1
|
Wu H, Hui Y, Qian X, Wang X, Xu J, Wang F, Pan S, Chen K, Liu Z, Gao W, Bai J, Liang G. Exosomes derived from mesenchymal stem cells ameliorate impaired glucose metabolism in myocardial Ischemia/reperfusion injury through miR-132-3p/PTEN/AKT pathway. Cell Cycle 2025:1-20. [PMID: 40181235 DOI: 10.1080/15384101.2025.2485834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 04/05/2025] Open
Abstract
Exosomes secreted by mesenchymal stem cells (MSCs) have been considered as a novel biological therapy for myocardial ischemia/reperfusion injury (MIRI). However, the underlying mechanism of exosomes has not been completely established, especially in the early stage of MIRI. In this study, we primarily investigated the protective effect of exosomes on MIRI from both in vitro and ex vivo perspectives. Bioinformatic analysis was conducted to identify exosomal miRNA associated with myocardial protection, Genes and proteins related to functional studies and myocardial energy metabolism were analyzed and evaluated using techniques such as Polymerase Chain Re-action (PCR), Western blotting, double luciferase biochemical techniques, flow cytometry assay, etc. It was discovered that exosomes ameliorated cardiomyocyte injury t by delivery of miR-132-3p.This process reduced the expression of Phosphatase and tensin homolog (PTEN) mRNA and protein, enhanced the expression of phosphorylated protein kinase (pAKT), regulated the insulin signaling pathway, facilitated intracellular Glucose transporter 4 (GLUT4) protein membrane translocation, and enhanced glucose uptake and Adenosine Triphosphate (ATP) production. This study confirmed, for the first time, that MSC-EXO can provide myocardial protection in the early stages of MIRI through miR-132/PTEN/AKT pathway. This research establishes a theoretical and experimental foundation for the clinical application of MSC-derived exosomes.
Collapse
Affiliation(s)
- Hongkun Wu
- School of basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Cardiac Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yongpeng Hui
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xingkai Qian
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xueting Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, University Town, Guiyang, China
| | - Jianwei Xu
- National Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Center for Tissue Engineering and Stem Cell Research, Guizhou Province Key Laboratory of Regenerative Medicine, Guizhou Medical University, Guiyang, China
| | - Feng Wang
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Sisi Pan
- Department of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Kaiyuan Chen
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhou Liu
- School of basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Weilong Gao
- School of basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Center for Translational Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jue Bai
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Guiyou Liang
- Department of Cardiac Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
2
|
Kim Y, Kim D, Kim J, Yun M, Kang ES. Association between appendicular skeletal muscle mass and myocardial glucose uptake measured by 18F-FDG PET. ESC Heart Fail 2025; 12:467-476. [PMID: 39344859 PMCID: PMC11769618 DOI: 10.1002/ehf2.15086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/18/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Low muscle mass is associated with high insulin resistance and an increased risk of cardiovascular disease. This study aims to determine whether low muscle mass affects the alterations in myocardial substrate metabolism that are associated with the development of cardiovascular disease. METHOD The study included 299 individuals (182 men and 117 women) who underwent examination at the Severance Health Check-up Center between January 2018 and February 2019. Myocardial glucose uptake was assessed using [18F]-fluorodeoxyglucose-positron emission tomography (18F-FDG PET/CT) scanning. Direct segmental bioimpedance analysis was used to measure appendicular skeletal muscle mass (ASM). RESULTS We analysed men and women separately owing to sex-related body composition differences. ASM/Ht2 was significantly positively correlated with myocardial glucose uptake measured by 18F-FDG PET/CT [ln (SUVheart/liver)] only in men (r = 0.154, P = 0.038 in men; r = -0.042, P = 0.652 in women, respectively). In men, myocardial glucose uptake was significantly associated with ASM/Ht2 even after adjusting for multiple confounders in a multivariable linear regression model (standardized β = 0.397, P = 0.004, in men; β = - 0.051, P = 0.698, in women). In women, age (β = -0.424 P = 0.029) was independent determinants of myocardial glucose uptake. CONCLUSIONS In men, ASM was strongly associated with myocardial glucose uptake as measured by 18F-FDG PET/CT. In women, age was significantly correlated with myocardial substrate utilization, but not with ASM.
Collapse
Affiliation(s)
- Young‐eun Kim
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Anam HospitalKorea University College of MedicineSeoulRepublic of Korea
| | - Dongwoo Kim
- Department of Nuclear MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Jiwon Kim
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Department of Internal Medicine, National Health Insurance Service Ilsan HospitalGoyangRepublic of Korea
| | - Mijin Yun
- Department of Nuclear MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Eun Seok Kang
- Division of Endocrinology and Metabolism, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
3
|
Chong L, Dushaj N, Rakoubian A, Yarbro J, Kobayashi S, Liang Q. Unraveling the Roles of HIF-1, HO-1, GLUT-1 and GLUT-4 in Myocardial Protection. INTERNATIONAL JOURNAL OF DRUG DISCOVERY AND PHARMACOLOGY 2024; 3:100016. [PMID: 40376262 PMCID: PMC12080592 DOI: 10.53941/ijddp.2024.100016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Cardiomyocytes are highly dependent on oxygen for optimal function. Disruption of oxygen availability, as in the case of ischemic heart disease, can significantly impair heart function. Moreover, comorbidities like diabetes, hyperlipidemia, and hypertension can exacerbate ischemic cardiac injury. However, cardiomyocytes possess inherent protective mechanisms that can be activated to enhance myocardial survival under such conditions. Understanding the functions and regulatory mechanisms of these cardioprotective genes is crucial for advancing our knowledge of cardiovascular health and for developing therapeutic strategies. This review examines the intricate mechanisms of cardioprotection, with a focus on key genes and proteins, including hypoxia-inducible factor-1 (HIF-1), heme oxygenase-1 (HO-1), glucose transporter 1 (GLUT-1), and GLUT-4. In addition, the review explores the roles and regulation of these factors in the heart under ischemic stress, shedding light on their relevance in conditions like diabetes, hypertension, and hyperlipidemia/atherosclerosis. Moreover, it highlights the complex interplay among their mechanisms and suggests opportunities for developing targeted therapiesfor the treatment of ischemic heart disease, hypertension, and hyperlipidemia.
Collapse
Affiliation(s)
- Lionel Chong
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568-8000, USA
| | - Nicholas Dushaj
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568-8000, USA
| | - Ani Rakoubian
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568-8000, USA
| | - Johnathan Yarbro
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568-8000, USA
| | - Satoru Kobayashi
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568-8000, USA
| | - Qiangrong Liang
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568-8000, USA
| |
Collapse
|
4
|
Caturano A, Galiero R, Vetrano E, Sardu C, Rinaldi L, Russo V, Monda M, Marfella R, Sasso FC. Insulin-Heart Axis: Bridging Physiology to Insulin Resistance. Int J Mol Sci 2024; 25:8369. [PMID: 39125938 PMCID: PMC11313400 DOI: 10.3390/ijms25158369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Insulin signaling is vital for regulating cellular metabolism, growth, and survival pathways, particularly in tissues such as adipose, skeletal muscle, liver, and brain. Its role in the heart, however, is less well-explored. The heart, requiring significant ATP to fuel its contractile machinery, relies on insulin signaling to manage myocardial substrate supply and directly affect cardiac muscle metabolism. This review investigates the insulin-heart axis, focusing on insulin's multifaceted influence on cardiac function, from metabolic regulation to the development of physiological cardiac hypertrophy. A central theme of this review is the pathophysiology of insulin resistance and its profound implications for cardiac health. We discuss the intricate molecular mechanisms by which insulin signaling modulates glucose and fatty acid metabolism in cardiomyocytes, emphasizing its pivotal role in maintaining cardiac energy homeostasis. Insulin resistance disrupts these processes, leading to significant cardiac metabolic disturbances, autonomic dysfunction, subcellular signaling abnormalities, and activation of the renin-angiotensin-aldosterone system. These factors collectively contribute to the progression of diabetic cardiomyopathy and other cardiovascular diseases. Insulin resistance is linked to hypertrophy, fibrosis, diastolic dysfunction, and systolic heart failure, exacerbating the risk of coronary artery disease and heart failure. Understanding the insulin-heart axis is crucial for developing therapeutic strategies to mitigate the cardiovascular complications associated with insulin resistance and diabetes.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy;
| | - Vincenzo Russo
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA;
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| |
Collapse
|
5
|
Troise D, Infante B, Mercuri S, Piccoli C, Lindholm B, Stallone G. Hypoxic Inducible Factor Stabilization in Pericytes beyond Erythropoietin Production: The Good and the Bad. Antioxidants (Basel) 2024; 13:537. [PMID: 38790642 PMCID: PMC11118908 DOI: 10.3390/antiox13050537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The paracrine signaling pathways for the crosstalk between pericytes and endothelial cells are essential for the coordination of cell responses to challenges such as hypoxia in both healthy individuals and pathological conditions. Ischemia-reperfusion injury (IRI), one of the causes of cellular dysfunction and death, is associated with increased expression of genes involved in cellular adaptation to a hypoxic environment. Hypoxic inducible factors (HIFs) have a central role in the response to processes initiated by IRI not only linked to erythropoietin production but also because of their participation in inflammation, angiogenesis, metabolic adaptation, and fibrosis. While pericytes have an essential physiological function in erythropoietin production, a lesser-known role of HIF stabilization during IRI is that pericytes' HIF expression could influence vascular remodeling, cell loss and organ fibrosis. Better knowledge of mechanisms that control functions and consequences of HIF stabilization in pericytes beyond erythropoietin production is advisable for the development of therapeutic strategies to influence disease progression and improve treatments. Thus, in this review, we discuss the dual roles-for good or bad-of HIF stabilization during IRI, focusing on pericytes, and consequences in particular for the kidneys.
Collapse
Affiliation(s)
- Dario Troise
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Silvia Mercuri
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Bengt Lindholm
- Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
6
|
Silva-Velasco DL, Cervantes-Pérez LG, Sánchez-Mendoza A. ACE inhibitors and their interaction with systems and molecules involved in metabolism. Heliyon 2024; 10:e24655. [PMID: 38298628 PMCID: PMC10828069 DOI: 10.1016/j.heliyon.2024.e24655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
The main function of the renin-angiotensin-aldosterone system (RAAS) is the regulation of blood pressure; therefore, researchers have focused on its study to treat cardiovascular and renal diseases. One of the most widely used treatments derived from the study of RAAS, is the use of angiotensin-converting enzyme inhibitors (ACEi). Since it was discovered, the main target of ACEi has been the cardiovascular and renal systems. However, being the RAAS expressed locally in several specialized tissues and cells such as pneumocytes, hepatocytes, spleenocytes, enterocytes, adipocytes, and neurons the effect of inhibitors has expanded, because it is expected that RAAS has a role in the specific function of those cells. Many chronic degenerative diseases compromise the correct function of those organs, and in most of them, the RAAS is overactivated. Therefore, the use of ACEi must exert a benefit on an impaired system. Accordingly, the objective of this review is to present a brief overview of the cardiovascular and renal actions of ACEi and its effects in organs that are not the classic targets of ACEi that carry on glucose and lipid metabolism.
Collapse
Affiliation(s)
| | - Luz G. Cervantes-Pérez
- Departamento de Farmacología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Alicia Sánchez-Mendoza
- Departamento de Farmacología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
7
|
Shi P, Wu J, Li M, Cao Y, Wu J, Ren P, Liu K, Zhou J, Sha Y, Zhang Q, Sun H. Upregulation of Hsp27 via further inhibition of histone H2A ubiquitination confers protection against myocardial ischemia/reperfusion injury by promoting glycolysis and enhancing mitochondrial function. Cell Death Discov 2023; 9:466. [PMID: 38114486 PMCID: PMC10730859 DOI: 10.1038/s41420-023-01762-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023] Open
Abstract
Research suggests that ischemic glycolysis improves myocardial tolerance to anoxia and low-flow ischemia. The rate of glycolysis during ischemia reflects the severity of the injury caused by ischemia and subsequent functional recovery following reperfusion. Histone H2AK119 ubiquitination (H2Aub) is a common modification that is primarily associated with gene silencing. Recent studies have demonstrated that H2Aub contributes to the development of cardiovascular diseases. However, the underlying mechanism remains unclear. This study identified Hsp27 (heat shock protein 27) as a H2Aub binding protein and explored its involvement in mediating glycolysis and mitochondrial function. Functional studies revealed that inhibition of PRC1 (polycomb repressive complex 1) decreased H2Aub occupancy and promoted Hsp27 expression through inhibiting ubiquitination. Additionally, it increased glycolysis by activating the NF-κB/PFKFB3 signaling pathway during myocardial ischemia. Furthermore, Hsp27 reduced mitochondrial ROS production by chaperoning COQ9, and suppressed ferroptosis during reperfusion. A delivery system was developed based on PCL-PEG-MAL (PPM)-PCM-SH (CWLSEAGPVVTVRALRGTGSW) to deliver PRT4165 (PRT), a potent inhibitor of PRC1, to damaged myocardium, resulting in decreased H2Aub. These findings revealed a novel epigenetic mechanism connecting glycolysis and ferroptosis in protecting the myocardium against ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Jiawei Wu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Minghui Li
- Department of Pharmaceutics, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Jiabi Wu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Ping Ren
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Kai Liu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Jiajun Zhou
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Yuetong Sha
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Qianhui Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang, 163319, China.
| |
Collapse
|
8
|
Wang Y, Liu Z, Bian X, Zhao C, Zhang X, Liu X, Wang N. Function and regulation of ubiquitin-like SUMO system in heart. Front Cell Dev Biol 2023; 11:1294717. [PMID: 38033852 PMCID: PMC10687153 DOI: 10.3389/fcell.2023.1294717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
The small ubiquitin-related modifier (SUMOylation) system is a conserved, reversible, post-translational protein modification pathway covalently attached to the lysine residues of proteins in eukaryotic cells, and SUMOylation is catalyzed by SUMO-specific activating enzyme (E1), binding enzyme (E2) and ligase (E3). Sentrin-specific proteases (SENPs) can cleave the isopeptide bond of a SUMO conjugate and catalyze the deSUMOylation reaction. SUMOylation can regulate the activity of proteins in many important cellular processes, including transcriptional regulation, cell cycle progression, signal transduction, DNA damage repair and protein stability. Biological experiments in vivo and in vitro have confirmed the key role of the SUMO conjugation/deconjugation system in energy metabolism, Ca2+ cycle homeostasis and protein quality control in cardiomyocytes. In this review, we summarized the research progress of the SUMO conjugation/deconjugation system and SUMOylation-mediated cardiac actions based on related studies published in recent years, and highlighted the further research areas to clarify the role of the SUMO system in the heart by using emerging technologies.
Collapse
Affiliation(s)
- Ying Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Zhihao Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiyun Bian
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Chenxu Zhao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Xin Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Xiaozhi Liu
- Central Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Nan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
9
|
Vanni E, Lindner K, Gavin AC, Montessuit C. Differential intracellular management of fatty acids impacts on metabolic stress-stimulated glucose uptake in cardiomyocytes. Sci Rep 2023; 13:14805. [PMID: 37684349 PMCID: PMC10491837 DOI: 10.1038/s41598-023-42072-7] [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: 03/03/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Stimulation of glucose uptake in response to ischemic metabolic stress is important for cardiomyocyte function and survival. Chronic exposure of cardiomyocytes to fatty acids (FA) impairs the stimulation of glucose uptake, whereas induction of lipid droplets (LD) is associated with preserved glucose uptake. However, the mechanisms by which LD induction prevents glucose uptake impairment remain elusive. We induced LD with either tetradecanoyl phorbol acetate (TPA) or 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR). Triacylglycerol biosynthesis enzymes were inhibited in cardiomyocytes exposed to FA ± LD inducers, either upstream (glycerol-3-phosphate acyltransferases; GPAT) or downstream (diacylglycerol acyltransferases; DGAT) of the diacylglycerol step. Although both inhibitions reduced LD formation in cardiomyocytes treated with FA and LD inducers, only DGAT inhibition impaired metabolic stress-stimulated glucose uptake. DGAT inhibition in FA plus TPA-treated cardiomyocytes reduced triacylglycerol but not diacylglycerol content, thus increasing the diacylglycerol/triacylglycerol ratio. In cardiomyocytes exposed to FA alone, GPAT inhibition reduced diacylglycerol but not triacylglycerol, thus decreasing the diacylglycerol/triacylglycerol ratio, prevented PKCδ activation and improved metabolic stress-stimulated glucose uptake. Changes in AMP-activated Protein Kinase activity failed to explain variations in metabolic stress-stimulated glucose uptake. Thus, LD formation regulates metabolic stress-stimulated glucose uptake in a manner best reflected by the diacylglycerol/triacylglycerol ratio.
Collapse
Affiliation(s)
- Ettore Vanni
- Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | - Karina Lindner
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, Geneva, Switzerland
| | - Anne-Claude Gavin
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, Geneva, Switzerland
| | - Christophe Montessuit
- Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland.
| |
Collapse
|
10
|
Balatskyi VV, Sowka A, Dobrzyn P, Piven OO. WNT/β-catenin pathway is a key regulator of cardiac function and energetic metabolism. Acta Physiol (Oxf) 2023; 237:e13912. [PMID: 36599355 DOI: 10.1111/apha.13912] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/24/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
The WNT/β-catenin pathway is a master regulator of cardiac development and growth, and its activity is low in healthy adult hearts. However, even this low activity is essential for maintaining normal heart function. Acute activation of the WNT/β-catenin signaling cascade is considered to be cardioprotective after infarction through the upregulation of prosurvival genes and reprogramming of metabolism. Chronically high WNT/β-catenin pathway activity causes profibrotic and hypertrophic effects in the adult heart. New data suggest more complex functions of β-catenin in metabolic maturation of the perinatal heart, establishing an adult pattern of glucose and fatty acid utilization. Additionally, low basal activity of the WNT/β-catenin cascade maintains oxidative metabolism in the adult heart, and this pathway is reactivated by physiological or pathological stimuli to meet the higher energy needs of the heart. This review summarizes the current state of knowledge of the organization of canonical WNT signaling and its function in cardiogenesis, heart maturation, adult heart function, and remodeling. We also discuss the role of the WNT/β-catenin pathway in cardiac glucose, lipid metabolism, and mitochondrial physiology.
Collapse
Affiliation(s)
- Volodymyr V Balatskyi
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Adrian Sowka
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Oksana O Piven
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Department of Human Genetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
11
|
Al-Kouh A, Babiker F, Al-Bader M. Renin-Angiotensin System Antagonism Protects the Diabetic Heart from Ischemia/Reperfusion Injury in Variable Hyperglycemia Duration Settings by a Glucose Transporter Type 4-Mediated Pathway. Pharmaceuticals (Basel) 2023; 16:238. [PMID: 37259385 PMCID: PMC9967344 DOI: 10.3390/ph16020238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/15/2023] [Accepted: 02/01/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a risk factor for cardiovascular diseases, specifically, the ischemic heart diseases (IHD). The renin-angiotensin system (RAS) affects the heart directly and indirectly. However, its role in the protection of the heart against I/R injury is not completely understood. The aim of the current study was to evaluate the efficacy of the angiotensin-converting enzyme (ACE) inhibitor and Angiotensin II receptor (AT1R) blocker or a combination thereof in protection of the heart from I/R injury. METHODS Hearts isolated from adult male Wistar rats (n = 8) were subjected to high glucose levels; acute hyperglycemia or streptozotocin (STZ)-induced diabetes were used in this study. Hearts were subjected to I/R injury, treated with Captopril, an ACE inhibitor; Losartan, an AT1R antagonist; or a combination thereof. Hemodynamics data were measured using a suitable software for that purpose. Additionally, infarct size was evaluated using 2,3,5-Triphenyltetrazolium chloride (TTC) staining. The levels of apoptosis markers (caspase-3 and -8), antioxidant enzymes, superoxide dismutase (SOD) and catalase (CAT), nitric oxide synthase (eNOS), and glucose transporter type 4 (GLUT-4) protein levels were evaluated by Western blotting. Pro-inflammatory and anti-inflammatory cytokines levels were evaluated by enzyme-linked immunosorbent assay (ELISA). RESULTS Captopril and Losartan alone or in combination abolished the effect of I/R injury in hearts subjected to acute hyperglycemia or STZ-induced diabetes. There was a significant (p < 0.05) recovery in hemodynamics, infarct size, and apoptosis markers following the treatment with Captopril, Losartan, or their combination. Treatment with Captopril, Losartan, or their combination significantly (p < 0.05) reduced pro-inflammatory cytokines and increased GLUT-4 protein levels. CONCLUSIONS The blockade of the RAS system protected the diabetic heart from I/R injury. This protection followed a pathway that utilizes GLUT-4 to decrease the apoptosis markers, pro-inflammatory cytokines, and to increase the anti-inflammatory cytokines. This protection seems to employ a pathway which is not involving ERK1/2 and eNOS.
Collapse
Affiliation(s)
| | - Fawzi Babiker
- Department of Physiology, Faculty of Medicine, Kuwait University, P.O. Box 24923, Kuwait City 13110, Kuwait
| | | |
Collapse
|
12
|
Binsch C, Barbosa DM, Hansen-Dille G, Hubert M, Hodge SM, Kolasa M, Jeruschke K, Weiß J, Springer C, Gorressen S, Fischer JW, Lienhard M, Herwig R, Börno S, Timmermann B, Cremer AL, Backes H, Chadt A, Al-Hasani H. Deletion of Tbc1d4/As160 abrogates cardiac glucose uptake and increases myocardial damage after ischemia/reperfusion. Cardiovasc Diabetol 2023; 22:17. [PMID: 36707786 PMCID: PMC9881301 DOI: 10.1186/s12933-023-01746-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Type 2 Diabetes mellitus (T2DM) is a major risk factor for cardiovascular disease and associated with poor outcome after myocardial infarction (MI). In T2DM, cardiac metabolic flexibility, i.e. the switch between carbohydrates and lipids as energy source, is disturbed. The RabGTPase-activating protein TBC1D4 represents a crucial regulator of insulin-stimulated glucose uptake in skeletal muscle by controlling glucose transporter GLUT4 translocation. A human loss-of-function mutation in TBC1D4 is associated with impaired glycemic control and elevated T2DM risk. The study's aim was to investigate TBC1D4 function in cardiac substrate metabolism and adaptation to MI. METHODS Cardiac glucose metabolism of male Tbc1d4-deficient (D4KO) and wild type (WT) mice was characterized using in vivo [18F]-FDG PET imaging after glucose injection and ex vivo basal/insulin-stimulated [3H]-2-deoxyglucose uptake in left ventricular (LV) papillary muscle. Mice were subjected to cardiac ischemia/reperfusion (I/R). Heart structure and function were analyzed until 3 weeks post-MI using echocardiography, morphometric and ultrastructural analysis of heart sections, complemented by whole heart transcriptome and protein measurements. RESULTS Tbc1d4-knockout abolished insulin-stimulated glucose uptake in ex vivo LV papillary muscle and in vivo cardiac glucose uptake after glucose injection, accompanied by a marked reduction of GLUT4. Basal cardiac glucose uptake and GLUT1 abundance were not changed compared to WT controls. D4KO mice showed mild impairments in glycemia but normal cardiac function. However, after I/R D4KO mice showed progressively increased LV endsystolic volume and substantially increased infarction area compared to WT controls. Cardiac transcriptome analysis revealed upregulation of the unfolded protein response via ATF4/eIF2α in D4KO mice at baseline. Transmission electron microscopy revealed largely increased extracellular matrix (ECM) area, in line with decreased cardiac expression of matrix metalloproteinases of D4KO mice. CONCLUSIONS TBC1D4 is essential for insulin-stimulated cardiac glucose uptake and metabolic flexibility. Tbc1d4-deficiency results in elevated cardiac endoplasmic reticulum (ER)-stress response, increased deposition of ECM and aggravated cardiac damage following MI. Hence, impaired TBC1D4 signaling contributes to poor outcome after MI.
Collapse
Affiliation(s)
- C. Binsch
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - D. M. Barbosa
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - G. Hansen-Dille
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - M. Hubert
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - S. M. Hodge
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - M. Kolasa
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - K. Jeruschke
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - J. Weiß
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - C. Springer
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - S. Gorressen
- grid.411327.20000 0001 2176 9917Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - J. W. Fischer
- grid.411327.20000 0001 2176 9917Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - M. Lienhard
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - R. Herwig
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - S. Börno
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - B. Timmermann
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - A. L. Cremer
- grid.418034.a0000 0004 4911 0702Max Planck Institute for Metabolism Research, Cologne, Germany
| | - H. Backes
- grid.418034.a0000 0004 4911 0702Max Planck Institute for Metabolism Research, Cologne, Germany
| | - A. Chadt
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany ,grid.452622.5German Center for Diabetes Research, Partner Düsseldorf, Munich-Neuherberg, Germany
| | - H. Al-Hasani
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany ,grid.452622.5German Center for Diabetes Research, Partner Düsseldorf, Munich-Neuherberg, Germany
| |
Collapse
|
13
|
She Y, Ge R, Gu X, Fang P, Zhang Z. Cardioprotective effects of neuropeptide galanin: Focusing on its roles against diabetic heart. Peptides 2023; 159:170918. [PMID: 36435275 DOI: 10.1016/j.peptides.2022.170918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/09/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022]
Abstract
Following an unprecedented rise in the number of the aged, the incidence of age-related diseases, such as diabetes and cardiovascular disease, is consequently increasing in the world. Type 2 diabetes mellitus (T2DM) is associated with excess cardiovascular morbidity and mortality. The diabetic heart is characterized by increased cardiomyocyte stiffness and fibrotic changes. Despite many factors resulting in cardiomyocyte injury and dysfunction in diabetes, insulin resistance is still a critical etiology of diabetic cardiomyopathy. Preclinical and clinical studies have revealed an intriguing role for galanin in the pathogenesis of insulin resistance and diabetic heart disease. A significant change in plasma galanin levels occurred in patients suffering from type 2 diabetes or cardiomyocyte injury. In turn, galanin may also distinctly mitigate hyperglycemia and insulin resistance in diabetes as well as increase glucose metabolism and mitochondrial biogenesis in cardiac muscle. Here, we critically review current data about the multivariate relationship among galanin, insulin resistance, and cardiac muscle to comprehensively evaluate the protective role of galanin and its receptors for the diabetic heart and to determine whether galanin receptor 2 agonists potentially represent a feasible way to treat diabetic cardiomyopathy in the future.
Collapse
Affiliation(s)
- Yuqing She
- Department of Endocrinology, Pukou Branch of Jiangsu People's Hospital, Nanjing 211899, China
| | - Ran Ge
- Key Laboratory for Metabolic Diseases in Chinese Medicine & Hanlin College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xuewen Gu
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine & Hanlin College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
14
|
Nakayama Y, Mukai N, Kreitzer G, Patwari P, Yoshioka J. Interaction of ARRDC4 With GLUT1 Mediates Metabolic Stress in the Ischemic Heart. Circ Res 2022; 131:510-527. [PMID: 35950500 PMCID: PMC9444972 DOI: 10.1161/circresaha.122.321351] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/01/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND An ancient family of arrestin-fold proteins, termed alpha-arrestins, may have conserved roles in regulating nutrient transporter trafficking and cellular metabolism as adaptor proteins. One alpha-arrestin, TXNIP (thioredoxin-interacting protein), is known to regulate myocardial glucose uptake. However, the in vivo role of the related alpha-arrestin, ARRDC4 (arrestin domain-containing protein 4), is unknown. METHODS We first tested whether interaction with GLUTs (glucose transporters) is a conserved function of the mammalian alpha-arrestins. To define the in vivo function of ARRDC4, we generated and characterized a novel Arrdc4 knockout (KO) mouse model. We then analyzed the molecular interaction between arrestin domains and the basal GLUT1. RESULTS ARRDC4 binds to GLUT1, induces its endocytosis, and blocks cellular glucose uptake in cardiomyocytes. Despite the closely shared protein structure, ARRDC4 and its homologue TXNIP operate by distinct molecular pathways. Unlike TXNIP, ARRDC4 does not increase oxidative stress. Instead, ARRDC4 uniquely mediates cardiomyocyte death through its effects on glucose deprivation and endoplasmic reticulum stress. At baseline, Arrdc4-KO mice have mild fasting hypoglycemia. Arrdc4-KO hearts exhibit a robust increase in myocardial glucose uptake and glycogen storage. Accordingly, deletion of Arrdc4 improves energy homeostasis during ischemia and protects cardiomyocytes against myocardial infarction. Furthermore, structure-function analyses of the interaction of ARRDC4 with GLUT1 using both scanning mutagenesis and deep-learning Artificial Intelligence identify specific residues in the C-terminal arrestin-fold domain as the interaction interface that regulates GLUT1 function, revealing a new molecular target for potential therapeutic intervention against myocardial ischemia. CONCLUSIONS These results uncover a new mechanism of ischemic injury in which ARRDC4 drives glucose deprivation-induced endoplasmic reticulum stress leading to cardiomyocyte death. Our findings establish ARRDC4 as a new scaffold protein for GLUT1 that regulates cardiac metabolism in response to ischemia and provide insight into the therapeutic strategy for ischemic heart disease.
Collapse
Affiliation(s)
- Yoshinobu Nakayama
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, New York, New York
| | - Nobuhiro Mukai
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, New York, New York
| | - Geri Kreitzer
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, New York, New York
| | - Parth Patwari
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jun Yoshioka
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, New York, New York
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Xu B, Li F, Zhang W, Su Y, Tang L, Li P, Joshi J, Yang A, Li D, Wang Z, Wang S, Xie J, Gu H, Zhu W. Identification of metabolic pathways underlying FGF1 and CHIR99021-mediated cardioprotection. iScience 2022; 25:104447. [PMID: 35707727 PMCID: PMC9189130 DOI: 10.1016/j.isci.2022.104447] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/16/2022] [Accepted: 05/18/2022] [Indexed: 12/05/2022] Open
Abstract
Acute myocardial infarction is a leading cause of death worldwide. We have previously identified two cardioprotective molecules — FGF1 and CHIR99021— that confer cardioprotection in mouse and pig models of acute myocardial infarction. Here, we aimed to determine if improved myocardial metabolism contributes to this cardioprotection. Nanofibers loaded with FGF1 and CHIR99021 were intramyocardially injected to ischemic myocardium of adult mice immediately following surgically induced myocardial infarction. Animals were euthanized 3 and 7 days later. Our data suggested that FGF1/CHIR99021 nanofibers enhanced the heart’s capacity to utilize glycolysis as an energy source and reduced the accumulation of branched-chain amino acids in ischemic myocardium. The impact of FGF1/CHIR99021 on metabolism was more obvious in the first three days post myocardial infarction. Taken together, these findings suggest that FGF1/CHIR99021 protects the heart against ischemic injury via improving myocardial metabolism which may be exploited for treatment of acute myocardial infarction in humans. FGF1/CHIR confer cardioprotection in myocardial infarction animals FGF1/CHIR enhance the capability of ischemic hearts to produce energy via glycolysis FGF1/CHIR reduce the abundance of branched chain amino acids in ischemic hearts This study reveals a novel approach to correct metabolic disorders in ischemic hearts
Collapse
Affiliation(s)
- Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259.,Department of Cardiology, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Fan Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259.,Department of Kinesiology, South China Normal University, Guangzhou 510631, China
| | - Wenjing Zhang
- Center for Translational Science, Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL 34987, USA.,College of Health Solutions, Arizona State University, Phoenix, AZ 85287, USA
| | - Yajuan Su
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Jyotsna Joshi
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Dong Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Zhao Wang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Shu Wang
- College of Health Solutions, Arizona State University, Phoenix, AZ 85287, USA
| | - Jingwei Xie
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Haiwei Gu
- Center for Translational Science, Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL 34987, USA.,College of Health Solutions, Arizona State University, Phoenix, AZ 85287, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| |
Collapse
|
16
|
Exogenous Galanin Reduces Hyperglycemia and Myocardial Metabolic Disorders Induced by Streptozotocin in Rats. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10412-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
17
|
Studneva IM, Veselova OM, Dobrokhotov IV, Serebryakova LI, Palkeeva ME, Molokoedov AS, Azmuko AA, Ovchinnikov MV, Sidorova MV, Pisarenko OI. Chimeric Agonist of Galanin Receptor GALR2 Reduces Heart Damage in Rats with Streptozotocin-Induced Diabetes. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:346-355. [PMID: 35527373 DOI: 10.1134/s0006297922040046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 06/14/2023]
Abstract
Neuropeptide galanin and its N-terminal fragments reduce the generation of reactive oxygen species and normalize metabolic and antioxidant states of myocardium in experimental cardiomyopathy and ischemia/reperfusion injury. The aim of this study was to elucidate the effect of WTLNSAGYLLGPβAH-OH (peptide G), a pharmacological agonist of the galanin receptor GalR2, on the cardiac injury induced by administration of streptozotocin (STZ) in rats. Peptide G was prepared by solid phase peptide synthesis using the Fmoc strategy and purified by preparative HPLC; its structure was confirmed by 1H-NMR spectroscopy and MALDI-TOF mass spectrometry. Experimental animals were randomly distributed into five groups: C, control; S, STZ-treated; SG10, STZ + peptide G (10 nmol/kg/day); SG50, STZ + peptide G (50 nmol/kg/day); G, peptide G (50 nmol/kg/day). Administration of peptide G prevented hyperglycemia in SG50 rats. By the end of the experiment, the ATP content, total pool of adenine nucleotides, phosphocreatine (PCr) content, and PCr/ATP ratio in the myocardium of animals of the SG50 group were significantly higher than in rats of the S group. In the SG50 and SG10 groups, the content of lactate and lactate/pyruvate ratio in the myocardium were reduced, while the glucose content was increased vs. the S group. Both doses of peptide G reduced the activation of creatine kinase-MB and lactate dehydrogenase, as well as the concentration of thiobarbituric acid reactive products in the blood plasma of STZ-treated rats to the control values. Taken together, these results suggest that peptide G has cardioprotective properties in type 1 diabetes mellitus. Possible mechanisms of peptide G action in the STZ-induced diabetes are discussed.
Collapse
Affiliation(s)
- Irina M Studneva
- National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Oksana M Veselova
- National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | | | | | - Marina E Palkeeva
- National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | | | - Andrey A Azmuko
- National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | | | - Maria V Sidorova
- National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Oleg I Pisarenko
- National Medical Research Center for Cardiology, Moscow, 121552, Russia.
| |
Collapse
|
18
|
Exploring the Pattern of Metabolic Alterations Causing Energy Imbalance via PPARα Dysregulation in Cardiac Muscle During Doxorubicin Treatment. Cardiovasc Toxicol 2022; 22:436-461. [PMID: 35157213 DOI: 10.1007/s12012-022-09725-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 02/02/2022] [Indexed: 12/14/2022]
Abstract
Cardiotoxicity by anthracycline antineoplastic drug doxorubicin is one of the systemic toxicity of the cardiovascular system. The mechanism responsible for doxorubicin cardiotoxicity and lipid metabolism remains elusive. The current study tested the hypotheses that the role of peroxisome proliferator-activated receptor α (PPARα) in the progress of doxorubicin-induced cardiomyopathy and its mechanism behind lipid metabolism. In the present study, male rats were subjected to intraperitoneal injection (5-week period) of doxorubicin with different dosages such as low dosage (1.5 mg/kg body weight) and high dosage (15 mg/kg body weight) to induce doxorubicin cardiomyopathy. Myocardial PPARα was impaired in both low dosage and high dosage of doxorubicin-treated rats in a dose-dependent manner. The attenuated level of PPARα impairs the expression of the genes involved in mitochondrial transporter, fatty acid transportation, lipolysis, lipid metabolism, and fatty acid oxidation. Moreover, it disturbs the reverse triacylglycerol transporter apolipoprotein B-100 (APOB) in the myocardium. Doxorubicin elevates the circulatory lipid profile and glucose. Further aggravated lipid profile in circulation impedes the metabolism of lipid in cardiac tissue, which causes a lipotoxic condition in the heart and subsequently associated disease for the period of doxorubicin treatment. Elevated lipids in the circulation translocate into the heart dysregulates lipid metabolism in the heart, which causes augmented oxidative stress and necro-apoptosis and mediates lipotoxic conditions. This finding determines the mechanistic role of doxorubicin-disturbed lipid metabolism via PPARα, which leads to cardiac dysfunction.
Collapse
|
19
|
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: 32] [Impact Index Per Article: 8.0] [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
|
20
|
Sodium Glucose Cotransporter 1 (SGLT1) Inhibitors in Cardiovascular Protection: Mechanism Progresses and Challenges. Pharmacol Res 2021; 176:106049. [PMID: 34971725 DOI: 10.1016/j.phrs.2021.106049] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/15/2021] [Accepted: 12/26/2021] [Indexed: 12/20/2022]
Abstract
In recent years, multiple clinical trials have shown that sodium glucose cotransporter 1 (SGLT1) inhibitors have significant beneficial cardiovascular effects. This includes reducing the incidence of cardiovascular deaths and heart failure hospitalizations in people with and without diabetes, as well as those with and without generalized heart failure. The exact mechanism responsible for these beneficial effects is not completely understood. To explain the cardiovascular protective effects of SGLT1 inhibitors, several potential arguments have been proposed, including decreasing oxidative stress, regulating cardiac glucose uptake, preventing ischemia/reperfusion injury, alleviating the activation of cardiac fibroblasts, attenuating apoptosis, reducing intermittent high glucose-induced pyroptosis, ameliorating cardiac hypertrophy, attenuating arrhythmic vulnerabilities, and improving left ventricular systolic disorder. This article reviews the advantages and disadvantages of these mechanisms, and attempts to synthesize and prioritize mechanisms related to the reduction of clinical events.
Collapse
|
21
|
Pisarenko OI, Studneva IM, Veselova OM. Modified N-Terminal Fragments of Galanin: Cardioprotective Properties and Mechanisms of Action. BIOCHEMISTRY. BIOKHIMIIA 2021; 86:1342-1351. [PMID: 34903156 DOI: 10.1134/s000629792110014x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The design of new drugs for treatment of cardiovascular diseases based on endogenous peptide hormones is of undoubted interest and stimulates intensive experimental research. One of the approaches for development in this area is synthesis of the short bioactive peptides that mimic effects of the larger peptide molecules and have improved physicochemical characteristics. In recent years, it has been found that the N-terminal fragments of the neuropeptide galanin reduce metabolic and functional disorders in the experimental heart damage. The review presents literature data and generalized results of our own experiments on the effects of the full-size galanin and its chemically modified N-terminal fragments (2-11) and (2-15) on the heart in normal conditions and in modeling pathophysiological conditions in vitro and in vivo. It has been shown that the spectrum of the peptide actions on the damaged myocardium includes decrease in the necrotic death of cardiomyocytes, decrease in the damage of sarcolemma, improvement in the metabolic state of myocardium, decrease in the formation of reactive oxygen species (ROS) and lipid peroxidation (LPO) products. Mechanisms of the protective action of the modified galanin fragments associated with activation of the GalR2 receptor subtype and manifestation of antioxidant properties are discussed. The data summarized in the review indicate that the molecular design of pharmacological agonists of the GalR2 receptor is a promising approach, because they can serve as a basis for the development of cardioprotectors influencing processes of free radical oxidation and metabolic adaptation.
Collapse
Affiliation(s)
- Oleg I Pisarenko
- National Medical Research Center for Cardiology, Moscow, 121552, Russia.
| | - Irina M Studneva
- National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Oxana M Veselova
- National Medical Research Center for Cardiology, Moscow, 121552, Russia
| |
Collapse
|
22
|
Morgunova GV, Shilovsky GA, Khokhlov AN. Effect of Caloric Restriction on Aging: Fixing the Problems of Nutrient Sensing in Postmitotic Cells? BIOCHEMISTRY. BIOKHIMIIA 2021; 86:1352-1367. [PMID: 34903158 DOI: 10.1134/s0006297921100151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The review discusses the role of metabolic disorders (in particular, insulin resistance) in the development of age-related diseases and normal aging with special emphasis on the changes in postmitotic cells of higher organisms. Caloric restriction helps to prevent such metabolic disorders, which could probably explain its ability to prolong the lifespan of laboratory animals. Maintaining metabolic homeostasis is especially important for the highly differentiated long-lived body cells, whose lifespan is comparable to the lifespan of the organism itself. Normal functioning of these cells can be ensured only upon correct functioning of the cytoplasm clean-up system and availability of all required nutrients and energy sources. One of the central problems in gerontology is the age-related disruption of glucose metabolism leading to obesity, diabetes, metabolic syndrome, and other related pathologies. Along with the adipose tissue, skeletal muscles are the main consumers of insulin; hence the physical activity of muscles, which supports their energy metabolism, delays the onset of insulin resistance. Insulin resistance disrupts the metabolism of cardiomyocytes, so that they fail to utilize the nutrients to perform their functions even being surrounded by a nutrient-rich environment, which contributes to the development of age-related cardiovascular diseases. Metabolic pathologies also alter the nutrient sensitivity of neurons, thus disrupting the action of insulin in the central nervous system. In addition, there is evidence that neurons can develop insulin resistance as well. It has been suggested that affecting nutritional sensors (e.g., AMPK) in postmitotic cells might improve the state of the entire multicellular organism, slow down its aging, and increase the lifespan.
Collapse
Affiliation(s)
- Galina V Morgunova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Gregory A Shilovsky
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | | |
Collapse
|
23
|
Pisarenko OI, Studneva IM, Serebryakova LI, Timoshin AA, Konovalova GG, Lankin VZ, Tihaze AK, Veselova OM, Dobrokhotov IV, Lyubimov RO, Sidorova MV, Palkeeva ME, Molokoedov AS. Antioxidant Properties of Galanin and Its N-Terminal Fragments in in vitro and in vivo Oxidative Stress Modeling. BIOCHEMISTRY (MOSCOW) 2021; 86:496-505. [PMID: 33941070 DOI: 10.1134/s0006297921040106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Antioxidant properties of rat galanin GWTLNSAGYLLGPHAIDNHRSFSDKHGLT-NH2 (Gal), N-terminal fragment of galanin (2-15 aa) WTLNSAGYLLGPHA (G1), and its modified analogue WTLNSAGYLLGPβAH (G2) were studied in vivo in the rat model of regional myocardial ischemia and reperfusion and in vitro in the process of Cu2+-induced free radical oxidation of human blood plasma low-density lipoproteins. Intravenous administration of G1, G2, and Gal to rats after ischemia induction reduced the infarction size and activities of the necrosis markers, creatine kinase-MB and lactate dehydrogenase, in blood plasma at the end of reperfusion. G1, G2, and Gal reduced formation of the spin adducts of hydroxyl radicals in the interstitium of the area at risk during reperfusion, moreover, G2 and Gal also reduced formation of the secondary products of lipid peroxidation in the reperfused myocardium. It was shown in the in vivo experiments and in the in vitro model system that the ability of galanin peptides to reduce formation of ROS and attenuate lipid peroxidation during myocardial reperfusion injury was not associated directly with their effects on activities of the antioxidant enzymes of the heart: Cu,Zn-superoxide dismutase, catalase, and glutathione peroxidase. The peptides G1, G2, and Gal at concentrations of 0.01 and 0.1 mM inhibited Cu2+-induced free radical oxidation of human low-density lipoproteins in vitro. The results of oxidative stress modeling demonstrated that the natural and synthetic agonists of galanin receptors reduced formation of the short-lived ROS in the reperfused myocardium, as well as of lipid radicals in blood plasma. Thus, galanin receptors could be a promising therapeutic target for cardiovascular diseases.
Collapse
Affiliation(s)
- Oleg I Pisarenko
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia.
| | - Irina M Studneva
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Larisa I Serebryakova
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Alexandr A Timoshin
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Galina G Konovalova
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Vadim Z Lankin
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Alla K Tihaze
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Oksana M Veselova
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Igor V Dobrokhotov
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Roman O Lyubimov
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Mariya V Sidorova
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Marina E Palkeeva
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Alexandr S Molokoedov
- National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| |
Collapse
|
24
|
Abstract
Insulin receptors are highly expressed in the heart and vasculature. Insulin signaling regulates cardiac growth, survival, substrate uptake, utilization, and mitochondrial metabolism. Insulin signaling modulates the cardiac responses to physiological and pathological stressors. Altered insulin signaling in the heart may contribute to the pathophysiology of ventricular remodeling and heart failure progression. Myocardial insulin signaling adapts rapidly to changes in the systemic metabolic milieu. What may initially represent an adaptation to protect the heart from carbotoxicity may contribute to amplifying the risk of heart failure in obesity and diabetes. This review article presents the multiple roles of insulin signaling in cardiac physiology and pathology and discusses the potential therapeutic consequences of modulating myocardial insulin signaling.
Collapse
Affiliation(s)
- E Dale Abel
- Division of Endocrinology, Metabolism and Diabetes and Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
25
|
Abstract
Alterations in cardiac energy metabolism contribute to the severity of heart failure. However, the energy metabolic changes that occur in heart failure are complex and are dependent not only on the severity and type of heart failure present but also on the co-existence of common comorbidities such as obesity and type 2 diabetes. The failing heart faces an energy deficit, primarily because of a decrease in mitochondrial oxidative capacity. This is partly compensated for by an increase in ATP production from glycolysis. The relative contribution of the different fuels for mitochondrial ATP production also changes, including a decrease in glucose and amino acid oxidation, and an increase in ketone oxidation. The oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in heart failure associated with diabetes and obesity, myocardial fatty acid oxidation increases, while in heart failure associated with hypertension or ischemia, myocardial fatty acid oxidation decreases. Combined, these energy metabolic changes result in the failing heart becoming less efficient (ie, a decrease in cardiac work/O2 consumed). The alterations in both glycolysis and mitochondrial oxidative metabolism in the failing heart are due to both transcriptional changes in key enzymes involved in these metabolic pathways, as well as alterations in NAD redox state (NAD+ and nicotinamide adenine dinucleotide levels) and metabolite signaling that contribute to posttranslational epigenetic changes in the control of expression of genes encoding energy metabolic enzymes. Alterations in the fate of glucose, beyond flux through glycolysis or glucose oxidation, also contribute to the pathology of heart failure. Of importance, pharmacological targeting of the energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac efficiency, decreasing the energy deficit and improving cardiac function in the failing heart.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington, Seattle (R.T.)
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.)
| | - E Dale Abel
- Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City (E.D.A.).,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City (E.D.A.)
| |
Collapse
|
26
|
Kuspriyanti NP, Ariyanto EF, Syamsunarno MRAA. Role of Warburg Effect in Cardiovascular Diseases: A Potential Treatment Option. Open Cardiovasc Med J 2021. [DOI: 10.2174/1874192402115010006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background:
Under normal conditions, the heart obtains ATP through the oxidation of fatty acids, glucose, and ketones. While fatty acids are the main source of energy in the heart, under certain conditions, the main source of energy shifts to glucose where pyruvate converts into lactate, to meet the energy demand. The Warburg effect is the energy shift from oxidative phosphorylation to glycolysis in the presence of oxygen. This effect is observed in tumors as well as in diseases, including cardiovascular diseases. If glycolysis is more dominant than glucose oxidation, the two pathways uncouple, contributing to the severity of the heart condition. Recently, several studies have documented changes in metabolism in several cardiovascular diseases; however, the specific mechanisms remain unclear.
Methods:
This literature review was conducted by an electronic database of Pub Med, Google Scholar, and Scopus published until 2020. Relevant papers are selected based on inclusion and exclusion criteria.
Results:
A total of 162 potentially relevant articles after the title and abstract screening were screened for full-text. Finally, 135 papers were included for the review article.
Discussion:
This review discusses the effects of alterations in glucose metabolism, particularly the Warburg effect, on cardiovascular diseases, including heart failure, atrial fibrillation, and cardiac hypertrophy.
Conclusion:
Reversing the Warburg effect could become a potential treatment option for cardiovascular diseases.
Collapse
|
27
|
Pasqua T, Rocca C, Giglio A, Angelone T. Cardiometabolism as an Interlocking Puzzle between the Healthy and Diseased Heart: New Frontiers in Therapeutic Applications. J Clin Med 2021; 10:721. [PMID: 33673114 PMCID: PMC7918460 DOI: 10.3390/jcm10040721] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiac metabolism represents a crucial and essential connecting bridge between the healthy and diseased heart. The cardiac muscle, which may be considered an omnivore organ with regard to the energy substrate utilization, under physiological conditions mainly draws energy by fatty acids oxidation. Within cardiomyocytes and their mitochondria, through well-concerted enzymatic reactions, substrates converge on the production of ATP, the basic chemical energy that cardiac muscle converts into mechanical energy, i.e., contraction. When a perturbation of homeostasis occurs, such as an ischemic event, the heart is forced to switch its fatty acid-based metabolism to the carbohydrate utilization as a protective mechanism that allows the maintenance of its key role within the whole organism. Consequently, the flexibility of the cardiac metabolic networks deeply influences the ability of the heart to respond, by adapting to pathophysiological changes. The aim of the present review is to summarize the main metabolic changes detectable in the heart under acute and chronic cardiac pathologies, analyzing possible therapeutic targets to be used. On this basis, cardiometabolism can be described as a crucial mechanism in keeping the physiological structure and function of the heart; furthermore, it can be considered a promising goal for future pharmacological agents able to appropriately modulate the rate-limiting steps of heart metabolic pathways.
Collapse
Affiliation(s)
- Teresa Pasqua
- Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
| | - Anita Giglio
- Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy;
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
- National Institute of Cardiovascular Research (I.N.R.C.), 40126 Bologna, Italy
| |
Collapse
|
28
|
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: 2.5] [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
|
29
|
|
30
|
Arkadievich OD. Metabolic markers of myocardium insulin resistance in dogs with heart failure. Open Vet J 2021; 10:363-370. [PMID: 33614430 PMCID: PMC7830177 DOI: 10.4314/ovj.v10i4.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background Heart failure syndrome is an aspect of primary or secondary heart disease and is associated with decompensation, formation, and activation of pathological interactions between regulation systems. This results in myocardial energy metabolism alteration. This study was carried out to defy some metabolic aspects of myocardial tissue insulin resistance (IRM) development in canine heart failure. Aim To investigate the myocardial tissue concentration of adenosine triphosphate (ATP), glucose transporters 1 and 4, pyruvate dehydrogenase (PDH), hexokinase 2, insulin receptor (InsR), and adropin (ADR) protein and to screen metabolic changes and IRM in canine myocardium with heart failure. Methods We studied 28 dogs of different sexes, ages, and breeds. Groups were formed according to primary pathology: apparently healthy dogs (HD, n = 6); dogs with CDVD (CDVDD, n = 8); dogs with DCM (DCMD, n = 6); and dogs with doxorubicin chemotherapy and doxorubicin-induced cardiomyopathy (DoxCMD, n = 8). Animals in the study were diagnosed for primary disease by standard methods and algorithms. Animals were euthanized due to incurable neurological disease, refractory heart failure, or by owners will. The material was obtained immediately after death, fixed in liquid nitrogen, and stored in -80°C refrigerator. Studied proteins concentrations were analyzed in a specialized research laboratory, using ELISA kits, provided by Cloud-Clone Corp. Results ATP, GLUT1, and GLUT4 concentrations in myocardial tissue from the valvular disease group did not differ from the HD group. In CDVD, we found depression of PDH, hexokinase II (HX2), and ADR concentrations in comparison to HD. InsR was significantly lower in the CDVD and DoxCMD groups in comparison to the HD group, but in the DCM group, it was twofold higher than in the HD group. In the DCMD and DoxCMD groups, all parameters were lower than in the HD group. ATP, HX2, ADR, GLUT1, and GLUT4 were higher in the CDVD group, than in the DCM and DoxCM groups. PDH in the CDVD and DoxCM groups did not differ. PDH was depleted in the DCM to CDVD and DoxCM groups. InsR did not differ between the CDVD and DoxCM groups, but was upregulated in the DCM to CDVD and DoxCM groups. Conclusion Development of myocardial tissue IRM is a part of the structural, functional and metabolic remodeling in dogs with heart failure of different etiology. At the late stages, we found significant changes in energy supply availability and production in the myocardium.
Collapse
|
31
|
Houson H, Hedrick A, Awasthi V. Drug-induced cardiomyopathy: Characterization of a rat model by [ 18F]FDG/PET and [ 99mTc]MIBI/SPECT. Animal Model Exp Med 2020; 3:295-303. [PMID: 33532704 PMCID: PMC7824964 DOI: 10.1002/ame2.12136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/10/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Drug-induced cardiomyopathy is a significant medical problem. Clinical diagnosis of myocardial injury is based on initial electrocardiogram, levels of circulating biomarkers, and perfusion imaging with single photon emission computed tomography (SPECT). Positron emission tomography (PET) is an alternative imaging modality that provides better resolution and sensitivity than SPECT, improves diagnostic accuracy, and allows therapeutic monitoring. The objective of this study was to assess the detection of drug-induced cardiomyopathy by PET using 2-deoxy-2-[18F]fluoro-D-glucose (FDG) and compare it with the conventional SPECT technique with [99mTc]-Sestamibi (MIBI). METHODS Cardiomyopathy was induced in Sprague Dawley rats using high-dose isoproterenol. Nuclear [18F]FDG/PET and [99mTc]MIBI/SPECT were performed before and after isoproterenol administration. [18F]FDG (0.1 mCi, 200-400 µL) and [99mTc]MIBI (2 mCi, 200-600 µL) were administered via the tail vein and imaging was performed 1 hour postinjection. Isoproterenol-induced injury was confirmed by the plasma level of cardiac troponin and triphenyltetrazolium chloride (TTC) staining. RESULTS Isoproterenol administration resulted in an increase in circulating cardiac troponin I and showed histologic damage in the myocardium. Visually, preisoproterenol and postisoproterenol images showed alterations in cardiac accumulation of [18F]FDG, but not of [99mTc]MIBI. Image analysis revealed that myocardial uptake of [18F]FDG reduced by 60% after isoproterenol treatment, whereas that of [99mTc]MIBI decreased by 45%. CONCLUSION We conclude that [18F]FDG is a more sensitive radiotracer than [99mTc]MIBI for imaging of drug-induced cardiomyopathy. We theorize that isoproterenol-induced cardiomyopathy impacts cellular metabolism more than perfusion, which results in more substantial changes in [18F]FDG uptake than in [99mTc]MIBI accumulation in cardiac tissue.
Collapse
Affiliation(s)
- Hailey Houson
- Research Imaging FacilityDepartment of Pharmaceutical SciencesCollege of PharmacyUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
| | - Andria Hedrick
- Research Imaging FacilityDepartment of Pharmaceutical SciencesCollege of PharmacyUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
| | - Vibhudutta Awasthi
- Research Imaging FacilityDepartment of Pharmaceutical SciencesCollege of PharmacyUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
- Hexakit, Inc.EdmondOKUSA
| |
Collapse
|
32
|
Tsg101 Is Involved in the Sorting and Re-Distribution of Glucose Transporter-4 to the Sarcolemma Membrane of Cardiac Myocytes. Cells 2020; 9:cells9091936. [PMID: 32839388 PMCID: PMC7565110 DOI: 10.3390/cells9091936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 11/17/2022] Open
Abstract
Cardiac cells can adapt to pathological stress-induced energy crisis by shifting from fatty acid oxidation to glycolysis. However, the use of glucose-insulin-potassium (GIK) solution in patients undergoing cardiac surgery does not alleviate ischemia/reperfusion (I/R)-induced energy shortage. This indicates that insulin-mediated translocation of glucose transporter-4 (Glut-4) is impaired in ischemic hearts. Indeed, cardiac myocytes contain two intracellular populations of Glut-4: an insulin-dependent non-endosomal pool (also referred to as Glut-4 storage vesicles, GSVs) and an insulin-independent endosomal pool. Tumor susceptibility gene 101 (Tsg101) has been implicated in the endosomal recycling of membrane proteins. In this study, we aimed to examine whether Tsg101 regulated the sorting and re-distribution of Glut-4 to the sarcolemma membrane of cardiomyocytes under basal and ischemic conditions, using gain- and loss-of-function approaches. Forced overexpression of Tsg101 in mouse hearts and isolated cardiomyocytes could promote Glut-4 re-distribution to the sarcolemma, leading to enhanced glucose entry and adenosine triphosphate (ATP) generation in I/R hearts which in turn, attenuation of I/R-induced cardiac dysfunction. Conversely, knockdown of Tsg101 in cardiac myocytes exhibited opposite effects. Mechanistically, we identified that Tsg101 could interact and co-localize with Glut-4 in the sarcolemma membrane of cardiomyocytes. Our findings define Tsg101 as a novel regulator of cardiac Glut-4 trafficking, which may provide a new therapeutic strategy for the treatment of ischemic heart disease.
Collapse
|
33
|
Bertrand L, Auquier J, Renguet E, Angé M, Cumps J, Horman S, Beauloye C. Glucose transporters in cardiovascular system in health and disease. Pflugers Arch 2020; 472:1385-1399. [PMID: 32809061 DOI: 10.1007/s00424-020-02444-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022]
Abstract
Glucose transporters are essential for the heart to sustain its function. Due to its nature as a high energy-consuming organ, the heart needs to catabolize a huge quantity of metabolic substrates. For optimized energy production, the healthy heart constantly switches between various metabolites in accordance with substrate availability and hormonal status. This metabolic flexibility is essential for the maintenance of cardiac function. Glucose is part of the main substrates catabolized by the heart and its use is fine-tuned via complex molecular mechanisms that include the regulation of the glucose transporters GLUTs, mainly GLUT4 and GLUT1. Besides GLUTs, glucose can also be transported by cotransporters of the sodium-glucose cotransporter (SGLT) (SLC5 gene) family, in which SGLT1 and SMIT1 were shown to be expressed in the heart. This SGLT-mediated uptake does not seem to be directly linked to energy production but is rather associated with intracellular signalling triggering important processes such as the production of reactive oxygen species. Glucose transport is markedly affected in cardiac diseases such as cardiac hypertrophy, diabetic cardiomyopathy and heart failure. These alterations are not only fingerprints of these diseases but are involved in their onset and progression. The present review will depict the importance of glucose transport in healthy and diseased heart, as well as proposed therapies targeting glucose transporters.
Collapse
Affiliation(s)
- Luc Bertrand
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium.
| | - Julien Auquier
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Edith Renguet
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Marine Angé
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Julien Cumps
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Sandrine Horman
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium
| | - Christophe Beauloye
- Institut de Recherche Expérimentale et Clinique, Pole of Cardiovascular Research, Université catholique de Louvain, Avenue Hippocrate 55, B1.55.05, B-1200, Brussels, Belgium.,Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
34
|
Kang BS, Choi BY, Kho AR, Lee SH, Hong DK, Jeong JH, Kang DH, Park MK, Suh SW. An Inhibitor of the Sodium-Hydrogen Exchanger-1 (NHE-1), Amiloride, Reduced Zinc Accumulation and Hippocampal Neuronal Death after Ischemia. Int J Mol Sci 2020; 21:ijms21124232. [PMID: 32545865 PMCID: PMC7352629 DOI: 10.3390/ijms21124232] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Acidosis in the brain plays an important role in neuronal injury and is a common feature of several neurological diseases. It has been reported that the sodium–hydrogen exchanger-1 (NHE-1) is a key mediator of acidosis-induced neuronal injury. It modulates the concentration of intra- and extra-cellular sodium and hydrogen ions. During the ischemic state, excessive sodium ions enter neurons and inappropriately activate the sodium–calcium exchanger (NCX). Zinc can also enter neurons through voltage-gated calcium channels and NCX. Here, we tested the hypothesis that zinc enters the intracellular space through NCX and the subsequent zinc accumulation induces neuronal cell death after global cerebral ischemia (GCI). Thus, we conducted the present study to confirm whether inhibition of NHE-1 by amiloride attenuates zinc accumulation and subsequent hippocampus neuronal death following GCI. Mice were subjected to GCI by bilateral common carotid artery (BCCA) occlusion for 30 min, followed by restoration of blood flow and resuscitation. Amiloride (10 mg/kg, intraperitoneally (i.p.)) was immediately injected, which reduced zinc accumulation and neuronal death after GCI. Therefore, the present study demonstrates that amiloride attenuates GCI-induced neuronal injury, likely via the prevention of intracellular zinc accumulation. Consequently, we suggest that amiloride may have a high therapeutic potential for the prevention of GCI-induced neuronal death.
Collapse
Affiliation(s)
- Beom Seok Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Bo Young Choi
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - A Ra Kho
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Dae Ki Hong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Jeong Hyun Jeong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Dong Hyeon Kang
- Department of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Korea;
| | - Min Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
- Correspondence: ; Tel.: +82-10-8573-6364
| |
Collapse
|
35
|
Toll-Like Receptor-Mediated Cardiac Injury during Experimental Sepsis. Mediators Inflamm 2020; 2020:6051983. [PMID: 32410859 PMCID: PMC7199613 DOI: 10.1155/2020/6051983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
Sepsis is associated with global cardiac dysfunction and with high mortality rate. The development of septic cardiomyopathy is due to complex interactions of damage-associated molecular patters, cytokines, and complement activation products. The aim of this study was to define the effects of sepsis on cardiac structure, gap junction, and tight junction (TJ) proteins. Sepsis was induced by cecal ligation and puncture in male C57BL/6 mice. After a period of 24 h, the expression of cardiac structure, gap junction, and TJ proteins was determined. Murine HL-1 cells were stimulated with LPS, and mRNA expression of cardiac structure and gap junction proteins, intracellular reactive oxygen species, and troponin I release was analyzed. Furthermore, pyrogenic receptor subtype 7 (P2X7) expression and troponin I release of human cardiomyocytes (iPS) were determined after LPS exposure. In vivo, protein expression of connexin43 and α-actinin was decreased after the onset of polymicrobial sepsis, whereas in HL-1 cells, mRNA expression of connexin43, α-actinin, and desmin was increased in the presence of LPS. Expression of TJ proteins was not affected in vivo during sepsis. Although the presence of LPS and nigericin resulted in a significant troponin I release from HL-1 cells. Sepsis affected cardiac structure and gap junction proteins in mice, potentially contributing to compromised cardiac function.
Collapse
|
36
|
Lackner I, Weber B, Baur M, Fois G, Gebhard F, Pfeifer R, Cinelli P, Halvachizadeh S, Lipiski M, Cesarovic N, Schrezenmeier H, Huber-Lang M, Pape HC, Kalbitz M. Complement Activation and Organ Damage After Trauma-Differential Immune Response Based on Surgical Treatment Strategy. Front Immunol 2020; 11:64. [PMID: 32117238 PMCID: PMC7025487 DOI: 10.3389/fimmu.2020.00064] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/10/2020] [Indexed: 12/27/2022] Open
Abstract
Background: The complement system is part of the innate immunity, is activated immediately after trauma and is associated with adult respiratory distress syndrome, acute lung injury, multiple organ failure, and with death of multiply injured patients. The aim of the study was to investigate the complement activation in multiply injured pigs as well as its effects on the heart in vivo and in vitro. Moreover, the impact of reamed vs. non-reamed intramedullary nailing was examined with regard to the complement activation after multiple trauma in pigs. Materials and Methods: Male pigs received multiple trauma, followed by femoral nailing with/without prior conventional reaming. Systemic complement hemolytic activity (CH-50 and AH-50) as well as the local cardiac expression of C3a receptor, C5a receptors1/2, and the deposition of the fragments C3b/iC3b/C3c was determined in vivo after trauma. Human cardiomyocytes were exposed to C3a or C5a and analyzed regarding calcium signaling and mitochondrial respiration. Results: Systemic complement activation increased within 6 h after trauma and was mediated via the classical and the alternative pathway. Furthermore, complement activation correlated with invasiveness of fracture treatment. The expression of receptors for complement activation were altered locally in vivo in left ventricles. C3a and C5a acted detrimentally on human cardiomyocytes by affecting their functionality and their mitochondrial respiration in vitro. Conclusion: After multiple trauma, an early activation of the complement system is triggered, affecting the heart in vivo as well as in vitro, leading to complement-induced cardiac dysfunction. The intensity of complement activation after multiple trauma might correlate with the invasiveness of fracture treatment. Reaming of the femoral canal might contribute to an enhanced “second hit” response after trauma. Consequently, the choice of fracture treatment might imply the clinical outcome of the critically injured patients and might be therefore crucial for their survival.
Collapse
Affiliation(s)
- Ina Lackner
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| | - Birte Weber
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| | - Meike Baur
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| | - Giorgio Fois
- Institute of General Physiology, University of Ulm, Ulm, Germany
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| | - Roman Pfeifer
- Department of Trauma, University Hospital of Zurich, Zurich, Switzerland
| | - Paolo Cinelli
- Department of Trauma, University Hospital of Zurich, Zurich, Switzerland
| | | | - Miriam Lipiski
- Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Nikola Cesarovic
- Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Hubert Schrezenmeier
- Institute of Transfusion Medicine, University of Ulm and Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, Ulm, Germany.,German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen and University Hospital Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute for Clinical- and Experimental Trauma-Immunology, University of Ulm, Ulm, Germany
| | | | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University of Ulm, Ulm, Germany
| |
Collapse
|
37
|
Ruiz-Velasco A, Zi M, Hille SS, Azam T, Kaur N, Jiang J, Nguyen B, Sekeres K, Binder P, Collins L, Pu F, Xiao H, Guan K, Frey N, Cartwright EJ, Müller OJ, Wang X, Liu W. Targeting mir128-3p alleviates myocardial insulin resistance and prevents ischemia-induced heart failure. eLife 2020; 9:54298. [PMID: 32223896 PMCID: PMC7124275 DOI: 10.7554/elife.54298] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/27/2020] [Indexed: 01/02/2023] Open
Abstract
Myocardial insulin resistance contributes to heart failure in response to pathological stresses, therefore, a therapeutic strategy to maintain cardiac insulin pathways requires further investigation. We demonstrated that insulin receptor substrate 1 (IRS1) was reduced in failing mouse hearts post-myocardial infarction (MI) and failing human hearts. The mice manifesting severe cardiac dysfunction post-MI displayed elevated mir128-3p in the myocardium. Ischemia-upregulated mir128-3p promoted Irs1 degradation. Using rat cardiomyocytes and human-induced pluripotent stem cell-derived cardiomyocytes, we elucidated that mitogen-activated protein kinase 7 (MAPK7, also known as ERK5)-mediated CCAAT/enhancer-binding protein beta (CEBPβ) transcriptionally represses mir128-3p under hypoxia. Therapeutically, functional studies demonstrated gene therapy-delivered cardiac-specific MAPK7 restoration or overexpression of CEBPβ impeded cardiac injury after MI, at least partly due to normalization of mir128-3p. Furthermore, inhibition of mir128-3p preserved Irs1 and ameliorated cardiac dysfunction post-MI. In conclusion, we reveal that targeting mir128-3p mitigates myocardial insulin resistance, thereafter slowing down the progression of heart failure post-ischemia.
Collapse
Affiliation(s)
- Andrea Ruiz-Velasco
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Min Zi
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Susanne S Hille
- Department of Internal Medicine III, University of KielKielGermany,DZHK, German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckKielGermany
| | - Tayyiba Azam
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Namrita Kaur
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Juwei Jiang
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Binh Nguyen
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Karolina Sekeres
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universitaet DresdenDresdenGermany
| | - Pablo Binder
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Lucy Collins
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Fay Pu
- Edinburgh University Medical SchoolEdinburghUnited Kingdom
| | - Han Xiao
- Institute of Vascular Medicine, Peking UniversityBeijingChina
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universitaet DresdenDresdenGermany
| | - Norbert Frey
- Department of Internal Medicine III, University of KielKielGermany,DZHK, German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckKielGermany
| | - Elizabeth J Cartwright
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Oliver J Müller
- Department of Internal Medicine III, University of KielKielGermany,DZHK, German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/LübeckKielGermany
| | - Xin Wang
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| | - Wei Liu
- Faculty of Biology, Medicine, and Health, the University of ManchesterManchesterUnited Kingdom
| |
Collapse
|
38
|
Studneva IM, Veselova OM, Bahtin AA, Konovalova GG, Lankin VZ, Pisarenko OI. The Mechanisms of Cardiac Protection Using a Synthetic Agonist of Galanin Receptors during Chronic Administration of Doxorubicin. Acta Naturae 2020; 12:89-98. [PMID: 32477603 PMCID: PMC7245963 DOI: 10.32607/actanaturae.10945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The use of the anticancer drug doxorubicin (Dox) is limited by its cardiotoxic
effect. The aim of this work was to study the effect of a new synthetic agonist
of the galanin receptor GalR1-3 [βAla14, His15]-galanine (2–15) (G)
on the metabolism, antioxidant enzyme activity, and cardiac function in rats
with cardiomyopathy (CM) caused by chronic administration of Dox.
Coadministration of peptide G and Dox significantly increased the fractional
shortening (FS) and ejection fraction (EF) by an average of 30 ± 4%
compared with the indices in the Dox group. The reduced severity of cardiac
dysfunction under the action of G was accompanied by a 2.5-fold decrease in the
activity of creatine kinase-MB (CK-MB) in blood plasma. The protective
mechanism of the action of peptide G is caused by a reduced lipid peroxidation
(LP) that is due to the increased activity of Cu,Zn superoxide dismutase
(Cu,Zn-SOD) and glutathione peroxidase (GSH-Px) in the damaged heart.
Administration of peptide G significantly increased the adenine nucleotide pool
(ΣAH), ATP content, and the levels of phosphocreatine (PCr) and total
creatine (ΣCr) in the damaged myocardium. It also reduced lactate
accumulation relative to its content in the Dox group. The better energy supply
of cardiomyocytes after treatment with peptide G prevented the accumulation of
cytotoxic ammonia and disruption in the metabolism of the key myocardial amino
acids (glutamic acid (Glu), aspartic acid (Asp), and alanine (Ala)). Peptide G
significantly improved the morphological parameters of the heart in rats
treated with Dox. The results show promise in using peptide G to efficiently
correct functional, morphological, and metabolic damage to the heart caused by
anthracycline chemotherapy.
Collapse
Affiliation(s)
- I. M. Studneva
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| | - O. M. Veselova
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| | - A. A. Bahtin
- Research and Clinical Center of Otorhinolaryngology, Moscow, 123182 Russia
| | - G. G. Konovalova
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| | - V. Z. Lankin
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| | - O. I. Pisarenko
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| |
Collapse
|
39
|
Studneva I, Serebryakova L, Veselova O, Pal'keeva M, Molokoedov A, Ovchinnikov M, Konovalova G, Lankin V, Sidorova M, Pisarenko O. Galanin receptors activation modulates myocardial metabolic and antioxidant responses to ischaemia/reperfusion stress. Clin Exp Pharmacol Physiol 2019; 46:1174-1182. [PMID: 31429479 DOI: 10.1111/1440-1681.13164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/01/2019] [Accepted: 08/12/2019] [Indexed: 01/23/2023]
Abstract
The mechanisms of protective action of the neuropeptide galanin and its N-terminal fragments against myocardial ischaemia/reperfusion (I/R) injury remain obscure. The aim of this work was to study effects of a novel peptide agonist of galanin receptors [βAla14, His15]-galanin (2-15) (G1) and the full-length galanin (G2) on energy and antioxidant status of the heart with acute infarction. The peptides were synthesized by the automatic solid phase method using Fmoc technology. Their structure was identified by 1 H-NMR spectroscopy and MALDI-TOF mass spectrometry. Experiments were performed on anaesthetized open-chest rats subjected to myocardial regional ischaemia and reperfusion. Intravenous (iv) administration of optimal doses of peptides G1 and G2 (1.0 and 0.5 mg/kg, respectively, at the onset of reperfusion significantly reduced infarct size (on average by 40% compared with control) and the plasma activity of creatine kinase-MB (CK-MB) and lactate dehydrogenase (LDH). These effects were associated with augmented preservation of aerobic energy metabolism, increased activity of Cu,Zn superoxide dismutase (Cu,Zn-SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) and decreased lipid peroxidation in the area at risk (AAR) at the end of reperfusion. Peptide G1 showed more efficient recovery of the majority of metabolic and antioxidant parameters. The results provide evidence that the galaninergic system can be considered a promising target to reduce energy dysregulation and oxidative damage in myocardial I/R injury.
Collapse
Affiliation(s)
- Irina Studneva
- National Medical Research Center for Cardiology, Moscow, Russia
| | | | - Oksana Veselova
- National Medical Research Center for Cardiology, Moscow, Russia
| | | | | | | | | | - Vadim Lankin
- National Medical Research Center for Cardiology, Moscow, Russia
| | - Maria Sidorova
- National Medical Research Center for Cardiology, Moscow, Russia
| | - Oleg Pisarenko
- National Medical Research Center for Cardiology, Moscow, Russia
| |
Collapse
|
40
|
Serebryakova LI, Studneva IM, Ovchinnikov MV, Veselova OM, Molokoedov AS, Arzamastsev EV, Afanasyeva EY, Terekhova OA, Sidorova MV, Pisarenko OI. [Cardiometabolic efficacy and toxicological evaluation of a pharmacological galanin receptor agonist]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 65:231-238. [PMID: 31258147 DOI: 10.18097/pbmc20196503231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The goal of this study was to examine effects of a novel galanin receptor agonist GalR1-3 [bAla14, His15]-galanine 2-15 (G), obtained by automatic solid-phase synthesis, on the metabolic state of the area at risk and the size of acute myocardial infarction (MI) in rats in vivo and evaluate its toxicity in BALB /c mice. In anesthetized rats, regional ischemia was simulated by coronary artery occlusion and then coronary blood flow was restored. The peptide G was administered intravenously (i.v.) with a bolus after a period of regional ischemia in the dose range of 0.25-3.0 mg/kg. The sizes of MI and the activities of creatine kinase-MB (СK-MB) and lactate dehydrogenase (LDH) in blood plasma were estimated. The effect of administration of the optimal dose of G (1.0 mg/kg) on myocardial content of adenine nucleotides (AN), phosphocreatine (PCr), creatine (Cr) and lactate was studied. I.v. administration of G to rats at a dose of 1.0 mg/kg slightly affected hemodynamic parameters, but reduced MI size by 40% and decreased plasma LDH and CK-MB activity by the end of reperfusion compared to control. These effects were accompanied by a significant improvement in energy state of area at risk (AAR) - an increase in myocardial content of ATP, åAN, PCr and åCr, and combined with a decrease in myocardial lactate level compared with the control. Toxicity of peptide G was studied with a single intraperitoneal injection of 0.5-3.0% solution of the peptide substance to mice. The absence of signs of intoxication and death of animals after G injection in the maximum possible dose did not allow determining the value of the average lethal dose. The results indicate therapeutic potential of the peptide G for preventing myocardial ischemia and reperfusion injury and feasibility for further study of its pharmacological properties and mechanisms of action.
Collapse
Affiliation(s)
| | - I M Studneva
- National Medical Research Center for Cardiology, Moscow, Russia
| | - M V Ovchinnikov
- National Medical Research Center for Cardiology, Moscow, Russia
| | - O M Veselova
- National Medical Research Center for Cardiology, Moscow, Russia
| | - A S Molokoedov
- National Medical Research Center for Cardiology, Moscow, Russia
| | - E V Arzamastsev
- National Medical Research Center for Cardiology, Moscow, Russia
| | - E Yu Afanasyeva
- National Medical Research Center for Cardiology, Moscow, Russia
| | - O A Terekhova
- National Medical Research Center for Cardiology, Moscow, Russia
| | - M V Sidorova
- National Medical Research Center for Cardiology, Moscow, Russia
| | - O I Pisarenko
- National Medical Research Center for Cardiology, Moscow, Russia
| |
Collapse
|
41
|
Yoshii A, Nagoshi T, Kashiwagi Y, Kimura H, Tanaka Y, Oi Y, Ito K, Yoshino T, Tanaka TD, Yoshimura M. Cardiac ischemia-reperfusion injury under insulin-resistant conditions: SGLT1 but not SGLT2 plays a compensatory protective role in diet-induced obesity. Cardiovasc Diabetol 2019; 18:85. [PMID: 31262297 PMCID: PMC6604374 DOI: 10.1186/s12933-019-0889-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/23/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent large-scale clinical trials have shown that SGLT2-inhibitors reduce cardiovascular events in diabetic patients. However, the regulation and functional role of cardiac sodium-glucose cotransporter (SGLT1 is the dominant isoform) compared with those of other glucose transporters (insulin-dependent GLUT4 is the major isoform) remain incompletely understood. Given that glucose is an important preferential substrate for myocardial energy metabolism under conditions of ischemia-reperfusion injury (IRI), we hypothesized that SGLT1 contributes to cardioprotection during the acute phase of IRI via enhanced glucose transport, particularly in insulin-resistant phenotypes. METHODS AND RESULTS The hearts from mice fed a high-fat diet (HFD) for 12 weeks or a normal-fat diet (NFD) were perfused with either the non-selective SGLT-inhibitor phlorizin or selective SGLT2-inhibitors (tofogliflozin, ipragliflozin, canagliflozin) during IRI using Langendorff model. After ischemia-reperfusion, HFD impaired left ventricular developed pressure (LVDP) recovery compared with the findings in NFD. Although phlorizin-perfusion impaired LVDP recovery in NFD, a further impaired LVDP recovery and a dramatically increased infarct size were observed in HFD with phlorizin-perfusion. Meanwhile, none of the SGLT2-inhibitors significantly affected cardiac function or myocardial injury after ischemia-reperfusion under either diet condition. The plasma membrane expression of GLUT4 was significantly increased after IRI in NFD but was substantially attenuated in HFD, the latter of which was associated with a significant reduction in myocardial glucose uptake. In contrast, SGLT1 expression at the plasma membrane remained constant during IRI, regardless of the diet condition, whereas SGLT2 was not detected in the hearts of any mice. Of note, phlorizin considerably reduced myocardial glucose uptake after IRI, particularly in HFD. CONCLUSIONS Cardiac SGLT1 but not SGLT2 plays a compensatory protective role during the acute phase of IRI via enhanced glucose uptake, particularly under insulin-resistant conditions, in which IRI-induced GLUT4 upregulation is compromised.
Collapse
Affiliation(s)
- Akira Yoshii
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan.
| | - Yusuke Kashiwagi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Haruka Kimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yoshiro Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yuhei Oi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Keiichi Ito
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takuya Yoshino
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Toshikazu D Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| |
Collapse
|
42
|
Affiliation(s)
- Diem H Tran
- 1 Division of Cardiology Department of Internal Medicine University of Texas Southwestern Medical Center Dallas TX
| | - Zhao V Wang
- 1 Division of Cardiology Department of Internal Medicine University of Texas Southwestern Medical Center Dallas TX
| |
Collapse
|
43
|
Abstract
Inflammatory processes underlie many diseases associated with injury of the heart muscle, including conditions without an obvious inflammatory pathogenic component such as hypertensive and diabetic cardiomyopathy. Persistence of cardiac inflammation can cause irreversible structural and functional deficits. Some are induced by direct damage of the heart muscle by cellular and soluble mediators but also by metabolic adaptations sustained by the inflammatory microenvironment. It is well established that both cardiomyocytes and immune cells undergo metabolic reprogramming in the site of inflammation, which allow them to deal with decreased availability of nutrients and oxygen. However, like in cancer, competition for nutrients and increased production of signalling metabolites such as lactate initiate a metabolic cross-talk between immune cells and cardiomyocytes which, we propose, might tip the balance between resolution of the inflammation versus adverse cardiac remodeling. Here we review our current understanding of the metabolic reprogramming of both heart tissue and immune cells during inflammation, and we discuss potential key mechanisms by which these metabolic responses intersect and influence each other and ultimately define the prognosis of the inflammatory process in the heart.
Collapse
Affiliation(s)
- Federica M Marelli-Berg
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Dunja Aksentijevic
- School of Biological and Chemical Sciences, Queen Mary University of London, G.E. Fogg Building, Mile End Road, London E1 4NS, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
44
|
Chronic AICAR treatment prevents metabolic changes in cardiomyocytes exposed to free fatty acids. Pflugers Arch 2019; 471:1219-1234. [PMID: 31152240 DOI: 10.1007/s00424-019-02285-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/27/2019] [Accepted: 05/15/2019] [Indexed: 01/09/2023]
Abstract
The stimulation of glucose transport by metabolic stress is an important determinant of myocardial susceptibility to ischemia and reperfusion injury. Stimulation of glucose transport is markedly impaired in cardiomyocytes chronically exposed to excess free fatty acids (FFA), as occurs in vivo in type 2 diabetes. To determine whether chronic low-grade activation of AMP-activated kinase (AMPK) improves substrate metabolism in cardiomyocytes exposed to FFA, isolated cultured cardiomyocytes were exposed for 7 days to FFA ± the AMPK agonist 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR). Glucose transport and glycolysis were then measured during acute metabolic stress provoked by oligomycin. Chronic treatment with AICAR improved basal and oligomycin-stimulated glucose transport in FFA-exposed but not in control cardiomyocytes. Similarly, basal and oligomycin-stimulated glycolysis was reduced in FFA-exposed cardiomyocytes but restored by chronic AICAR treatment. Conversely, fatty acid oxidation was increased in FFA-exposed cardiomyocytes and reduced by chronic AICAR treatment. Chronic AICAR treatment induced in FFA-exposed cardiomyocytes the biogenesis of numerous lipid droplets. Curiously, whereas acute treatment of cardiomyocytes with AICAR increased phosphorylation of the AMPKα subunit on T172, a classical marker of AMPK activation, chronic AICAR treatment almost completely obliterated T172 phosphorylation. However, phosphorylation of the AMPK target protein raptor on S792 was reduced in FFA-exposed cardiomyocytes but restored by AICAR treatment. In conclusion, chronic AICAR treatment induces a metabolic shift in FFA-exposed cardiomyocytes, characterized by improved glucose transport and glycolysis and redirection of fatty acids towards neutral storage. Such metabolic changes in vivo could protect the hearts of patients with type 2 diabetes against ischemia-reperfusion injury.
Collapse
|
45
|
Mahmod M, Pal N, Rayner J, Holloway C, Raman B, Dass S, Levelt E, Ariga R, Ferreira V, Banerjee R, Schneider JE, Rodgers C, Francis JM, Karamitsos TD, Frenneaux M, Ashrafian H, Neubauer S, Rider O. The interplay between metabolic alterations, diastolic strain rate and exercise capacity in mild heart failure with preserved ejection fraction: a cardiovascular magnetic resonance study. J Cardiovasc Magn Reson 2018; 20:88. [PMID: 30580760 PMCID: PMC6304764 DOI: 10.1186/s12968-018-0511-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/27/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Heart failure (HF) is characterized by altered myocardial substrate metabolism which can lead to myocardial triglyceride accumulation (steatosis) and lipotoxicity. However its role in mild HF with preserved ejection fraction (HFpEF) is uncertain. We measured myocardial triglyceride content (MTG) in HFpEF and assessed its relationships with diastolic function and exercise capacity. METHODS Twenty seven HFpEF (clinical features of HF, left ventricular EF >50%, evidence of mild diastolic dysfunction and evidence of exercise limitation as assessed by cardiopulmonary exercise test) and 14 controls underwent 1H-cardiovascular magnetic resonance spectroscopy (1H-CMRS) to measure MTG (lipid/water, %), 31P-CMRS to measure myocardial energetics (phosphocreatine-to-adenosine triphosphate - PCr/ATP) and feature-tracking cardiovascular magnetic resonance (CMR) imaging for diastolic strain rate. RESULTS When compared to controls, HFpEF had 2.3 fold higher in MTG (1.45 ± 0.25% vs. 0.64 ± 0.16%, p = 0.009) and reduced PCr/ATP (1.60 ± 0.09 vs. 2.00 ± 0.10, p = 0.005). HFpEF had significantly reduced diastolic strain rate and maximal oxygen consumption (VO2 max), which both correlated significantly with elevated MTG and reduced PCr/ATP. On multivariate analyses, MTG was independently associated with diastolic strain rate while diastolic strain rate was independently associated with VO2 max. CONCLUSIONS Myocardial steatosis is pronounced in mild HFpEF, and is independently associated with impaired diastolic strain rate which is itself related to exercise capacity. Steatosis may adversely affect exercise capacity by indirect effect occurring via impairment in diastolic function. As such, myocardial triglyceride may become a potential therapeutic target to treat the increasing number of patients with HFpEF.
Collapse
Affiliation(s)
- Masliza Mahmod
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
- National University of Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Nikhil Pal
- Divisions of Experimental Therapeutics and Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Jennifer Rayner
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Cameron Holloway
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Betty Raman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Sairia Dass
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Eylem Levelt
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Rina Ariga
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Vanessa Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Rajarshi Banerjee
- 1st Department of Cardiology, Aristotle University, Thessaloniki, Greece
| | - Jurgen E. Schneider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Christopher Rodgers
- Department of Medicine, John Radcliffe Hospital, Oxford, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Jane M. Francis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Theodoros D. Karamitsos
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
- 1st Department of Cardiology, Aristotle University, Thessaloniki, Greece
| | - Michael Frenneaux
- Norwich Medical School, Bob Champion Research and Education Building, James Watson Road, University of East Anglia Norwich Research Park, Norwich, NR4 7UQ UK
| | - Houman Ashrafian
- Divisions of Experimental Therapeutics and Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| | - Oliver Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU UK
| |
Collapse
|
46
|
Meagher P, Adam M, Civitarese R, Bugyei-Twum A, Connelly KA. Heart Failure With Preserved Ejection Fraction in Diabetes: Mechanisms and Management. Can J Cardiol 2018; 34:632-643. [PMID: 29731023 DOI: 10.1016/j.cjca.2018.02.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 02/25/2018] [Accepted: 02/25/2018] [Indexed: 12/20/2022] Open
Abstract
Diabetes mellitus (DM) is a major cause of heart failure in the Western world, either secondary to coronary artery disease or from a distinct entity known as "diabetic cardiomyopathy." Furthermore, heart failure with preserved ejection fraction (HFpEF) is emerging as a significant clinical problem for patients with DM. Current clinical data suggest that between 30% and 40% of patients with HFpEF suffer from DM. The typical structural phenotype of the HFpEF heart consists of endothelial dysfunction, increased interstitial and perivascular fibrosis, cardiomyocyte stiffness, and hypertrophy along with advanced glycation end products deposition. There is a myriad of mechanisms that result in the phenotypical HFpEF heart including impaired cardiac metabolism and substrate utilization, altered insulin signalling leading to protein kinase C activation, advanced glycated end products deposition, prosclerotic cytokine activation (eg, transforming growth factor-β activation), along with impaired nitric oxide production from the endothelium. Moreover, recent investigations have focused on the role of endothelial-myocyte interactions. Despite intense research, current therapeutic strategies have had little effect on improving morbidity and mortality in patients with DM and HFpEF. Possible explanations for this include a limited understanding of the role that direct cell-cell communication or indirect cell-cell paracrine signalling plays in the pathogenesis of DM and HFpEF. Additionally, integrins remain another important mediator of signals from the extracellular matrix to cells within the failing heart and might play a significant role in cell-cell cross-talk. In this review we discuss the characteristics and mechanisms of DM and HFpEF to stimulate potential future research for patients with this common, and morbid condition.
Collapse
Affiliation(s)
- Patrick Meagher
- Keenan Research Centre for Biomedical Science, St Michael's Hospital; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Mohamed Adam
- Keenan Research Centre for Biomedical Science, St Michael's Hospital; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Robert Civitarese
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Antoinette Bugyei-Twum
- Keenan Research Centre for Biomedical Science, St Michael's Hospital; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St Michael's Hospital; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St Michael's Hospital; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Division of Cardiology, St Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Chen YP, Kuo WW, Baskaran R, Day CH, Chen RJ, Wen SY, Ho TJ, Padma VV, Kuo CH, Huang CY. Acute hypoxic preconditioning prevents palmitic acid-induced cardiomyocyte apoptosis via switching metabolic GLUT4-glucose pathway back to CD36-fatty acid dependent. J Cell Biochem 2018; 119:3363-3372. [PMID: 29130531 DOI: 10.1002/jcb.26501] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 11/09/2017] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome is a risk factor for the development of cardiovascular diseases. Myocardial cell damage leads to an imbalance of energy metabolism, and many studies have indicated that short-term hypoxia during myocardial cell injury has a protective effect. In our previous animal studies, we found that short-term hypoxia in the heart has a protective effect, but long-term hypoxia increases myocardial cell injury. Palmitic acid (PA)-treated H9c2 cardiomyoblasts and neonatal rat ventricle cardiomyocytes were used to simulate hyperlipidemia model, which suppress cluster of differentiation 36 (CD36) and activate glucose transporter type 4 (GLUT4). We exposed the cells to short- and long-term hypoxia and investigated the protective effects of hypoxic preconditioning on PA-induced lipotoxicity in H9c2 cardiomyoblasts and neonatal rat cardiomyocytes. Preconditioning with short-term hypoxia enhanced both CD36 and GLUT4 metabolism pathway protein levels. Expression levels of phospho-PI3K, phospho-Akt, phospho-AMPK, SIRT1, PGC1α, PPARα, CD36, and CPT1β induced by PA was reversed by short-term hypoxia in a time-dependent manner. PA-induced increased GLUT4 membrane protein level was reduced in the cells exposed to short-term hypoxia and si-PKCζ. Short-term hypoxia, resveratrol and si-PKCζ rescue H9c2 cells from apoptosis induced by PA and switch the metabolic pathway from GLUT4 dependent to CD36 dependent. We demonstrate short-term hypoxic preconditioning as a more efficient way as resveratrol in maintaining the energy metabolism of hearts during hyperlipidemia and can be used as a therapeutic strategy.
Collapse
Affiliation(s)
- Yeh-Peng Chen
- PhD Program for Aging, China Medical University, Taichung, Taiwan.,Division of Cardiology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Rathinasamy Baskaran
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | | | - Ray-Jade Chen
- Department of Surgery, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Su-Ying Wen
- Department of Dermatology, Taipei City Hospital, Renai Branch, Taipei, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, China Medical University Beigang Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
| | | | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan.,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
48
|
Riddy DM, Delerive P, Summers RJ, Sexton PM, Langmead CJ. G Protein-Coupled Receptors Targeting Insulin Resistance, Obesity, and Type 2 Diabetes Mellitus. Pharmacol Rev 2018; 70:39-67. [PMID: 29233848 DOI: 10.1124/pr.117.014373] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/13/2017] [Indexed: 03/21/2025] Open
Abstract
G protein-coupled receptors (GPCRs) continue to be important discovery targets for the treatment of type 2 diabetes mellitus (T2DM). Many GPCRs are directly involved in the development of insulin resistance and β-cell dysfunction, and in the etiology of inflammation that can lead to obesity-induced T2DM. This review summarizes the current literature describing a number of well-validated GPCR targets, but also outlines several new and promising targets for drug discovery. We highlight the importance of understanding the role of these receptors in the disease pathology, and their basic pharmacology, which will pave the way to the development of novel pharmacological probes that will enable these targets to fulfill their promise for the treatment of these metabolic disorders.
Collapse
Affiliation(s)
- Darren M Riddy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Philippe Delerive
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Patrick M Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| | - Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (D.M.R., R.J.S., P.M.S., C.J.L.); and Institut de Recherches Servier, Pôle d'Innovation Thérapeutique Métabolisme, Suresnes, France (P.D.)
| |
Collapse
|
49
|
Drozd K, Ahmadi A, Deng Y, Jiang B, Petryk J, Thorn S, Stewart D, Beanlands R, deKemp RA, DaSilva JN, Mielniczuk LM. Effects of an endothelin receptor antagonist, Macitentan, on right ventricular substrate utilization and function in a Sugen 5416/hypoxia rat model of severe pulmonary arterial hypertension. J Nucl Cardiol 2017; 24:1979-1989. [PMID: 27688036 DOI: 10.1007/s12350-016-0663-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 07/25/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Altered myocardial energy metabolism has been linked to worsening of RV function in pulmonary arterial hypertension (PAH). The aim of this study was to evaluate RV glucose and fatty acid metabolism in vivo in a rat model of PAH using positron emission tomography (PET) and investigate the effects of Macitentan on RV substrate utilization. METHODS PAH was induced in male Sprague-Dawley rats by a single subcutaneous injection of Sugen 5416 (20 mg/kg) followed by 3 weeks of hypoxia (10% oxygen). At week 5 post-injection, the PAH rats were randomized to Macitentan (30 mg/kg daily) treatment or no treatment. Substrate utilization was serially assessed 5 and 8 weeks post-injection with 2-[18F]fluoro-2-deoxyglucose (FDG) and 14(R,S)-[18F]fluoro-6-thia-heptadecanoic acid (FTHA) PET for glucose and fatty acid metabolism respectively and correlated with in vivo functional measurements. RESULTS PAH induction resulted in a 2.5-fold increase in RV FDG uptake (standardized uptake value (SUV) of normal control: 1.6 ± 0.4, week 5: 4.1 ± 1.9, week 8: 4.0 ± 1.6, P < 0.05 for all groups vs. control). RV FTHA showed twofold increased uptake at week 5 (SUV control: 1.50 ± 0.39, week 5: 3.06 ± 1.10, P = 0.03). Macitentan significantly decreased RV FDG uptake at 8 weeks (SUV: 2.5 ± 0.9, P = 0.04), associated with improved RV ejection fraction and reduced RV systolic pressure, while FTHA uptake was maintained. CONCLUSION PAH is associated with metabolic changes in the RV, characterized by a marked increase in FDG and FTHA uptake. Macitentan treatment reduced PAH severity and was associated with a decrease in RV FDG uptake and improved RV function.
Collapse
Affiliation(s)
- Katarzyna Drozd
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y 4W7, Canada
| | - Ali Ahmadi
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y 4W7, Canada
| | - Yupu Deng
- Ottawa Hospital Research Institute, 1053 Carling Ave, Ottawa, ON, Canada
| | - Baohua Jiang
- Ottawa Hospital Research Institute, 1053 Carling Ave, Ottawa, ON, Canada
| | - Julia Petryk
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y 4W7, Canada
| | - Stephanie Thorn
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y 4W7, Canada
| | - Duncan Stewart
- Ottawa Hospital Research Institute, 1053 Carling Ave, Ottawa, ON, Canada
| | - Rob Beanlands
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y 4W7, Canada
| | - Robert A deKemp
- Department of Cardiac Imaging, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Jean N DaSilva
- Department of Radiology, Radio-Oncology and Nuclear Medicine, University of Montreal, Montreal, QC, Canada
- University of Montreal Hospital Research Centre (CRCHUM), Montreal, QC, Canada
| | - Lisa M Mielniczuk
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON, K1Y 4W7, Canada.
| |
Collapse
|
50
|
Sato M, Evans BA, Sandström AL, Chia LY, Mukaida S, Thai BS, Nguyen A, Lim L, Tan CYR, Baltos JA, White PJ, May LT, Hutchinson DS, Summers RJ, Bengtsson T. α 1A-Adrenoceptors activate mTOR signalling and glucose uptake in cardiomyocytes. Biochem Pharmacol 2017; 148:27-40. [PMID: 29175420 DOI: 10.1016/j.bcp.2017.11.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/22/2017] [Indexed: 12/20/2022]
Abstract
The capacity of G protein-coupled receptors to modulate mechanistic target of rapamycin (mTOR) activity is a newly emerging paradigm with the potential to link cell surface receptors with cell survival. Cardiomyocyte viability is linked to signalling pathways involving Akt and mTOR, as well as increased glucose uptake and utilization. Our aim was to determine whether the α1A-adrenoceptor (AR) couples to these protective pathways, and increased glucose uptake. We characterised α1A-AR signalling in CHO-K1 cells co-expressing the human α1A-AR and GLUT4 (CHOα1AGLUT4myc) and in neonatal rat ventricular cardiomyocytes (NRVM), and measured glucose uptake, intracellular Ca2+ mobilization, and phosphorylation of mTOR, Akt, 5' adenosine monophosphate-activated kinase (AMPK) and S6 ribosomal protein (S6rp). In both systems, noradrenaline and the α1A-AR selective agonist A61603 stimulated glucose uptake by parallel pathways involving mTOR and AMPK, whereas another α1-AR agonist oxymetazoline increased glucose uptake predominantly by mTOR. All agonists promoted phosphorylation of mTOR at Ser2448 and Ser2481, indicating activation of both mTORC1 and mTORC2, but did not increase Akt phosphorylation. In CHOα1AGLUT4myc cells, siRNA directed against rictor but not raptor suppressed α1A-AR mediated glucose uptake. We have thus identified mTORC2 as a key component in glucose uptake stimulated by α1A-AR agonists. Our findings identify a novel link between the α1A-AR, mTORC2 and glucose uptake, that have been implicated separately in cardiomyocyte survival. Our studies provide an improved framework for examining the utility of α1A-AR selective agonists as tools in the treatment of cardiac dysfunction.
Collapse
Affiliation(s)
- Masaaki Sato
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Bronwyn A Evans
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Anna L Sandström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Ling Yeong Chia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Saori Mukaida
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Bui San Thai
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Anh Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Linzi Lim
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Christina Y R Tan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Paul J White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden.
| |
Collapse
|