1
|
Xie J, Yu Z, Zhu Y, Zheng M, Zhu Y. Functions of Coenzyme A and Acyl-CoA in Post-Translational Modification and Human Disease. FRONT BIOSCI-LANDMRK 2024; 29:331. [PMID: 39344325 DOI: 10.31083/j.fbl2909331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 05/24/2024] [Accepted: 07/17/2024] [Indexed: 10/01/2024]
Abstract
Coenzyme A (CoA) is synthesized from pantothenate, L-cysteine and adenosine triphosphate (ATP), and plays a vital role in diverse physiological processes. Protein acylation is a common post-translational modification (PTM) that modifies protein structure, function and interactions. It occurs via the transfer of acyl groups from acyl-CoAs to various amino acids by acyltransferase. The characteristics and effects of acylation vary according to the origin, structure, and location of the acyl group. Acetyl-CoA, formyl-CoA, lactoyl-CoA, and malonyl-CoA are typical acyl group donors. The major acyl donor, acyl-CoA, enables modifications that impart distinct biological functions to both histone and non-histone proteins. These modifications are crucial for regulating gene expression, organizing chromatin, managing metabolism, and modulating the immune response. Moreover, CoA and acyl-CoA play significant roles in the development and progression of neurodegenerative diseases, cancer, cardiovascular diseases, and other health conditions. The goal of this review was to systematically describe the types of commonly utilized acyl-CoAs, their functions in protein PTM, and their roles in the progression of human diseases.
Collapse
Affiliation(s)
- Jumin Xie
- Hubei Key Laboratory of Renal Disease Occurrence and Intervention, Medical School, Hubei Polytechnic University, 435003 Huangshi, Hubei, China
| | - Zhang Yu
- Hubei Key Laboratory of Renal Disease Occurrence and Intervention, Medical School, Hubei Polytechnic University, 435003 Huangshi, Hubei, China
| | - Ying Zhu
- Hubei Key Laboratory of Renal Disease Occurrence and Intervention, Medical School, Hubei Polytechnic University, 435003 Huangshi, Hubei, China
| | - Mei Zheng
- Hubei Key Laboratory of Renal Disease Occurrence and Intervention, Medical School, Hubei Polytechnic University, 435003 Huangshi, Hubei, China
| | - Yanfang Zhu
- Department of Critical Care Medicine, Huangshi Hospital of TCM (Infectious Disease Hospital), 435003 Huangshi, Hubei, China
| |
Collapse
|
2
|
Wong A, Sun Q, Latif II, Karwi QG. Metabolic flux in macrophages in obesity and type-2 diabetes. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13210. [PMID: 38988822 PMCID: PMC11233469 DOI: 10.3389/jpps.2024.13210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024]
Abstract
Recent literature extensively investigates the crucial role of energy metabolism in determining the inflammatory response and polarization status of macrophages. This rapidly expanding area of research highlights the importance of understanding the link between energy metabolism and macrophage function. The metabolic pathways in macrophages are intricate and interdependent, and they can affect the polarization of macrophages. Previous studies suggested that glucose flux through cytosolic glycolysis is necessary to trigger pro-inflammatory phenotypes of macrophages, and fatty acid oxidation is crucial to support anti-inflammatory responses. However, recent studies demonstrated that this understanding is oversimplified and that the metabolic control of macrophage polarization is highly complex and not fully understood yet. How the metabolic flux through different metabolic pathways (glycolysis, glucose oxidation, fatty acid oxidation, ketone oxidation, and amino acid oxidation) is altered by obesity- and type 2 diabetes (T2D)-associated insulin resistance is also not fully defined. This mini-review focuses on the impact of insulin resistance in obesity and T2D on the metabolic flux through the main metabolic pathways in macrophages, which might be linked to changes in their inflammatory responses. We closely evaluated the experimental studies and methodologies used in the published research and highlighted priority research areas for future investigations.
Collapse
Affiliation(s)
- Angela Wong
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Qiuyu Sun
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ismail Ibrahim Latif
- Department of Microbiology, College of Medicine, University of Diyala, Baqubaa, Diyala, Iraq
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, NL, Canada
| |
Collapse
|
3
|
Han SC, Kang JI, Choi YK, Boo HJ, Yoon WJ, Kang HK, Yoo ES. Intermittent Fasting Modulates Immune Response by Generating Tregs via TGF-β Dependent Mechanisms in Obese Mice with Allergic Contact Dermatitis. Biomol Ther (Seoul) 2024; 32:136-145. [PMID: 37424516 PMCID: PMC10762271 DOI: 10.4062/biomolther.2023.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/25/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
People with obesity maintain low levels of inflammation; therefore, their exposure to foreign antigens can trigger an excessive immune response. In people with obesity or allergic contact dermatitis (ACD), symptoms are exacerbated by a reduction in the number of regulatory T cells (Tregs) and IL-10/TGF-β-modified macrophages (M2 macrophages) at the inflammatory site. Benefits of intermittent fasting (IF) have been demonstrated for many diseases; however, the immune responses regulated by macrophages and CD4+T cells in obese ACD animal models are poorly understood. Therefore, we investigated whether IF suppresses inflammatory responses and upregulates the generation of Tregs and M2 macrophages in experimental ACD animal models of obese mice. The IF regimen relieved various ACD symptoms in inflamed and adipose tissues. We showed that the IF regimen upregulates Treg generation in a TGF-β-dependent manner and induces CD4+T cell hypo-responsiveness. IF-M2 macrophages, which strongly express TGF-β and inhibit CD4+T cell proliferation, directly regulated Treg differentiation from CD4+T cells. These results indicate that the IF regimen enhances the TGF-β-producing ability of M2 macrophages and that the development of Tregs keeps mice healthy against ACD exacerbated by obesity. Therefore, the IF regimen may ameliorate inflammatory immune disorders caused by obesity.
Collapse
Affiliation(s)
- Sang-Chul Han
- Department of Medicine, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jung-Il Kang
- Department of Medicine, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Youn Kyung Choi
- Department of Medicine, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Hye-Jin Boo
- Department of Medicine, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Weon-Jong Yoon
- Jeju Biodiversity Research Institute (JBRI), Jeju Technopark (JTP), Jeju 63208, Republic of Korea
| | - Hee-Kyoung Kang
- Department of Medicine, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Eun-Sook Yoo
- Department of Medicine, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
4
|
Zou L, Yang Y, Wang Z, Fu X, He X, Song J, Li T, Ma H, Yu T. Lysine Malonylation and Its Links to Metabolism and Diseases. Aging Dis 2023; 14:84-98. [PMID: 36818560 PMCID: PMC9937698 DOI: 10.14336/ad.2022.0711] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/11/2022] [Indexed: 11/18/2022] Open
Abstract
Malonylation is a recently identified post-translational modification with malonyl-coenzyme A as the donor. It conserved both in prokaryotes and eukaryotes. Recent advances in the identification and quantification of lysine malonylation by bioinformatic analysis have improved our understanding of its role in the regulation of protein activity, interaction, and localization and have elucidated its involvement in many biological processes. Malonylation has been linked to diverse physiological processes, including metabolic disorders, inflammation, and immune regulation. This review discusses malonylation in theory, describes the underlying mechanism, and summarizes the recent progress in malonylation research. The latest findings point to novel functions of malonylation and highlight the mechanisms by which malonylation regulates a variety of cellular processes. Our review also marks the association between lysine malonylation, the enzymes involved, and various diseases, and discusses promising diagnostic and therapeutic biomolecular targets for future clinical applications.
Collapse
Affiliation(s)
- Lu Zou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, Qingdao, China.
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Xiangqin He
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Jiayi Song
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Tianxiang Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Huibo Ma
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China.,Correspondence should be addressed to: Dr. Tao Yu, Center for Regenerative Medicine, Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
5
|
Alsabeelah N, Kumar V. Protective Effect of Triclosan in Monocrotaline-Induced Pulmonary Arterial Hypertension: FASN Inhibition a Novel Approach. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2022; 14:171-177. [PMID: 37051426 PMCID: PMC10084994 DOI: 10.4103/jpbs.jpbs_307_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/10/2022] [Accepted: 09/27/2022] [Indexed: 02/17/2023] Open
Abstract
Background Novel pharmacological approaches are needed to improve the outcomes of patients with idiopathic pulmonary hypertension. Fatty acid synthase (FASN) inhibitors have shown beneficial effects in preclinical models of pulmonary arterial hypertension (PAH), because of their role in the regulation of pulmonary artery vasoconstrictor tone and remodeling. Objective We compared a Triclosan (FASN inhibitor), for the first time with the dual endothelin receptor antagonist, macitentan, in a monocrotaline-induced rat pulmonary hypertension model. Methods Different methods (hemodynamics, histology of right ventricle and pulmonary vessels, and circulating biomarkers) showed consistently that 30 mg/kg daily of Triclosan (FASN inhibitor) and 10 mg/kg daily of macitentan slowed the progression of PAH both at the functional and structural levels. Results Treatments started on day 14 after monocrotaline injection and lasted 14 days. The findings of all experimental methods show that the FASN inhibitor has more similar effects as compared to macitentan. Conclusion Our study reveals that inhibition of FAS decreases RV hypertrophy and improves cardiac function associated with PAH with the regulation of metabolic functions and governs further studies to establish "FASN inhibitor as a potential therapeutic approach" for the management of PAH.
Collapse
Affiliation(s)
- Nimer Alsabeelah
- Pharmacy Practice Department, Pharmacy College, University of Hafr Al Batin, Saudi Arabia
| | - Vinay Kumar
- Department of Pharmacology, KIET Group of Institutions (KIET School of Pharmacy), Delhi-NCR, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
6
|
Jiang M, Xie X, Cao F, Wang Y. Mitochondrial Metabolism in Myocardial Remodeling and Mechanical Unloading: Implications for Ischemic Heart Disease. Front Cardiovasc Med 2021; 8:789267. [PMID: 34957264 PMCID: PMC8695728 DOI: 10.3389/fcvm.2021.789267] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Ischemic heart disease refers to myocardial degeneration, necrosis, and fibrosis caused by coronary artery disease. It can lead to severe left ventricular dysfunction (LVEF ≤ 35–40%) and is a major cause of heart failure (HF). In each contraction, myocardium is subjected to a variety of mechanical forces, such as stretch, afterload, and shear stress, and these mechanical stresses are clinically associated with myocardial remodeling and, eventually, cardiac outcomes. Mitochondria produce 90% of ATP in the heart and participate in metabolic pathways that regulate the balance of glucose and fatty acid oxidative phosphorylation. However, altered energetics and metabolic reprogramming are proved to aggravate HF development and progression by disturbing substrate utilization. This review briefly summarizes the current insights into the adaptations of cardiomyocytes to mechanical stimuli and underlying mechanisms in ischemic heart disease, with focusing on mitochondrial metabolism. We also discuss how mechanical circulatory support (MCS) alters myocardial energy metabolism and affects the detrimental metabolic adaptations of the dysfunctional myocardium.
Collapse
Affiliation(s)
- Min Jiang
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaoye Xie
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China.,Department of Cadre Ward, The 960 Hospital of Chinese People's Liberation Army, Jinan, China
| | - Feng Cao
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yabin Wang
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
7
|
Hala D, Petersen LH, Huggett DB, Puchowicz MA, Brunengraber H, Zhang GF. Overcompensation of CoA Trapping by Di(2-ethylhexyl) Phthalate (DEHP) Metabolites in Livers of Wistar Rats. Int J Mol Sci 2021; 22:ijms222413489. [PMID: 34948286 PMCID: PMC8709406 DOI: 10.3390/ijms222413489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is commonly used as a plasticizer in various industrial and household plastic products, ensuring widespread human exposures. Its routine detection in human bio-fluids and the propensity of its monoester metabolite to activate peroxisome proliferator activated receptor-α (PPARα) and perturb lipid metabolism implicate it as a metabolic disrupter. In this study we evaluated the effects of DEHP exposure on hepatic levels of free CoA and various CoA esters, while also confirming the metabolic activation to CoA esters and partial β-oxidation of a DEHP metabolite (2-ethyhexanol). Male Wistar rats were exposed via diet to 2% (w/w) DEHP for fourteen-days, following which hepatic levels of free CoA and various CoA esters were identified using liquid chromatography-mass spectrometry. DEHP exposed rats showed significantly elevated free CoA and increased levels of physiological, DEHP-derived and unidentified CoA esters. The physiological CoA ester of malonyl-CoA and DEHP-derived CoA ester of 3-keto-2-ethylhexanoyl-CoA were the most highly elevated, at eighteen- and ninety eight-times respectively. We also detected sixteen unidentified CoA esters which may be derivative of DEHP metabolism or induction of other intermediary metabolism metabolites. Our results demonstrate that DEHP is a metabolic disrupter which affects production and sequestration of CoA, an essential cofactor of oxidative and biosynthetic reactions.
Collapse
Affiliation(s)
- David Hala
- Department of Biology, University of North Texas, Denton, TX 76203, USA; (L.H.P.); (D.B.H.)
- Department of Marine Biology, Texas A&M at Galveston, Galveston, TX 77554, USA
- Correspondence: ; Tel.: +1-409-740-4535
| | - Lene H. Petersen
- Department of Biology, University of North Texas, Denton, TX 76203, USA; (L.H.P.); (D.B.H.)
- Department of Marine Biology, Texas A&M at Galveston, Galveston, TX 77554, USA
| | - Duane B. Huggett
- Department of Biology, University of North Texas, Denton, TX 76203, USA; (L.H.P.); (D.B.H.)
- Boehringer Ingelheim Animal Health, Athens, GA 30601, USA
| | - Michelle A. Puchowicz
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA; (M.A.P.); (H.B.)
- Department of Pediatrics, The University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Henri Brunengraber
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA; (M.A.P.); (H.B.)
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27705, USA;
- Department of Medicine, Division of Endocrinology, Metabolism Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
8
|
Dong S, Qian L, Cheng Z, Chen C, Wang K, Hu S, Zhang X, Wu T. Lactate and Myocadiac Energy Metabolism. Front Physiol 2021; 12:715081. [PMID: 34483967 PMCID: PMC8415870 DOI: 10.3389/fphys.2021.715081] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/29/2021] [Indexed: 12/05/2022] Open
Abstract
The myocardium is capable of utilizing different energy substrates, which is referred to as “metabolic flexibility.” This process assures ATP production from fatty acids, glucose, lactate, amino acids, and ketones, in the face of varying metabolic contexts. In the normal physiological state, the oxidation of fatty acids contributes to approximately 60% of energy required, and the oxidation of other substrates provides the rest. The accumulation of lactate in ischemic and hypoxic tissues has traditionally be considered as a by-product, and of little utility. However, recent evidence suggests that lactate may represent an important fuel for the myocardium during exercise or myocadiac stress. This new paradigm drives increasing interest in understanding its role in cardiac metabolism under both physiological and pathological conditions. In recent years, blood lactate has been regarded as a signal of stress in cardiac disease, linking to prognosis in patients with myocardial ischemia or heart failure. In this review, we discuss the importance of lactate as an energy source and its relevance to the progression and management of heart diseases.
Collapse
Affiliation(s)
- Shuohui Dong
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Linhui Qian
- Department of Colorectal and Anal Surgery, Feicheng Hospital Affiliated to Shandong First Medical University, Feicheng, China
| | - Zhiqiang Cheng
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Chang Chen
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Kexin Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Sanyuan Hu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Tongzhi Wu
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, Australia.,Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
9
|
Guo N, Chen Y, Yang X, Yan H, Fan B, Quan J, Wang M, Yang H. Urinary metabolomic profiling reveals difference between two traditional Chinese medicine subtypes of coronary heart disease. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1179:122808. [PMID: 34218095 DOI: 10.1016/j.jchromb.2021.122808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/21/2021] [Accepted: 05/24/2021] [Indexed: 11/28/2022]
Abstract
The World Health Organization has shown that coronary heart disease (CHD) is a more common cause of death than cancer. In traditional Chinese medicine (TCM), CHD is classified as a form of thoracic obstruction that can be divided in different subtypes including Qi stagnation with blood stasis (QS) and Qi deficiency with blood stasis (QD). Different treatment strategies are used based on this subtyping. Owing to the lack of scientific markers in the diagnosis of these subtypes, subjective judgments made by clinicians have limited the objective manner for utility of TCM in the treatment of CHD. Untargeted (UHPLC-QTOF-MS) and targeted (UHPLC-MS/MS) metabolomics approaches were employed to search significantly different metabolites related to the QS or QD subtypes of CHD with angina pectoris in this study. A total of 42 metabolites were obtained in the untargeted metabolomics analysis and 34 amino acids were detected in the targeted metabolomics analysis. In total, 16 metabolites were found significantly different among different groups. The results showed distinct metabolic profiles of urine samples not only between CHD patients and healthy controls, but also between the two subtypes of CHD. Pathway analysis of the significantly varied metabolites revealed that there were subtype-related differences in the activity of pathways. Therefore, urinary metabolomics can reveal the pathological changes of CHD in different subtypes, make the diagnosis of CHD in different subtypes in an objective manner and comprehensive and contribute to personalized treatment by providing scientific evidence.
Collapse
Affiliation(s)
- Na Guo
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, Center for Post-doctoral Research, China Academy of Chinese Medical Sciences, Beijing 100700, China; State Key Laboratory of Generic Manufacture Technology of Traditional Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Shandong 276006, China
| | - Yangan Chen
- LU-European Center for Chinese Medicine and Natural Compounds, Institute of Biology, Leiden University, Sylviusweg 72, 2333 BE Leiden, the Netherlands
| | - Xiaofang Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Han Yan
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Bin Fan
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jianye Quan
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Mei Wang
- LU-European Center for Chinese Medicine and Natural Compounds, Institute of Biology, Leiden University, Sylviusweg 72, 2333 BE Leiden, the Netherlands; SU BioMedicine, Post Bus 546, 2300 AM Leiden, the Netherlands.
| | - Hongjun Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
10
|
Wells MA, See Hoe LE, Heather LC, Molenaar P, Suen JY, Peart J, McGiffin D, Fraser JF. Peritransplant Cardiometabolic and Mitochondrial Function: The Missing Piece in Donor Heart Dysfunction and Graft Failure. Transplantation 2021; 105:496-508. [PMID: 33617201 DOI: 10.1097/tp.0000000000003368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Primary graft dysfunction is an important cause of morbidity and mortality after cardiac transplantation. Donor brain stem death (BSD) is a significant contributor to donor heart dysfunction and primary graft dysfunction. There remain substantial gaps in the mechanistic understanding of peritransplant cardiac dysfunction. One of these gaps is cardiac metabolism and metabolic function. The healthy heart is an "omnivore," capable of utilizing multiple sources of nutrients to fuel its enormous energetic demand. When this fails, metabolic inflexibility leads to myocardial dysfunction. Data have hinted at metabolic disturbance in the BSD donor and subsequent heart transplantation; however, there is limited evidence demonstrating specific metabolic or mitochondrial dysfunction. This review will examine the literature surrounding cardiometabolic and mitochondrial function in the BSD donor, organ preservation, and subsequent cardiac transplantation. A more comprehensive understanding of this subject may then help to identify important cardioprotective strategies to improve the number and quality of donor hearts.
Collapse
Affiliation(s)
- Matthew A Wells
- School of medical Science, Griffith University Gold Coast, Australia
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Australia
| | - Louise E See Hoe
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Australia
- Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, St Lucia, Australia
| | - Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Peter Molenaar
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane City, Australia
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Australia
- Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, St Lucia, Australia
| | - Jason Peart
- School of medical Science, Griffith University Gold Coast, Australia
| | - David McGiffin
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Australia
- Cardiothoracic Surgery and Transplantation, The Alfred Hospital, Melbourne, Australia
| | - John F Fraser
- School of medical Science, Griffith University Gold Coast, Australia
- Critical Care Research Group, The Prince Charles Hospital, Chermside, Australia
- Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, St Lucia, Australia
| |
Collapse
|
11
|
Liu XS, Zeng J, Yang YX, Qi CL, Xiong T, Wu GZ, Zeng CY, Wang DX. DRD4 Mitigates Myocardial Ischemia/Reperfusion Injury in Association With PI3K/AKT Mediated Glucose Metabolism. Front Pharmacol 2021; 11:619426. [PMID: 33584304 PMCID: PMC7873565 DOI: 10.3389/fphar.2020.619426] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
Ischemia-reperfusion (I/R) could cause heart irreversible damage, which is tightly combined with glucose metabolism disorder. It is demonstrated that GLUT4 (glucose transporter 4) translocation is critical for glucose metabolism in the cardiomyocytes under I/R injury. Moreover, DRD4 (dopamine receptor D4) modulate glucose metabolism, and protect neurocytes from anoxia/reoxygenation (A/R) injury. Thus, DRD4 might regulate myocardial I/R injury in association with GLUT4-mediated glucose metabolism. However, the effects and mechanisms are largely unknown. In the present study, the effect of DRD4 in heart I/R injury were studied ex vivo and in vitro. For I/R injury ex vivo, DRD4 agonist (PD168077) was perfused by Langendorff system in the isolated rat heart. DRD4 activated by PD168077 improved cardiac function in the I/R-injured heart as determined by the left ventricular developed pressure (LVDP), +dp/dt, and left ventricular end diastolic pressure (LVEDP), and reduced heart damage evidenced by infarct size, the release of troponin T (TNT) and lactate dehydrogenase (LDH). DRD4 activation diminished I/R injury induced apoptosis and enhanced cell viability impaired by I/R injury in cardiomyocyte, showed by TUNEL staining, flow cytometer and CCK8 assay. Furthermore, DRD4 activation did not change total GULT4 protein expression level but increased the membrane GULT4 localization determined by western blot. In terms of mechanism, DRD4 activation increased pPI3K/p-AKT but not the total PI3K/AKT during anoxia/reoxygenation (A/R) injury in vitro. Interestingly, PI3K inhibitor, Wortmannin, blocked PI3K/AKT pathway and depleted the membrane GULT4, and further promoted apoptosis showed by TUNEL staining, flow cytometer, western blot of cleaved caspase 3, BAX and BCL2 expression. Thus, DRD4 activation exerted a protective effect against I/R injury by promoting GLUT4 translocation depended on PI3K/AKT pathway, which enhanced the ability of glucose uptake, and ultimately reduced the apoptosis in cardiomyocytes.
Collapse
Affiliation(s)
- Xue-Song Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jing Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Yu-Xue Yang
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Chun-Lei Qi
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ting Xiong
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Geng-Ze Wu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Chun-Yu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Da-Xin Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou people's Hospital), Taizhou, China
| |
Collapse
|
12
|
Yehualashet AS, Belachew TF, Kifle ZD, Abebe AM. Targeting Cardiac Metabolic Pathways: A Role in Ischemic Management. Vasc Health Risk Manag 2020; 16:353-365. [PMID: 32982263 PMCID: PMC7501978 DOI: 10.2147/vhrm.s264130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Among the vast number of noncommunicable diseases encountered worldwide, cardiovascular diseases accounted for about 17.8 million deaths in 2017 and ischemic heart disease (IHD) remains the single-largest cause of death in countries across all income groups. Because conventional medications are not without shortcomings and patients still refractory to these medications, scientific investigation is ongoing to advance the management of IHD, and shows a great promise for better treatment modalities, but additional research can warrant improvement in terms of the quality of life of patients. Metabolic modulation is one promising strategy for the treatment of IHD, because alterations in energy metabolism are involved in progression of the disease. Therefore, the purpose of this review was to strengthen attention toward the use of metabolic modulators and to review the current level of knowledge on cardiac energy metabolic pathways.
Collapse
Affiliation(s)
- Awgichew Shewasinad Yehualashet
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Health Sciences, Debre Berhan University, Debre Berhan, Ethiopia
| | | | - Zemene Demelash Kifle
- School of Pharmacy, Department of Pharmacology, University of Gondar, Gondar, Ethiopia
| | - Ayele Mamo Abebe
- Department of Nursing, College of Health Sciences, Debre Berhan University, Debre Berhan, Ethiopia
| |
Collapse
|
13
|
Liu T, Gou L, Yan S, Huang T. Inhibition of acetyl-CoA carboxylase by PP-7a exerts beneficial effects on metabolic dysregulation in a mouse model of diet-induced obesity. Exp Ther Med 2020; 20:521-529. [PMID: 32550887 PMCID: PMC7296295 DOI: 10.3892/etm.2020.8700] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Acetyl-coenzyme A carboxylase (ACC) is a critical regulator of fatty acid metabolism and represents a promising therapeutic target for metabolic diseases, including obesity, type 2 diabetes and non-alcoholic fatty liver disease. Recently, a novel ACC inhibitor, PP-7a, was developed by our group by utilizing a structure-based drug design. In the present study, the pharmacological effects of PP-7a on the metabolic dysregulation in mice with high-fat diet (HFD)-induced obesity and the underlying mechanisms were investigated. The inhibitory effect on ACC activities was confirmed by assessing the level of malonyl-CoA, a product synthesized by the catalyzation of ACC. Following 16 weeks of being fed an HFD, the mice were administered PP-7a (15, 45 or 75 mg/kg) for 4 weeks. The effects of PP-7a on weight gain, glucose intolerance, hepatic lipid accumulation and the increase of serum triglyceride (TG), total cholesterol (TC) and free fatty acids (FFA) in mice were assessed. CP-640186 was used as a positive control drug and administered in the same manner as PP-7a. Chronic administration of PP-7a lowered the malonyl-CoA levels in liver and heart tissues of mice in the HFD group. In addition, HFD-induced weight gain and glucose intolerance were improved by PP-7a treatment in the mice fed the HFD. Furthermore, PP-7a suppressed hepatic lipid accumulation and the increase in TG, TC and FFA levels. Taken together, these results suggest that ACC inhibition by PP-7a may have a beneficial effect on metabolic dysregulation in obese mice.
Collapse
Affiliation(s)
- Tianya Liu
- Department of Pharmacy, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- Jiangsu Province Key Laboratory of Anesthesiology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Lingshan Gou
- Center for Genetic Medicine, Maternity and Child Health Care Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Shirong Yan
- Jiangsu Province Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Tonghui Huang
- Jiangsu Province Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| |
Collapse
|
14
|
Snyder J, Zhai R, Lackey AI, Sato PY. Changes in Myocardial Metabolism Preceding Sudden Cardiac Death. Front Physiol 2020; 11:640. [PMID: 32612538 PMCID: PMC7308560 DOI: 10.3389/fphys.2020.00640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Heart disease is widely recognized as a major cause of death worldwide and is the leading cause of mortality in the United States. Centuries of research have focused on defining mechanistic alterations that drive cardiac pathogenesis, yet sudden cardiac death (SCD) remains a common unpredictable event that claims lives in every age group. The heart supplies blood to all tissues while maintaining a constant electrical and hormonal feedback communication with other parts of the body. As such, recent research has focused on understanding how myocardial electrical and structural properties are altered by cardiac metabolism and the various signaling pathways associated with it. The importance of cardiac metabolism in maintaining myocardial function, or lack thereof, is exemplified by shifts in cardiac substrate preference during normal development and various pathological conditions. For instance, a shift from fatty acid (FA) oxidation to oxygen-sparing glycolytic energy production has been reported in many types of cardiac pathologies. Compounded by an uncoupling of glycolysis and glucose oxidation this leads to accumulation of undesirable levels of intermediate metabolites. The resulting accumulation of intermediary metabolites impacts cardiac mitochondrial function and dysregulates metabolic pathways through several mechanisms, which will be reviewed here. Importantly, reversal of metabolic maladaptation has been shown to elicit positive therapeutic effects, limiting cardiac remodeling and at least partially restoring contractile efficiency. Therein, the underlying metabolic adaptations in an array of pathological conditions as well as recently discovered downstream effects of various substrate utilization provide guidance for future therapeutic targeting. Here, we will review recent data on alterations in substrate utilization in the healthy and diseased heart, metabolic pathways governing cardiac pathogenesis, mitochondrial function in the diseased myocardium, and potential metabolism-based therapeutic interventions in disease.
Collapse
Affiliation(s)
- J Snyder
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - R Zhai
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - A I Lackey
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - P Y Sato
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
15
|
Wen J, Tian L, Xu M, Zhou X, Zhang Y, Cai M. A Synthetic Malonyl-CoA Metabolic Oscillator in Komagataella phaffii. ACS Synth Biol 2020; 9:1059-1068. [PMID: 32227991 DOI: 10.1021/acssynbio.9b00378] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Malonyl-CoA is a key metabolic molecule that participates in a diverse range of physiological responses and can act as a building block for a variety of value-added pharmaceuticals and chemicals. The cytosolic malonyl-CoA concentration is usually very low, and thus dynamic metabolic control of malonyl-CoA variation will aid its stable formation and efficient consumption. We developed a synthetic malonyl-CoA metabolic oscillator in yeast. A synthetic regulatory protein, Prm1-FapR, was constructed by fusing a yeast transcriptional activator, Prm1, with a bacterial malonyl-CoA-sensitive transcription repressor, FapR. Two oppositely regulated biosensors were then engineered. A total of 18 hybrid promoter variants were designed, each carrying the operator sequence (fapO) of FapR and the core promoter of PAOX1 (cPAOX1), which is naturally regulated by Prm1. The promoter activities of these variants, regulated by Prm1-FapR, were tested. Through this process, a sensor for Prm1-FapR/(-52)fapO-PAOX1 carrying an activation/deactivation regulation module was built. Meanwhile, 24 promoter variants of PGAP with fapO inserted were designed and tested using the fusion regulator, giving a sensor for Prm1-FapR/PGAP-(+22) fapO that contained a repression/derepression regulation module. Both sensors were subsequently integrated into a single cell, which exhibited correct metabolic switching of eGFP and mCherry reporters following manipulation of cytosolic malonyl-CoA levels. The Prm1-FapR/(-52)fapO-PAOX1 and the Prm1-FapR/PGAP-(+22)fapO were also used to control the malonyl-CoA source and sink pathways, respectively, for the synthesis of 6-methylsalicylic acid. This finally led to an oscillatory metabolic mode of cytosolic malonyl-CoA. Such a metabolator is useful in exploring potential industrial and biomedical applications not limited by natural cellular behavior.
Collapse
Affiliation(s)
- Jiao Wen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Lin Tian
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Mingqiang Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiangshan Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- Shanghai Collaborative Innovation Center for Biomanufacturing, 130 Meilong Road, Shanghai 200237, China
| | - Menghao Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- Shanghai Collaborative Innovation Center for Biomanufacturing, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
16
|
Involvement of fatty acid synthase in right ventricle dysfunction in pulmonary hypertension. Exp Cell Res 2019; 383:111569. [DOI: 10.1016/j.yexcr.2019.111569] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 08/09/2019] [Accepted: 08/20/2019] [Indexed: 10/26/2022]
|
17
|
Karwi QG, Uddin GM, Ho KL, Lopaschuk GD. Loss of Metabolic Flexibility in the Failing Heart. Front Cardiovasc Med 2018; 5:68. [PMID: 29928647 PMCID: PMC5997788 DOI: 10.3389/fcvm.2018.00068] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022] Open
Abstract
To maintain its high energy demand the heart is equipped with a highly complex and efficient enzymatic machinery that orchestrates ATP production using multiple energy substrates, namely fatty acids, carbohydrates (glucose and lactate), ketones and amino acids. The contribution of these individual substrates to ATP production can dramatically change, depending on such variables as substrate availability, hormonal status and energy demand. This "metabolic flexibility" is a remarkable virtue of the heart, which allows utilization of different energy substrates at different rates to maintain contractile function. In heart failure, cardiac function is reduced, which is accompanied by discernible energy metabolism perturbations and impaired metabolic flexibility. While it is generally agreed that overall mitochondrial ATP production is impaired in the failing heart, there is less consensus as to what actual switches in energy substrate preference occur. The failing heart shift toward a greater reliance on glycolysis and ketone body oxidation as a source of energy, with a decrease in the contribution of glucose oxidation to mitochondrial oxidative metabolism. The heart also becomes insulin resistant. However, there is less consensus as to what happens to fatty acid oxidation in heart failure. While it is generally believed that fatty acid oxidation decreases, a number of clinical and experimental studies suggest that fatty acid oxidation is either not changed or is increased in heart failure. Of importance, is that any metabolic shift that does occur has the potential to aggravate cardiac dysfunction and the progression of the heart failure. An increasing body of evidence shows that increasing cardiac ATP production and/or modulating cardiac energy substrate preference positively correlates with heart function and can lead to better outcomes. This includes increasing glucose and ketone oxidation and decreasing fatty acid oxidation. In this review we present the physiology of the energy metabolism pathways in the heart and the changes that occur in these pathways in heart failure. We also look at the interventions which are aimed at manipulating the myocardial metabolic pathways toward more efficient substrate utilization which will eventually improve cardiac performance.
Collapse
Affiliation(s)
| | | | | | - Gary D. Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
18
|
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
|
19
|
Design, synthesis and in vitro evaluation of some small molecules malonyl CoA decarboxylase inhibitors containing pyrazoline scaffold and study of their binding interactions with malonyl CoA decarboxylase via preliminary docking simulation. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1917-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
20
|
Zlobine I, Gopal K, Ussher JR. Lipotoxicity in obesity and diabetes-related cardiac dysfunction. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1555-68. [DOI: 10.1016/j.bbalip.2016.02.011] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/15/2016] [Indexed: 12/11/2022]
|
21
|
Ussher JR, Fillmore N, Keung W, Zhang L, Mori J, Sidhu VK, Fukushima A, Gopal K, Lopaschuk DG, Wagg CS, Jaswal JS, Dyck JRB, Lopaschuk GD. Genetic and Pharmacological Inhibition of Malonyl CoA Decarboxylase Does Not Exacerbate Age-Related Insulin Resistance in Mice. Diabetes 2016; 65:1883-91. [PMID: 27207536 DOI: 10.2337/db15-1145] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 03/31/2016] [Indexed: 11/13/2022]
Abstract
Aging is associated with the development of chronic diseases such as insulin resistance and type 2 diabetes. A reduction in mitochondrial fat oxidation is postulated to be a key factor contributing to the progression of these diseases. Our aim was to investigate the contribution of impaired mitochondrial fat oxidation toward age-related disease. Mice deficient for malonyl CoA decarboxylase (MCD(-/-)), a mouse model of reduced fat oxidation, were allowed to age while life span and a number of physiological parameters (glucose tolerance, insulin tolerance, indirect calorimetry) were assessed. Decreased fat oxidation in MCD(-/-) mice resulted in the accumulation of lipid intermediates in peripheral tissues, but this was not associated with a worsening of age-associated insulin resistance and, conversely, improved longevity. This improvement was associated with reduced oxidative stress and reduced acetylation of the antioxidant enzyme superoxide dismutase 2 in muscle but not the liver of MCD(-/-) mice. These findings were recapitulated in aged mice treated with an MCD inhibitor (CBM-3001106), and these mice also demonstrated improvements in glucose and insulin tolerance. Therefore, our results demonstrate that in addition to decreasing fat oxidation, MCD inhibition also has novel effects on protein acetylation. These combined effects protect against age-related metabolic dysfunction, demonstrating that MCD inhibitors may have utility in the battle against chronic disease in the elderly.
Collapse
Affiliation(s)
- John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Natasha Fillmore
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Wendy Keung
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Liyan Zhang
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Jun Mori
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Vaninder K Sidhu
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Arata Fukushima
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - David G Lopaschuk
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Cory S Wagg
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Jagdip S Jaswal
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Jason R B Dyck
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Gary D Lopaschuk
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
22
|
Li R, Li F, Feng Q, Liu Z, Jie Z, Wen B, Xu X, Zhong S, Li G, He K. An LC-MS based untargeted metabolomics study identified novel biomarkers for coronary heart disease. MOLECULAR BIOSYSTEMS 2016; 12:3425-3434. [PMID: 27713999 DOI: 10.1039/c6mb00339g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This work leads to candidate diagnosis biomarker discovery in coronary heart disease which can be developed as a non-invasive and feasible method for CHD diagnosis.
Collapse
Affiliation(s)
- Ruijun Li
- Department of Cardiology
- Chinese PLA General Hospital
- Beijing
- China
| | - Fengyu Li
- BGI Education Center
- University of Chinese Academy of Sciences
- Shenzhen
- China
- BGI-Shenzhen
| | - Qiang Feng
- BGI-Shenzhen
- Shenzhen
- China
- Department of Biology
- University of Copenhagen
| | | | | | - Bo Wen
- BGI-Shenzhen
- Shenzhen
- China
| | - Xun Xu
- BGI-Shenzhen
- Shenzhen
- China
| | - Shilong Zhong
- Medical Research Center of Guangdong General Hospital
- Guangzhou
- China
- Guangdong Cardiovascular Institute
- Guangdong Academy of Medical Sciences
| | | | - Kunlun He
- Department of Cardiology
- Chinese PLA General Hospital
- Beijing
- China
| |
Collapse
|
23
|
Talati M, Hemnes A. Fatty acid metabolism in pulmonary arterial hypertension: role in right ventricular dysfunction and hypertrophy. Pulm Circ 2015; 5:269-78. [PMID: 26064451 DOI: 10.1086/681227] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 12/30/2014] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex, multifactorial disease in which an increase in pulmonary vascular resistance leads to increased afterload on the right ventricle (RV), causing right heart failure and death. Our understanding of the pathophysiology of RV dysfunction in PAH is limited but is constantly improving. Increasing evidence suggests that in PAH RV dysfunction is associated with various components of metabolic syndrome, such as insulin resistance, hyperglycemia, and dyslipidemia. The relationship between RV dysfunction and fatty acid/glucose metabolites is multifaceted, and in PAH it is characterized by a shift in utilization of energy sources toward increased glucose utilization and reduced fatty acid consumption. RV dysfunction may be caused by maladaptive fatty acid metabolism resulting from an increase in fatty acid uptake by fatty acid transporter molecule CD36 and an imbalance between glucose and fatty acid oxidation in mitochondria. This leads to lipid accumulation in the form of triglycerides, diacylglycerol, and ceramides in the cytoplasm, hallmarks of lipotoxicity. Current interventions in animal models focus on improving RV dysfunction through altering fatty acid oxidation rates and limiting lipid accumulation, but more specific and effective therapies may be available in the coming years based on current research. In conclusion, a deeper understanding of the complex mechanisms of the metabolic remodeling of the RV will aid in the development of targeted treatments for RV failure in PAH.
Collapse
Affiliation(s)
- Megha Talati
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anna Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
24
|
Sankaralingam S, Lopaschuk GD. Cardiac energy metabolic alterations in pressure overload-induced left and right heart failure (2013 Grover Conference Series). Pulm Circ 2015; 5:15-28. [PMID: 25992268 DOI: 10.1086/679608] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 07/29/2014] [Indexed: 01/07/2023] Open
Abstract
Pressure overload of the heart, such as seen with pulmonary hypertension and/or systemic hypertension, can result in cardiac hypertrophy and the eventual development of heart failure. The development of hypertrophy and heart failure is accompanied by numerous molecular changes in the heart, including alterations in cardiac energy metabolism. Under normal conditions, the high energy (adenosine triphosphate [ATP]) demands of the heart are primarily provided by the mitochondrial oxidation of fatty acids, carbohydrates (glucose and lactate), and ketones. In contrast, the hypertrophied failing heart is energy deficient because of its inability to produce adequate amounts of ATP. This can be attributed to a reduction in mitochondrial oxidative metabolism, with the heart becoming more reliant on glycolysis as a source of ATP production. If glycolysis is uncoupled from glucose oxidation, a decrease in cardiac efficiency can occur, which can contribute to the severity of heart failure due to pressure-overload hypertrophy. These metabolic changes are accompanied by alterations in the enzymes that are involved in the regulation of fatty acid and carbohydrate metabolism. It is now becoming clear that optimizing both energy production and the source of energy production are potential targets for pharmacological intervention aimed at improving cardiac function in the hypertrophied failing heart. In this review, we will focus on what alterations in energy metabolism occur in pressure overload induced left and right heart failure. We will also discuss potential targets and pharmacological approaches that can be used to treat heart failure occurring secondary to pulmonary hypertension and/or systemic hypertension.
Collapse
Affiliation(s)
| | - Gary D Lopaschuk
- Department of Pediatrics, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
25
|
Sun W, Liu Q, Leng J, Zheng Y, Li J. The role of Pyruvate Dehydrogenase Complex in cardiovascular diseases. Life Sci 2014; 121:97-103. [PMID: 25498896 DOI: 10.1016/j.lfs.2014.11.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/10/2014] [Accepted: 11/28/2014] [Indexed: 12/23/2022]
Abstract
The regulation of mammalian myocardial carbohydrate metabolism is complex; many factors such as arterial substrate and hormone levels, coronary flow, inotropic state and the nutritional status of the tissue play a role in regulating mammalian myocardial carbohydrate metabolism. The Pyruvate Dehydrogenase Complex (PDHc), a mitochondrial matrix multienzyme complex, plays an important role in energy homeostasis in the heart by providing the link between glycolysis and the tricarboxylic acid (TCA) cycle. In TCA cycle, PDHc catalyzes the conversion of pyruvate into acetyl-CoA. This review determines that there is altered cardiac glucose in various pathophysiological states consequently causing PDC to be altered. This review further summarizes evidence for the metabolism mechanism of the heart under normal and pathological conditions including ischemia, diabetes, hypertrophy and heart failure.
Collapse
Affiliation(s)
- Wanqing Sun
- Division of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Quan Liu
- Division of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Jiyan Leng
- Division of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Yang Zheng
- Division of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Ji Li
- Division of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
26
|
Ussher JR. Sweet as sugar: excessive glucose metabolism in the failing heart. Future Cardiol 2014; 10:465-8. [PMID: 25301309 DOI: 10.2217/fca.14.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
27
|
Hu J, Wang X, Liu Q, Zhang K, Xiong W, Xu C, Wang P, Leung AW. Antitumor Effect of Sinoporphyrin Sodium-Mediated Photodynamic Therapy on Human Esophageal Cancer Eca-109 Cells. Photochem Photobiol 2014; 90:1404-12. [DOI: 10.1111/php.12333] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 08/13/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Jianmin Hu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry; Ministry of Education; National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China; College of Life Sciences; Shaanxi Normal University; Xi'an Shaanxi China
| | - Xiaobing Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry; Ministry of Education; National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China; College of Life Sciences; Shaanxi Normal University; Xi'an Shaanxi China
| | - Quanhong Liu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry; Ministry of Education; National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China; College of Life Sciences; Shaanxi Normal University; Xi'an Shaanxi China
| | - Kun Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry; Ministry of Education; National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China; College of Life Sciences; Shaanxi Normal University; Xi'an Shaanxi China
| | - Wenli Xiong
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry; Ministry of Education; National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China; College of Life Sciences; Shaanxi Normal University; Xi'an Shaanxi China
| | - Chuanshan Xu
- School of Chinese Medicine; Faculty of Medicine; The Chinese University of Hong Kong; Shatin Hong Kong China
| | - Pan Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry; Ministry of Education; National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China; College of Life Sciences; Shaanxi Normal University; Xi'an Shaanxi China
- School of Chinese Medicine; Faculty of Medicine; The Chinese University of Hong Kong; Shatin Hong Kong China
| | - Albert Wingnang Leung
- School of Chinese Medicine; Faculty of Medicine; The Chinese University of Hong Kong; Shatin Hong Kong China
| |
Collapse
|
28
|
Ussher JR, Keung W, Fillmore N, Koves TR, Mori J, Zhang L, Lopaschuk DG, Ilkayeva OR, Wagg CS, Jaswal JS, Muoio DM, Lopaschuk GD. Treatment with the 3-ketoacyl-CoA thiolase inhibitor trimetazidine does not exacerbate whole-body insulin resistance in obese mice. J Pharmacol Exp Ther 2014; 349:487-96. [PMID: 24700885 DOI: 10.1124/jpet.114.214197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
There is a growing need to understand the underlying mechanisms involved in the progression of cardiovascular disease during obesity and diabetes. Although inhibition of fatty acid oxidation has been proposed as a novel approach to treat ischemic heart disease and heart failure, reduced muscle fatty acid oxidation rates may contribute to the development of obesity-associated insulin resistance. Our aim was to determine whether treatment with the antianginal agent trimetazidine, which inhibits fatty acid oxidation in the heart secondary to inhibition of 3-ketoacyl-CoA thiolase (3-KAT), may have off-target effects on glycemic control in obesity. We fed C57BL/6NCrl mice a high-fat diet (HFD) for 10 weeks before a 22-day treatment with the 3-KAT inhibitor trimetazidine (15 mg/kg per day). Insulin resistance was assessed via glucose/insulin tolerance testing, and lipid metabolite content was assessed in gastrocnemius muscle. Trimetazidine-treatment led to a mild shift in substrate preference toward carbohydrates as an oxidative fuel source in obese mice, evidenced by an increase in the respiratory exchange ratio. This shift in metabolism was accompanied by an accumulation of long-chain acyl-CoA and a trend to an increase in triacylglycerol content in gastrocnemius muscle, but did not exacerbate HFD-induced insulin resistance compared with control-treated mice. It is noteworthy that trimetazidine treatment reduced palmitate oxidation rates in the isolated working mouse heart and neonatal cardiomyocytes but not C2C12 skeletal myotubes. Our findings demonstrate that trimetazidine therapy does not adversely affect HFD-induced insulin resistance, suggesting that treatment with trimetazidine would not worsen glycemic control in obese patients with angina.
Collapse
Affiliation(s)
- John R Ussher
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada (J.R.U., W.K., N.F., J.M., L.Z., D.G.L., C.S.W., J.S.J., G.D.L.); and Sarah W. Stedman Nutrition and Metabolism Center (T.R.K., O.R.I., D.M.M.), Department of Medicine (T.R.K., O.R.I., D.M.M.), Department of Pharmacology and Cancer Biology (D.M.M.), Duke University, Durham, North Carolina
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Cardiomyopathy, the presence of cardiac dysfunction independent of ischemic heart disease and/or hypertension, is becoming a more prominent condition in our diabetic patient population. Unfortunately, we do not yet understand the mechanism(s) responsible for causing diabetic cardiomyopathy. With the recent explosion in the obesity and Type 2 diabetes epidemic, our understanding of dyslipidemia and the adverse effects of lipid surplus on cellular and organ function has grown considerably. Numerous studies now illustrate that excess lipid accumulation may exert direct toxic effects on cellular function, a term coined 'lipotoxicity'. As obesity and Type 2 diabetes are significant risk factors for cardiovascular disease, cardiac lipotoxicity may represent a significant component mediating the diabetic cardiomyopathy phenotype. Therefore, a more complete understanding of how cardiac lipotoxicity is regulated and how different lipid metabolites cause cellular dysfunction may lead to the discovery of novel targets to treat cardiomyopathy in our diabetic patient population.
Collapse
Affiliation(s)
- John R Ussher
- Lunenfeld-Tanenbaum, Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Lee CT, Ussher JR, Mohammad A, Lam A, Lopaschuk GD. 5'-AMP-activated protein kinase increases glucose uptake independent of GLUT4 translocation in cardiac myocytes. Can J Physiol Pharmacol 2014; 92:307-14. [PMID: 24708213 DOI: 10.1139/cjpp-2013-0107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glucose uptake and glycolysis are increased in the heart during ischemia, and this metabolic alteration constitutes an important contributing factor towards ischemic injury. Therefore, it is important to understand glucose uptake regulation in the ischemic heart. There are primarily 2 glucose transporters controlling glucose uptake into cardiac myocytes: GLUT1 and GLUT4. In the non-ischemic heart, insulin stimulates GLUT4 translocation to the sarcolemmal membrane, while both GLUT1 and GLUT4 translocation can occur following AMPK stimulation. Using a newly developed technique involving [(3)H]2-deoxyglucose, we measured glucose uptake in H9c2 ventricular myoblasts, and demonstrated that while insulin has no detectable effect on glucose uptake, phenformin-induced AMPK activation increases glucose uptake 2.5-fold. Furthermore, insulin treatment produced no discernible effect on either Akt serine 473 phosphorylation or AMPKα threonine 172 phosphorylation, while treatment with phenformin results in an increase in AMPKα threonine 172 phosphorylation, and a decrease in Akt serine 473 phosphorylation. Visualization of a dsRed-GLUT4 fusion construct in H9c2 cells by laser confocal microscopy showed that unlike insulin, AMPK activation did not redistribute GLUT4 to the sarcolemmal membrane, suggesting that AMPK may regulate glucose uptake via another glucose transporter. These studies suggest that AMPK is a major regulator of glucose uptake in cardiac myocytes.
Collapse
Affiliation(s)
- Christopher T Lee
- a Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, 8440 112 Street NW, Edmonton, AB T6G 2P4, Canada
| | | | | | | | | |
Collapse
|
31
|
Wu H, Zhu Q, Cai M, Tong X, Liu D, Huang J, Yang G, Jiang Y. Effect of Inhibiting Malonyl-CoA Decarboxylase on Cardiac Remodeling after Myocardial Infarction in Rats. Cardiology 2014; 127:236-44. [DOI: 10.1159/000356471] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 10/09/2013] [Indexed: 11/19/2022]
|
32
|
Li Q, Zhang S, Berthiaume JM, Simons B, Zhang GF. Novel approach in LC-MS/MS using MRM to generate a full profile of acyl-CoAs: discovery of acyl-dephospho-CoAs. J Lipid Res 2013; 55:592-602. [PMID: 24367045 DOI: 10.1194/jlr.d045112] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
A metabolomic approach to selectively profile all acyl-CoAs was developed using a programmed multiple reaction monitoring (MRM) method in LC-MS/MS and was employed in the analysis of various rat organs. The programmed MRM method possessed 300 mass ion transitions with the mass difference of 507 between precursor ion (Q1) and product ion (Q3), and the precursor ion started from m/z 768 and progressively increased one mass unit at each step. Acyl-dephospho-CoAs resulting from the dephosphorylation of acyl-CoAs were identified by accurate MS and fragmentation. Acyl-dephospho-CoAs were also quantitatively scanned by the MRM method with the mass difference of 427 between Q1 and Q3 mass ions. Acyl-CoAs and dephospho-CoAs were assayed with limits of detection ranging from 2 to 133 nM. The accuracy of the method was demonstrated by assaying a range of concentrations of spiked acyl-CoAs with the results of 80-114%. The distribution of acyl-CoAs reflects the metabolic status of each organ. The physiological role of dephosphorylation of acyl-CoAs remains to be further characterized. The methodology described herein provides a novel strategy in metabolomic studies to quantitatively and qualitatively profile all potential acyl-CoAs and acyl-dephospho-CoAs.
Collapse
Affiliation(s)
- Qingling Li
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106
| | | | | | | | | |
Collapse
|
33
|
Ussher JR, Lopaschuk GD, Arduini A. Gut microbiota metabolism of L-carnitine and cardiovascular risk. Atherosclerosis 2013; 231:456-61. [PMID: 24267266 DOI: 10.1016/j.atherosclerosis.2013.10.013] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/12/2013] [Accepted: 10/14/2013] [Indexed: 02/07/2023]
Abstract
In recent years, a number of studies have alluded to the importance of the intestinal microflora in controlling whole-body metabolic homeostasis and organ physiology. In particular, it has been suggested that the hepatic production of trimethylamine-N-oxide (TMAO) from gut microbiota-derived trimethylamine (TMA) may enhance cardiovascular risk via promoting atherosclerotic lesion development. The source of TMA production via the gut microbiota appears to originate from 2 principle sources, either phosphatidylcholine/choline and/or L-carnitine. Therefore, it has been postulated that consumption of these dietary sources, which are often found in large quantities in red meats, may be critical factors promoting cardiovascular risk. In contrast, a number of studies demonstrate beneficial properties for l-carnitine consumption against metabolic diseases including skeletal muscle insulin resistance and ischemic heart disease. Furthermore, fish are a significant source of TMAO, but dietary fish consumption and fish oil supplementation may exhibit positive effects on cardiovascular health. In this mini-review we will discuss the discrepancies regarding L-carnitine supplementation and its possible negative effects on cardiovascular risk through potential increases in TMAO production, as well as its positive effects on metabolic health via increasing glucose metabolism in the muscle and heart.
Collapse
Affiliation(s)
- John R Ussher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, 5-1001-DD, 25 Orde Street, Toronto, Ontario M5T 3H7, Canada.
| | | | | |
Collapse
|
34
|
García-Ruiz C, Baulies A, Mari M, García-Rovés PM, Fernandez-Checa JC. Mitochondrial dysfunction in non-alcoholic fatty liver disease and insulin resistance: Cause or consequence? Free Radic Res 2013; 47:854-68. [DOI: 10.3109/10715762.2013.830717] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
35
|
Wei B, You MG, Ling JJ, Wei LL, Wang K, Li WW, Chen T, Du QM, Ji H. Regulation of antioxidant system, lipids and fatty acid β-oxidation contributes to the cardioprotective effect of sodium tanshinone IIA sulphonate in isoproterenol-induced myocardial infarction in rats. Atherosclerosis 2013; 230:148-56. [PMID: 23958267 DOI: 10.1016/j.atherosclerosis.2013.07.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 06/28/2013] [Accepted: 07/09/2013] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Myocardial infarction (MI) is a cause of high morbidity and mortality in the world. Sodium tanshinone IIA sulphonate (STS) has been well used in Oriental medicine for treating cardiovascular diseases, however, the underlying mechanisms remain unclear. Alterations of circulating lipid profiles, increased fatty acid β-oxidation and oxidative stress play most important roles in the pathogenesis of MI. The present study aims to elucidate whether STS possesses cardioprotective effect against MI driven by isoproterenol (ISO), and to investigate its potential mechanisms of action. METHODS MI was induced by subcutaneous injection of ISO (85 mg/kg at interval of 24 h for 2 consecutive days) to rats. The rats were randomly divided into 6 groups: (1) control; (2) ISO; (3) STS (16 mg/kg) +control; (4-6) STS (16, 8, 4 mg/kg) +ISO. RESULTS Our study showed that STS could ameliorate cardiac dysfunction and variation of myocardial zymogram, up-regulate antioxidant systems, and maintain the levels of circulating lipids driven by supramaximal doses ISO as well. Moreover, modulation of redox-sensitive extracellular signal-regulated kinase1/2 (ERK1/2)/Nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) and AMP-activated protein kinase (AMPK)/acetyl CoA carboxylase (ACC)/carnitine palmitoyltransferase (CPT) 1 pathways were involved in STS induced cardioprotection. CONCLUSIONS STS exerts strong favorable cardioprotective action. Additionally, the properties of STS, such as anti-dyslipidemia, anti-oxidant and inhibition of fatty acid β-oxidation, may be the mechanisms underlying the observed results.
Collapse
Affiliation(s)
- Bo Wei
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu Province, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ussher JR, Sutendra G, Jaswal JS. The impact of current and novel anti-diabetic therapies on cardiovascular risk. Future Cardiol 2013. [PMID: 23176691 DOI: 10.2217/fca.12.68] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) has become an overwhelming health condition that is no longer just a threat to developed nations, but to undeveloped nations as well. Current therapies for T2DM are relatively effective in controlling hyperglycemia; examples include metformin, thiazolidinediones, sulfonylurea derivatives, α-glucosidase inhibitors, glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Despite their efficacy in controlling hyperglycemia, due to recent findings of increased cardiovascular risk following treatment with either rosiglitazone or intensive glucose lowering, new guidelines from the US FDA recommend that new therapies for diabetes not only improve glycemia, but exert no adverse cardiovascular effects. Based on cardiovascular risk profiles, metformin appears to be the superior anti-diabetic therapy, although studies in humans with glucagon-like peptide-1 receptor agonists are encouraging. As patients with T2DM also often have cardiovascular disease, the increased rigor in drug development should ultimately reduce the health burden of both of these conditions.
Collapse
Affiliation(s)
- John R Ussher
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Canada.
| | | | | |
Collapse
|
37
|
Grossman AN, Opie LH, Beshansky JR, Ingwall JS, Rackley CE, Selker HP. Glucose-insulin-potassium revived: current status in acute coronary syndromes and the energy-depleted heart. Circulation 2013; 127:1040-8. [PMID: 23459576 DOI: 10.1161/circulationaha.112.130625] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
38
|
Keung W, Ussher JR, Jaswal JS, Raubenheimer M, Lam VH, Wagg CS, Lopaschuk GD. Inhibition of carnitine palmitoyltransferase-1 activity alleviates insulin resistance in diet-induced obese mice. Diabetes 2013; 62:711-20. [PMID: 23139350 PMCID: PMC3581198 DOI: 10.2337/db12-0259] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Impaired skeletal muscle fatty acid oxidation has been suggested to contribute to insulin resistance and glucose intolerance. However, increasing muscle fatty acid oxidation may cause a reciprocal decrease in glucose oxidation, which might impair insulin sensitivity and glucose tolerance. We therefore investigated what effect inhibition of mitochondrial fatty acid uptake has on whole-body glucose tolerance and insulin sensitivity in obese insulin-resistant mice. C57BL/6 mice were fed a high-fat diet (60% calories from fat) for 12 weeks to develop insulin resistance. Subsequent treatment of mice for 4 weeks with the carnitine palmitoyltransferase-1 inhibitor, oxfenicine (150 mg/kg i.p. daily), resulted in improved whole-body glucose tolerance and insulin sensitivity. Exercise capacity was increased in oxfenicine-treated mice, which was accompanied by an increased respiratory exchange ratio. In the gastrocnemius muscle, oxfenicine increased pyruvate dehydrogenase activity, membrane GLUT4 content, and insulin-stimulated Akt phosphorylation. Intramyocellular levels of lipid intermediates, including ceramide, long-chain acyl CoA, and diacylglycerol, were also decreased. Our results demonstrate that inhibition of mitochondrial fatty acid uptake improves insulin sensitivity in diet-induced obese mice. This is associated with increased carbohydrate utilization and improved insulin signaling in the skeletal muscle, suggestive of an operating Randle Cycle in muscle.
Collapse
|
39
|
Ku HC, Chen WP, Su MJ. DPP4 deficiency exerts protective effect against H2O2 induced oxidative stress in isolated cardiomyocytes. PLoS One 2013; 8:e54518. [PMID: 23359639 PMCID: PMC3554719 DOI: 10.1371/journal.pone.0054518] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/12/2012] [Indexed: 01/01/2023] Open
Abstract
Apart from the antihyperglycemic effects, DPP4 inhibitors and GLP-1 molecules are involved in the preservation of cardiac functions. We have demonstrated that DPP4-deficient rats possess resistance to endotoxemia and ischemia/reperfusion stress. However, whether the decrease of DPP4 activity simply augmented the GLP-1 signaling or that such decrease resulted in a change of cellular function remain unclear. Accordingly, we investigated the responses of H2O2-induced oxidative stress in adult wild-type and DPP4-deficient rats isolated cardiomyocytes. The coadministration of GLP-1 or DPP4 inhibitor was also performed to define the mechanisms. Cell viability, ROS concentration, catalase activity, glucose uptake, prosurvival, proapoptotic signaling, and contractile function were examined after cells exposed to H2O2. DPP4-deficient cardiomyocytes were found to be resistant to H2O2-induced cell death via activating AKT signaling, enhancing glucose uptake, preserving catalase activity, diminishing ROS level and proapoptotic signaling. GLP-1 concentration-dependently improved cell viability in wild-type cardiomyocyte against ROS stress, and the ceiling response concentration (200 nM) was chosen for studies. GLP-1 was shown to decrease H2O2-induced cell death by its receptor-dependent AKT pathway in wild-type cardiomyocytes, but failed to cause further activation of AKT in DPP4-deficient cardiomyocytes. Acute treatment of DPP4 inhibitor only augmented the protective effect of low dose GLP-1, but failed to alter fuel utilization or ameliorate cell viability in wild-type cardiomyocytes after H2O2 exposure. The improvement of cell viability after H2O2 exposure was correlated with the alleviation of cellular contractile dysfunction in both DPP4-deficient and GLP-1 treated wild-type cardiomyocytes. These findings demonstrated that GLP-1 receptor-dependent pathway is important and exert protective effect in wild-type cardiomyocyte. Long term loss of DPP4 activity increased the capability against ROS stress, which was more than GLP-1 dependent pathway.
Collapse
Affiliation(s)
- Hui-Chun Ku
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Pin Chen
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Jai Su
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
40
|
Singh KK, Shukla PC, Yanagawa B, Quan A, Lovren F, Pan Y, Wagg CS, Teoh H, Lopaschuk GD, Verma S. Regulating cardiac energy metabolism and bioenergetics by targeting the DNA damage repair protein BRCA1. J Thorac Cardiovasc Surg 2013; 146:702-9. [PMID: 23317938 DOI: 10.1016/j.jtcvs.2012.12.046] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/15/2012] [Accepted: 12/11/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Alterations in cardiac energy and substrate metabolism play a critical role in the development and clinical course of heart failure. We hypothesized that the cardioprotective role of the breast cancer 1, early onset (BRCA1) gene might be mediated in part by alterations in cardiac bioenergetics. METHODS We generated cardiomyocyte-specific BRCA1 homozygous and heterozygous knockout mice using the Cre-loxP technology and evaluated the key molecules and pathways involved in glucose metabolism, fatty acid metabolism, and mitochondrial bioenergetics. RESULTS Cardiomyocyte-specific BRCA1-deficient mice showed reduced cardiac expression of glucose and fatty acid transporters, reduced acetyl-coenzyme A carboxylase 2 and malonyl-coenzyme A decarboxylase (key enzymes that control malonyl coenzyme A, which in turn controls fatty acid oxidation), and reduced carnitine palmitoyltransferase I, a rate-limiting enzyme for mitochondrial fatty acid uptake. Peroxisome proliferator-activated receptor α and γ and carnitine palmitoyltransferase I levels were also downregulated in these hearts. Rates of glucose and fatty acid oxidation were reduced in the hearts of heterozygous cardiomyocyte-restricted BRCA1-deficient mice, resulting in a decrease in the rate of adenosine triphosphate production. This decrease in metabolism and adenosine triphosphate production occurred despite an increase in 5'-adenosine monophosphate-activated protein kinase and AKT activation in the heart. CONCLUSIONS Cardiomyocyte-specific loss of BRCA1 alters critical pathways of fatty acid and glucose metabolism, leading to an energy starved heart. BRCA1-based cell or gene therapy might serve as a novel target to improve cardiac bioenergetics in patients with heart failure.
Collapse
Affiliation(s)
- Krishna K Singh
- Division of Cardiac Surgery and Endocrinology, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhabyeyev P, Gandhi M, Mori J, Basu R, Kassiri Z, Clanachan A, Lopaschuk GD, Oudit GY. Pressure-overload-induced heart failure induces a selective reduction in glucose oxidation at physiological afterload. Cardiovasc Res 2012; 97:676-85. [PMID: 23257023 DOI: 10.1093/cvr/cvs424] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Development of heart failure is known to be associated with changes in energy substrate metabolism. Information on the changes in energy substrate metabolism that occur in heart failure is limited and results vary depending on the methods employed. Our aim is to characterize the changes in energy substrate metabolism associated with pressure overload and ischaemia-reperfusion (I/R) injury. METHODS AND RESULTS We used transverse aortic constriction (TAC) in mice to induce pressure overload-induced heart failure. Metabolic rates were measured in isolated working hearts perfused at physiological afterload (80 mmHg) using (3)H- or (14)C-labelled substrates. As a result of pressure-overload injury, murine hearts exhibited: (i) hypertrophy, systolic, and diastolic dysfunctions; (ii) reduction in LV work, (iii) reduced rates of glucose and lactate oxidations, with no change in glycolysis or fatty acid oxidation and a small decrease in triacylglycerol oxidation, and (iv) increased phosphorylation of AMPK and a reduction in malonyl-CoA levels. Sham hearts produced more acetyl CoA from carbohydrates than from fats, whereas TAC hearts showed a reverse trend. I/R in sham group produced a metabolic switch analogous to the TAC-induced shift to fatty acid oxidation, whereas I/R in TAC hearts greatly exacerbated the existing imbalance, and was associated with a poorer recovery during reperfusion. CONCLUSIONS Pressure overload-induced heart failure and I/R shift the preference of substrate oxidation from glucose and lactate to fatty acid due to a selective reduction in carbohydrate oxidation. Normalizing the balance between metabolic substrate utilization may alleviate pressure-overload-induced heart failure and ischaemia.
Collapse
Affiliation(s)
- Pavel Zhabyeyev
- Division of Cardiology, Department of Medicine, University of Alberta, 8440 112 Street NW, Edmonton, AB, Canada T6G 2B7
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Samokhvalov V, Ussher JR, Fillmore N, Armstrong IKG, Keung W, Moroz D, Lopaschuk DG, Seubert J, Lopaschuk GD. Inhibition of malonyl-CoA decarboxylase reduces the inflammatory response associated with insulin resistance. Am J Physiol Endocrinol Metab 2012; 303:E1459-68. [PMID: 23074239 DOI: 10.1152/ajpendo.00018.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We previously showed that genetic inactivation of malonyl-CoA decarboxylase (MCD), which regulates fatty acid oxidation, protects mice against high-fat diet-induced insulin resistance. Development of insulin resistance has been associated with activation of the inflammatory response. Therefore, we hypothesized that the protective effect of MCD inhibition might be caused by a favorable effect on the inflammatory response. We examined if pharmacological inhibition of MCD protects neonatal cardiomyocytes and peritoneal macrophages against inflammatory-induced metabolic perturbations. Cardiomyocytes and macrophages were treated with LPS to induce an inflammatory response, in the presence or absence of an MCD inhibitor (CBM-301106, 10 μM). Inhibition of MCD attenuated the LPS-induced inflammatory response in cardiomyocytes and macrophages. MCD inhibition also prevented LPS impairment of insulin-stimulated glucose uptake in cardiomyocytes and increased phosphorylation of Akt. Additionally, inhibition of MCD strongly diminished LPS-induced activation of palmitate oxidation. We also found that treatment with an MCD inhibitor prevented LPS-induced collapse of total cellular antioxidant capacity. Interestingly, treatment with LPS or an MCD inhibitor did not alter intracellular triacylglycerol content. Furthermore, inhibition of MCD prevented LPS-induced increases in the level of ceramide in cardiomyocytes and macrophages while also ameliorating LPS-initiated decreases in PPAR binding. This suggests that the anti-inflammatory effect of MCD inhibition is mediated via accumulation of long-chain acyl-CoA, which in turn stimulates PPAR binding. Our results also demonstrate that pharmacological inhibition of MCD is a novel and promising approach to treat insulin resistance and its associated metabolic complications.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Biological Transport/drug effects
- Carboxy-Lyases/antagonists & inhibitors
- Carboxy-Lyases/metabolism
- Cardiotonic Agents/pharmacology
- Cells, Cultured
- Ceramides/metabolism
- Enzyme Inhibitors/pharmacology
- Glucose/metabolism
- Insulin Resistance
- Lipid Metabolism/drug effects
- Macrophage Activation/drug effects
- Macrophages, Peritoneal/cytology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/immunology
- Myocytes, Cardiac/metabolism
- Phenylurea Compounds/pharmacology
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
Collapse
Affiliation(s)
- Victor Samokhvalov
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
The pyridine nucleotides NAD(+) and NADP(+) play a pivotal role in regulating intermediary metabolism in the heart. The intracellular NAD(+)/NADH ratio controls flux through various dehydrogenase enzymes involved in both anaerobic and aerobic metabolism and also regulates posttranslational protein modification. The intracellular NADP(+)/NADPH ratio controls flux through the pentose phosphate pathway (PPP) and the polyol pathway, while also regulating ion channel function and oxidative stress. Not only does the NAD(+)/NADH ratio regulate the rates of ATP production, it can also modify energy substrate preference. For instance, in many forms of heart disease a greater contribution from fatty acids for oxidative energy metabolism increases fatty acid β-oxidation-derived NADH, which can activate pyruvate dehydrogenase (PDH) kinase isoforms that inhibit PDH and subsequent glucose oxidation. As such, novel therapies that overcome fatty acid β-oxidation-induced inhibition of PDH improve cardiac efficiency and subsequent function during ischemia/reperfusion and in heart failure. Furthermore, recent studies have implicated a pivotal role for increased PPP-derived NADPH in mediating oxidative stress observed in heart failure. In this article, we review the multiple actions of NAD(+)/NADH and NADP(+)/NADPH in regulating intermediary metabolism in the heart. A better understanding of the roles of NAD(+)/NADH and NADP(+)/NADPH in cellular physiology and pathology could potentially be used to exploit pyridine nucleotide modification in the treatment of a number of different forms of heart disease.
Collapse
Affiliation(s)
- John R Ussher
- 423 Heritage Medical Research Center, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
44
|
Guo C, Zeng X, Song J, Zhang M, Wang H, Xu X, Du F, Chen B. A soluble receptor for advanced glycation end-products inhibits hypoxia/reoxygenation-induced apoptosis in rat cardiomyocytes via the mitochondrial pathway. Int J Mol Sci 2012; 13:11923-11940. [PMID: 23109892 PMCID: PMC3472784 DOI: 10.3390/ijms130911923] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 09/05/2012] [Accepted: 09/11/2012] [Indexed: 12/14/2022] Open
Abstract
Severe myocardial dysfunction and tissue damage resulting from ischemia/reperfusion (I/R) is a common clinical scenario in patients with certain types of heart diseases and therapies such as thrombolysis, percutaneous coronary intervention, coronary artery bypass grafting, and cardiac transplantation. The underlining mechanism of endogenous cardiac protection after I/R injury has been a focus of current research. Growing evidences suggests that soluble receptor for advanced glycation end-products (sRAGE) has a cardioprotective effect; however, its role in I/R injury remains unclear. We hypothesized that exogenous administration of sRAGE during hypoxia/reoxygenation (H/R) induces cardioprotection by inhibiting cardiomyocyte apoptosis via multiple signals, involving mitochondrial membrane potential (MMP), the mitochondrial permeability transition pore (mPTP), mitochondrial cytochrome c, caspase-3, Bcl-2 and Bax. Neonatal rat cardiomyocytes underwent hypoxia for 3-h followed by 2-h reoxygenation or were treated with sRAGE for 10 min before H/R. Compared with H/R alone, sRAGE pretreatment reduced H/R-induced cardiomyocyte apoptosis from 27.9% ± 5.9% to 9.4% ± 0.7% (p < 0.05). In addition, sRAGE treatment significantly inhibited H/R-induced mitochondrial depolarization and mPTP opening, reduced mitochondrial cytochrome c leakage, caspase-3 and caspase-9 activity, and decreased the ratio of Bax to Bcl-2. Therefore, we conclude that the exogenous administration of sRAGE during H/R is involved in cardioprotection by inhibiting apoptosis via the mitochondrial pathway, which, if further confirmed in vivo, may have important clinical implications during H/R.
Collapse
Affiliation(s)
- Caixia Guo
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
- Author to whom correspondence should be addressed. E-Mail: ; Tel.: +86-10-6709-6562; Fax: +86-10-6709-6567
| | - Xiangjun Zeng
- Department of Pathophysiology, Capital Medical University, Beijing 100069, China; E-Mails: (X.Z.); (H.W.)
| | - Juanjuan Song
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| | - Min Zhang
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| | - Hongxia Wang
- Department of Pathophysiology, Capital Medical University, Beijing 100069, China; E-Mails: (X.Z.); (H.W.)
| | - Xiaowei Xu
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| | - Fenghe Du
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| | - Buxing Chen
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| |
Collapse
|
45
|
Abstract
The average human life span has markedly increased in modern society largely attributed to advances in medical and therapeutic sciences that have successfully reduced important health risks. However, advanced age results in numerous alterations to cellular and subcellular components that can impact the overall health and function of an individual. Not surprisingly, advanced age is a major risk factor for the development of heart disease in which elderly populations observe increased morbidity and mortality. Even healthy individuals that appear to have normal heart function under resting conditions, actually have an increased susceptibility and vulnerability to stress. This is confounded by the impact that stress and disease can have over time to both the heart and vessels. Although, there is a rapidly growing body of literature investigating the effects of aging on the heart and how age-related alterations affect cardiac function, the biology of aging and underlying mechanisms remain unclear. In this review, we summarize effects of aging on the heart and discuss potential theories of cellular aging with special emphasis on mitochondrial dysfunction.
Collapse
|
46
|
Wang HX, Zhang DM, Zeng XJ, Mu J, Yang H, Lu LQ, Zhang LK. Upregulation of cytochrome P450 2J3/11,12-epoxyeicosatrienoic acid inhibits apoptosis in neonatal rat cardiomyocytes by a caspase-dependent pathway. Cytokine 2012; 60:360-8. [PMID: 22717287 DOI: 10.1016/j.cyto.2012.04.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 02/10/2012] [Accepted: 04/18/2012] [Indexed: 11/19/2022]
Abstract
Short, nonlethal ischemic episodes administered to hearts directly after ischemic events (ischemic postconditioning, IPost) have an advantage over ischemic preconditioning (IPC). The endogenous cytochrome P450 2J3/11,12-epoxyeicosatrienoic acid (CYP2J3/11,12-EET) is upregulated by IPost, but not IPC, in the rat heart. The CYP epoxygenase inhibitor N-methylsulphonyl-6-(2-propargyloxyphenyl) hexanamide (MS-PPOH) reduces the cardioprotective effects of IPost, but not IPC. We proposed that upregulation of CYP2J3/11,12-EET during IPost induces cardioprotection by inhibiting cardiomyocyte apoptosis and that multiple apoptotic signals, including changes in mitochondrial membrane potential (MMP) and mitochondrial permeability transition pore (mPTP) opening, mitochondrial cytochrome c leakage, caspase-3 levels, and levels of protective kinases such as Bcl-2 and Bax, are involved in the process. Neonatal rat cardiomyocytes underwent 3-h hypoxia followed by 2-, 5-, or 6-h reoxygenation (H/R) or three cycles of 5-min reoxygenation followed by 5-min hypoxia before 90-min reoxygenation (HPost); or were transfected with pcDNA3.1-CYP2J3 for 48 h before H/R; or were treated with MS-PPOH for 10 min before HPost. For HPost alone, pcDNA3.1-CYP2J3 transfection attenuated cardiomyocyte apoptosis to 68.4% (p<0.05) of that with H/R. pcDNA3.1-CYP2J3 transfection significantly decreased MMP and inhibited mPTP opening induced by H/R, reduced mitochondrial cytochrome c leakage, cleaved caspase-3 protein expression, and increased the ratio of Bcl-2 to Bax expression. MS-PPOH abolished this effect. Therefore, upregulation of CYP2J3/11,12-EET during HPost is involved in cardioprotection by inhibiting apoptosis via a caspase-dependent pathway, and the apoptosis-suppressive effect may have important clinical implications during HPost.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/metabolism
- Amides/pharmacology
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Caspase 3/metabolism
- Cell Survival/drug effects
- Cytochrome P-450 Enzyme Inhibitors
- Cytochrome P-450 Enzyme System/metabolism
- Cytochromes c/metabolism
- Hypoxia/enzymology
- Hypoxia/pathology
- Membrane Potential, Mitochondrial/drug effects
- Mitochondrial Membrane Transport Proteins/metabolism
- Mitochondrial Permeability Transition Pore
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Oxygen/metabolism
- Rats
- Rats, Wistar
- Up-Regulation/drug effects
- bcl-2-Associated X Protein/metabolism
Collapse
Affiliation(s)
- Hong-Xia Wang
- Department of Pathophysiology, Capital Medical University, Beijing 100069, China
| | | | | | | | | | | | | |
Collapse
|
47
|
Ussher JR, Folmes CDL, Keung W, Fillmore N, Jaswal JS, Cadete VJ, Beker DL, Lam VH, Zhang L, Lopaschuk GD. Inhibition of serine palmitoyl transferase I reduces cardiac ceramide levels and increases glycolysis rates following diet-induced insulin resistance. PLoS One 2012; 7:e37703. [PMID: 22629445 PMCID: PMC3358297 DOI: 10.1371/journal.pone.0037703] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 04/23/2012] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Diet-induced obesity (DIO) leads to an accumulation of intra-myocardial lipid metabolites implicated in causing cardiac insulin resistance and contractile dysfunction. One such metabolite is ceramide, and our aim was to determine the effects of inhibiting de novo ceramide synthesis on cardiac function and insulin stimulated glucose utilization in mice subjected to DIO. MATERIALS AND METHODS C57BL/6 mice were fed a low fat diet or subjected to DIO for 12 weeks, and then treated for 4 weeks with either vehicle control or the serine palmitoyl transferase I (SPT I) inhibitor, myriocin. In vivo cardiac function was assessed via ultrasound echocardiography, while glucose metabolism was assessed in isolated working hearts. RESULTS DIO was not associated with an accumulation of intra-myocardial ceramide, but rather, an accumulation of intra-myocardial DAG (2.63±0.41 vs. 4.80±0.97 nmol/g dry weight). Nonetheless, treatment of DIO mice with myriocin decreased intra-myocardial ceramide levels (50.3±7.7 vs. 26.9±2.7 nmol/g dry weight) and prevented the DIO-associated increase in intra-myocardial DAG levels. Interestingly, although DIO impaired myocardial glycolysis rates (7789±1267 vs. 2671±326 nmol/min/g dry weight), hearts from myriocin treated DIO mice exhibited an increase in glycolysis rates. CONCLUSIONS Our data reveal that although intra-myocardial ceramide does not accumulate following DIO, inhibition of de novo ceramide synthesis nonetheless reduces intra-myocardial ceramide levels and prevents the accumulation of intra-myocardial DAG. These effects improved the DIO-associated impairment of cardiac glycolysis rates, suggesting that SPT I inhibition increases cardiac glucose utilization.
Collapse
Affiliation(s)
- John R. Ussher
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Clifford D. L. Folmes
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Wendy Keung
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Natasha Fillmore
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Jagdip S. Jaswal
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Virgilio J. Cadete
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Donna L. Beker
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Victoria H. Lam
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Liyan Zhang
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Gary D. Lopaschuk
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
- * E-mail:
| |
Collapse
|
48
|
Witham W, Yester K, O'Donnell CP, McGaffin KR. Restoration of glucose metabolism in leptin-resistant mouse hearts after acute myocardial infarction through the activation of survival kinase pathways. J Mol Cell Cardiol 2012; 53:91-100. [PMID: 22507542 DOI: 10.1016/j.yjmcc.2012.03.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 03/26/2012] [Accepted: 03/28/2012] [Indexed: 02/07/2023]
Abstract
In the normal heart, leptin modulates cardiac metabolism. It is unknown, however, what effect leptin has on cardiac metabolism and outcomes in acute myocardial infarction (MI). This study was performed to test the hypothesis that leptin signaling increases glucose metabolism and attenuates injury in the acutely infarcted heart. Mice with (ObR(+/+)) and without (ObR(-/-)) cardiomyocyte specific expression of leptin receptor (ObR) were randomly assigned to experimental MI or sham procedure, and studied 3 days later. ObR(+/+) and ObR(-/-) sham mice were not significantly different in any measured outcome. However, after MI, ObR(-/-) mice had greater cardiac dysfunction, left ventricular dilation, and levels of oxidative stress. These worse indices of cardiac injury in ObR(-/-) mice were associated with attenuated signal transducer and activator of transcription (STAT) 3, phosphatidylinositol-3-kinase (PI3K), and Akt signaling, decreased malonyl CoA content, and reduced mitochondrial pyruvate dehydrogenase and electron transport Complex I, II and IV activities. Furthermore, ObR(-/-) mice maintained high rates of cardiac fatty acid oxidation after MI, whereas ObR(+/+) mice demonstrated a switch away from fatty acid oxidation to glucose metabolism. Restoration of cardiac STAT3, PI3K and Akt activity and mitochondrial function in ObR(-/-) mice post-MI was accomplished by ciliary neurotrophic factor (CNTF), an established STAT3 activator, administered immediately after MI. Moreover, CNTF therapy resulted in mitigation of cardiac structural and functional injury, attenuated levels of oxidative stress, and rescued glucose metabolism in the infarcted ObR(-/-) heart. These data demonstrate that impaired cardiac leptin signaling results in metabolic inflexibility for glucose utilization in the face of cardiac stress, and greater morbidity after MI. Further, these studies show that cardiac glucose metabolism can be restored in leptin-resistant hearts by CNTF-mediated activation of survival kinases, resulting in multiple improved structural and functional outcomes post-MI.
Collapse
Affiliation(s)
- William Witham
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
49
|
Ussher JR, Wang W, Gandhi M, Keung W, Samokhvalov V, Oka T, Wagg CS, Jaswal JS, Harris RA, Clanachan AS, Dyck JRB, Lopaschuk GD. Stimulation of glucose oxidation protects against acute myocardial infarction and reperfusion injury. Cardiovasc Res 2012; 94:359-69. [PMID: 22436846 DOI: 10.1093/cvr/cvs129] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIMS During reperfusion of the ischaemic myocardium, fatty acid oxidation rates quickly recover, while glucose oxidation rates remain depressed. Direct stimulation of glucose oxidation via activation of pyruvate dehydrogenase (PDH), or secondary to an inhibition of malonyl CoA decarboxylase (MCD), improves cardiac functional recovery during reperfusion following ischaemia. However, the effects of such interventions on the evolution of myocardial infarction are unknown. The purpose of this study was to determine whether infarct size is decreased in response to increased glucose oxidation. METHODS AND RESULTS In vivo, direct stimulation of PDH in mice with the PDH kinase (PDHK) inhibitor, dichloroacetate, significantly decreased infarct size following temporary ligation of the left anterior descending coronary artery. These results were recapitulated in PDHK 4-deficient (PDHK4-/-) mice, which have enhanced myocardial PDH activity. These interventions also protected against ischaemia/reperfusion injury in the working heart, and dichloroacetate failed to protect in PDHK4-/- mice. In addition, there was a dramatic reduction in the infarct size in malonyl CoA decarboxylase-deficient (MCD-/-) mice, in which glucose oxidation rates are enhanced (secondary to an inhibition of fatty acid oxidation) relative to their wild-type littermates (10.8 ± 3.8 vs. 39.5 ± 4.7%). This cardioprotective effect in MCD-/- mice was associated with increased PDH activity in the ischaemic area at risk (1.89 ± 0.18 vs. 1.52 ± 0.05 μmol/g wet weight/min). CONCLUSION These findings demonstrate that stimulating glucose oxidation via targeting either PDH or MCD decreases the infarct size, validating the concept that optimizing myocardial metabolism is a novel therapy for ischaemic heart disease.
Collapse
Affiliation(s)
- John R Ussher
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tuder RM, Davis LA, Graham BB. Targeting energetic metabolism: a new frontier in the pathogenesis and treatment of pulmonary hypertension. Am J Respir Crit Care Med 2012; 185:260-6. [PMID: 22077069 PMCID: PMC3297113 DOI: 10.1164/rccm.201108-1536pp] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 10/26/2011] [Indexed: 01/01/2023] Open
Abstract
This perspective highlights advances in the understanding of the role of cellular metabolism in the pathogenesis of pulmonary hypertension. Insights gained in the past 20 years have revealed several similarities between the cellular processes underlying the pulmonary vascular remodeling in pulmonary hypertension and those seen in cancer processes. In line with these insights, there is increasing recognition that abnormal cellular metabolism, notably of aerobic glycolysis (the "Warburg effect"), the potential involvement of hypoxia-inducible factor in this process, and alterations in mitochondrial function, are key elements in the pathogenesis of this disease. The glycolytic shift may underlie the resistance to apoptosis and increased vascular cell proliferation, which are hallmarks of pulmonary hypertension. These investigations have led to novel approaches in the diagnosis and therapy of pulmonary hypertension.
Collapse
Affiliation(s)
- Rubin M Tuder
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, USA.
| | | | | |
Collapse
|