is also discussed.
Collapse
|
10
|
Brewer RA, Collins HE, Berry RD, Brahma MK, Tirado BA, Peliciari-Garcia RA, Stanley HL, Wende AR, Taegtmeyer H, Rajasekaran NS, Darley-Usmar V, Zhang J, Frank SJ, Chatham JC, Young ME. Temporal partitioning of adaptive responses of the murine heart to fasting. Life Sci 2018; 197:30-39. [PMID: 29410090 DOI: 10.1016/j.lfs.2018.01.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 12/16/2022]
Abstract
Recent studies suggest that the time of day at which food is consumed dramatically influences clinically-relevant cardiometabolic parameters (e.g., adiposity, insulin sensitivity, and cardiac function). Meal feeding benefits may be the result of daily periods of feeding and/or fasting, highlighting the need for improved understanding of the temporal adaptation of cardiometabolic tissues (e.g., heart) to fasting. Such studies may provide mechanistic insight regarding how time-of-day-dependent feeding/fasting cycles influence cardiac function. We hypothesized that fasting during the sleep period elicits beneficial adaptation of the heart at transcriptional, translational, and metabolic levels. To test this hypothesis, temporal adaptation was investigated in wild-type mice fasted for 24-h, or for either the 12-h light/sleep phase or the 12-h dark/awake phase. Fasting maximally induced fatty acid responsive genes (e.g., Pdk4) during the dark/active phase; transcriptional changes were mirrored at translational (e.g., PDK4) and metabolic flux (e.g., glucose/oleate oxidation) levels. Similarly, maximal repression of myocardial p-mTOR and protein synthesis rates occurred during the dark phase; both parameters remained elevated in the heart of fasted mice during the light phase. In contrast, markers of autophagy (e.g., LC3II) exhibited peak responses to fasting during the light phase. Collectively, these data show that responsiveness of the heart to fasting is temporally partitioned. Autophagy peaks during the light/sleep phase, while repression of glucose utilization and protein synthesis is maximized during the dark/active phase. We speculate that sleep phase fasting may benefit cardiac function through augmentation of protein/cellular constituent turnover.
Collapse
Affiliation(s)
- Rachel A Brewer
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Helen E Collins
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ryan D Berry
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Manoja K Brahma
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brian A Tirado
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rodrigo A Peliciari-Garcia
- Morphophysiology & Pathology Sector, Department of Biological Sciences, Federal University of São Paulo, Diadema, SP, Brazil
| | - Haley L Stanley
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Adam R Wende
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School UT Health Science Center, Houston, TX, USA
| | - Namakkal Soorappan Rajasekaran
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor Darley-Usmar
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianhua Zhang
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stuart J Frank
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Endocrinology Section, Birmingham VAMC Medical Service, Birmingham, AL, USA
| | - John C Chatham
- Division of Molecular Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
11
|
DuSablon A, Parks J, Whitehurst K, Estes H, Chase R, Vlahos E, Sharma U, Wert D, Virag J. EphrinA1-Fc attenuates myocardial ischemia/reperfusion injury in mice. PLoS One 2017; 12:e0189307. [PMID: 29236774 PMCID: PMC5728502 DOI: 10.1371/journal.pone.0189307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/22/2017] [Indexed: 12/19/2022] Open
Abstract
EphrinA1, a membrane-bound receptor tyrosine kinase ligand expressed in healthy cardiomyocytes, is lost in injured cells following myocardial infarction. Previously, we have reported that a single intramyocardial injection of chimeric ephrinA1-Fc at the time of ischemia reduced injury in the nonreperfused myocardium by 50% at 4 days post-MI by reducing apoptosis and inflammatory cell infiltration. In a clinically relevant model of acute ischemia (30min)/reperfusion (24hr or 4 days) injury, we now demonstrate that ephrinA1-Fc reduces infarct size by 46% and completely preserves cardiac function (ejection fraction, fractional shortening, and chamber dimensions) in the short-term (24hrs post-MI) as well as long-term (4 days). At 24 hours post-MI, diminished serum inflammatory cell chemoattractants in ephrinA1-Fc-treated mice reduces recruitment of neutrophils and leukocytes into the myocardium. Differences in relative expression levels of EphA-Rs are described in the context of their putative role in mediating cardioprotection. Validation by Western blotting of selected targets from mass spectrometry analyses of pooled samples of left ventricular tissue homogenates from mice that underwent 30min ischemia and 24hr of reperfusion (I/R) indicates that ephrinA1-Fc administration alters several regulators of signaling pathways that attenuate apoptosis, promote autophagy, and shift from FA metabolism in favor of increased glycolysis to optimize anaerobic ATP production. Taken together, reduced injury is due a combination of adaptive metabolic reprogramming, improved cell survival, and decreased inflammatory cell recruitment, suggesting that ephrinA1-Fc enhances the capacity of the heart to withstand an ischemic insult.
Collapse
Affiliation(s)
- Augustin DuSablon
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Justin Parks
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - K’Shylah Whitehurst
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Heather Estes
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Robert Chase
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Eleftherios Vlahos
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Uma Sharma
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - David Wert
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Jitka Virag
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
12
|
PPARγ Modulates Long Chain Fatty Acid Processing in the Intestinal Epithelium. Int J Mol Sci 2017; 18:ijms18122559. [PMID: 29182565 PMCID: PMC5751162 DOI: 10.3390/ijms18122559] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/26/2022] Open
Abstract
Nuclear receptor PPARγ affects lipid metabolism in several tissues, but its role in intestinal lipid metabolism has not been explored. As alterations have been observed in the plasma lipid profile of ad libitum fed intestinal epithelium-specific PPARγ knockout mice (iePPARγKO), we submitted these mice to lipid gavage challenges. Within hours after gavage with long chain unsaturated fatty acid (FA)-rich canola oil, the iePPARγKO mice had higher plasma free FA levels and lower gastric inhibitory polypeptide levels than their wild-type (WT) littermates, and altered expression of incretin genes and lipid metabolism-associated genes in the intestinal epithelium. Gavage with the medium chain saturated FA-rich coconut oil did not result in differences between the two genotypes. Furthermore, the iePPARγKO mice did not exhibit defective lipid uptake and stomach emptying; however, their intestinal transit was more rapid than in WT mice. When fed a canola oil-rich diet for 4.5 months, iePPARγKO mice had higher body lean mass than the WT mice. We conclude that intestinal epithelium PPARγ is activated preferentially by long chain unsaturated FAs compared to medium chain saturated FAs. Furthermore, we hypothesize that the iePPARγKO phenotype originates from altered lipid metabolism and release in epithelial cells, as well as changes in intestinal motility.
Collapse
|
13
|
Abstract
Enlarged fat cells in obese adipose tissue diminish capacity to store fat and are resistant to the anti-lipolytic effect of insulin. Insulin resistance (IR)-associated S-nitrosylation of insulin-signaling proteins increases in obesity. In accordance with the inhibition of insulin-mediated anti-lipolytic action, plasma free fatty acid (FFA) levels increase. Additionally, endoplasmic reticulum stress stimuli induce lipolysis by activating cyclic adenosine monophosphate/Protein kinase A (cAMP/PKA) and extracellular signal-regulated kinase ½ (ERK1/2) signaling in adipocytes. Failure of packaging of excess lipid into lipid droplets causes chronic elevation of circulating fatty acids, which can reach to toxic levels within non-adipose tissues. Deleterious effects of lipid accumulation in non-adipose tissues are known as lipotoxicity. In fact, triglycerides may also serve a storage function for long-chain non-esterified fatty acids and their products such as ceramides and diacylglycerols (DAGs). Thus, excess DAG, ceramide and saturated fatty acids in obesity can induce chronic inflammation and have harmful effect on multiple organs and systems. In this context, chronic adipose tissue inflammation, mitochondrial dysfunction and IR have been discussed within the scope of lipotoxicity.
Collapse
|
14
|
Abstract
The heart is a biological pump that converts chemical to mechanical energy. This process of energy conversion is highly regulated to the extent that energy substrate metabolism matches energy use for contraction on a beat-to-beat basis. The biochemistry of cardiac metabolism includes the biochemistry of energy transfer, metabolic regulation, and transcriptional, translational as well as posttranslational control of enzymatic activities. Pathways of energy substrate metabolism in the heart are complex and dynamic, but all of them conform to the First Law of Thermodynamics. The perspectives expand on the overall idea that cardiac metabolism is inextricably linked to both physiology and molecular biology of the heart. The article ends with an outlook on emerging concepts of cardiac metabolism based on new molecular models and new analytical tools. © 2016 American Physiological Society. Compr Physiol 6:1675-1699, 2016.
Collapse
Affiliation(s)
- Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| | - Truong Lam
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| | - Giovanni Davogustto
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| |
Collapse
|
15
|
Fukushima A, Lopaschuk GD. Cardiac fatty acid oxidation in heart failure associated with obesity and diabetes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1525-34. [PMID: 26996746 DOI: 10.1016/j.bbalip.2016.03.020] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 12/01/2022]
Abstract
Obesity and diabetes are major public health problems, and are linked to the development of heart failure. Emerging data highlight the importance of alterations in cardiac energy metabolism as a major contributor to cardiac dysfunction related to obesity and diabetes. Increased rates of fatty acid oxidation and decreased rates of glucose utilization are two prominent changes in cardiac energy metabolism that occur in obesity and diabetes. This metabolic profile is probably both a cause and consequence of a prominent cardiac insulin resistance, which is accompanied by a decrease in both cardiac function and efficiency, and by the accumulation of potentially toxic lipid metabolites in the heart that can further exaggerate insulin resistance and cardiac dysfunction. The high cardiac fatty acid oxidation rates seen in obesity and diabetes are attributable to several factors, including: 1) increased fatty acid supply and uptake into the cardiomyocyte, 2) increased transcription of fatty acid metabolic enzymes, 3) decreased allosteric control of mitochondrial fatty acid uptake and fatty acid oxidation, and 4) increased post-translational acetylation control of various fatty acid oxidative enzymes. Emerging evidence suggests that therapeutic approaches aimed at switching the balance of cardiac energy substrate preference from fatty acid oxidation to glucose use can prevent cardiac dysfunction associated with obesity and diabetes. Modulating acetylation control of fatty acid oxidative enzymes is also a potentially attractive strategy, although presently this is limited to precursors of nicotinamide adenine or nonspecific activators of deacetylation such as resveratrol. This review will focus on the metabolic alterations in the heart that occur in obesity and diabetes, as well as on the molecular mechanisms controlling these metabolic changes. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.
Collapse
Affiliation(s)
- Arata Fukushima
- Cardiovascular Translational Science Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Cardiovascular Translational Science Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
16
|
Abstract
Reversible acetylation was initially described as an epigenetic mechanism regulating DNA accessibility. Since then, this process has emerged as a controller of histone and nonhistone acetylation that integrates key physiological processes such as metabolism, circadian rhythm and cell cycle, along with gene regulation in various organisms. The widespread and reversible nature of acetylation also revitalized interest in the mechanisms that regulate lysine acetyltransferases (KATs) and deacetylases (KDACs) in health and disease. Changes in protein or histone acetylation are especially relevant for many common diseases including obesity, diabetes mellitus, neurodegenerative diseases and cancer, as well as for some rare diseases such as mitochondrial diseases and lipodystrophies. In this Review, we examine the role of reversible acetylation in metabolic control and how changes in levels of metabolites or cofactors, including nicotinamide adenine dinucleotide, nicotinamide, coenzyme A, acetyl coenzyme A, zinc and butyrate and/or β-hydroxybutyrate, directly alter KAT or KDAC activity to link energy status to adaptive cellular and organismal homeostasis.
Collapse
Affiliation(s)
- Keir J Menzies
- Interdisciplinary School of Health Sciences, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | - Hongbo Zhang
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Station 15, 1015 Lausanne, Switzerland
| | - Elena Katsyuba
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Station 15, 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Station 15, 1015 Lausanne, Switzerland
| |
Collapse
|
17
|
Cheng W, Wu P, Du Y, Wang Y, Zhou N, Ge Y, Yang Z. Puerarin improves cardiac function through regulation of energy metabolism in Streptozotocin-Nicotinamide induced diabetic mice after myocardial infarction. Biochem Biophys Res Commun 2015; 463:1108-14. [PMID: 26079885 DOI: 10.1016/j.bbrc.2015.06.067] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/09/2015] [Indexed: 12/29/2022]
Abstract
It is well recognized that the incidence of heart failure and the risk of death is high in diabetic patients after myocardial infarction (MI). Accumulating evidence showed that puerarin (PUE) has protecting function on both cardiovascular disease and diabetes. The aim of this study is to explore whether puerarin could improve cardiac function in diabetic mice after MI and the underlying mechanism. The left anterior of Streptozotocin (STZ)-Nicotinamide (NA) induced diabetic mice were ligated permanently except for the Shame group. Then the operated mice were randomly treated with PUE or saline. Cardiac function was evaluated by echocardiograph before and at 1, 2, 4 weeks after MI. GLUT4/CD36/p-Akt/PPAR α of the heart was examined after treatment for 4 weeks. The results indicated that PUE significantly increased survival rate, improved cardiac function compared with MI group. Moreover, PUE increased expression and translocation of GLUT4 while attenuated expression and translocation of CD36. Western blot analysis showed that PUE enhanced phosphorylation of Akt and decreased PPAR α. This study demonstrated that PUE improved cardiac function after MI in diabetic mice through regulation of energy metabolism, the possible mechanism responsible for the effect of PUE was increasing the expression and translocation of GLUT4 while attenuating the expression and translocation of CD36.
Collapse
Affiliation(s)
- Weili Cheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peng Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yingqiang Du
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunle Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ningtian Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yingbin Ge
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Zhijian Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
18
|
Role of CoA and acetyl-CoA in regulating cardiac fatty acid and glucose oxidation. Biochem Soc Trans 2015; 42:1043-51. [PMID: 25110000 DOI: 10.1042/bst20140094] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CoA (coenzyme A) and its derivatives have a critical role in regulating cardiac energy metabolism. This includes a key role as a substrate and product in the energy metabolic pathways, as well as serving as an allosteric regulator of cardiac energy metabolism. In addition, the CoA ester malonyl-CoA has an important role in regulating fatty acid oxidation, secondary to inhibiting CPT (carnitine palmitoyltransferase) 1, a key enzyme involved in mitochondrial fatty acid uptake. Alterations in malonyl-CoA synthesis by ACC (acetyl-CoA carboxylase) and degradation by MCD (malonyl-CoA decarboxylase) are important contributors to the high cardiac fatty acid oxidation rates seen in ischaemic heart disease, heart failure, obesity and diabetes. Additional control of fatty acid oxidation may also occur at the level of acetyl-CoA involvement in acetylation of mitochondrial fatty acid β-oxidative enzymes. We find that acetylation of the fatty acid β-oxidative enzymes, LCAD (long-chain acyl-CoA dehydrogenase) and β-HAD (β-hydroxyacyl-CoA dehydrogenase) is associated with an increase in activity and fatty acid oxidation in heart from obese mice with heart failure. This is associated with decreased SIRT3 (sirtuin 3) activity, an important mitochondrial deacetylase. In support of this, cardiac SIRT3 deletion increases acetylation of LCAD and β-HAD, and increases cardiac fatty acid oxidation. Acetylation of MCD is also associated with increased activity, decreases malonyl-CoA levels and an increase in fatty acid oxidation. Combined, these data suggest that malonyl-CoA and acetyl-CoA have an important role in mediating the alterations in fatty acid oxidation seen in heart failure.
Collapse
|
19
|
Benítez R, Núñez Y, Fernández A, Isabel B, Fernández AI, Rodríguez C, Barragán C, Martín-Palomino P, López-Bote C, Silió L, Óvilo C. Effects of dietary fat saturation on fatty acid composition and gene transcription in different tissues of Iberian pigs. Meat Sci 2014; 102:59-68. [PMID: 25549539 DOI: 10.1016/j.meatsci.2014.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 10/24/2022]
Abstract
The effect of two diets, respectively enriched with SFA (S) and PUFA (P), on FA tissue composition and gene expression was studied in fattened Iberian pigs. The FA composition of adipose, muscular and liver tissues was affected by dietary treatment. S group showed higher MUFA and MUFA/SFA ratio and lower PUFA and n-6/n-3 ratio than P group in all analyzed tissues. In muscle and liver the extracted lipids were separated into neutral lipids and polar lipid fractions which showed significantly different responses to the dietary treatment, especially in liver where no significant effect of diet was observed in NL fraction. The expression of six candidate genes related to lipogenesis and FA oxidation was analyzed by qPCR. In liver, stearoyl CoA desaturase (SCD), acetyl CoA carboxylase alpha (ACACA) and malic enzyme 1 (ME1) genes showed higher expression in S group. SCD, ACACA, ME1, and fatty acid synthase (FASN) gene expression levels showed a wide variation across the tested tissues, with much higher expression levels observed in adipose tissue than other tissues. Tissue FA profile and gene expression results support the deposition of dietary FA, the lipogenic effect of dietary saturated fat in liver and the employment of saturated dietary fat for endogenous synthesis of MUFA in all the analyzed tissues.
Collapse
Affiliation(s)
- R Benítez
- Departamento Mejora Genética Animal, INIA, Ctra. Coruña Km 7.5, 28040 Madrid, Spain.
| | - Y Núñez
- Departamento Mejora Genética Animal, INIA, Ctra. Coruña Km 7.5, 28040 Madrid, Spain.
| | - A Fernández
- Departamento Mejora Genética Animal, INIA, Ctra. Coruña Km 7.5, 28040 Madrid, Spain.
| | - B Isabel
- Departamento Producción Animal, Facultad de Veterinaria, UCM, 28040 Madrid, Spain.
| | - A I Fernández
- Departamento Mejora Genética Animal, INIA, Ctra. Coruña Km 7.5, 28040 Madrid, Spain.
| | - C Rodríguez
- Departamento Mejora Genética Animal, INIA, Ctra. Coruña Km 7.5, 28040 Madrid, Spain.
| | - C Barragán
- Departamento Mejora Genética Animal, INIA, Ctra. Coruña Km 7.5, 28040 Madrid, Spain.
| | - P Martín-Palomino
- Universidad Alfonso X El Sabio, Campus de Villanueva de la Cañada, Avda. de la Universidad 1, 28691 Villanueva de la Cañada, Madrid, Spain.
| | - C López-Bote
- Departamento Producción Animal, Facultad de Veterinaria, UCM, 28040 Madrid, Spain.
| | - L Silió
- Departamento Mejora Genética Animal, INIA, Ctra. Coruña Km 7.5, 28040 Madrid, Spain.
| | - C Óvilo
- Departamento Mejora Genética Animal, INIA, Ctra. Coruña Km 7.5, 28040 Madrid, Spain.
| |
Collapse
|
20
|
Abstract
Diabetic cardiomyopathy (DCM) is defined as cardiac disease independent of vascular complications during diabetes. The number of new cases of DCM is rising at epidemic rates in proportion to newly diagnosed cases of diabetes mellitus (DM) throughout the world. DCM is a heart failure syndrome found in diabetic patients that is characterized by left ventricular hypertrophy and reduced diastolic function, with or without concurrent systolic dysfunction, occurring in the absence of hypertension and coronary artery disease. DCM and other diabetic complications are caused in part by elevations in blood glucose and lipids, characteristic of DM. Although there are pathological consequences to hyperglycemia and hyperlipidemia, the combination of the two metabolic abnormalities potentiates the severity of diabetic complications. A natural competition exists between glucose and fatty acid metabolism in the heart that is regulated by allosteric and feedback control and transcriptional modulation of key limiting enzymes. Inhibition of these glycolytic enzymes not only controls flux of substrate through the glycolytic pathway, but also leads to the diversion of glycolytic intermediate substrate through pathological pathways, which mediate the onset of diabetic complications. The present review describes the limiting steps involved in the development of these pathological pathways and the factors involved in the regulation of these limiting steps. Additionally, therapeutic options with demonstrated or postulated effects on DCM are described.
Collapse
Affiliation(s)
- Michael Isfort
- The Ohio State University College of Medicine, Columbus, OH, USA
| | | | | | | | | |
Collapse
|
21
|
Fillmore N, Lopaschuk GD. Malonyl CoA: A promising target for the treatment of cardiac disease. IUBMB Life 2014; 66:139-146. [DOI: 10.1002/iub.1253] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/14/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Natasha Fillmore
- Cardiovascular Research Centre; Mazankowski Alberta Heart Institute; University of Alberta; Edmonton AB Canada
| | - Gary D. Lopaschuk
- Cardiovascular Research Centre; Mazankowski Alberta Heart Institute; University of Alberta; Edmonton AB Canada
| |
Collapse
|
22
|
Óvilo C, Benítez R, Fernández A, Isabel B, Núñez Y, Fernández AI, Rodríguez C, Daza A, Silió L, López-Bote C. Dietary energy source largely affects tissue fatty acid composition but has minor influence on gene transcription in Iberian pigs. J Anim Sci 2014; 92:939-54. [PMID: 24492573 DOI: 10.2527/jas.2013-6988] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A trial was performed to compare the effects of different dietary sources of MUFA on the fatty acid (FA) composition, lipid metabolism, and gene transcription in different tissues of Iberian pigs. Twenty-seven Iberian male pigs of 28 kg live weight (LW) were divided in 2 groups and fed with 1 of 2 isocaloric diets: a standard diet with carbohydrates as energy source (CH) and a diet enriched with high-oleic sunflower oil (HO). Ham adipose tissue was sampled by biopsy at 44 and 70 kg LW. At 110 kg LW pigs were slaughtered and backfat, loin, and liver tissues were sampled. Animals of the HO group showed higher MUFA content and lower SFA in all the analyzed tissues (P < 0.001). These main effects were established early during the treatment and increased only slightly along time. Small diet effects were also detected on PUFA, which showed differences according to sampling time, tissue, and lipid fraction. Effects of diet on gene expression were explored with a combined approach analyzing adipose tissue transcriptome and quantifying the expression of a panel of key genes implicated in lipogenesis and lipid metabolism processes in backfat, muscle, and liver. Backfat transcriptome showed small effects of diet on gene expression, in number and magnitude. According to the posterior probabilities (PP) of the probe-specific expression differences between dietary groups (PP < 0.01), 37 genes were considered differentially expressed (DE). Gene ontology allowed relating them with several biological functions including lipid metabolic processes. Quantitative PCR confirmed several DE genes in adipose tissue (RXRG, LEP, and ME1; P < 0.0001, P < 0.05, and P < 0.0001, respectively), but no DE gene was found in loin or liver tissues. Joint results agree with a metabolic adjustment of adipose tissue FA levels by the subtle effect of the diet on the regulation of several lipid metabolism pathways, mainly FA oxidation and prostanoid synthesis, with LEP, RXRG, and PTGS2 genes playing mayor roles.
Collapse
Affiliation(s)
- C Óvilo
- Departamento de Mejora Genética Animal, INIA, 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
MIZUNOYA W, SAWANO S, IWAMOTO Y, SATO Y, TATSUMI R, IKEUCHI Y. Effect of 48-h Food Deprivation on the Expressions of Myosin Heavy-Chain Isoforms and Fiber Type-Related Factors in Rats. J Nutr Sci Vitaminol (Tokyo) 2013; 59:289-98. [DOI: 10.3177/jnsv.59.289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
24
|
Soares FLP, de Oliveira Matoso R, Teixeira LG, Menezes Z, Pereira SS, Alves AC, Batista NV, de Faria AMC, Cara DC, Ferreira AVM, Alvarez-Leite JI. Gluten-free diet reduces adiposity, inflammation and insulin resistance associated with the induction of PPAR-alpha and PPAR-gamma expression. J Nutr Biochem 2012; 24:1105-11. [PMID: 23253599 DOI: 10.1016/j.jnutbio.2012.08.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 07/20/2012] [Accepted: 08/13/2012] [Indexed: 12/20/2022]
Abstract
Gluten exclusion (protein complex present in many cereals) has been proposed as an option for the prevention of diseases other than coeliac disease. However, the effects of gluten-free diets on obesity and its mechanisms of action have not been studied. Thus, our objective was to assess whether gluten exclusion can prevent adipose tissue expansion and its consequences. C57BL/6 mice were fed a high-fat diet containing 4.5% gluten (Control) or no gluten (GF). Body weight and adiposity gains, leukocyte rolling and adhesion, macrophage infiltration and cytokine production in adipose tissue were assessed. Blood lipid profiles, glycaemia, insulin resistance and adipokines were measured. Expression of the PPAR-α and γ, lipoprotein lipase (LPL), hormone sensitive lipase (HSL), carnitine palmitoyl acyltransferase-1 (CPT-1), insulin receptor, GLUT-4 and adipokines were assessed in epidydimal fat. Gluten-free animals showed a reduction in body weight gain and adiposity, without changes in food intake or lipid excretion. These results were associated with up-regulation of PPAR-α, LPL, HSL and CPT-1, which are related to lipolysis and fatty acid oxidation. There was an improvement in glucose homeostasis and pro-inflammatory profile-related overexpression of PPAR-γ. Moreover, intravital microscopy showed a lower number of adhered cells in the adipose tissue microvasculature. The overexpression of PPAR-γ is related to the increase of adiponectin and GLUT-4. Our data support the beneficial effects of gluten-free diets in reducing adiposity gain, inflammation and insulin resistance. The data suggests that diet gluten exclusion should be tested as a new dietary approach to prevent the development of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Fabíola Lacerda Pires Soares
- Departamento de Alimentos, Faculdade de Farmácia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
McAinch AJ, Lee JS, Bruce CR, Tunstall RJ, Hawley JA, Cameron-Smith D. Dietary Regulation of Fat Oxidative Gene Expression in Different Skeletal Muscle Fiber Types. ACTA ACUST UNITED AC 2012; 11:1471-9. [PMID: 14694211 DOI: 10.1038/oby.2003.197] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To determine the effect of a high-fat diet on the expression of genes important for fat oxidation, the protein abundance of the transcription factors peroxisome proliferator-activated receptor (PPAR) isoforms alpha and gamma, and selected enzyme activities in type I and II skeletal muscle. RESEARCH METHODS AND PROCEDURES Sprague-Dawley rats consumed either a high-fat (HF: 78% energy, n = 8) or high-carbohydrate (64% energy, n = 8) diet for 8 weeks while remaining sedentary. RESULTS The expression of genes important for fat oxidation tended to increase in both type I (soleus) and type II (extensor digitorum longus) fiber types after an HF dietary intervention. However, the expression of muscle type carnitine palmitoyltransferase I was not increased in extensor digitorum longus. Analysis of the gene expression of both peroxisome proliferator-activated receptor-gamma coactivator and fork-head transcription factor O1 demonstrated no alteration in response to the HF diet. Similarly, PPARalpha and PPARgamma protein levels were also not altered by the HF diet. DISCUSSION An HF diet increased the expression of an array of genes involved in lipid metabolism, with only subtle differences evident in the response within differing skeletal muscle fiber types. Despite changes in gene expression, there were no effects of diet on peroxisome proliferator-activated receptor-gamma coactivator and fork-head transcription factor O1 mRNA and the protein abundance of PPARalpha and PPARgamma.
Collapse
MESH Headings
- 3-Hydroxyacyl CoA Dehydrogenases/genetics
- 3-Hydroxyacyl CoA Dehydrogenases/metabolism
- Animals
- Blotting, Western
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- Carnitine O-Palmitoyltransferase/genetics
- Carnitine O-Palmitoyltransferase/metabolism
- DNA-Binding Proteins
- Dietary Fats/metabolism
- Dietary Fats/pharmacology
- Female
- Forkhead Transcription Factors
- Gene Expression Regulation, Enzymologic/physiology
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Nerve Tissue Proteins
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Protein Isoforms
- Protein Kinases/genetics
- Protein Kinases/metabolism
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Andrew J McAinch
- School of Health Sciences, Deakin University, Burwood, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
26
|
Hussein AAM, Abdel-Aziz A, Gabr M, Hemmaid KZ. Myocardial and metabolic dysfunction in type 2 diabetic rats: impact of ghrelin. Can J Physiol Pharmacol 2012; 90:99-111. [DOI: 10.1139/y11-103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Diabetes mellitus (DM) is commonly associated with metabolic and cardiac dysfunctions. The aim of this study was to examine the effect of ghrelin on metabolic and cardiac dysfunctions in a type-2 diabetes mellitus (T2DM) rat model. For this, 48 male adult Sprague–Dawley rats were divided equally into 4 groups: Group I, fed normal chow, served as normal control group; Groups II–IV, were fed a high-fat diet for 2 weeks followed by injection of streptozotocin (STZ) (35 mg/kg body mass) to create a model of T2DM; Group II, were not treated; Group III, were treated with the vehicle (saline); Group IV, were treated with ghrelin (40 µg/kg body mass) twice daily for 10 days. The untreated diabetic rats showed a significant increase in serum fasting blood glucose, insulin homeostasis model assessment (HOMA) index, triglycerides (TGs), low-density lipoprotein cholesterol (LDL-C), total serum cholesterol (TC), and body mass, with a decrease in high-density lipoprotein cholesterol (HDL-C) (p < 0.05). Hearts isolated from diabetic rats showed a significant increase in myocardial fat content, a significant decrease in GLUT4, and an increase in acyl-CoA oxidase enzyme mRNA (p < 0.05). Ghrelin administration for 10 days caused a significant improvement in lipid profile, HOMA index, and body mass, and significantly corrected the myocardial mass, significantly reduced the fat content of the myocardium, significantly increased GLUT4, and decreased acyl CoA oxidase mRNA (p < 0.05). Thus, ghrelin improves both the metabolic functions and the disturbed energy metabolism in the cardiac muscle of obese diabetic rats.
Collapse
Affiliation(s)
| | - Azza Abdel-Aziz
- Department of Pathology, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Mahmoud Gabr
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Kamel Z. Hemmaid
- Department of Zoology, Faculty of Science, Zagazig University, Egypt
| |
Collapse
|
27
|
Zeng T, Xie KQ. Ethanol and liver: recent advances in the mechanisms of ethanol-induced hepatosteatosis. Arch Toxicol 2011; 83:1075-81. [PMID: 19588123 DOI: 10.1007/s00204-009-0457-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 06/29/2009] [Indexed: 12/20/2022]
Abstract
Ethanol-induced fatty liver is a worldwide health problem without effective therapeutic methods. The underlying mechanisms are extremely complex and not fully understood. The hepatosteatosis caused by ethanol can be attributed to many factors, including the changes of the redox condition, transportation impairment of the synthesized lipid, inhibition of fatty acid oxidation, and the enhancement of the lipogenesis. Recent studies focus on the reduced oxidation of fatty acid and the enhancement of the do novo lipogenesis, and several factors are sequentially revealed. Two important nuclear transcription factors, peroxisome proliferators-activated receptor α (PPARα) and sterol regulatory element binding protein-1 (SREBP-1), and the lipid metabolism-associated enzymes regulated by the two molecules, are shown to be involved in ethanol-induced steatosis. The AMP-dependent protein kinase, adiponectin, and tumor necrosis factor α (TNF-α) may mediate the modulation of ethanol on PPARα and SREBP-1. In addition, a number of studies demonstrate that plasminogen activator inhibitor-1 (PAI-1) is also involved in ethanol- induced fatty liver, and its effects may be associated with the TNF-α production. Furthermore, the role of CYP2E1 has also been investigated. Some studies showed that CYP2E1 played a critical role in the development of alcoholic fatty liver, which was denied by other reports. As such, the exact role of CYP2E1 needs to be further studied.
Collapse
Affiliation(s)
- Tao Zeng
- Institute of Toxicology, Shandong University, 44 Wenhua West Road, Jinan, Shandong 250012, People's Republic of China.
| | | |
Collapse
|
28
|
Lopaschuk GD, Ussher JR, Jaswal JS. Targeting intermediary metabolism in the hypothalamus as a mechanism to regulate appetite. Pharmacol Rev 2010; 62:237-64. [PMID: 20392806 DOI: 10.1124/pr.109.002428] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The central nervous system mediates energy balance (energy intake and energy expenditure) in the body; the hypothalamus has a key role in this process. Recent evidence has demonstrated an important role for hypothalamic malonyl CoA in mediating energy balance. Malonyl CoA is generated by the carboxylation of acetyl CoA by acetyl CoA carboxylase and is then either incorporated into long-chain fatty acids by fatty acid synthase, or converted back to acetyl-CoA by malonyl CoA decarboxylase. Increased hypothalamic malonyl CoA is an indicator of energy surplus, resulting in a decrease in food intake and an increase in energy expenditure. In contrast, a decrease in hypothalamic malonyl CoA signals an energy deficit, resulting in an increased appetite and a decrease in body energy expenditure. A number of hormonal and neural orexigenic and anorexigenic signaling pathways have now been shown to be associated with changes in malonyl CoA levels in the arcuate nucleus (ARC) of the hypothalamus. Despite compelling evidence that malonyl CoA is an important mediator in the hypothalamic ARC control of food intake and regulation of energy balance, the mechanism(s) by which this occurs has not been established. Malonyl CoA inhibits carnitine palmitoyltransferase-1 (CPT-1), and it has been proposed that the substrate of CPT-1, long-chain acyl CoA(s), may act as a mediator(s) of appetite and energy balance. However, recent evidence has challenged the role of long-chain acyl CoA(s) in this process, as well as the involvement of CPT-1 in hypothalamic malonyl CoA signaling. A better understanding of how malonyl CoA regulates energy balance should provide novel approaches to targeting intermediary metabolism in the hypothalamus as a mechanism to control appetite and body weight. Here, we review the data supporting an important role for malonyl CoA in mediating hypothalamic control of energy balance, and recent evidence suggesting that targeting malonyl CoA synthesis or degradation may be a novel approach to favorably modify appetite and weight gain.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- 423 Heritage Medical Research Center, University of Alberta, Edmonton, Canada T6G2S2.
| | | | | |
Collapse
|
29
|
Lopaschuk GD, Ussher JR, Folmes CDL, Jaswal JS, Stanley WC. Myocardial fatty acid metabolism in health and disease. Physiol Rev 2010; 90:207-58. [PMID: 20086077 DOI: 10.1152/physrev.00015.2009] [Citation(s) in RCA: 1468] [Impact Index Per Article: 104.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
There is a constant high demand for energy to sustain the continuous contractile activity of the heart, which is met primarily by the beta-oxidation of long-chain fatty acids. The control of fatty acid beta-oxidation is complex and is aimed at ensuring that the supply and oxidation of the fatty acids is sufficient to meet the energy demands of the heart. The metabolism of fatty acids via beta-oxidation is not regulated in isolation; rather, it occurs in response to alterations in contractile work, the presence of competing substrates (i.e., glucose, lactate, ketones, amino acids), changes in hormonal milieu, and limitations in oxygen supply. Alterations in fatty acid metabolism can contribute to cardiac pathology. For instance, the excessive uptake and beta-oxidation of fatty acids in obesity and diabetes can compromise cardiac function. Furthermore, alterations in fatty acid beta-oxidation both during and after ischemia and in the failing heart can also contribute to cardiac pathology. This paper reviews the regulation of myocardial fatty acid beta-oxidation and how alterations in fatty acid beta-oxidation can contribute to heart disease. The implications of inhibiting fatty acid beta-oxidation as a potential novel therapeutic approach for the treatment of various forms of heart disease are also discussed.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Group, Mazankowski Alberta Heart Institute, University of Alberta, Alberta T6G 2S2, Canada.
| | | | | | | | | |
Collapse
|
30
|
Abstract
In 1963, Lancet published a paper by Randle et al. that proposed a "glucose-fatty acid cycle" to describe fuel flux between and fuel selection by tissues. The original biochemical mechanism explained the inhibition of glucose oxidation by fatty acids. Since then, the principle has been confirmed by many investigators. At the same time, many new mechanisms controlling the utilization of glucose and fatty acids have been discovered. Here, we review the known short- and long-term mechanisms involved in the control of glucose and fatty acid utilization at the cytoplasmic and mitochondrial level in mammalian muscle and liver under normal and pathophysiological conditions. They include allosteric control, reversible phosphorylation, and the expression of key enzymes. However, the complexity is formidable. We suggest that not all chapters of the Randle cycle have been written.
Collapse
Affiliation(s)
- Louis Hue
- Université Catholique de Louvain and de Duve Institute, Hormone and Metabolic Research Unit, Brussels, Belgium.
| | | |
Collapse
|
31
|
Bashir A, Gropler RJ. Translation of myocardial metabolic imaging concepts into the clinics. Cardiol Clin 2009; 27:291-310, Table of Contents. [PMID: 19306771 DOI: 10.1016/j.ccl.2008.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Flexibility in myocardial substrate metabolism for energy production is fundamental to cardiac health. This loss in plasticity or flexibility leads to overdependence on the metabolism of an individual category of substrates, with the predominance in fatty acid metabolism characteristic of diabetic heart disease and the accelerated glucose use associated with pressure-overload left ventricular hypertrophy being prime examples. There is a strong demand for accurate noninvasive imaging approaches of myocardial substrate metabolism that can facilitate the crosstalk between the bench and the bedside, leading to improved patient management paradigms. In this article potential future applications of metabolic imaging, particularly radionuclide approaches, for assessment of cardiovascular disease are discussed.
Collapse
Affiliation(s)
- Adil Bashir
- Division of Radiological Sciences, Cardiovascular Imaging Laboratory, Edward Mallinckrodt Institute of Radiology, St Louis, MO 63110, USA
| | | |
Collapse
|
32
|
Zhao Z, Lee YJ, Kim SK, Kim HJ, Shim WS, Ahn CW, Lee HC, Cha BS, Ma ZA. Rosiglitazone and fenofibrate improve insulin sensitivity of pre-diabetic OLETF rats by reducing malonyl-CoA levels in the liver and skeletal muscle. Life Sci 2009; 84:688-95. [PMID: 19250943 DOI: 10.1016/j.lfs.2009.02.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Revised: 12/04/2008] [Accepted: 02/11/2009] [Indexed: 12/13/2022]
Affiliation(s)
- Zhengshan Zhao
- Department of Internal Medicine, Yonsei University College of Medicine, 134 Shinchon-Dong, Seodaemoon-Ku, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ussher JR, Lopaschuk GD. Targeting malonyl CoA inhibition of mitochondrial fatty acid uptake as an approach to treat cardiac ischemia/reperfusion. Basic Res Cardiol 2009; 104:203-10. [PMID: 19242641 DOI: 10.1007/s00395-009-0003-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Revised: 01/22/2009] [Accepted: 01/30/2009] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease is the major cause of death and disability in the world, with ischemic heart disease accounting for the vast majority of this health problem. Current treatments for ischemic heart disease are primarily aimed at either increasing blood and oxygen supply to the heart or decreasing the heart's oxygen demand. A novel treatment strategy involves increasing the efficiency of oxygen use by the heart. During and following ischemia, the heart can become inefficient in using oxygen, due in part to an excessive use of fatty acids as a source of fuel. One potential strategy to increase cardiac efficiency is to inhibit this use of fatty acid oxidation as a fuel source, while stimulating the use of glucose oxidation as a fuel source, which allows the heart to produce energy more efficiently and reduces the acidosis associated with ischemia/reperfusion, both of which are beneficial to the heart. Malonyl CoA is a potent endogenous inhibitor of cardiac fatty acid oxidation, secondary to inhibition of carnitine palmitoyl transferase-I, the gatekeeper of mitochondrial fatty acid uptake. Malonyl CoA is synthesized in the heart by acetyl CoA carboxylase and degraded by malonyl CoA decarboxylase (MCD). Strategies aimed at increasing cardiac malonyl CoA levels, such as via inhibition of MCD, are associated with a decrease in fatty acid oxidation rates, and a parallel increase in glucose oxidation rates. This is associated with a decrease in acidosis and an improvement in cardiac function and efficiency during and following ischemia. Therefore, targeting malonyl CoA is a novel exciting approach for the treatment of cardiac ischemia/reperfusion.
Collapse
Affiliation(s)
- John R Ussher
- Department of Pediatrics, 423 Heritage Medical Research Center, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
34
|
Raher MJ, Thibault HB, Buys ES, Kuruppu D, Shimizu N, Brownell AL, Blake SL, Rieusset J, Kaneki M, Derumeaux G, Picard MH, Bloch KD, Scherrer-Crosbie M. A short duration of high-fat diet induces insulin resistance and predisposes to adverse left ventricular remodeling after pressure overload. Am J Physiol Heart Circ Physiol 2008; 295:H2495-502. [PMID: 18978196 DOI: 10.1152/ajpheart.00139.2008] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Insulin resistance is an increasingly prevalent condition in humans that frequently clusters with disorders characterized by left ventricular (LV) pressure overload, such as systemic hypertension. To investigate the impact of insulin resistance on LV remodeling and functional response to pressure overload, C57BL6 male mice were fed a high-fat (HFD) or a standard diet (SD) for 9 days and then underwent transverse aortic constriction (TAC). LV size and function were assessed in SD- and HFD-fed mice using serial echocardiography before and 7, 21, and 28 days after TAC. Serial echocardiography was also performed on nonoperated SD- and HFD-fed mice over a period of 6 wk. LV perfusion was assessed before and 7 and 28 days after TAC. Nine days of HFD induced systemic and myocardial insulin resistance (assessed by myocardial 18F-fluorodeoxyglucose uptake), and myocardial perfusion response to acetylcholine was impaired. High-fat feeding for 28 days did not change LV size and function in nonbanded mice; however, TAC induced greater hypertrophy, more marked LV systolic and diastolic dysfunction, and decreased survival in HFD-fed compared with SD-fed mice. Compared with SD-fed mice, myocardial perfusion reserve was decreased 7 days after TAC, and capillary density was decreased 28 days after TAC in HFD-fed mice. A short duration of HFD induces insulin resistance in mice. These metabolic changes are accompanied by increased LV remodeling and dysfunction after TAC, highlighting the impact of insulin resistance in the development of pressure-overload-induced heart failure.
Collapse
Affiliation(s)
- Michael J Raher
- Cardiac Ultrasound Laboratory, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St., Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Malonyl-CoA can be formed within the mitochondria, peroxisomes, and cytosol of mammalian cells. Besides being an intermediate in the pathways of de novo fatty acid biosynthesis and fatty acid elongation, malonyl-CoA has an important signaling function through its allosteric inhibition of carnitine palmitoyltransferase 1, the enzyme that normally exerts flux control over mitochondrial beta-oxidation. Malonyl-CoA is rapidly turned over in mammalian cells, and the activities of acetyl-CoA carboxylase and malonyl-CoA decarboxylase are important determinants of its cytosolic concentration. It is now recognized that malonyl-CoA participates in a diverse range of physiological or pathological responses and systems. These include the ketogenic response of the liver to fasting and diabetes, carbohydrate versus fat fuel selection in muscle tissues, metabolic changes in muscle during contracture, alterations in fatty acid metabolism during cardiac ischemia and postischemic reperfusion, stimulation of B cell insulin secretion by glucose, and the hypothalamic control of appetite.
Collapse
Affiliation(s)
- David Saggerson
- Institute of Structural & Molecular Biology, Darwin Building, University College London, Gower Street, WC1E 6BT, Great Britain.
| |
Collapse
|
36
|
Durgan DJ, Young ME. Linking the cardiomyocyte circadian clock to myocardial metabolism. Cardiovasc Drugs Ther 2008; 22:115-24. [PMID: 18274886 DOI: 10.1007/s10557-008-6086-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Accepted: 01/17/2008] [Indexed: 02/02/2023]
Abstract
INTRODUCTION The energetic demands imposed upon the heart vary dramatically over the course of the day. In the face of equally commanding oscillations in the neurohumoral mileu, the heart must respond both rapidly and appropriately to its diurnal environment, for the survival of the organism. A major response of the heart to alterations in workload, nutrients, and various neurohumoral stimuli is at the level of metabolism. Failure of the heart to achieve adequate metabolic adaptation results in contractile dysfunction. DISCUSSION Substantial evidence is accumulating which suggests that a transcriptionally based timekeeping mechanism known as the circadian clock plays a role in mediating myocardial metabolic rhythms. Here, we provide an overview of our current knowledge regarding the interplay between the circadian clock within the cardiomyocyte and myocardial metabolism. This includes a particular focus on circadian clock mediated regulation of endogenous energy stores, as well as those mechanisms orchestrating circadian rhythms in metabolic gene expression. CONCLUSION An essential need to elucidate fully the functions of this molecular mechanism in the heart remains.
Collapse
Affiliation(s)
- David J Durgan
- Department of Pediatrics, USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, 1100 Bates Street, Houston, TX 77030, USA
| | | |
Collapse
|
37
|
Ussher JR, Lopaschuk GD. The malonyl CoA axis as a potential target for treating ischaemic heart disease. Cardiovasc Res 2008; 79:259-68. [PMID: 18499682 DOI: 10.1093/cvr/cvn130] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease is the leading cause of death and disability for people living in western societies, with ischaemic heart disease accounting for the majority of this health burden. The primary treatment for ischaemic heart disease consists of either improving blood and oxygen supply to the heart or reducing the heart's oxygen demand. Unfortunately, despite recent advances with these approaches, ischaemic heart disease still remains a major health problem. Therefore, the development of new treatment strategies is still required. One exciting new approach is to optimize cardiac energy metabolism, particularly by decreasing the use of fatty acids as a fuel and by increasing the use of glucose as a fuel. This approach is beneficial in the setting of ischaemic heart disease, as it allows the heart to produce energy more efficiently and it reduces the degree of acidosis associated with ischaemia/reperfusion. Malonyl CoA is a potent endogenous inhibitor of cardiac fatty acid oxidation, secondary to inhibiting carnitine palmitoyl transferase-I, the rate-limiting enzyme in the mitochondrial uptake of fatty acids. Malonyl CoA is synthesized in the heart by acetyl CoA carboxylase, which in turn is phosphorylated and inhibited by 5'AMP-activated protein kinase. The degradation of myocardial malonyl CoA occurs via malonyl CoA decarboxylase (MCD). Previous studies have shown that inhibiting MCD will significantly increase cardiac malonyl CoA levels. This is associated with an increase in glucose oxidation, a decrease in acidosis, and an improvement in cardiac function and efficiency during and following ischaemia. Hence, the malonyl CoA axis represents an exciting new target for the treatment of ischaemic heart disease.
Collapse
Affiliation(s)
- John R Ussher
- Cardiovascular Research Group, Department of Pediatrics, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
38
|
Leichman JG, Lavis VR, Aguilar D, Wilson CR, Taegtmeyer H. The metabolic syndrome and the heart--a considered opinion. Clin Res Cardiol 2008; 95 Suppl 1:i134-41. [PMID: 16598541 DOI: 10.1007/s00392-006-1119-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The metabolic syndrome (MS) is a multifactorial, heterogeneous group of risk factors for the development of cardiovascular disease. Here we review the evidence in support of the hypothesis that metabolic dysregulation of the body as a whole leads to contractile dysfunction of the heart due to an imbalance of substrate uptake (increased) and substrate oxidation (decreased). The consequences of this imbalance were already recognized 150 years ago by Virchow when he described "fatty atrophy" of the heart as a "true metamorphosis of the heart muscle cell."
Collapse
Affiliation(s)
- J G Leichman
- The University of Texas Houston Medical School, Department of Internal Medicine, Division of Cardiology, 6431 Fannin Street, MSB 1.246, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
39
|
Lockridge JB, Sailors ML, Durgan DJ, Egbejimi O, Jeong WJ, Bray MS, Stanley WC, Young ME. Bioinformatic profiling of the transcriptional response of adult rat cardiomyocytes to distinct fatty acids. J Lipid Res 2008; 49:1395-408. [PMID: 18387886 DOI: 10.1194/jlr.m700517-jlr200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Diabetes mellitus, obesity, and dyslipidemia increase risk for cardiovascular disease, and expose the heart to high plasma fatty acid (FA) levels. Recent studies suggest that distinct FA species are cardiotoxic (e.g., palmitate), while others are cardioprotective (e.g., oleate), although the molecular mechanisms mediating these observations are unclear. The purpose of the present study was to investigate the differential effects of distinct FA species (varying carbon length and degree of saturation) on adult rat cardiomyocyte (ARC) gene expression. ARCs were initially challenged with 0.4 mM octanoate (8:0), palmitate (16:0), stearate (18:0), oleate (18:1), or linoleate (18:2) for 24 h. Microarray analysis revealed differential regulation of gene expression by the distinct FAs; the order regarding the number of genes whose expression was influenced by a specific FA was octanoate (1,188) > stearate (740) > palmitate (590) > oleate (83) > linoleate (65). In general, cardioprotective FAs (e.g., oleate) increased expression of genes promoting FA oxidation to a greater extent than cardiotoxic FAs (e.g., palmitate), whereas the latter induced markers of endoplasmic reticulum and oxidative stress. Subsequent RT-PCR analysis revealed distinct time- and concentration-dependent effects of these FA species, in a gene-specific manner. For example, stearate- and palmitate-mediated ucp3 induction tended to be transient (i.e., initial high induction, followed by subsequent repression), whereas oleate-mediated induction was sustained. These findings may provide insight into why diets high in unsaturated FAs (e.g., oleate) are cardioprotective, whereas diets rich in saturated FAs (e.g., palmitate) are not.
Collapse
Affiliation(s)
- Joseph B Lockridge
- University of Texas Health Science Center at Houston, Brown Foundation Institute of Molecular Medicine, Houston TX, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Cree MG, Aarsland A, Herndon DN, Wolfe RR. Role of fat metabolism in burn trauma-induced skeletal muscle insulin resistance. Crit Care Med 2007; 35:S476-83. [PMID: 17713396 DOI: 10.1097/01.ccm.0000278066.05354.53] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Review current evidence on the role of fat in post-trauma insulin resistance, in reference to new studies with peroxisome proliferating activating receptor-alpha agonists. DESIGN Review. SETTING University laboratory. PATIENTS Thirty pediatric burn trauma patients. INTERVENTIONS Fourteen days of peroxisome proliferating activating receptor-alpha agonist immediately following burn trauma. MEASUREMENTS AND MAIN RESULTS We measured glucose metabolism and fat metabolism via tracer methodology and intracellular measurements. Insulin-stimulated glucose uptake is impaired following burn trauma, as is intracellular insulin signaling, palmitate oxidation, and mitochondrial oxidative capacity. Furthermore, levels of intracellular lipids are increased. Two weeks of peroxisome proliferating activating receptor-alpha treatment significantly reverses these pathologic changes incurred from burn injury. CONCLUSIONS Severe burn injury seriously affects multiple aspects of glucose and fat metabolism within the muscle, which can adversely affect clinical outcomes. Treatment with a peroxisome proliferating activating receptor-alpha drug may be a potential new therapeutic option.
Collapse
Affiliation(s)
- Melanie G Cree
- University of Arkansas Medical Sciences, Donald W. Reynolds Center on Aging, Nutrition, Metabolism and Exercise Lab, Little Rock, AR, USA
| | | | | | | |
Collapse
|
41
|
Xu Y, Wang Q, Cook TJ, Knipp GT. Effect of Placental Fatty Acid Metabolism and Regulation by Peroxisome Proliferator Activated Receptor on Pregnancy and Fetal Outcomes. J Pharm Sci 2007; 96:2582-606. [PMID: 17549724 DOI: 10.1002/jps.20973] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fatty acids, particularly the omega-3 and omega-6 essential fatty acids (EFAs), are considered critical nutritional sources for the developing fetus. The placenta governs the fetal supply of fatty acids via two processes: transport and metabolism. Placental fatty acid metabolism can play a critical role in guiding pregnancy and fetal outcome. EFAs can be metabolized to important cell signaling molecules in placenta by several major isoform families including: the Cytochrome P450 subfamily 4A (CYP4A); Cyclooxygenases (COXs); and Lipoxygenases (LOXs). Peroxisome proliferator-activated nuclear receptors (PPARs) have been demonstrated to regulate a number of placental fatty acid/lipid homeostasis-related proteins (e.g., metabolizing enzymes and transporters). The present review summarizes research on the molecular and functional relevance of fatty acid metabolizing enzymes and the role of PPARs in regulating their expression in the mammalian placenta. Elucidating the pathways of placental fatty acid metabolism and the regulatory processes governing these pathways is critical for advancing our understanding of the role of placenta in supplying EFAs to the developing fetus and the potential implications on pregnancy and fetal outcome. A more complete understanding of placental fatty acid disposition may also provide a basis for nutritional/pharmacological interventions to ameliorate the risk of adverse pregnancy and/or fetal outcomes.
Collapse
Affiliation(s)
- Yan Xu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | | | |
Collapse
|
42
|
Cree MG, Newcomer BR, Herndon DN, Qian T, Sun D, Morio B, Zwetsloot JJ, Dohm GL, Fram RY, Mlcak RP, Aarsland A, Wolfe RR. PPAR-alpha agonism improves whole body and muscle mitochondrial fat oxidation, but does not alter intracellular fat concentrations in burn trauma children in a randomized controlled trial. Nutr Metab (Lond) 2007; 4:9. [PMID: 17451602 PMCID: PMC1868739 DOI: 10.1186/1743-7075-4-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Accepted: 04/23/2007] [Indexed: 11/18/2022] Open
Abstract
Background Insulin resistance is often associated with increased levels of intracellular triglycerides, diacylglycerol and decreased fat β-oxidation. It was unknown if this relationship was present in patients with acute insulin resistance induced by trauma. Methods A double blind placebo controlled trial was conducted in 18 children with severe burn injury. Metabolic studies to assess whole body palmitate oxidation and insulin sensitivity, muscle biopsies for mitochondrial palmitate oxidation, diacylglycerol, fatty acyl Co-A and fatty acyl carnitine concentrations, and magnetic resonance spectroscopy for muscle and liver triglycerides were compared before and after two weeks of placebo or PPAR-α agonist treatment. Results Insulin sensitivity and basal whole body palmitate oxidation as measured with an isotope tracer increased significantly (P = 0.003 and P = 0.004, respectively) after PPAR-α agonist treatment compared to placebo. Mitochondrial palmitate oxidation rates in muscle samples increased significantly after PPAR-α treatment (P = 0.002). However, the concentrations of muscle triglyceride, diacylglycerol, fatty acyl CoA, fatty acyl carnitine, and liver triglycerides did not change with either treatment. PKC-θ activation during hyper-insulinemia decreased significantly following PPAR-α treatment. Conclusion PPAR-α agonist treatment increases palmitate oxidation and decreases PKC activity along with reduced insulin sensitivity in acute trauma, However, a direct link between these responses cannot be attributed to alterations in intracellular lipid concentrations.
Collapse
Affiliation(s)
- Melanie G Cree
- Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, USA
| | | | - David N Herndon
- Surgery, University of Texas Medical Branch, Galveston, USA
- Shriners Hospitals for Children, Galveston, USA
| | - Ting Qian
- Surgery, University of Texas Medical Branch, Galveston, USA
| | - Dayoung Sun
- Surgery, University of Texas Medical Branch, Galveston, USA
| | - Beatrice Morio
- UMPE- Laboratoire de Nutrition Humain, Clermont-Ferrand, France
| | - Jennifer J Zwetsloot
- Department of Physiology, Brody SOM, Eastern Carolina University, Greenville, USA
| | - G Lynis Dohm
- Department of Physiology, Brody SOM, Eastern Carolina University, Greenville, USA
| | - Ricki Y Fram
- Surgery, University of Texas Medical Branch, Galveston, USA
| | | | - Asle Aarsland
- Anesthesiology, University of Texas Medical Branch, Galveston, USA
- Shriners Hospitals for Children, Galveston, USA
| | - Robert R Wolfe
- Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, USA
- Surgery, University of Texas Medical Branch, Galveston, USA
- Shriners Hospitals for Children, Galveston, USA
| |
Collapse
|
43
|
Abstract
Control of energy metabolism in the heart is closely linked to cardiac performance. Dysregulation of energy-generating pathways occurs in many forms of heart disease, including heart failure. Uncertainty exists as to whether these alterations in the way adenosine triphosphate (ATP) is produced serve to protect the heart from excessive oxygen demands or have untoward long-term consequences. Regulation of fatty acid beta-oxidation (FAO), the principal source of ATP in the healthy heart, occurs at multiple levels, including a strong gene transcriptional component. In the heart, members of the peroxisome proliferator-activated receptor (PPAR) family of transcription factors are the primary regulators of FAO gene expression. PPARs are ligand activated by endogenous lipids and synthetic small molecules, thus providing attractive targets for pharmaceutical intervention. This article discusses controversies surrounding our understanding of cardiac energy metabolism in heart failure and the role that PPAR family members may play, either as contributors to or as potential adjunctive therapy for cardiac disease.
Collapse
|
44
|
Wang X, Stanley WC, Darrow CJ, Brunengraber H, Kasumov T. Assay of the activity of malonyl-coenzyme A decarboxylase by gas chromatography-mass spectrometry. Anal Biochem 2007; 363:169-74. [PMID: 17316539 DOI: 10.1016/j.ab.2007.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Revised: 01/08/2007] [Accepted: 01/10/2007] [Indexed: 11/21/2022]
Abstract
We developed a gas chromatography-mass spectrometry (GC-MS) assay to measure the activity of malonyl-coenzyme A (CoA) decarboxylase (MCD) in crude tissue homogenates. Liver extracts are incubated with [U-(13)C(3)]malonyl-CoA to form [U-(13)C(2)]acetyl-CoA by the action of MCD. The reaction mixture contains 2 mM ADP to prevent the hydrolysis of [1,2-(13)C(2)]acetyl-CoA by acetyl-CoA hydrolase present in the extracts. Newly formed [U-(13)C(2)]acetyl-CoA and internal standard of [(2)H(3),1-(13)C]acetyl-CoA are analyzed as thiophenol derivatives by GC-MS. This assay was applied to a study of the kinetics of MCD in rat liver. Using the Lineweaver-Burke plot of MCD kinetics, K(m) of 202microM and V(max) of 3.3micromol min(-1) (g liver)(-1) were calculated. The liver MCD activities (micromol min(-1) g(-1)+/-SD) in three groups of rats with different nutritional statuses-fed, 1-day fasted, and 2-day fasted-were 1.80+/-0.41, 2.59+/-0.37 (P<0.05), and 3.07+/-0.70 (P<0.05), respectively. We report a practical, nonradioactive, sensitive assay of MCD in crude tissue extract.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | |
Collapse
|
45
|
|
46
|
Pruimboom-Brees I, Haghpassand M, Royer L, Brees D, Aldinger C, Reagan W, Singh J, Kerlin R, Kane C, Bagley S, Hayward C, Loy J, O'Brien P, Francone OL. A critical role for peroxisomal proliferator-activated receptor-alpha nuclear receptors in the development of cardiomyocyte degeneration and necrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:750-60. [PMID: 16936252 PMCID: PMC1698838 DOI: 10.2353/ajpath.2006.051110] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Peroxisomal proliferator-activated receptor (PPAR)-alpha is a ligand-activated transcriptional factor that regulates genes involved in lipid metabolism and energy homeostasis. PPAR-alpha activators, including fibrates, have been used to treat dyslipidemia for several decades. In contrast to their known effects on lipids, the pharmacological consequences of PPAR-alpha activation on cardiac metabolism and function are not well understood. Therefore, we evaluated the role that PPAR-alpha receptors play in the heart. Our studies demonstrate that activation of PPAR-alpha receptors using a selective PPAR-alpha ligand results in cardiomyocyte necrosis in mice. Studies in PPAR-alpha-deficient mice demonstrated that cardiomyocyte necrosis is a consequence of the activation of PPAR-alpha receptors. Cardiac fatty acyl-CoA oxidase mRNA levels increased at doses in which cardiac damage was observed and temporally preceded cardiomyocyte degeneration, suggesting that peroxisomal beta-oxidation correlates with the appearance of microscopic injury and cardiac injury biomarkers. Increased myocardial oxidative stress was evident in mice treated with the PPAR-alpha agonists coinciding with increased peroxisomal biomarkers of fatty acid oxidation. These findings suggest that activation of PPAR-alpha leads to increased cardiac fatty acid oxidation and subsequent accumulation of oxidative stress intermediates resulting in cardiomyocyte necrosis.
Collapse
Affiliation(s)
- Ingrid Pruimboom-Brees
- Pfizer Global Research and Development, Department of Cardiovascular and Metabolic Diseases, Eastern Point Rd., Groton, CT 06340, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nam HW, Lee GY, Kim YS. Mass spectrometric identification of K210 essential for rat malonyl-CoA decarboxylase catalysis. J Proteome Res 2006; 5:1398-406. [PMID: 16739991 DOI: 10.1021/pr050487g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Proteomic technology provides useful tools to detect protein modification sites in vivo and in vitro. In this work, we applied proteomics to identify an essential amino acid residue involved in Malonyl-CoA Decarboxylase (MCD) catalysis. A reaction with acetic anhydride and MCD, under mild conditions without acetyl CoA as a substrate, resulted in the acetylation of six lysyl residues, K210, K58, K167, K316, K388, and K444. When acetyl CoA was added to the reaction, K210 was protected from acetylation, indicating a potential role for this residue in catalysis. In addition, K210 was the only lysyl residue, out of six, that was not endogenously acetylated. Because K210, K308, and K388 are conserved across species, they were site-specifically mutated to methionine which is size-wise similar to lysine but not protonated. The K308M and K388M MCD mutants retained 60% of their enzyme activities, whereas the K210M mutant was completely inactive. These results strongly suggest that K210 is an essential residue in rat MCD catalysis and is a likely proton donor to the alpha carbon of malonyl-CoA. Therapeutic inhibition of MCD may be a viable approach to treating various clinical pathologies associated with defective fatty acid metabolism.
Collapse
Affiliation(s)
- Hyung Wook Nam
- Department of Biochemistry, College of Science, Protein Network Research Center, Yonsei University, Seoul, Korea 120-749
| | | | | |
Collapse
|
48
|
An D, Rodrigues B. Role of changes in cardiac metabolism in development of diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol 2006; 291:H1489-506. [PMID: 16751293 DOI: 10.1152/ajpheart.00278.2006] [Citation(s) in RCA: 333] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In patients with diabetes, an increased risk of symptomatic heart failure usually develops in the presence of hypertension or ischemic heart disease. However, a predisposition to heart failure might also reflect the effects of underlying abnormalities in diastolic function that can occur in asymptomatic patients with diabetes alone (termed diabetic cardiomyopathy). Evidence of cardiomyopathy has also been demonstrated in animal models of both Type 1 (streptozotocin-induced diabetes) and Type 2 diabetes (Zucker diabetic fatty rats and ob/ob or db/db mice). During insulin resistance or diabetes, the heart rapidly modifies its energy metabolism, resulting in augmented fatty acid and decreased glucose consumption. Accumulating evidence suggests that this alteration of cardiac metabolism plays an important role in the development of cardiomyopathy. Hence, a better understanding of this dysregulation in cardiac substrate utilization during insulin resistance and diabetes could provide information as to potential targets for the treatment of cardiomyopathy. This review is focused on evaluating the acute and chronic regulation and dysregulation of cardiac metabolism in normal and insulin-resistant/diabetic hearts and how these changes could contribute toward the development of cardiomyopathy.
Collapse
MESH Headings
- Animals
- Cardiomyopathies/etiology
- Cardiomyopathies/metabolism
- Cardiomyopathies/pathology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Disease Models, Animal
- Energy Metabolism/physiology
- Fatty Acids/metabolism
- Glucose/metabolism
- Humans
- Insulin Resistance/physiology
- Mice
- Mice, Obese
- Myocardium/metabolism
- Myocardium/pathology
- Rats
- Rats, Zucker
Collapse
Affiliation(s)
- Ding An
- Div. of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, The Univ. of British Columbia, 2146 East Mall, Vancouver, BC, Canada
| | | |
Collapse
|
49
|
Durgan DJ, Smith JK, Hotze MA, Egbejimi O, Cuthbert KD, Zaha VG, Dyck JRB, Abel ED, Young ME. Distinct transcriptional regulation of long-chain acyl-CoA synthetase isoforms and cytosolic thioesterase 1 in the rodent heart by fatty acids and insulin. Am J Physiol Heart Circ Physiol 2006; 290:H2480-97. [PMID: 16428347 DOI: 10.1152/ajpheart.01344.2005] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The molecular mechanism(s) responsible for channeling long-chain fatty acids (LCFAs) into oxidative versus nonoxidative pathways is (are) poorly understood in the heart. Intracellular LCFAs are converted to long-chain fatty acyl-CoAs (LCFA-CoAs) by a family of long-chain acyl-CoA synthetases (ACSLs). Cytosolic thioesterase 1 (CTE1) hydrolyzes cytosolic LCFA-CoAs to LCFAs, generating a potential futile cycle at the expense of ATP utilization. We hypothesized that ACSL isoforms and CTE1 are differentially regulated in the heart during physiological and pathophysiological conditions. Using quantitative RT-PCR, we report that the five known acsl isoforms ( acsl1, acsl3, acsl4, acsl5, and acsl6) and cte1 are expressed in whole rat and mouse hearts, as well as adult rat cardiomyocytes (ARCs). Streptozotocin-induced insulin-dependent diabetes (4 wk) and fasting (≤24 h) both dramatically induced cte1 and repressed acsl6 mRNA, with no significant effects on the other acsl isoforms. In contrast, high-fat feeding (4 wk) induced cte1 without affecting expression of the acsl isoforms in the heart. Investigation into the mechanism(s) responsible for these transcriptional changes uncovered roles for peroxisome proliferator-activated receptor-α (PPARα) and insulin as regulators of specific acsl isoforms and cte1 in the heart. Culturing ARCs with oleate (0.1–0.4 mM) or the PPARα agonists WY-14643 (1 μM) and fenofibrate (10 μM) consistently induced acsl1 and cte1. Conversely, PPARα null mouse hearts exhibited decreased acsl1 and cte1 expression. Culturing ARCs with insulin (10 nM) induced acsl6, whereas specific loss of insulin signaling within the heart (cardiac-specific insulin receptor knockout mice) caused decreased acsl6 expression. Our data expose differential regulation of acsl isoforms and cte1 in the heart, where acsl1 and cte1 are PPARα-regulated genes, whereas acsl6 is an insulin-regulated gene.
Collapse
Affiliation(s)
- David J Durgan
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Department of Pediatrics, 1100 Bates Street, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cuthbert KD, Dyck JRB. Malonyl-CoA decarboxylase is a major regulator of myocardial fatty acid oxidation. Curr Hypertens Rep 2006; 7:407-11. [PMID: 16386195 DOI: 10.1007/s11906-005-0034-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The energy demands of the heart are normally met by oxidation of both glucose and fatty acids. Fatty acid oxidation is limited by the uptake of fatty acyl coenzyme A (CoA) into the mitochondria, a process regulated by carnitine palmitoyltransferase (CPT)1. Malonyl CoA is a potent endogenous inhibitor of CPT1, and therefore plays an integral role in the control of myocardial fatty acid oxidation. Malonyl-CoA decarboxylase (MCD) is responsible for the removal of malonyl CoA and may control myocardial fatty acid oxidation. Indeed, strategies using MCD inhibitors and MCD knockout mice have provided the first evidence for a direct role of MCD in the control of myocardial fatty acid oxidation. Based on these studies, pharmacologic inhibition of MCD has been proposed to be a viable approach for the treatment of ischemic heart disease resulting from a variety of pathologic conditions, including coronary artery diseases, pathologic hypertrophy, and hypertension.
Collapse
Affiliation(s)
- Karalyn D Cuthbert
- Department of Pediatrics, 474 Heritage Medical Research Centre, Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada, T6G 2S2
| | | |
Collapse
|