1
|
Sun S, Guo H, Chen G, Zhang H, Zhang Z, Wang X, Li D, Li X, Zhao G, Lin F. Peroxisome proliferator‑activated receptor γ coactivator‑1α in heart disease (Review). Mol Med Rep 2025; 31:17. [PMID: 39513608 PMCID: PMC11551696 DOI: 10.3892/mmr.2024.13382] [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: 07/29/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
Heart disease (HD) is a general term for various diseases affecting the heart. An increasing body of evidence suggests that the pathogenesis of HD is closely related to mitochondrial dysfunction. Peroxisome proliferator‑activated receptor γ coactivator‑1α (PGC‑1α) is a transcriptional coactivator that plays an important role in mitochondrial function by regulating mitochondrial biogenesis, energy metabolism and oxidative stress. The present review shows that PGC‑1α expression and activity in the heart are controlled by multiple signaling pathways, including adenosine monophosphate‑activated protein kinase, sirtuin 1/3 and nuclear factor κB. These can mediate the activation or inhibition of transcription and post‑translational modifications (such as phosphorylation and acetylation) of PGC‑1α. Furthermore, it highlighted the recent progress of PGC‑1α in HD, including heart failure, coronary heart disease, diabetic cardiomyopathy, drug‑induced cardiotoxicity and arrhythmia. Understanding the mechanisms underlying PGC‑1α in response to pathological stimulation may prove to be beneficial in developing new ideas and strategies for preventing and treating HDs. Meanwhile, the present review explored why the opposite results occurred when PGC‑1α was used as a target therapy.
Collapse
Affiliation(s)
- Siyu Sun
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Huige Guo
- Department of Cardiology, Xinxiang Central Hospital, Xinxiang, Henan 453000, P.R. China
| | - Guohui Chen
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Hui Zhang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Zhanrui Zhang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Xiulong Wang
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Dongxu Li
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Xuefang Li
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Guoan Zhao
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| | - Fei Lin
- Department of Cardiology, Life Science Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
- Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, Henan 453100, P.R. China
| |
Collapse
|
2
|
Fitts RH, Wang X, Kwok WM, Camara AKS. Cardiomyocyte Adaptation to Exercise: K+ Channels, Contractility and Ischemic Injury. Int J Sports Med 2024; 45:791-803. [PMID: 38648799 DOI: 10.1055/a-2296-7604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality, and exercise-training (TRN) is known to reduce risk factors and protect the heart from ischemia and reperfusion injury. Though the cardioprotective effects of exercise are well-documented, underlying mechanisms are not well understood. This review highlights recent findings and focuses on cardiac factors with emphasis on K+ channel control of the action potential duration (APD), β-adrenergic and adenosine regulation of cardiomyocyte function, and mitochondrial Ca2+ regulation. TRN-induced prolongation and shortening of the APD at low and high activation rates, respectively, is discussed in the context of a reduced response of the sarcolemma delayed rectifier potassium channel (IK) and increased content and activation of the sarcolemma KATP channel. A proposed mechanism underlying the latter is presented, including the phosphatidylinositol-3kinase/protein kinase B pathway. TRN induced increases in cardiomyocyte contractility and the response to adrenergic agonists are discussed. The TRN-induced protection from reperfusion injury is highlighted by the increased content and activation of the sarcolemma KATP channel and the increased phosphorylated glycogen synthase kinase-3β, which aid in preventing mitochondrial Ca2+ overload and mitochondria-triggered apoptosis. Finally, a brief section is presented on the increased incidences of atrial fibrillation associated with age and in life-long exercisers.
Collapse
Affiliation(s)
- Robert H Fitts
- Biological Sciences, Marquette University, Milwaukee, United States
| | - Xinrui Wang
- Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
| | - Wai-Meng Kwok
- Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
- Anesthesiology, Medical College of Wisconsin, Milwaukee, United States
- Cancer Center, Medical College of Wisconsin, Milwaukee, United States
| | - Amadou K S Camara
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
- Anesthesiology, Medical College of Wisconsin, Milwaukee, United States
- Cancer Center, Medical College of Wisconsin, Milwaukee, United States
- Physiology, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
3
|
Yu T, Gao Q, Zhang G, Li T, Liu X, Li C, Zheng L, Sun X, Wu J, Cao H, Bi F, Wang R, Liang H, Li X, Zhou Y, Lv L, Shan H. lncRNA Gm20257 alleviates pathological cardiac hypertrophy by modulating the PGC-1α-mitochondrial complex IV axis. Front Med 2024; 18:664-677. [PMID: 38926249 DOI: 10.1007/s11684-024-1065-7] [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: 09/11/2023] [Accepted: 01/17/2024] [Indexed: 06/28/2024]
Abstract
Pathological cardiac hypertrophy, a major contributor to heart failure, is closely linked to mitochondrial function. The roles of long noncoding RNAs (lncRNAs), which regulate mitochondrial function, remain largely unexplored in this context. Herein, a previously unknown lncRNA, Gm20257, was identified. It markedly increased under hypertrophic stress in vivo and in vitro. The suppression of Gm20257 by using small interfering RNAs significantly induced cardiomyocyte hypertrophy. Conversely, the overexpression of Gm20257 through plasmid transfection or adeno-associated viral vector-9 mitigated angiotensin II-induced hypertrophic phenotypes in neonatal mouse ventricular cells or alleviated cardiac hypertrophy in a mouse TAC model respectively, thus restoring cardiac function. Importantly, Gm20257 restored mitochondrial complex IV level and enhanced mitochondrial function. Bioinformatics prediction showed that Gm20257 had a high binding score with peroxisome proliferator-activated receptor coactivator-1 (PGC-1α), which could increase mitochondrial complex IV. Subsequently, Western blot analysis results revealed that Gm20257 substantially affected the expression of PGC-1α. Further analyses through RNA immunoprecipitation and immunoblotting following RNA pull-down indicated that PGC-1α was a direct downstream target of Gm20257. This interaction was demonstrated to rescue the reduction of mitochondrial complex IV induced by hypertrophic stress and promote the generation of mitochondrial ATP. These findings suggest that Gm20257 improves mitochondrial function through the PGC-1α-mitochondrial complex IV axis, offering a novel approach for attenuating pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Tong Yu
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Qiang Gao
- Department of Physiology, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Guofang Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Tianyu Li
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiaoshan Liu
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Chao Li
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Lan Zheng
- Department of Physiology, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China
| | - Xiang Sun
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jianbo Wu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Huiying Cao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Fangfang Bi
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ruifeng Wang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Haihai Liang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xuelian Li
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yuhong Zhou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Lifang Lv
- Department of Physiology, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China.
- The Center of Functional Experiment Teaching, School of Basic Medicine, Harbin Medical University, Harbin, 150081, China.
| | - Hongli Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, China.
| |
Collapse
|
4
|
El-Meanawy SK, Dooge H, Wexler AC, Kosmach AC, Serban L, Santos EA, Alvarado FJ, Hacker TA, Ramratnam M. Overexpression of a Short Sulfonylurea Splice Variant Increases Cardiac Glucose Uptake and Uncouples Mitochondria by Regulating ROMK Activity. Life (Basel) 2023; 13:1015. [PMID: 37109544 PMCID: PMC10146620 DOI: 10.3390/life13041015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The mitochondrial splice variant of the sulfonylurea receptor (SUR2A-55) is associated with protection from myocardial ischemia-reperfusion (IR) injury, increased mitochondrial ATP sensitive K+ channel activity (mitoKATP) and altered glucose metabolism. While mitoKATP channels composed of CCDC51 and ABCB8 exist, the mitochondrial K+ pore regulated by SUR2A-55 is unknown. We explored whether SUR2A-55 regulates ROMK to form an alternate mitoKATP. We assessed glucose uptake in mice overexpressing SUR2A-55 (TGSUR2A-55) compared with WT mice during IR injury. We then examined the expression level of ROMK and the effect of ROMK modulation on mitochondrial membrane potential (Δψm) in WT and TGSUR2A-55 mice. TGSUR2A-55 had increased glucose uptake compared to WT mice during IR injury. The expression of ROMK was similar in WT compared to TGSUR2A-55 mice. ROMK inhibition hyperpolarized resting cardiomyocyte Δψm from TGSUR2A-55 mice but not from WT mice. In addition, TGSUR2A-55 and ROMK inhibitor treated WT isolated cardiomyocytes had enhanced mitochondrial uncoupling. ROMK inhibition blocked diazoxide induced Δψm depolarization and prevented preservation of Δψm from FCCP perfusion in WT and to a lesser degree TGSUR2A-55 mice. In conclusion, cardio-protection from SUR2A-55 is associated with ROMK regulation, enhanced mitochondrial uncoupling and increased glucose uptake.
Collapse
Affiliation(s)
- Sarah K. El-Meanawy
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Holly Dooge
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Allison C. Wexler
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Anna C. Kosmach
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
| | - Lara Serban
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
| | - Elizabeth A. Santos
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
| | - Francisco J. Alvarado
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Timothy A. Hacker
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Mohun Ramratnam
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (S.K.E.-M.)
- Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| |
Collapse
|
5
|
Werbner B, Tavakoli-Rouzbehani OM, Fatahian AN, Boudina S. The dynamic interplay between cardiac mitochondrial health and myocardial structural remodeling in metabolic heart disease, aging, and heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2023; 3:9. [PMID: 36742465 PMCID: PMC9894375 DOI: 10.20517/jca.2022.42] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This review provides a holistic perspective on the bi-directional relationship between cardiac mitochondrial dysfunction and myocardial structural remodeling in the context of metabolic heart disease, natural cardiac aging, and heart failure. First, a review of the physiologic and molecular drivers of cardiac mitochondrial dysfunction across a range of increasingly prevalent conditions such as metabolic syndrome and cardiac aging is presented, followed by a general review of the mechanisms of mitochondrial quality control (QC) in the heart. Several important mechanisms by which cardiac mitochondrial dysfunction triggers or contributes to structural remodeling of the heart are discussed: accumulated metabolic byproducts, oxidative damage, impaired mitochondrial QC, and mitochondrial-mediated cell death identified as substantial mechanistic contributors to cardiac structural remodeling such as hypertrophy and myocardial fibrosis. Subsequently, the less studied but nevertheless important reverse relationship is explored: the mechanisms by which cardiac structural remodeling feeds back to further alter mitochondrial bioenergetic function. We then provide a condensed pathogenesis of several increasingly important clinical conditions in which these relationships are central: diabetic cardiomyopathy, age-associated declines in cardiac function, and the progression to heart failure, with or without preserved ejection fraction. Finally, we identify promising therapeutic opportunities targeting mitochondrial function in these conditions.
Collapse
Affiliation(s)
- Benjamin Werbner
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Amir Nima Fatahian
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
6
|
Aggarwal R, Potel KN, McFalls EO, Butterick TA, Kelly RF. Novel Therapeutic Approaches Enhance PGC1-alpha to Reduce Oxidant Stress-Inflammatory Signaling and Improve Functional Recovery in Hibernating Myocardium. Antioxidants (Basel) 2022; 11:2155. [PMID: 36358527 PMCID: PMC9686496 DOI: 10.3390/antiox11112155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/29/2022] [Accepted: 10/30/2022] [Indexed: 09/02/2023] Open
Abstract
Ischemic heart disease affects millions of people around the world. Current treatment options, including coronary artery bypass grafting, do not result in full functional recovery, highlighting the need for novel adjunctive therapeutic approaches. Hibernation describes the myocardial response to prolonged ischemia and involves a set of complex cytoprotective metabolic and functional adaptations. PGC1-alpha, a key regulator of mitochondrial energy metabolism and inhibitor of oxidant-stress-inflammatory signaling, is known to be downregulated in hibernating myocardium. PGC1-alpha is a critical component of cellular stress responses and links cellular metabolism with inflammation in the ischemic heart. While beneficial in the acute setting, a chronic state of hibernation can be associated with self-perpetuating oxidant stress-inflammatory signaling which leads to tissue injury. It is likely that incomplete functional recovery following revascularization of chronically ischemic myocardium is due to persistence of metabolic changes as well as prooxidant and proinflammatory signaling. Enhancement of PGC1-alpha signaling has been proposed as a possible way to improve functional recovery in patients with ischemic heart disease. Adjunctive mesenchymal stem cell therapy has been shown to induce PGC1-alpha signaling in hibernating myocardium and could help improve clinical outcomes for patients undergoing bypass surgery.
Collapse
Affiliation(s)
- Rishav Aggarwal
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Koray N. Potel
- School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK
| | - Edward O. McFalls
- Division of Cardiology, Richmond VA Medical Center, Richmond, VA 23249-4915, USA
| | - Tammy A. Butterick
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Research, Center for Veterans Research and Education, Minneapolis, MN 55417, USA
| | - Rosemary F. Kelly
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
7
|
Chen L, Qin Y, Liu B, Gao M, Li A, Li X, Gong G. PGC-1 α-Mediated Mitochondrial Quality Control: Molecular Mechanisms and Implications for Heart Failure. Front Cell Dev Biol 2022; 10:871357. [PMID: 35721484 PMCID: PMC9199988 DOI: 10.3389/fcell.2022.871357] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/27/2022] [Indexed: 12/26/2022] Open
Abstract
Mitochondria with structural and functional integrity are essential for maintaining mitochondrial function and cardiac homeostasis. It is involved in the pathogenesis of many diseases. Peroxisome proliferator-activated receptor γ coactivator 1 α (PGC-1α), acted as a transcriptional cofactor, is abundant in the heart, which modulates mitochondrial biogenesis and mitochondrial dynamics and mitophagy to sustain a steady-state of mitochondria. Cumulative evidence suggests that dysregulation of PGC-1α is closely related to the onset and progression of heart failure. PGC-1α deficient-mice can lead to worse cardiac function under pressure overload compared to sham. Here, this review mainly focuses on what is known about its regulation in mitochondrial functions, as well as its crucial role in heart failure.
Collapse
Affiliation(s)
- Lei Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuan Qin
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Department of Pharmacy, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Bilin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Meng Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Anqi Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xue Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Guohua Gong
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
8
|
Arrell DK, Park S, Yamada S, Alekseev AE, Garmany A, Jeon R, Vuckovic I, Lindor JZ, Terzic A. K ATP channel dependent heart multiome atlas. Sci Rep 2022; 12:7314. [PMID: 35513538 PMCID: PMC9072320 DOI: 10.1038/s41598-022-11323-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/21/2022] [Indexed: 11/09/2022] Open
Abstract
Plasmalemmal ATP sensitive potassium (KATP) channels are recognized metabolic sensors, yet their cellular reach is less well understood. Here, transgenic Kir6.2 null hearts devoid of the KATP channel pore underwent multiomics surveillance and systems interrogation versus wildtype counterparts. Despite maintained organ performance, the knockout proteome deviated beyond a discrete loss of constitutive KATP channel subunits. Multidimensional nano-flow liquid chromatography tandem mass spectrometry resolved 111 differentially expressed proteins and their expanded network neighborhood, dominated by metabolic process engagement. Independent multimodal chemometric gas and liquid chromatography mass spectrometry unveiled differential expression of over one quarter of measured metabolites discriminating the Kir6.2 deficient heart metabolome. Supervised class analogy ranking and unsupervised enrichment analysis prioritized nicotinamide adenine dinucleotide (NAD+), affirmed by extensive overrepresentation of NAD+ associated circuitry. The remodeled metabolome and proteome revealed functional convergence and an integrated signature of disease susceptibility. Deciphered cardiac patterns were traceable in the corresponding plasma metabolome, with tissue concordant plasma changes offering surrogate metabolite markers of myocardial latent vulnerability. Thus, Kir6.2 deficit precipitates multiome reorganization, mapping a comprehensive atlas of the KATP channel dependent landscape.
Collapse
Affiliation(s)
- D Kent Arrell
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Sungjo Park
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Satsuki Yamada
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Division of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Alexey E Alekseev
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Institute of Theoretical and Experimental Biophysics, Russian Academy of Science, Pushchino, Moscow Region, Russia
| | - Armin Garmany
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ryounghoon Jeon
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ivan Vuckovic
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Metabolomics Core, Mayo Clinic, Rochester, MN, USA
| | - Jelena Zlatkovic Lindor
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Andre Terzic
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA. .,Marriott Family Comprehensive Cardiac Regenerative Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA. .,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA. .,Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Zhang F, Zhou GH, An Q, Yang J, Wang YH, Zhu JQ, Jin SN, Wen JF. Decreased gene expression of K ACh and K ATP channels in hyperthyroid rabbit atria. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2022; 15:145-151. [PMID: 35414842 PMCID: PMC8986473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 02/12/2022] [Indexed: 06/14/2023]
Abstract
Cardiac hypertrophy is a common myocardial structural abnormality which may cause heart failure. Many studies have shown that cardiac hypertrophy can be induced by hyperthyroidism. Ligand-gated potassium channels have been reported to be involved in various biological processes in the cardiovascular system, such as GPCR coupled KACh and metabolism sensor KATP channel. It is unclear whether the gene expression of KACh and KATP was altered in hyperthyroid rabbit atria. We aimed to investigate the expression of KACh and KATP genes in rabbit atria in our experimental model. We established an effective hyperthyroidism-induced cardiac hypertrophy animal model through an injection of T4. H&E staining and RT-PCR were used to observe the histomorphological damages and alteration of gene expression. The results showed that the heart weight, heart rate significantly increased in T4-treated rabbits. The systolic pressure increased from 115.60 mmHg to 152.6 mmHg in T4-treated rabbits. The expression of KACh and KATP genes was decreased in the atria of hyperthyroidism-induced cardiac hypertrophied rabbits. These findings indicated that the decreased gene expression of KACh and KATP may be related to hyperthyroidism-induced cardiac hypertrophy and atrial fibrillation.
Collapse
Affiliation(s)
- Feng Zhang
- School of Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical ScienceJinan 250117, Shandong, China
| | - Guang Hai Zhou
- School of Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical ScienceJinan 250117, Shandong, China
| | - Qi An
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical SciencesTaian 271000, Shandong, China
| | - Jie Yang
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical SciencesTaian 271000, Shandong, China
| | - Yu Hao Wang
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical SciencesTaian 271000, Shandong, China
| | - Jia Quan Zhu
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical SciencesTaian 271000, Shandong, China
| | - Song Nan Jin
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical SciencesTaian 271000, Shandong, China
| | - Jin Fu Wen
- School of Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical ScienceJinan 250117, Shandong, China
| |
Collapse
|
10
|
Wang J, Bai J, Duan P, Wang H, Li Y, Zhu Q. Kir6.1 improves cardiac dysfunction in diabetic cardiomyopathy via the AKT-FoxO1 signalling pathway. J Cell Mol Med 2021; 25:3935-3949. [PMID: 33547878 PMCID: PMC8051713 DOI: 10.1111/jcmm.16346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/07/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Previous studies have shown that the expression of inwardly rectifying potassium channel 6.1 (Kir6.1) in heart mitochondria is significantly reduced in type 1 diabetes. However, whether its expression and function are changed and what role it plays in type 2 diabetic cardiomyopathy (DCM) have not been reported. This study investigated the role and mechanism of Kir6.1 in DCM. We found that the cardiac function and the Kir6.1 expression in DCM mice were decreased. We generated mice overexpressing or lacking Kir6.1 gene specifically in the heart. Kir6.1 overexpression improved cardiac dysfunction in DCM. Cardiac‐specific Kir6.1 knockout aggravated cardiac dysfunction. Kir6.1 regulated the phosphorylation of AKT and Foxo1 in DCM. We further found that Kir6.1 overexpression also improved cardiomyocyte dysfunction and up‐regulated the phosphorylation of AKT and FoxO1 in neonatal rat ventricular cardiomyocytes with insulin resistance. Furthermore, FoxO1 activation down‐regulated the expression of Kir6.1 and decreased the mitochondrial membrane potential (ΔΨm) in cardiomyocytes. FoxO1 inactivation up‐regulated the expression of Kir6.1 and increased the ΔΨm in cardiomyocytes. Chromatin immunoprecipitation assay demonstrated that the Kir6.1 promoter region contains a functional FoxO1‐binding site. In conclusion, Kir6.1 improves cardiac dysfunction in DCM, probably through the AKT‐FoxO1 signalling pathway.
Collapse
Affiliation(s)
- Jinxin Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China.,Department of Geriatric Cardiology, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jing Bai
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Peng Duan
- Department of Cardiology, Chinese PLA No. 371 Hospital, Henan, China
| | - Hao Wang
- Department of Geriatric Cardiology, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yang Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qinglei Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
11
|
Yu W, Chen C, Cheng J. The role and molecular mechanism of FoxO1 in mediating cardiac hypertrophy. ESC Heart Fail 2020; 7:3497-3504. [PMID: 33089967 PMCID: PMC7755013 DOI: 10.1002/ehf2.13065] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 02/05/2023] Open
Abstract
Cardiac hypertrophy can lead to heart failure and cardiovascular events and has become a research hotspot in the field of cardiovascular disease. Despite extensive and in-depth research, the pathogenesis of cardiac hypertrophy is far from being fully understood. Increasing evidence has shown that the transcription factor forkhead box protein O 1 (FoxO1) is closely related to the occurrence and development of cardiac hypertrophy. This review summarizes the current literature on the role and molecular mechanism of FoxO1 in cardiac hypertrophy. We searched the database MEDLINE via PubMed for available evidence on the effect of FoxO1 on cardiac hypertrophy. FoxO1 has many effects on multiple diseases, including cardiovascular diseases, diabetes, cancer, aging, and stem cell activity. Recent studies have shown that FoxO1 plays a critical role in the development of cardiac hypertrophy. Evidence for this relationship includes the following. (i) FoxO1 can regulate cardiac growth/protein synthesis, calcium homeostasis, cell apoptosis, and autophagy and (ii) is controlled by several upstream signalling molecules (e.g. phosphatidylinositol 3-kinase/Akt, AMP-activated protein kinase, and sirtuins) and regulates many downstream transcription proteins (e.g. ubiquitin ligases muscle RING finger 1/muscle atrophy F-box, calcineurin/nuclear factor of activated T cells, and microRNAs). In response to stress or external stimulation (e.g. low energy, oxidative stress, or growth factor signalling), FoxO1 undergoes post-translational modification and transfers from the cytoplasm to nucleus, thus regulating the expression of a series of target genes in myocardium that are involved in cardiac growth/protein synthesis, calcium homeostasis, cell apoptosis, and autophagy. (iii) Finally, targeted regulation of FoxO1 is an effective method of intervening in myocardial hypertrophy. The information reviewed here should be significant for understanding the roles of FoxO1 in cardiac hypertrophy and should contribute to the design of further studies related to FoxO1 and the hypertrophic response. It should also shed light on a potential treatment for cardiac hypertrophy.
Collapse
Affiliation(s)
- Wei Yu
- Department of Internal MedicineXiang'an Hospital of Xiamen UniversityXiamen361102China
| | - Chunjuan Chen
- Department of CardiologySecond Affiliated Hospital of Shantou University Medical College69 Dong Xia North RoadShantou515041China
| | - Jidong Cheng
- Department of Internal MedicineXiang'an Hospital of Xiamen UniversityXiamen361102China
| |
Collapse
|
12
|
Yan D, Cai Y, Luo J, Liu J, Li X, Ying F, Xie X, Xu A, Ma X, Xia Z. FOXO1 contributes to diabetic cardiomyopathy via inducing imbalanced oxidative metabolism in type 1 diabetes. J Cell Mol Med 2020; 24:7850-7861. [PMID: 32450616 PMCID: PMC7348139 DOI: 10.1111/jcmm.15418] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/24/2020] [Accepted: 05/06/2020] [Indexed: 02/06/2023] Open
Abstract
Forkhead box protein O1 (FOXO1), a nuclear transcription factor, is preferably activated in the myocardium of diabetic mice. However, its role and mechanism in the development of diabetic cardiomyopathy in non-obese insulin-deficient diabetes are unclear. We hypothesized that cardiac FOXO1 over-activation was attributable to the imbalanced myocardial oxidative metabolism and mitochondrial and cardiac dysfunction in type 1 diabetes. FOXO1-selective inhibitor AS1842856 was administered to streptozotocin-induced diabetic (D) rats, and cardiac functions, mitochondrial enzymes PDK4 and CPT1 and mitochondrial function were assessed. Primary cardiomyocytes isolated from non-diabetic control (C) and D rats were treated with or without 1 µM AS1842856 and underwent Seahorse experiment to determine the effects of glucose, palmitate and pyruvate on cardiomyocyte bioenergetics. The results showed diabetic hearts displayed elevated FOXO1 nuclear translocation, concomitant with cardiac and mitochondrial dysfunction (manifested as elevated mtROS level and reduced mitochondrial membrane potential) and increased cell apoptosis (all P < .05, D vs C). Diabetic myocardium showed impaired glycolysis, glucose oxidation and elevated fatty acid oxidation and enhanced PDK4 and CPT1 expression. AS1842856 attenuated or prevented all these changes except for glycolysis. We concluded that FOXO1 activation, through stimulating PDK4 and CPT1, shifts substrate selection from glucose to fatty acid and causes mitochondrial and cardiac dysfunction.
Collapse
Affiliation(s)
- Dan Yan
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China.,Diabetes Center, Shenzhen University, Shenzhen, China
| | - Yin Cai
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Jierong Luo
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Jingjin Liu
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Xia Li
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Ying
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Xiang Xie
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaosong Ma
- Diabetes Center, Shenzhen University, Shenzhen, China
| | - Zhengyuan Xia
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
13
|
Oka SI, Sabry AD, Cawley KM, Warren JS. Multiple Levels of PGC-1α Dysregulation in Heart Failure. Front Cardiovasc Med 2020; 7:2. [PMID: 32083094 PMCID: PMC7002390 DOI: 10.3389/fcvm.2020.00002] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022] Open
Abstract
Metabolic adaption is crucial for the heart to sustain its contractile activity under various physiological and pathological conditions. At the molecular level, the changes in energy demand impinge on the expression of genes encoding for metabolic enzymes. Among the major components of an intricate transcriptional circuitry, peroxisome proliferator-activated receptor γ coactivator 1 alpha (PGC-1α) plays a critical role as a metabolic sensor, which is responsible for the fine-tuning of transcriptional responses to a plethora of stimuli. Cumulative evidence suggests that energetic impairment in heart failure is largely attributed to the dysregulation of PGC-1α. In this review, we summarize recent studies revealing how PGC-1α is regulated by a multitude of mechanisms, operating at different regulatory levels, which include epigenetic regulation, the expression of variants, post-transcriptional inhibition, and post-translational modifications. We further discuss how the PGC-1α regulatory cascade can be impaired in the failing heart.
Collapse
Affiliation(s)
- Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Amira D Sabry
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Keiko M Cawley
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Junco S Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States.,Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States.,Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
14
|
Single-Channel Properties of the ROMK-Pore-Forming Subunit of the Mitochondrial ATP-Sensitive Potassium Channel. Int J Mol Sci 2019; 20:ijms20215323. [PMID: 31731540 PMCID: PMC6862428 DOI: 10.3390/ijms20215323] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/11/2022] Open
Abstract
An increased flux of potassium ions into the mitochondrial matrix through the ATP-sensitive potassium channel (mitoKATP) has been shown to provide protection against ischemia-reperfusion injury. Recently, it was proposed that the mitochondrial-targeted isoform of the renal outer medullary potassium channel (ROMK) protein creates a pore-forming subunit of mitoKATP in heart mitochondria. Our research focuses on the properties of mitoKATP from heart-derived H9c2 cells. For the first time, we detected single-channel activity and describe the pharmacology of mitoKATP in the H9c2 heart-derived cells. The patch-clamping of mitoplasts from wild type (WT) and cells overexpressing ROMK2 revealed the existence of a potassium channel that exhibits the same basic properties previously attributed to mitoKATP. ROMK2 overexpression resulted in a significant increase of mitoKATP activity. The conductance of both channels in symmetric 150/150 mM KCl was around 97 ± 2 pS in WT cells and 94 ± 3 pS in cells overexpressing ROMK2. The channels were inhibited by 5-hydroxydecanoic acid (a mitoKATP inhibitor) and by Tertiapin Q (an inhibitor of both the ROMK-type channels and mitoKATP). Additionally, mitoKATP from cells overexpressing ROMK2 were inhibited by ATP/Mg2+ and activated by diazoxide. We used an assay based on proteinase K to examine the topology of the channel in the inner mitochondrial membrane and found that both termini of the protein localized to the mitochondrial matrix. We conclude that the observed activity of the channel formed by the ROMK protein corresponds to the electrophysiological and pharmacological properties of mitoKATP.
Collapse
|
15
|
Shang L, Weng X, Wang D, Yue W, Mernaugh R, Amarnath V, Weir EK, Dudley SC, Xu Y, Hou M, Chen Y. Isolevuglandin scavenger attenuates pressure overload-induced cardiac oxidative stress, cardiac hypertrophy, heart failure and lung remodeling. Free Radic Biol Med 2019; 141:291-298. [PMID: 31254620 DOI: 10.1016/j.freeradbiomed.2019.06.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 12/20/2022]
Abstract
Increased levels of reactive isolevuglandins (IsoLGs) are associated with vascular inflammation and hypertension, two important factors affect heart failure (HF) development. The role of IsoLGs in HF development is unknown. Here we studied the role of IsoLG scavenger 2-hydroxybenzylamine (2-HOBA) in transverse aortic constriction (TAC) induced heart failure. We observed that TAC caused a significant increase of IsoLG protein adducts in cardiac and lung tissues in mice. Both IsoLG scavenger 2-hydroxybenzylamine (2-HOBA) and its less reactive isomer 4-hydroxybenzylamine (4-HOBA) significantly attenuated the left ventricular (LV) and lung IsoLGs in mice after TAC. 2-HOBA and 4-HOBA attenuated TAC-induced LV hypertrophy, heart failure, and the increase of lung weight in mice, and also improved TAC-induced LV dysfunction. Moreover, both 2-HOBA and 4-HOBA effectively attenuated LV cardiomyocyte hypertrophy, lung inflammation, lung fibrosis. These findings suggest that methods to reduce IsoLGs may be useful for HF therapy.
Collapse
Affiliation(s)
- Linlin Shang
- Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyu Weng
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongzhi Wang
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenhui Yue
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ray Mernaugh
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
| | | | - E Kenneth Weir
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Samuel C Dudley
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Mingxiao Hou
- Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| | - Yingjie Chen
- Lillehei Heart Institute and Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
16
|
Bhat S, Chin A, Shirakabe A, Ikeda Y, Ikeda S, Zhai P, Hsu CP, Sayed D, Abdellatif M, Byun J, Schesing K, Tang F, Tian Y, Babu G, Ralda G, Warren JS, Cho J, Sadoshima J, Oka SI. Recruitment of RNA Polymerase II to Metabolic Gene Promoters Is Inhibited in the Failing Heart Possibly Through PGC-1α (Peroxisome Proliferator-Activated Receptor-γ Coactivator-1α) Dysregulation. Circ Heart Fail 2019; 12:e005529. [PMID: 30798619 PMCID: PMC6392084 DOI: 10.1161/circheartfailure.118.005529] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/29/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND Proper dynamics of RNA polymerase II, such as promoter recruitment and elongation, are essential for transcription. PGC-1α (peroxisome proliferator-activated receptor [PPAR]-γ coactivator-1α), also termed PPARGC1a, is a transcriptional coactivator that stimulates energy metabolism, and PGC-1α target genes are downregulated in the failing heart. However, whether the dysregulation of polymerase II dynamics occurs in PGC-1α target genes in heart failure has not been defined. METHODS AND RESULTS Chromatin immunoprecipitation-sequencing revealed that reduced promoter occupancy was a major form of polymerase II dysregulation on PGC-1α target metabolic gene promoters in the pressure-overload-induced heart failure model. PGC-1α-cKO (cardiac-specific PGC-1α knockout) mice showed phenotypic similarity to the pressure-overload-induced heart failure model in wild-type mice, such as contractile dysfunction and downregulation of PGC-1α target genes, even under basal conditions. However, the protein levels of PGC-1α were neither changed in the pressure-overload model nor in human failing hearts. Chromatin immunoprecipitation assays revealed that the promoter occupancy of polymerase II and PGC-1α was consistently reduced both in the pressure-overload model and PGC-1α-cKO mice. In vitro DNA binding assays using an endogenous PGC-1α target gene promoter sequence confirmed that PGC-1α recruits polymerase II to the promoter. CONCLUSIONS These results suggest that PGC-1α promotes the recruitment of polymerase II to the PGC-1α target gene promoters. Downregulation of PGC-1α target genes in the failing heart is attributed, in part, to a reduction of the PGC-1α occupancy and the polymerase II recruitment to the promoters, which might be a novel mechanism of metabolic perturbations in the failing heart.
Collapse
Affiliation(s)
- Santosh Bhat
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Adave Chin
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Akihiro Shirakabe
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Yoshiyuki Ikeda
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Shohei Ikeda
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Chiao-po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan
| | - Danish Sayed
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Maha Abdellatif
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Kevin Schesing
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Fan Tang
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Yimin Tian
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Gopal Babu
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Guersom Ralda
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Junco S. Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112
| | - Jaeyeaon Cho
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea, 03722
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| | - Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical Health Sciences, Newark, NJ 07103
| |
Collapse
|
17
|
Lv J, Deng C, Jiang S, Ji T, Yang Z, Wang Z, Yang Y. Blossoming 20: The Energetic Regulator's Birthday Unveils its Versatility in Cardiac Diseases. Am J Cancer Res 2019; 9:466-476. [PMID: 30809287 PMCID: PMC6376194 DOI: 10.7150/thno.29130] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/03/2018] [Indexed: 12/20/2022] Open
Abstract
The peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α) was first identified in 1998 as a PGC-1 family member that regulates adaptive thermogenesis and mitochondrial function following cold exposure in brown adipose tissue. The PGC-1 family has drawn widespread attention over the past two decades as the energetic regulator. We recently summarized a review regarding PGC-1 signaling pathway and its mechanisms in cardiac metabolism. In this review, we elaborate upon the PGC-1 signaling network and highlight the recent progress of its versatile roles in cardiac diseases, including myocardial hypertrophy, peripartum and diabetic cardiomyopathy, and heart failure. The information reviewed here may be useful in future studies, which may increase the potential of this energetic regulator as a therapeutic target.
Collapse
|
18
|
Tinker A, Aziz Q, Li Y, Specterman M. ATP‐Sensitive Potassium Channels and Their Physiological and Pathophysiological Roles. Compr Physiol 2018; 8:1463-1511. [DOI: 10.1002/cphy.c170048] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
19
|
Strutynskyi RB, Voronkov LG, Nagibin VS, Mazur ID, Stroy D, Dosenko VE. Changes of the echocardiographic parameters in chronic heart failure patients with Ile337val, Glu23lys, and Ser1369ala polymorphisms of genes encoding the ATP‐sensitive potassium channels subunits in the Ukrainian population. Ann Hum Genet 2018; 82:272-279. [DOI: 10.1111/ahg.12250] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/29/2017] [Accepted: 03/01/2018] [Indexed: 11/28/2022]
Affiliation(s)
- R. B. Strutynskyi
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine Kyiv Ukraine
| | - L. G. Voronkov
- Department of Heart Failure State Institution «National scientific center «M.D. Strazhesko Institute of Cardiology» National Academy of Medical Sciences of Ukraine Kyiv Ukraine
| | - V. S. Nagibin
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine Kyiv Ukraine
| | - I. D. Mazur
- Department of Heart Failure State Institution «National scientific center «M.D. Strazhesko Institute of Cardiology» National Academy of Medical Sciences of Ukraine Kyiv Ukraine
| | - D. Stroy
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine Kyiv Ukraine
| | - V. E. Dosenko
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine Kyiv Ukraine
| |
Collapse
|
20
|
Mutikainen M, Tuomainen T, Naumenko N, Huusko J, Smirin B, Laidinen S, Kokki K, Hynynen H, Ylä-Herttuala S, Heinäniemi M, Ruas JL, Tavi P. Peroxisome proliferator-activated receptor-γ coactivator 1 α1 induces a cardiac excitation-contraction coupling phenotype without metabolic remodelling. J Physiol 2017; 594:7049-7071. [PMID: 27716916 DOI: 10.1113/jp272847] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/21/2016] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS Transcriptional co-activator PGC-1α1 has been shown to regulate energy metabolism and to mediate metabolic adaptations in pathological and physiological cardiac hypertrophy but other functional implications of PGC-1α1 expression are not known. Transgenic PGC-1α1 overexpression within the physiological range in mouse heart induces purposive changes in contractile properties, electrophysiology and calcium signalling but does not induce substantial metabolic remodelling. The phenotype of the PGC-1α1 transgenic mouse heart recapitulates most of the functional modifications usually associated with the exercise-induced heart phenotype, but does not protect the heart against load-induced pathological hypertrophy. Transcriptional effects of PGC-1α1 show clear dose-dependence with diverse changes in genes in circadian clock, heat shock, excitability, calcium signalling and contraction pathways at low overexpression levels, while metabolic genes are recruited at much higher PGC-1α1 expression levels. These results imply that the physiological role of PGC-1α1 is to promote a beneficial excitation-contraction coupling phenotype in the heart. ABSTRACT The transcriptional coactivator PGC-1α1 has been identified as a central factor mediating metabolic adaptations of the heart. However, to what extent physiological changes in PGC-1α1 expression levels actually contribute to the functional adaptation of the heart is still mostly unresolved. The aim of this study was to characterize the transcriptional and functional effects of physiologically relevant, moderate PGC-1α1 expression in the heart. In vivo and ex vivo physiological analysis shows that expression of PGC-1α1 within a physiological range in mouse heart does not induce the expected metabolic alterations, but instead induces a unique excitation-contraction (EC) coupling phenotype recapitulating features typically seen in physiological hypertrophy. Transcriptional screening of PGC-1α1 overexpressing mouse heart and myocyte cultures with higher, acute adenovirus-induced PGC-1α1 expression, highlights PGC-1α1 as a transcriptional coactivator with a number of binding partners in various pathways (such as heat shock factors and the circadian clock) through which it acts as a pleiotropic transcriptional regulator in the heart, to both augment and repress the expression of its target genes in a dose-dependent fashion. At low levels of overexpression PGC-1α1 elicits a diverse transcriptional response altering the expression state of circadian clock, heat shock, excitability, calcium signalling and contraction pathways, while metabolic targets of PGC-1α1 are recruited at higher PGC-1α1 expression levels. Together these findings demonstrate that PGC-1α1 elicits a dual effect on cardiac transcription and phenotype. Further, our results imply that the physiological role of PGC-1α1 is to promote a beneficial EC coupling phenotype in the heart.
Collapse
Affiliation(s)
- Maija Mutikainen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tomi Tuomainen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nikolay Naumenko
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jenni Huusko
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Boris Smirin
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Svetlana Laidinen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Krista Kokki
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Heidi Hynynen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Pasi Tavi
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
21
|
Peterzan MA, Lygate CA, Neubauer S, Rider OJ. Metabolic remodeling in hypertrophied and failing myocardium: a review. Am J Physiol Heart Circ Physiol 2017. [PMID: 28646030 DOI: 10.1152/ajpheart.00731.2016] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The energy starvation hypothesis proposes that maladaptive metabolic remodeling antedates, initiates, and maintains adverse contractile dysfunction in heart failure (HF). Better understanding of the cardiac metabolic phenotype and metabolic signaling could help identify the role metabolic remodeling plays within HF and the conditions known to transition toward HF, including "pathological" hypertrophy. In this review, we discuss metabolic phenotype and metabolic signaling in the contexts of pathological hypertrophy and HF. We discuss the significance of alterations in energy supply (substrate utilization, oxidative capacity, and phosphotransfer) and energy sensing using observations from human and animal disease models and models of manipulated energy supply/sensing. We aim to provide ways of thinking about metabolic remodeling that center around metabolic flexibility, capacity (reserve), and efficiency rather than around particular substrate preferences or transcriptomic profiles. We show that maladaptive metabolic remodeling takes multiple forms across multiple energy-handling domains. We suggest that lack of metabolic flexibility and reserve (substrate, oxidative, and phosphotransfer) represents a final common denominator ultimately compromising efficiency and contractile reserve in stressful contexts.
Collapse
Affiliation(s)
- Mark A Peterzan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Oliver J Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
22
|
Preservation of CGRP in myocardium attenuates development of cardiac dysfunction in diabetic rats. Int J Cardiol 2016; 220:226-34. [PMID: 27389446 DOI: 10.1016/j.ijcard.2016.06.092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 06/01/2016] [Accepted: 06/21/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Calcitonin gene-related peptide (CGRP) plays an important role in cardiovascular regulation, which was found reduced in serum of diabetic patients. To test the hypothesis that lack of CGRP in myocardium is associated with diabetic cardiac dysfunction, which may be improved by preservation of CGRP in diabetic rats. METHODS AND RESULTS Diabetes was induced in male Sprague-Dawley rats by streptozotocin (50mg/kg). Two groups of the diabetic rats, one fed with standard laboratory chew and another with the laboratory food plus hot pepper (containing 0.0174% of capsaicin), to stimulate production and release of CGRP. Cardiac functions were evaluated by measurements of intraventricular pressures after 8weeks of development of diabetes. Transient receptor potential vanilloid type 1 (TRPV1), CGRP, β1-adreneregic receptor and norepinephrine were analyzed. Significantly lower levels of TRPV1 and CGRP were detected in the thoracic dorsal root ganglia (DRG) and myocardium of the diabetic animals, along with significant decline in left ventricular systolic pressure (by 24%) and heart rate (by 25%) and increase of the end-diastolic pressure (by 83%) with obvious reduction of CGRP in the DRG, by 41%, the myocardium (by 30%) and the serum (by 20%). The cardiac performance, the TRPV1 and the CGRP in the diabetic animals fed with hot pepper were well preserved. No any significant change in β1-adreneregic receptor and norepinephrine was detected. CONCLUSION The findings may suggest a novel mechanism underlying diabetic cardiac dysfunctions via impairing TRPV1-CGRP pathway in myocardium. Preservation of the TRPV1-CGRP mechanism may prevent the development of cardiac dysfunction in diabetes.
Collapse
|
23
|
Nichols CG. Adenosine Triphosphate-Sensitive Potassium Currents in Heart Disease and Cardioprotection. Card Electrophysiol Clin 2016; 8:323-35. [PMID: 27261824 DOI: 10.1016/j.ccep.2016.01.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The subunit makeup of the family of adenosine triphosphate-sensitive potassium channel (KATP) channels is more complex and labile than thought. The growing association of Kir6.1 and SUR2 variants with specific cardiovascular electrical and contractile derangements and the clear association with Cantu syndrome establish the importance of appropriate activity in normal function of the heart and vasculature. Further studies of such patients will reveal new mutations in KATP subunits and perhaps in proteins that regulate KATP synthesis, trafficking, or location, all of which may ultimately benefit therapeutically from the unique pharmacology of KATP channels.
Collapse
Affiliation(s)
- Colin G Nichols
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO 63110, USA.
| |
Collapse
|
24
|
Abstract
KATP channels are integral to the functions of many cells and tissues. The use of electrophysiological methods has allowed for a detailed characterization of KATP channels in terms of their biophysical properties, nucleotide sensitivities, and modification by pharmacological compounds. However, even though they were first described almost 25 years ago (Noma 1983, Trube and Hescheler 1984), the physiological and pathophysiological roles of these channels, and their regulation by complex biological systems, are only now emerging for many tissues. Even in tissues where their roles have been best defined, there are still many unanswered questions. This review aims to summarize the properties, molecular composition, and pharmacology of KATP channels in various cardiovascular components (atria, specialized conduction system, ventricles, smooth muscle, endothelium, and mitochondria). We will summarize the lessons learned from available genetic mouse models and address the known roles of KATP channels in cardiovascular pathologies and how genetic variation in KATP channel genes contribute to human disease.
Collapse
Affiliation(s)
- Monique N Foster
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - William A Coetzee
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| |
Collapse
|
25
|
Liu Z, Cai H, Dang Y, Qiu C, Wang J. Adenosine triphosphate-sensitive potassium channels and cardiomyopathies (Review). Mol Med Rep 2015; 13:1447-54. [PMID: 26707080 DOI: 10.3892/mmr.2015.4714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 06/05/2015] [Indexed: 11/06/2022] Open
Abstract
Cardiomyopathies have been indicated to be one of the leading causes of heart failure. Though it was indicated that genetic defects, viral infection and trace element deficiency were among the causes of cardiomyopathy, the etiology has remained to be fully elucidated. Cardiomyocytes require large amounts of energy to maintain their normal biological functions. Adenosine triphosphate-sensitive potassium channels (KATP), composed of inward-rectifier potassium ion channel and sulfonylurea receptor subunits, are present on the cell surface and mitochondrial membrane of cardiac muscle cells. As metabolic sensors sensitive to changes in intracellular energy levels, KATP adapt electrical activities to metabolic challenges, maintaining normal biological functions of myocytes. It is implied that malfunctions, mutations and altered expression of KATP are associated with the pathogenesis of conditions including c hypertrophy, diabetes as well as dilated, ischemic and endemic cardiomyopathy. However, the current knowledge is only the tip of the iceberg and the roles of KATP in cardiomyopathies largely remain to be elucidated in future studies.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Hui Cai
- Department of Anesthesiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yonghui Dang
- College of Medicine and Forensics, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Chuan Qiu
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans 70112‑2705, LA, USA
| | - Junkui Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
26
|
Tinker A, Aziz Q, Thomas A. The role of ATP-sensitive potassium channels in cellular function and protection in the cardiovascular system. Br J Pharmacol 2014; 171:12-23. [PMID: 24102106 DOI: 10.1111/bph.12407] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/30/2013] [Accepted: 08/26/2013] [Indexed: 12/14/2022] Open
Abstract
ATP-sensitive potassium channels (K(ATP)) are widely distributed and present in a number of tissues including muscle, pancreatic beta cells and the brain. Their activity is regulated by adenine nucleotides, characteristically being activated by falling ATP and rising ADP levels. Thus, they link cellular metabolism with membrane excitability. Recent studies using genetically modified mice and genomic studies in patients have implicated K(ATP) channels in a number of physiological and pathological processes. In this review, we focus on their role in cellular function and protection particularly in the cardiovascular system.
Collapse
Affiliation(s)
- Andrew Tinker
- William Harvey Heart Centre, Barts and The London School of Medicine and Dentistry, London, UK
| | | | | |
Collapse
|
27
|
Liu X, Kwak D, Lu Z, Xu X, Fassett J, Wang H, Wei Y, Cavener DR, Hu X, Hall J, Bache RJ, Chen Y. Endoplasmic reticulum stress sensor protein kinase R-like endoplasmic reticulum kinase (PERK) protects against pressure overload-induced heart failure and lung remodeling. Hypertension 2014; 64:738-44. [PMID: 24958502 DOI: 10.1161/hypertensionaha.114.03811] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Studies have reported that development of congestive heart failure is associated with increased endoplasmic reticulum stress. Double stranded RNA-activated protein kinase R-like endoplasmic reticulum kinase (PERK) is a major transducer of the endoplasmic reticulum stress response and directly phosphorylates eukaryotic initiation factor 2α, resulting in translational attenuation. However, the physiological effect of PERK on congestive heart failure development is unknown. To study the effect of PERK on ventricular structure and function, we generated inducible cardiac-specific PERK knockout mice. Under unstressed conditions, cardiac PERK knockout had no effect on left ventricular mass, or its ratio to body weight, cardiomyocyte size, fibrosis, or left ventricular function. However, in response to chronic transverse aortic constriction, PERK knockout mice exhibited decreased ejection fraction, increased left ventricular fibrosis, enhanced cardiomyocyte apoptosis, and exacerbated lung remodeling in comparison with wild-type mice. PERK knockout also dramatically attenuated cardiac sarcoplasmic reticulum Ca(2+)-ATPase expression in response to aortic constriction. Our findings suggest that PERK is required to protect the heart from pressure overload-induced congestive heart failure.
Collapse
Affiliation(s)
- Xiaoyu Liu
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Dongmin Kwak
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Zhongbing Lu
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Xin Xu
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - John Fassett
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Huan Wang
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Yidong Wei
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Douglas R Cavener
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Xinli Hu
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Jennifer Hall
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Robert J Bache
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.)
| | - Yingjie Chen
- From the Department of Chinese Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China (X.L., Y.W.); Cardiovascular Division, Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis (X.L., D.K., Z.L., X.X., J.F., H.W., J.H., R.J.B., Y.C.); College of Life Science, University of Chinese Academy of Science, Beijing, China (Z.L.); Department of Biology and The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park (D.R.C.); and Institute of Molecular Medicine, Peking University, Beijing, China (X.H.).
| |
Collapse
|
28
|
Pereira RO, Wende AR, Crum A, Hunter D, Olsen CD, Rawlings T, Riehle C, Ward WF, Abel ED. Maintaining PGC-1α expression following pressure overload-induced cardiac hypertrophy preserves angiogenesis but not contractile or mitochondrial function. FASEB J 2014; 28:3691-702. [PMID: 24776744 DOI: 10.1096/fj.14-253823] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
During pathological hypertrophy, peroxisome proliferator-activated receptor coactivator 1α (PGC-1α) is repressed in concert with reduced mitochondrial oxidative capacity and fatty acid oxidation (FAO). We therefore sought to determine if maintaining or increasing PGC-1α levels in the context of pressure overload hypertrophy (POH) would preserve mitochondrial function and prevent contractile dysfunction. Pathological cardiac hypertrophy was induced using 4 wk of transverse aortic constriction (TAC) in mice overexpressing the human PGC-1α genomic locus via a bacterial artificial chromosome (TG) and nontransgenic controls (Cont). PGC-1α levels were increased by 40% in TG mice and were sustained following TAC. Although TAC-induced repression of FAO genes and oxidative phosphorylation (oxphos) genes was prevented in TG mice, mitochondrial function and ATP synthesis were equivalently impaired in Cont and TG mice after TAC. Contractile function was also equally impaired in Cont and TG mice following TAC, as demonstrated by decreased +dP/dt and ejection fraction and increased left ventricular developed pressure and end diastolic pressure. Conversely, capillary density was preserved, in concert with increased VEGF expression, while apoptosis and fibrosis were reduced in TG relative to Cont mice after TAC. Hence, sustaining physiological levels of PGC-1α expression following POH, while preserving myocardial vascularity, does not prevent mitochondrial and contractile dysfunction.
Collapse
Affiliation(s)
- Renata O Pereira
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA; Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA; and
| | - Adam R Wende
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA; and
| | - Ashley Crum
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA; and
| | - Douglas Hunter
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA; and
| | - Curtis D Olsen
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA; and
| | - Tenley Rawlings
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA; and
| | - Christian Riehle
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA; Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA; and
| | - Walter F Ward
- University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA; Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA; and
| |
Collapse
|
29
|
Effects of hypertension and exercise on cardiac proteome remodelling. BIOMED RESEARCH INTERNATIONAL 2014; 2014:634132. [PMID: 24877123 PMCID: PMC4022191 DOI: 10.1155/2014/634132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/14/2014] [Indexed: 12/29/2022]
Abstract
Left ventricle hypertrophy is a common outcome of pressure overload stimulus closely associated with hypertension. This process is triggered by adverse molecular signalling, gene expression, and proteome alteration. Proteomic research has revealed that several molecular targets are associated with pathologic cardiac hypertrophy, including angiotensin II, endothelin-1 and isoproterenol. Several metabolic, contractile, and stress-related proteins are shown to be altered in cardiac hypertrophy derived by hypertension. On the other hand, exercise is a nonpharmacologic agent used for hypertension treatment, where cardiac hypertrophy induced by exercise training is characterized by improvement in cardiac function and resistance against ischemic insult. Despite the scarcity of proteomic research performed with exercise, healthy and pathologic heart proteomes are shown to be modulated in a completely different way. Hence, the altered proteome induced by exercise is mostly associated with cardioprotective aspects such as contractile and metabolic improvement and physiologic cardiac hypertrophy. The present review, therefore, describes relevant studies involving the molecular characteristics and alterations from hypertensive-induced and exercise-induced hypertrophy, as well as the main proteomic research performed in this field. Furthermore, proteomic research into the effect of hypertension on other target-demerged organs is examined.
Collapse
|
30
|
Liu ZW, Niu XL, Chen KL, Xing YJ, Wang X, Qiu C, Gao DF. Selenium attenuates adriamycin-induced cardiac dysfunction via restoring expression of ATP-sensitive potassium channels in rats. Biol Trace Elem Res 2013; 153:220-8. [PMID: 23475371 DOI: 10.1007/s12011-013-9641-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 02/27/2013] [Indexed: 02/07/2023]
Abstract
The possible mechanism of adriamycin (ADR) and/or selenium (Se) deficiency-induced cardiac dysfunction, and cardioprotective effects of Se against ADR-induced cardiac toxicity were investigated in this study. Cardiac function was evaluated by plasma brain natriuretic peptide level and echocardiographic and hemodynamic parameters. Cardiac glutathione peroxidase (GPx) activity was assessed spectrophotometrically. Expression of ATP-sensitive potassium channels (KATP) subunits-SUR2A and Kir6.2-were examined by real-time PCR and Western blotting. The results showed that cardiac function and cardiac GPx activity decreased remarkably after administration of ADR or Se deficiency; more dramatic impairment of cardiac function and cardiac GPx activity were observed after co-administration of ADR and Se deficiency. Mechanically, it is novel for us to find down-regulation of KATP subunits gene expression in cardiac tissue after administration of ADR or Se deficiency, and more significant inhibition of cardiac KATP gene expression was identified after co-administration of ADR and Se deficiency. Furthermore, cardiac toxicity of ADR was found alleviated by Se supplementation, accompanied by restoring of cardiac GPx activity and cardiac KATP gene expression. These results indicate that decreased expression of cardiac KATP is involved in adriamycin and/or Se deficiency-induced cardiac dysfunction; Se deficiency exacerbates adriamycin-induced cardiac dysfunction by future inhibition of KATP expression; Se supplementation seems to protect against adriamycin-induced cardiac dysfunction via restoring KATP expression, showing potential clinical application in cancer chemotherapy.
Collapse
Affiliation(s)
- Zhong-Wei Liu
- Department of Cardiology, Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi, Xi'an 710004, China
| | | | | | | | | | | | | |
Collapse
|
31
|
Wang J, Gao E, Chan TO, Zhang XQ, Song J, Shang X, Koch WJ, Feldman AM, Cheung JY. Induced overexpression of Na(+)/Ca(2+) exchanger does not aggravate myocardial dysfunction induced by transverse aortic constriction. J Card Fail 2013; 19:60-70. [PMID: 23273595 DOI: 10.1016/j.cardfail.2012.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 11/06/2012] [Accepted: 11/08/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alterations in expression and activity of cardiac Na(+)/Ca(2+) exchanger (NCX1) have been implicated in the pathogenesis of heart failure. METHODS AND RESULTS Using transgenic mice in which expression of rat NCX1 was induced at 5 weeks of age, we performed transverse aortic constriction (TAC) at 8 weeks and examined cardiac and myocyte function at 15-18 weeks after TAC (age 23-26 weeks). TAC induced left ventricular (LV) and myocyte hypertrophy and increased myocardial fibrosis in both wild-type (WT) and NCX1-overexpressed mice. NCX1 and phosphorylated ryanodine receptor expression was increased by TAC, whereas sarco(endo)plasmic reticulum Ca(2+)-ATPase levels were decreased by TAC. Action potential duration was prolonged by TAC, but to a greater extent in NCX1 myocytes. Na(+)/Ca(2+) exchange current was similar between WT-TAC and WT-sham myocytes, but was higher in NCX1-TAC myocytes. Both myocyte contraction and [Ca(2+)](i) transient amplitudes were reduced in WT-TAC myocytes, but restored to WT-sham levels in NCX1-TAC myocytes. Despite improvement in single myocyte contractility and Ca(2+) dynamics, induced NCX1 overexpression in TAC animals did not ameliorate LV hypertrophy, increase ejection fraction, or enhance inotropic (maximal first derivative of LV pressure rise, +dP/dt) responses to isoproterenol. CONCLUSIONS In pressure-overload hypertrophy, induced overexpression of NCX1 corrected myocyte contractile and [Ca(2+)](i) transient abnormalities but did not aggravate or improve myocardial dysfunction.
Collapse
Affiliation(s)
- Jufang Wang
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
ATP-sensitive potassium (KATP) channels were first discovered in the heart 30 years ago. Reconstitution of KATP channel activity by coexpression of members of the pore-forming inward rectifier gene family (Kir6.1, KCNJ8, and Kir6.2 KCNJ11) with sulfonylurea receptors (SUR1, ABCC8, and SUR2, ABCC9) of the ABCC protein subfamily has led to the elucidation of many details of channel gating and pore properties. In addition, the essential roles of Kir6.x and SURx subunits in generating cardiac and vascular KATP(2) and the detrimental consequences of genetic deletions or mutations in mice have been recognized. However, despite this extensive body of knowledge, there has been a paucity of defined roles of KATP subunits in human cardiovascular diseases, although there are reports of association of a single Kir6.1 variant with the J-wave syndrome in the ECG, and 2 isolated studies have reported association of loss of function mutations in SUR2 with atrial fibrillation and heart failure. Two new studies convincingly demonstrate that mutations in the SUR2 gene are associated with Cantu syndrome, a complex multi-organ disorder characterized by hypertrichosis, craniofacial dysmorphology, osteochondrodysplasia, patent ductus arteriosus, cardiomegaly, pericardial effusion, and lymphoedema. This realization of previously unconsidered consequences provides significant insight into the roles of the KATP channel in the cardiovascular system and suggests novel therapeutic possibilities.
Collapse
Affiliation(s)
- Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases and Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | | | | |
Collapse
|
33
|
Muravyeva M, Sedlic F, Dolan N, Bosnjak ZJ, Stadnicka A. Preconditioning by isoflurane elicits mitochondrial protective mechanisms independent of sarcolemmal KATP channel in mouse cardiomyocytes. J Cardiovasc Pharmacol 2013; 61:369-77. [PMID: 23318991 PMCID: PMC3648596 DOI: 10.1097/fjc.0b013e318285f55b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cardiac mitochondria and the sarcolemmal (sarc)KATP channels contribute to cardioprotective signaling of anesthetic-induced preconditioning. Changes in mitochondrial bioenergetics influence the sarcolemmal ATP-sensitive K (sarcKATP) channel function, but whether this channel has impacts on mitochondria is uncertain. We used the mouse model with deleted pore-forming Kir6.2 subunit of sarcKATP channel (Kir6.2 KO) to investigate whether the functional sarcKATP channels are necessary for isoflurane activation of mitochondrial protective mechanisms. Ventricular cardiomyocytes were isolated from C57Bl6 wild-type (WT) and Kir6.2 KO mouse hearts. Flavoprotein autofluorescence, mitochondrial reactive oxygen species production, and mitochondrial membrane potential were monitored by laser-scanning confocal microscopy in intact cardiomyocytes. Cell survival was assessed using H2O2-induced stress. Isoflurane (0.5 mM) increased flavoprotein fluorescence to 180% ± 14% and 190% ± 15% and reactive oxygen species production to 118% ± 2% and 124% ± 6% of baseline in WT and Kir6.2 KO myocytes, respectively. Tetramethylrhodamine ethyl ester fluorescence decreased to 84% ± 6% in WT and to 86% ± 4% in Kir6.2 KO myocytes. This effect was abolished by 5HD. Pretreatment with isoflurane decreased the stress-induced cell death from 31% ± 1% to 21% ± 1% in WT and from 44% ± 2% to 35% ± 2% in Kir6.2 KO myocytes. In conclusion, Kir6.2 deletion increases the sensitivity of intact cardiomyocytes to oxidative stress, but does not alter the isoflurane-elicited protective mitochondrial mechanisms, suggesting independent roles for cardiac mitochondria and sarcKATP channels in anesthetic-induced preconditioning by isoflurane.
Collapse
Affiliation(s)
- Maria Muravyeva
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | |
Collapse
|
34
|
Cardiac KATP channel alterations associated with acclimation to hypoxia in goldfish (Carassius auratus L.). Comp Biochem Physiol A Mol Integr Physiol 2013; 164:554-64. [DOI: 10.1016/j.cbpa.2012.12.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/24/2012] [Accepted: 12/25/2012] [Indexed: 01/21/2023]
|
35
|
Riehle C, Abel ED. PGC-1 proteins and heart failure. Trends Cardiovasc Med 2012; 22:98-105. [PMID: 22939990 DOI: 10.1016/j.tcm.2012.07.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/08/2012] [Accepted: 07/09/2012] [Indexed: 11/18/2022]
Abstract
The transcriptional coactivators PGC-1α and PGC-1β are master regulators of oxidative phosphorylation and fatty acid oxidation gene expression. Pressure overload hypertrophy and heart failure are associated with repressed PGC-1α and PGC-1β gene expression. Maintaining expression of PGC-1α and -β preserves contractile function in response to a pathological increase in workload. Here, we discuss the regulation of PGC-1 proteins under conditions of pressure overload hypertrophy and heart failure.
Collapse
Affiliation(s)
- Christian Riehle
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | |
Collapse
|
36
|
Kefaloyianni E, Bao L, Rindler MJ, Hong M, Patel T, Taskin E, Coetzee WA. Measuring and evaluating the role of ATP-sensitive K+ channels in cardiac muscle. J Mol Cell Cardiol 2012; 52:596-607. [PMID: 22245446 DOI: 10.1016/j.yjmcc.2011.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 12/06/2011] [Accepted: 12/23/2011] [Indexed: 11/27/2022]
Abstract
Since ion channels move electrical charge during their activity, they have traditionally been studied using electrophysiological approaches. This was sometimes combined with mathematical models, for example with the description of the ionic mechanisms underlying the initiation and propagation of action potentials in the squid giant axon by Hodgkin and Huxley. The methods for studying ion channels also have strong roots in protein chemistry (limited proteolysis, the use of antibodies, etc.). The advent of the molecular cloning and the identification of genes coding for specific ion channel subunits in the late 1980s introduced a multitude of new techniques with which to study ion channels and the field has been rapidly expanding ever since (e.g. antibody development against specific peptide sequences, mutagenesis, the use of gene targeting in animal models, determination of their protein structures) and new methods are still in development. This review focuses on techniques commonly employed to examine ion channel function in an electrophysiological laboratory. The focus is on the K(ATP) channel, but many of the techniques described are also used to study other ion channels.
Collapse
|
37
|
Graciotti L, Becker J, Granata AL, Procopio AD, Tessarollo L, Fulgenzi G. Dystrophin is required for the normal function of the cardio-protective K(ATP) channel in cardiomyocytes. PLoS One 2011; 6:e27034. [PMID: 22066028 PMCID: PMC3205025 DOI: 10.1371/journal.pone.0027034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 10/09/2011] [Indexed: 12/19/2022] Open
Abstract
Duchenne and Becker muscular dystrophy patients often develop a cardiomyopathy for which the pathogenesis is still unknown. We have employed the murine animal model of Duchenne muscular dystrophy (mdx), which develops a cardiomyopathy that includes some characteristics of the human disease, to study the molecular basis of this pathology. Here we show that the mdx mouse heart has defects consistent with alteration in compounds that regulate energy homeostasis including a marked decrease in creatine-phosphate (PC). In addition, the mdx heart is more susceptible to anoxia than controls. Since the cardio-protective ATP sensitive potassium channel (KATP) complex and PC have been shown to interact we investigated whether deficits in PC levels correlate with other molecular events including KATP ion channel complex presence, its functionality and interaction with dystrophin. We found that this channel complex is present in the dystrophic cardiac cell membrane but its ability to sense a drop in the intracellular ATP concentration and consequently open is compromised by the absence of dystrophin. We further demonstrate that the creatine kinase muscle isoform (CKm) is displaced from the plasma membrane of the mdx cardiac cells. Considering that CKm is a determinant of KATP channel complex function we hypothesize that dystrophin acts as a scaffolding protein organizing the KATP channel complex and the enzymes necessary for its correct functioning. Therefore, the lack of proper functioning of the cardio-protective KATP system in the mdx cardiomyocytes may be part of the mechanism contributing to development of cardiac disease in dystrophic patients.
Collapse
Affiliation(s)
- Laura Graciotti
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Jodi Becker
- Neural Development Group, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Anna Luisa Granata
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, INRCA, Ancona, Italy
| | - Lino Tessarollo
- Neural Development Group, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
- * E-mail: (GF); (LT)
| | - Gianluca Fulgenzi
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
- Neural Development Group, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
- * E-mail: (GF); (LT)
| |
Collapse
|
38
|
Hu X, Xu X, Lu Z, Zhang P, Fassett J, Zhang Y, Xin Y, Hall JL, Viollet B, Bache RJ, Huang Y, Chen Y. AMP activated protein kinase-α2 regulates expression of estrogen-related receptor-α, a metabolic transcription factor related to heart failure development. Hypertension 2011; 58:696-703. [PMID: 21825219 PMCID: PMC3182261 DOI: 10.1161/hypertensionaha.111.174128] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The normal expression of myocardial mitochondrial enzymes is essential to maintain the cardiac energy reserve and facilitate responses to stress, but the molecular mechanisms to maintain myocardial mitochondrial enzyme expression have been elusive. Here we report that congestive heart failure is associated with a significant decrease of myocardial estrogen-related receptor-α (ERRα), but not peroxisome proliferator-activated receptor-γ coactivator 1α, in human heart failure samples. In addition, chronic pressure overload in mice caused a decrease of ERRα expression that was significantly correlated to the degree of left ventricular dysfunction, pulmonary congestion, and decreases of a group of myocardial energy metabolism-related genes. We found that the metabolic sensor AMP activated protein kinase (AMPK) regulates ERRα expression in vivo and in vitro. AMPKα2 knockout decreased myocardial ERRα (both mRNA and protein) and its downstream targets under basal conditions, with no change in myocardial peroxisome proliferator-activated receptor-γ coactivator 1α expression. Using cultured rat neonatal cardiac myocytes, we found that overexpression of constitutively active AMPKα significantly induced ERRα mRNA, protein, and promoter activity. Conversely, selective gene silencing of AMPKα2 repressed ERRα and its target gene levels, indicating that AMPKα2 is involved in the regulation of ERRα expression. In addition, overexpression of ERRα in AMPKα2 knockout neonatal cardiac myocytes partially rescued the repressed expression of some energy metabolism-related genes. These data support an important role for AMPKα2 in regulating the expression of myocardial ERRα and its downstream mitochondrial enzymes.
Collapse
Affiliation(s)
- Xinli Hu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| | - Xin Xu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhongbing Lu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ping Zhang
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| | - John Fassett
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ying Zhang
- Molecular and Cellular Biology Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhenli, Chaoyang District, Beijing 100029, China
| | - Yi Xin
- Molecular and Cellular Biology Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhenli, Chaoyang District, Beijing 100029, China
| | - Jennifer L. Hall
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| | - Benoit Viollet
- Inserm, U1016, Institut Cochin; Cnrs, UMR8104; University Paris Descartes, Paris, France
| | - Robert J. Bache
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| | - Yimin Huang
- Molecular and Cellular Biology Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhenli, Chaoyang District, Beijing 100029, China
| | - Yingjie Chen
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
39
|
Jilkina O, Glogowski M, Kuzio B, Zhilkin PA, Gussakovsky E, Kupriyanov VV. Defects in myoglobin oxygenation in KATP-deficient mouse hearts under normal and stress conditions characterized by near infrared spectroscopy and imaging. Int J Cardiol 2011; 149:315-22. [DOI: 10.1016/j.ijcard.2010.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 11/22/2009] [Accepted: 02/06/2010] [Indexed: 10/19/2022]
|
40
|
Zingman LV, Zhu Z, Sierra A, Stepniak E, Burnett CML, Maksymov G, Anderson ME, Coetzee WA, Hodgson-Zingman DM. Exercise-induced expression of cardiac ATP-sensitive potassium channels promotes action potential shortening and energy conservation. J Mol Cell Cardiol 2011; 51:72-81. [PMID: 21439969 DOI: 10.1016/j.yjmcc.2011.03.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 02/19/2011] [Accepted: 03/14/2011] [Indexed: 12/25/2022]
Abstract
Physical activity is one of the most important determinants of cardiac function. The ability of the heart to increase delivery of oxygen and metabolic fuels relies on an array of adaptive responses necessary to match bodily demand while avoiding exhaustion of cardiac resources. The ATP-sensitive potassium (K(ATP)) channel has the unique ability to adjust cardiac membrane excitability in accordance with ATP and ADP levels, and up-regulation of its expression that occurs in response to exercise could represent a critical element of this adaption. However, the mechanism by which K(ATP) channel expression changes result in a beneficial effect on cardiac excitability and function remains to be established. Here, we demonstrate that an exercise-induced rise in K(ATP) channel expression enhanced the rate and magnitude of action potential shortening in response to heart rate acceleration. This adaptation in membrane excitability promoted significant reduction in cardiac energy consumption under escalating workloads. Genetic disruption of normal K(ATP) channel pore function abolished the exercise-related changes in action potential duration adjustment and caused increased cardiac energy consumption. Thus, an expression-driven enhancement in the K(ATP) channel-dependent membrane response to alterations in cardiac workload represents a previously unrecognized mechanism for adaptation to physical activity and a potential target for cardioprotection.
Collapse
Affiliation(s)
- Leonid V Zingman
- Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lu Z, Xu X, Hu X, Fassett J, Zhu G, Tao Y, Li J, Huang Y, Zhang P, Zhao B, Chen Y. PGC-1 alpha regulates expression of myocardial mitochondrial antioxidants and myocardial oxidative stress after chronic systolic overload. Antioxid Redox Signal 2010; 13:1011-22. [PMID: 20406135 PMCID: PMC2959178 DOI: 10.1089/ars.2009.2940] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Mitochondria are a principal site for generation of reactive oxygen species (ROS) in the heart. Peroxisome proliferator activated receptor gamma coactivator 1 alpha (PGC-1 alpha) plays an important role in regulating mitochondrial biogenesis and myocardial metabolism, but whether PGC-1 alpha can simultaneously upregulate myocardial mitochondrial antioxidants has not been studied. In the present study, we examined the effect of PGC-1 alpha deficiency (PGC-1 alpha(-/-)) on oxidative stress and expression of a group of mitochondrial antioxidants in normal hearts and in hearts exposed to chronic systolic pressure overload produced by transverse aortic constriction (TAC). We found that PGC-1 alpha(-/-) caused moderate but significant decreases of myocardial mitochondrial antioxidant enzymes such as SOD2, and thioredoxin (Trx2), but had no effect on expression of myocardial oxidative stress markers and left ventricular (LV) function under basal conditions. However, in response to TAC for 6 weeks, PGC-1 alpha(-/-) mice showed greater increases of myocardial oxidative stress markers 3'-nitrotyrosine and 4-hydroxynonenal, more severe LV hypertrophy and dilatation, pulmonary congestion, and a greater reduction of LV fractional shortening and dP/dt(max) than did wild-type hearts. SOD mimetic MnTMPyP treatment (6 mg/kg/day) significantly attenuated TAC-induced LV hypertrophy and dysfunction in PGC-1 alpha(-/-) mice. These data indicate that PGC-1 alpha plays an important role in regulating expression of myocardial mitochondrial antioxidants SOD2 and Trx2 and in protecting hearts against TAC-induced myocardial oxidative stress, hypertrophy, and dysfunction.
Collapse
Affiliation(s)
- Zhongbing Lu
- Cardiovascular Division, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Flagg TP, Enkvetchakul D, Koster JC, Nichols CG. Muscle KATP channels: recent insights to energy sensing and myoprotection. Physiol Rev 2010; 90:799-829. [PMID: 20664073 DOI: 10.1152/physrev.00027.2009] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
ATP-sensitive potassium (K(ATP)) channels are present in the surface and internal membranes of cardiac, skeletal, and smooth muscle cells and provide a unique feedback between muscle cell metabolism and electrical activity. In so doing, they can play an important role in the control of contractility, particularly when cellular energetics are compromised, protecting the tissue against calcium overload and fiber damage, but the cost of this protection may be enhanced arrhythmic activity. Generated as complexes of Kir6.1 or Kir6.2 pore-forming subunits with regulatory sulfonylurea receptor subunits, SUR1 or SUR2, the differential assembly of K(ATP) channels in different tissues gives rise to tissue-specific physiological and pharmacological regulation, and hence to the tissue-specific pharmacological control of contractility. The last 10 years have provided insights into the regulation and role of muscle K(ATP) channels, in large part driven by studies of mice in which the protein determinants of channel activity have been deleted or modified. As yet, few human diseases have been correlated with altered muscle K(ATP) activity, but genetically modified animals give important insights to likely pathological roles of aberrant channel activity in different muscle types.
Collapse
Affiliation(s)
- Thomas P Flagg
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | | | |
Collapse
|
43
|
Lu Z, Xu X, Hu X, Lee S, Traverse JH, Zhu G, Fassett J, Tao Y, Zhang P, dos Remedios C, Pritzker M, Hall JL, Garry DJ, Chen Y. Oxidative stress regulates left ventricular PDE5 expression in the failing heart. Circulation 2010; 121:1474-83. [PMID: 20308615 PMCID: PMC3110701 DOI: 10.1161/circulationaha.109.906818] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Phosphodiesterase type 5 (PDE5) inhibition has been shown to exert profound beneficial effects in the failing heart, suggesting a significant role for PDE5 in the development of congestive heart failure (CHF). The purpose of this study is to test the hypothesis that oxidative stress causes increased PDE5 expression in cardiac myocytes and that increased PDE5 contributes to the development of CHF. METHODS AND RESULTS Myocardial PDE5 expression and cellular distribution were determined in left ventricular samples from patients with end-stage CHF and normal donors and from mice after transverse aortic constriction (TAC)-induced CHF. Compared with donor human hearts, myocardial PDE5 protein was increased approximately equal 4.5-fold in CHF samples, and the increase of myocardial PDE5 expression was significantly correlated with myocardial oxidative stress markers 3'-nitrotyrosine or 4-hydroxynonenal expression (P<0.05). Histological examination demonstrated that PDE5 was mainly expressed in vascular smooth muscle in normal donor hearts, but its expression was increased in both cardiac myocytes and vascular smooth muscle of CHF hearts. Myocardial PDE5 protein content and activity also increased in mice after TAC-induced CHF (P<0.05). When the superoxide dismutase (SOD) mimetic M40401 was administered to attenuate oxidative stress, the increased PDE5 protein and activity caused by TAC was blunted, and the hearts were protected against left ventricular hypertrophy and CHF. Conversely, increased myocardial oxidative stress in superoxide dismutase 3 knockout mice caused a greater increase of PDE5 expression and CHF after TAC. In addition, administration of sildenafil to inhibit PDE5 attenuated TAC-induced myocardial oxidative stress, PDE5 expression, and CHF. CONCLUSIONS Myocardial oxidative stress increases PDE5 expression in the failing heart. Reducing oxidative stress by treatment with M40401 attenuated cardiomyocyte PDE5 expression. This and selective inhibition of PDE5 protected the heart against pressure overload-induced left ventricular hypertrophy and CHF.
Collapse
Affiliation(s)
- Zhongbing Lu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Vascular Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Xin Xu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Vascular Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Xinli Hu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sangjin Lee
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jay H. Traverse
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, Minneapolis
| | - Guangshuo Zhu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - John Fassett
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yi Tao
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ping Zhang
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Marc Pritzker
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jennifer L. Hall
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel J. Garry
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yingjie Chen
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Vascular Biology, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
44
|
Cardiac sarcolemmal K(ATP) channels: Latest twists in a questing tale! J Mol Cell Cardiol 2009; 48:71-5. [PMID: 19607836 DOI: 10.1016/j.yjmcc.2009.07.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 06/23/2009] [Accepted: 07/06/2009] [Indexed: 11/24/2022]
Abstract
Reconstitution of K(ATP) channel activity from coexpression of members of the pore-forming inward rectifier gene family (Kir6.1, KCNJ8, and Kir6.2 KCNJ11) with sulfonylurea receptors (SUR1, ABCC8, and SUR2, ABCC9) of the ABCC protein sub-family, has led to the elucidation of many details of channel gating and pore properties, as well as the essential roles of Kir6.2 and SUR2 subunits in generating cardiac ventricular K(ATP). However, despite this extensive body of knowledge, there remain significant holes in our understanding of the physiological role of the cardiac K(ATP) channel, and surprising new findings keep emerging. Recent findings from genetically modified animals include the apparent insensitivity of cardiac sarcolemmal channels to nucleotide levels, and unenvisioned complexities of the subunit make-up of the cardiac channels. This topical review focuses on these new findings and considers their implications.
Collapse
|