1
|
Wang XH, Ning ZH, Xie Z, Ou Y, Yang JY, Liu YX, Huang H, Tang HF, Jiang ZS, Hu HJ. SIRT3/AMPK Signaling Pathway Regulates Lipid Metabolism and Improves Vulnerability to Atrial Fibrillation in Dahl Salt-Sensitive Rats. Am J Hypertens 2024; 37:901-908. [PMID: 39023012 DOI: 10.1093/ajh/hpae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Hypertension may result in atrial fibrillation (AF) and lipid metabolism disorders. The Sirtuins3 (SIRT3)/AMP-activated protein kinase (AMPK) signaling pathway has the capacity to regulate lipid metabolism disorders and the onset of AF. We hypothesize that the SIRT3/AMPK signaling pathway suppresses lipid metabolism disorders, thereby mitigating salt-sensitive hypertension (SSHT)-induced susceptibility to AF. METHODS The study involved 7-week-old male Dahl salt-sensitive that were fed either a high-salt diet (8% NaCl; DSH group) or a normal diet (0.3% NaCl; DSN group). Then DSH group was administered either oral metformin (MET, an AMPK agonist) or intraperitoneal injection of Honokiol (HK, a SIRT3 agonist). This experimental model allowed for the measurement of Systolic blood pressure (SBP), the expression levels of lipid metabolism-related biomarkers, pathological examination of atrial fibrosis, and lipid accumulation, as well as AF inducibility and AF duration. RESULTS DSH decrease SIRT3, phosphorylation-AMPK, and very long-chain acyl-CoA dehydrogenase, (VLCAD) expression, increased FASN and FABP4 expression and concentrations of free fatty acid and triglyceride, atrial fibrosis and lipid accumulation in atrial tissue, enhanced level of SBP, promoted AF induction rate and prolonged AF duration, which are blocked by MET and HK. Our results also showed that the degree of atrial fibrosis was negatively correlated with VLCAD expression, but positively correlated with the expression of FASN and FABP4. CONCLUSIONS We have confirmed that a high-salt diet can result in hypertension, and associated atrial tissue lipid metabolism dysfunction. This condition is linked to the inhibition of the SIRT3/AMPK signaling pathway, which plays a significant role in the progression of susceptibility to AF in SSHT rats.
Collapse
Affiliation(s)
- Xiu-Heng Wang
- Department of Medical-Record, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Zhi-Hong Ning
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Zhong Xie
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Yun Ou
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Jia-Yang Yang
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Yun-Xi Liu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Hong Huang
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Hui-Fang Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Zhi-Sheng Jiang
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
- Department of Cardiovascular Disease and Key Lab for Atherosclerosis of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Heng-Jing Hu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
- Department of Cardiovascular Disease and Key Lab for Atherosclerosis of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| |
Collapse
|
2
|
Chakraborty P, Nattel S, Nanthakumar K, Connelly KA, Husain M, Po SS, Ha ACT. Sudden cardiac death due to ventricular arrhythmia in diabetes mellitus: A bench to bedside review. Heart Rhythm 2024; 21:1827-1837. [PMID: 38848857 DOI: 10.1016/j.hrthm.2024.05.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024]
Abstract
Diabetes mellitus (DM) confers an increased risk of sudden cardiac death (SCD) independent of its associated cardiovascular comorbidities. DM induces adverse structural, electrophysiologic, and autonomic cardiac remodeling that can increase one's risk of ventricular arrhythmias and SCD. Although glycemic control and prevention of microvascular and macrovascular complications are cornerstones in the management of DM, they are not adequate for the prevention of SCD. In this narrative review, we describe the contribution of DM to the pathophysiologic mechanism of SCD beyond its role in atherosclerotic cardiovascular disease and heart failure. On the basis of this pathophysiologic framework, we outline potential preventive and therapeutic strategies to mitigate the risk of SCD in this population of high-risk patients.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Kumaraswamy Nanthakumar
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kim A Connelly
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Mansoor Husain
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Sunny S Po
- Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andrew C T Ha
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Zhao Y, Du B, Chakraborty P, Denham N, Massé S, Lai PF, Azam MA, Billia F, Thavendiranathan P, Abdel‐Qadir H, Lopaschuk GD, Nanthakumar K. Impaired Cardiac AMPK (5'-Adenosine Monophosphate-Activated Protein Kinase) and Ca 2+-Handling, and Action Potential Duration Heterogeneity in Ibrutinib-Induced Ventricular Arrhythmia Vulnerability. J Am Heart Assoc 2024; 13:e032357. [PMID: 38842296 PMCID: PMC11255774 DOI: 10.1161/jaha.123.032357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/03/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND We recently demonstrated that acute administration of ibrutinib, a Bruton's tyrosine kinase inhibitor used in chemotherapy for blood malignancies, increases ventricular arrhythmia (VA) vulnerability. A pathway of ibrutinib-induced vulnerability to VA that can be modulated for cardioprotection remains unclear. METHODS AND RESULTS The effects of ibrutinib on cardiac electrical activity and Ca2+ dynamics were investigated in Langendorff-perfused hearts using optical mapping. We also conducted Western blotting analysis to evaluate the impact of ibrutinib on various regulatory and Ca2+-handling proteins in rat cardiac tissues. Treatment with ibrutinib (10 mg/kg per day) for 4 weeks was associated with an increased VA inducibility (72.2%±6.3% versus 38.9±7.0% in controls, P<0.002) and shorter action potential durations during pacing at various frequencies (P<0.05). Ibrutinib also decreased heart rate thresholds for beat-to-beat duration alternans of the cardiac action potential (P<0.05). Significant changes in myocardial Ca2+ transients included lower amplitude alternans ratios (P<0.05), longer times-to-peak (P<0.05), and greater spontaneous intracellular Ca2+ elevations (P<0.01). We also found lower abundance and phosphorylation of myocardial AMPK (5'-adenosine monophosphate-activated protein kinase), indicating reduced AMPK activity in hearts after ibrutinib treatment. An acute treatment with the AMPK activator 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside ameliorated abnormalities in action potential and Ca2+ dynamics, and significantly reduced VA inducibility (37.1%±13.4% versus 72.2%±6.3% in the absence of 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside, P<0.05) in hearts from ibrutinib-treated rats. CONCLUSIONS VA vulnerability inflicted by ibrutinib may be mediated in part by an impairment of myocardial AMPK activity. Pharmacological activation of AMPK may be a protective strategy against ibrutinib-induced cardiotoxicity.
Collapse
MESH Headings
- Animals
- Adenine/analogs & derivatives
- Adenine/pharmacology
- Piperidines/pharmacology
- Action Potentials/drug effects
- Pyrimidines/pharmacology
- AMP-Activated Protein Kinases/metabolism
- Pyrazoles/pharmacology
- Male
- Arrhythmias, Cardiac/chemically induced
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/prevention & control
- Protein Kinase Inhibitors/pharmacology
- Heart Rate/drug effects
- Isolated Heart Preparation
- Calcium/metabolism
- Rats
- Disease Models, Animal
- Rats, Sprague-Dawley
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Calcium Signaling/drug effects
- Time Factors
Collapse
Affiliation(s)
- Yanan Zhao
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
- China‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Beibei Du
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
- China‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Nathan Denham
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Patrick F.H. Lai
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Mohammed Ali Azam
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Filio Billia
- Toronto General Hospital Research InstituteTorontoCanada
- Ted Rogers Centre for Heart ResearchTorontoCanada
| | | | - Husam Abdel‐Qadir
- Toronto General Hospital Research InstituteTorontoCanada
- Ted Rogers Centre for Heart ResearchTorontoCanada
| | | | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| |
Collapse
|
4
|
Zhao Y, Chakraborty P, Tomassetti J, Subha T, Massé S, Thavendiranathan P, Billia F, Lai PFH, Abdel-Qadir H, Nanthakumar K. Arrhythmogenic Ventricular Remodeling by Next-Generation Bruton's Tyrosine Kinase Inhibitor Acalabrutinib. Int J Mol Sci 2024; 25:6207. [PMID: 38892396 PMCID: PMC11173147 DOI: 10.3390/ijms25116207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiac arrhythmias remain a significant concern with Ibrutinib (IBR), a first-generation Bruton's tyrosine kinase inhibitor (BTKi). Acalabrutinib (ABR), a next-generation BTKi, is associated with reduced atrial arrhythmia events. However, the role of ABR in ventricular arrhythmia (VA) has not been adequately evaluated. Our study aimed to investigate VA vulnerability and ventricular electrophysiology following chronic ABR therapy in male Sprague-Dawley rats utilizing epicardial optical mapping for ventricular voltage and Ca2+ dynamics and VA induction by electrical stimulation in ex-vivo perfused hearts. Ventricular tissues were snap-frozen for protein analysis for sarcoplasmic Ca2+ and metabolic regulatory proteins. The results show that both ABR and IBR treatments increased VA vulnerability, with ABR showing higher VA regularity index (RI). IBR, but not ABR, is associated with the abbreviation of action potential duration (APD) and APD alternans. Both IBR and ABR increased diastolic Ca2+ leak and Ca2+ alternans, reduced conduction velocity (CV), and increased CV dispersion. Decreased SERCA2a expression and AMPK phosphorylation were observed with both treatments. Our results suggest that ABR treatment also increases the risk of VA by inducing proarrhythmic changes in Ca2+ signaling and membrane electrophysiology, as seen with IBR. However, the different impacts of these two BTKi on ventricular electrophysiology may contribute to differences in VA vulnerability and distinct VA characteristics.
Collapse
Affiliation(s)
- Yanan Zhao
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Praloy Chakraborty
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Julianna Tomassetti
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Tasnia Subha
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Stéphane Massé
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Paaladinesh Thavendiranathan
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
- Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Filio Billia
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
- Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Patrick F. H. Lai
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Husam Abdel-Qadir
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
- Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Women’s College Hospital, Toronto, ON M5S 1B2, Canada
| | - Kumaraswamy Nanthakumar
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| |
Collapse
|
5
|
Lu YY, Cheng CC, Chen YC, Lin YK, Higa S, Kao YH, Chen YJ. Adenosine monophosphate-regulated protein kinase inhibition modulates electrophysiological characteristics and calcium homeostasis of rabbit right ventricular outflow tract. Fundam Clin Pharmacol 2024; 38:262-275. [PMID: 37664898 DOI: 10.1111/fcp.12953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 07/23/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND Metabolic stress predisposes to ventricular arrhythmias and sudden cardiac death. Right ventricular outflow tract (RVOT) is the common origin of ventricular arrhythmias. Adenosine monophosphate-regulated protein kinase (AMPK) activation is an important compensatory mechanism for cardiac remodeling during metabolic stress. OBJECTIVES The purpose of this study was to access whether AMPK inhibition would modulate RVOT electrophysiology, calcium (Ca2+ ) regulation, and RVOT arrhythmogenesis or not. METHODS Conventional microelectrodes were used to record electrical activity before and after compound C (10 µM, an AMPK inhibitor) in isoproterenol (1 µM)-treated rabbit RVOT tissue preparations under electrical pacing. Whole-cell patch-clamp and confocal microscopic examinations were performed in baseline and compound C-treated rabbit RVOT cardiomyocytes to investigate ionic currents and intracellular Ca2+ transients in isolated rabbit RVOT cardiomyocytes. RESULTS Compound C decreased RVOT contractility, and reversed isoproterenol increased RVOT contractility. Compound C decreased the incidence, rate, and duration of isoproterenol-induced RVOT burst firing under rapid pacing. Compared to baseline, compound C-treated RVOT cardiomyocytes had a longer action potential duration, smaller intracellular Ca2+ transients, late sodium (Na+ ), peak L-type Ca2+ current density, Na+ -Ca2+ exchanger, transient outward potassium (K+ ) current, and rapid and slow delayed rectifier K+ currents. CONCLUSION AMPK inhibition modulates RVOT electrophysiological characteristics and Ca2+ homeostasis, contributing to lower RVOT arrhythmogenic activity. Accordingly, AMPK inhibition might potentially reduce ventricular tachyarrhythmias.
Collapse
Affiliation(s)
- Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Chen-Chuan Cheng
- Department of Cardiology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, and Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Okinawa, Japan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
6
|
Chi J, Wu N, Li P, Hu J, Cai H, Lin C, Lai Y, Yang H, Huang J, Li M, Xu L. Hygrothermal stress increases malignant arrhythmias susceptibility by inhibiting the LKB1-AMPK-Cx43 pathway. Sci Rep 2024; 14:5010. [PMID: 38424223 PMCID: PMC10904738 DOI: 10.1038/s41598-024-55804-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024] Open
Abstract
High mortality due to hygrothermal stress during heat waves is mostly linked to cardiovascular malfunction, the most serious of which are malignant arrhythmias. However, the mechanism associated with hygrothermal stress leading to malignant arrhythmias remains unclear. The energy metabolism regulated by liver kinase B1 (LKB1) and adenosine monophosphate-activated protein kinase (AMPK) and the electrical signaling based on gap junction protein, connexin43 (Cx43), plays important roles in the development of cardiac arrhythmias. In order to investigate whether hygrothermal stress induces arrhythmias via the LKB1-AMPK-Cx43 pathway, Sprague-Dawley rats were exposed to high temperature and humidity for constructing the hygrothermal stress model. A final choice of 40 °C and 85% humidity was made by pre-exploration based on different gradient environmental conditions with reference to arrhythmia event-inducing stability and risk of sudden death. Then, the incidence of arrhythmic events, as well as the expression, phosphorylation at Ser368, and distribution of Cx43 in the myocardium, were examined. Meanwhile, the adenosine monophosphate-activated protein kinase activator, Acadesine, was also administered to investigate the role played by AMPK in the process. Our results showed that hygrothermal stress induced malignant arrhythmias such as ventricular tachycardia, ventricular fibrillation, and severe atrioventricular block. Besides, hygrothermal stress decreased the phosphorylation of Cx43 at Ser368, induced proarrhythmic redistribution of Cx43 from polar to lateral sides of the cardiomyocytes, and also caused LKB1 and phosphorylated-AMPK expression to be less abundant. While, pretreatment with Acadesine significantly actived the LKB1-AMPK-Cx43 pathway and thus ameliorated malignant arrhythmias, indicating that the hygrothermal stress-induced arrhythmias is associated with the redistribution of gap junctions in cardiomyocytes and the organism's energy metabolism.
Collapse
Affiliation(s)
- Jianing Chi
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
| | - Ningxia Wu
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pengfei Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Jiaman Hu
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hua Cai
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cailong Lin
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yingying Lai
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Han Yang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Jianyu Huang
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
| | - Min Li
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
| | - Lin Xu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China.
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China.
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China.
| |
Collapse
|
7
|
Chakraborty P, Po SS, Scherlag BJ, Dasari TW. The neurometabolic axis: A novel therapeutic target in heart failure. Life Sci 2023; 333:122122. [PMID: 37774940 DOI: 10.1016/j.lfs.2023.122122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
Abnormal cardiac metabolism or cardiac metabolic remodeling is reported before the onset of heart failure with reduced ejection fraction (HFrEF) and is known to trigger and maintain the mechanical dysfunction and electrical, and structural abnormalities of the ventricle. A dysregulated cardiac autonomic tone characterized by sympathetic overdrive with blunted parasympathetic activation is another pathophysiological hallmark of HF. Emerging evidence suggests a link between autonomic nervous system activity and cardiac metabolism. Chronic β-adrenergic activation promotes maladaptive metabolic remodeling whereas cholinergic activation attenuates the metabolic aberrations through favorable modulation of key metabolic regulatory molecules. Restoration of sympathovagal balance by neuromodulation strategies is emerging as a novel nonpharmacological treatment strategy in HF. The current review attempts to evaluate the 'neuro-metabolic axis' in HFrEF and whether neuromodulation can mitigate the adverse metabolic remodeling in HFrEF.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sunny S Po
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Benjamin J Scherlag
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tarun W Dasari
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
8
|
Keefe JA, Hulsurkar MM, Reilly S, Wehrens XHT. Mouse models of spontaneous atrial fibrillation. Mamm Genome 2023; 34:298-311. [PMID: 36173465 PMCID: PMC10898345 DOI: 10.1007/s00335-022-09964-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022]
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in adults, with a prevalence increasing with age. Current clinical management of AF is focused on tertiary prevention (i.e., treating the symptoms and sequelae) rather than addressing the underlying molecular pathophysiology. Robust animal models of AF, particularly those that do not require supraphysiologic stimuli to induce AF (i.e., showing spontaneous AF), enable studies that can uncover the underlying mechanisms of AF. Several mouse models of AF have been described to exhibit spontaneous AF, but pathophysiologic drivers of AF differ among models. Here, we describe relevant AF mechanisms and provide an overview of large and small animal models of AF. We then provide an in-depth review of the spontaneous mouse models of AF, highlighting the relevant AF mechanisms for each model.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mohit M Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
Altieri DI, Etzion Y, Anderson HD. Cannabinoid receptor agonist attenuates angiotensin II-induced enlargement and mitochondrial dysfunction in rat atrial cardiomyocytes. Front Pharmacol 2023; 14:1142583. [PMID: 37113758 PMCID: PMC10126395 DOI: 10.3389/fphar.2023.1142583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
Pathological remodeling of atrial tissue renders the atria more prone to arrhythmia upon arrival of electrical triggers. Activation of the renin-angiotensin system is an important factor that contributes to atrial remodeling, which may result in atrial hypertrophy and prolongation of P-wave duration. In addition, atrial cardiomyocytes are electrically coupled via gap junctions, and electrical remodeling of connexins may result in dysfunction of coordinated wave propagation within the atria. Currently, there is a lack of effective therapeutic strategies that target atrial remodeling. We previously proposed that cannabinoid receptors (CBR) may have cardioprotective qualities. CB13 is a dual cannabinoid receptor agonist that activates AMPK signaling in ventricular cardiomyocytes. We reported that CB13 attenuates tachypacing-induced shortening of atrial refractoriness and inhibition of AMPK signaling in the rat atria. Here, we evaluated the effects of CB13 on neonatal atrial rat cardiomyocytes (NRAM) stimulated by angiotensin II (AngII) in terms of atrial myocyte enlargement and mitochondrial function. CB13 inhibited AngII-induced enhancement of atrial myocyte surface area in an AMPK-dependent manner. CB13 also inhibited mitochondrial membrane potential deterioration in the same context. However, AngII and CB13 did not affect mitochondrial permeability transition pore opening. We further demonstrate that CB13 increased Cx43 compared to AngII-treated neonatal rat atrial myocytes. Overall, our results support the notion that CBR activation promotes atrial AMPK activation, and prevents myocyte enlargement (an indicator that suggests pathological hypertrophy), mitochondrial depolarization and Cx43 destabilization. Therefore, peripheral CBR activation should be further tested as a novel treatment strategy in the context of atrial remodeling.
Collapse
Affiliation(s)
- Danielle I. Altieri
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hope D. Anderson
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
- *Correspondence: Hope D. Anderson,
| |
Collapse
|
10
|
Lkhagva B, Lee TW, Lin YK, Chen YC, Chung CC, Higa S, Chen YJ. Disturbed Cardiac Metabolism Triggers Atrial Arrhythmogenesis in Diabetes Mellitus: Energy Substrate Alternate as a Potential Therapeutic Intervention. Cells 2022; 11:cells11182915. [PMID: 36139490 PMCID: PMC9497243 DOI: 10.3390/cells11182915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
Atrial fibrillation (AF) is the most common type of sustained arrhythmia in diabetes mellitus (DM). Its morbidity and mortality rates are high, and its prevalence will increase as the population ages. Despite expanding knowledge on the pathophysiological mechanisms of AF, current pharmacological interventions remain unsatisfactory; therefore, novel findings on the underlying mechanism are required. A growing body of evidence suggests that an altered energy metabolism is closely related to atrial arrhythmogenesis, and this finding engenders novel insights into the pathogenesis of the pathophysiology of AF. In this review, we provide comprehensive information on the mechanistic insights into the cardiac energy metabolic changes, altered substrate oxidation rates, and mitochondrial dysfunctions involved in atrial arrhythmogenesis, and suggest a promising advanced new therapeutic approach to treat patients with AF.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
| | - Cheng-Chih Chung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Okinawa 901-2131, Japan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Cardiovascular Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Correspondence:
| |
Collapse
|
11
|
Long C, Liu H, Zhan W, Chen L, Yu Z, Tian S, Xiang Y, Chen S, Tian X. Chronological attenuation of NPRA/PKG/AMPK signaling promotes vascular aging and elevates blood pressure. Aging Cell 2022; 21:e13699. [PMID: 36016499 PMCID: PMC9470896 DOI: 10.1111/acel.13699] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/09/2022] [Accepted: 08/02/2022] [Indexed: 01/25/2023] Open
Abstract
Hypertension is common in elderly population. We designed to search comprehensively for genes that are chronologically shifted in their expressions and to define their contributions to vascular aging and hypertension. RNA sequencing was conducted to search for senescence-shifted transcripts in human umbilical vein endothelial cells (HUVECs). Small interfering RNA (siRNA), small-molecule drugs, CRISPR/Cas9 techniques, and imaging were used to determine genes' function and contributions to age-related phenotypes of the endothelial cell and blood vessel. Of 25 genes enriched in the term of "regulation of blood pressure," NPRA was changed most significantly. The decreased NPRA expression was replicated in aortas of aged mice. The knockdown of NPRA promoted HUVEC senescence and it decreased expressions of protein kinase cGMP-dependent 1 (PKG), sirtuin 1 (SIRT1), and endothelial nitric oxide synthase (eNOS). Suppression of NPRA also decreased the phosphorylation of AMP-activated protein kinase (AMPK) as well as the ratio of oxidized nicotinamide adenine dinucleotide (NAD+ )/reduced nicotinamide adenine dinucleotide (NADH) but increased the production of reactive oxygen species (ROS). 8-Br-cGMP (analog of cGMP), or AICAR (AMPK activator), counteracted the observed changes in HUVECs. The Npr1+/- mice presented an elevated systolic blood pressure and their vessels became insensitive to endothelial-dependent vasodilators. Further, vessels from Npr1+/- mice increased Cdkn1a but decreased eNos expressions. These phenotypes were rescued by intravenously administrated 8-Br-cGMP and viral overexpression of human PKG, respectively. In conclusion, we demonstrate NPRA/PKG/AMPK as a novel and critical signaling axis in the modulation of endothelial cell senescence, vascular aging, and hypertension.
Collapse
Affiliation(s)
- Changkun Long
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Hongfei Liu
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Wenxing Zhan
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Liping Chen
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Zhenping Yu
- Institute of Translational MedicineNanchang UniversityNanchangChina,School of Life Science, Nanchang UniversityNanchangChina
| | - Shane Tian
- Department of Biochemistry/ChemistryOhio State UniversityColumbusOhioUSA
| | - Yang Xiang
- Metabolic Control and AgingHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Shenghan Chen
- Vascular Function LaboratoryHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| | - Xiao‐Li Tian
- Aging and Vascular DiseasesHuman Aging Research Institute and School of Life Science, Nanchang university, and Jiangxi Key Laboratory of Human AgingNanchangChina
| |
Collapse
|
12
|
Remme CA. Sudden cardiac death in diabetes and obesity: mechanisms and therapeutic strategies. Can J Cardiol 2022; 38:418-426. [PMID: 35017043 DOI: 10.1016/j.cjca.2022.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 02/07/2023] Open
Abstract
Ventricular arrhythmias and sudden cardiac death (SCD) occur most frequently in the setting of coronary artery disease, cardiomyopathy and heart failure, but are also increasingly observed in individuals suffering from diabetes mellitus and obesity. The incidence of these metabolic disorders is rising in Western countries, but adequate prevention and treatment of arrhythmias and SCD in affected patients is limited due to our incomplete knowledge of the underlying disease mechanisms. Here, an overview is presented of the prevalence of electrophysiological disturbances, ventricular arrhythmias and SCD in the clinical setting of diabetes and obesity. Experimental studies are reviewed, which have identified disease pathways and associated modulatory factors, in addition to pro-arrhythmic mechanisms. Key processes are discussed, including mitochondrial dysfunction, oxidative stress, cardiac structural derangements, abnormal cardiac conduction, ion channel dysfunction, prolonged repolarization and dysregulation of intracellular sodium and calcium homeostasis. In addition, the recently identified pro-arrhythmic effects of dysregulated branched chain amino acid metabolism, a common feature in patients with metabolic disorders, are addressed. Finally, current management options are discussed, in addition to the potential development of novel preventive and therapeutic strategies based on recent insight gained from translational studies.
Collapse
Affiliation(s)
- Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
13
|
Implications of SGLT Inhibition on Redox Signalling in Atrial Fibrillation. Int J Mol Sci 2021; 22:ijms22115937. [PMID: 34073033 PMCID: PMC8198069 DOI: 10.3390/ijms22115937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained (atrial) arrhythmia, a considerable global health burden and often associated with heart failure. Perturbations of redox signalling in cardiomyocytes provide a cellular substrate for the manifestation and maintenance of atrial arrhythmias. Several clinical trials have shown that treatment with sodium-glucose linked transporter inhibitors (SGLTi) improves mortality and hospitalisation in heart failure patients independent of the presence of diabetes. Post hoc analysis of the DECLARE-TIMI 58 trial showed a 19% reduction in AF in patients with diabetes mellitus (hazard ratio, 0.81 (95% confidence interval: 0.68-0.95), n = 17.160) upon treatment with SGLTi, regardless of pre-existing AF or heart failure and independent from blood pressure or renal function. Accordingly, ongoing experimental work suggests that SGLTi not only positively impact heart failure but also counteract cellular ROS production in cardiomyocytes, thereby potentially altering atrial remodelling and reducing AF burden. In this article, we review recent studies investigating the effect of SGLTi on cellular processes closely interlinked with redox balance and their potential effects on the onset and progression of AF. Despite promising insight into SGLTi effect on Ca2+ cycling, Na+ balance, inflammatory and fibrotic signalling, mitochondrial function and energy balance and their potential effect on AF, the data are not yet conclusive and the importance of individual pathways for human AF remains to be established. Lastly, an overview of clinical studies investigating SGLTi in the context of AF is provided.
Collapse
|
14
|
Lee DI, Murninkas M, Elyagon S, Etzion Y, Anderson HD. Cannabinoid Receptor Agonist Inhibits Atrial Electrical Remodeling in a Tachypaced Ex Vivo Rat Model. Front Pharmacol 2021; 12:642398. [PMID: 33967775 PMCID: PMC8100753 DOI: 10.3389/fphar.2021.642398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/16/2021] [Indexed: 12/17/2022] Open
Abstract
Introduction: Atrial fibrillation (AF) leads to rate-dependent atrial changes collectively defined as atrial remodelling (AR). Shortening of the atrial effective refractory period (AERP) and decreased conduction velocity are among the hallmarks of AR. Pharmacological strategies to inhibit AR, thereby reducing the self-perpetual nature of AF, are of great clinical value. Cannabinoid receptor (CBR) ligands may exert cardioprotective effects; CB13, a dual CBR agonist with limited brain penetration, protects cardiomyocytes from mitochondrial dysfunction induced by endothelin-1. Here, we examined the effects of CB13 on normal physiology of the rat heart and development of tachypacing-induced AR. Methods: Rat hearts were perfused in a Langendorff set-up with CB13 (1 µM) or vehicle. Hemodynamic properties of non-paced hearts were examined conventionally. In a different set of hearts, programmed stimulation protocol was performed before and after atrial tachypacing for 90 min using a mini-hook platinum quadrupole electrode inserted on the right atrium. Atrial samples were further assessed by western blot analysis. Results: CB13 had no effects on basal hemodynamic properties. However, the compound inhibited tachypacing-induced shortening of the AERP. Protein expression of PGC1α was significantly increased by CB13 compared to vehicle in paced and non-paced hearts. Phosphorylation of AMPKα at residue threonine 172 was increased suggesting upregulation of mitochondrial biogenesis. Connexin43 was downregulated by tachypacing. This effect was diminished in the presence of CB13. Conclusion: Our findings support the notion that peripheral activation of CBR may be a new treatment strategy to prevent AR in patients suffering from AF, and therefore warrants further study.
Collapse
Affiliation(s)
- Danielle I Lee
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
| | - Michael Murninkas
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sigal Elyagon
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hope D Anderson
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
| |
Collapse
|
15
|
Lkhagva B, Lin Y, Chen Y, Cheng W, Higa S, Kao Y, Chen Y. ZFHX3 knockdown dysregulates mitochondrial adaptations to tachypacing in atrial myocytes through enhanced oxidative stress and calcium overload. Acta Physiol (Oxf) 2021; 231:e13604. [PMID: 33332716 DOI: 10.1111/apha.13604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/06/2020] [Accepted: 12/11/2020] [Indexed: 01/02/2023]
Abstract
AIM To investigate the role of zinc finger homeobox 3 gene (ZFHX3) in tachypacing-induced mitochondrial dysfunction and explore its molecular mechanisms and potential as a therapeutic target in atrial fibrillation (AF). METHODS Through a bioluminescent assay, a patch clamp, confocal fluorescence and fluorescence microscopy, microplate enzyme activity assays and Western blotting, we studied ATP and ADP production, mitochondrial electron transfer chain complex activities, ATP-sensitive potassium channels (IKATP ), mitochondrial oxidative stress, Ca2+ content, and protein expression in control and ZFHX3 knockdown (KD) HL-1 cells subjected to 1 and 5-Hz pacing for 24 hours. RESULTS Compared with 1-Hz pacing, 5-Hz pacing increased ATP and ADP production, IKATP , phosphorylated adenosine monophosphate-activated protein kinase and inositol 1,4,5-triphosphate (IP3 ) receptor (IP3 R) protein expression. Tachypacing induced mitochondrial oxidative stress and Ca2+ overload in both cell types. Furthermore, under 1- and 5-Hz pacing, ZFHX3 KD cells showed higher IKATP , ATP and ADP production, mitochondrial oxidative stress and Ca2+ content than control cells. Under 5-Hz pacing, 2-aminoethoxydiphenyl borate (2-APB; 3 μmol/L, an IP3 R inhibitor) and MitoTEMPO (10 µmol/L, a mitochondria-targeted antioxidant) reduced ADP and increased ATP production in both cell types; however, only 2-APB significantly reduced mitochondrial Ca2+ overload in control cells. Under 5-Hz pacing, mitochondrial oxidative stress was significantly reduced by both MitoTEMPO and 2-APB and only by 2-APB in control and ZFHX3 KD cells respectively. CONCLUSION ZFHX3 KD cells modulate mitochondrial adaptations to tachypacing in HL-1 cardiomyocytes through Ca2+ overload, oxidative stress and metabolic disorder. Targeting IP3 R signalling or oxidative stress could reduce AF.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Yung‐Kuo Lin
- Division of Cardiology Department of Internal Medicine School of Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Yao‐Chang Chen
- Department of Biomedical Engineering National Defense Medical Center Taipei Taiwan
| | - Wan‐Li Cheng
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory Division of Cardiovascular Medicine Makiminato Central Hospital Okinawa Japan
| | - Yu‐Hsun Kao
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
- Department of Medical Education and Research Wan‐Fang Hospital Taipei Medical University Taipei Taiwan
| | - Yi‐Jen Chen
- Graduate Institute of Clinical Medicine College of Medicine Taipei Medical University Taipei Taiwan
- Cardiovascular Research CenterWan‐Fang HospitalTaipei Medical University Taipei Taiwan
| |
Collapse
|
16
|
Lee TW, Lee TI, Lin YK, Chen YC, Kao YH, Chen YJ. Effect of antidiabetic drugs on the risk of atrial fibrillation: mechanistic insights from clinical evidence and translational studies. Cell Mol Life Sci 2021; 78:923-934. [PMID: 32965513 PMCID: PMC11072414 DOI: 10.1007/s00018-020-03648-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/18/2020] [Accepted: 09/12/2020] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus (DM) is an independent risk factor for atrial fibrillation (AF), which is the most common sustained arrhythmia and is associated with substantial morbidity and mortality. Advanced glycation end product and its receptor activation, cardiac energy dysmetabolism, structural and electrical remodeling, and autonomic dysfunction are implicated in AF pathophysiology in diabetic hearts. Antidiabetic drugs have been demonstrated to possess therapeutic potential for AF. However, clinical investigations of AF in patients with DM have been scant and inconclusive. This article provides a comprehensive review of research findings on the association between DM and AF and critically analyzes the effect of different pharmacological classes of antidiabetic drugs on AF.
Collapse
Affiliation(s)
- Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ting-I Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
17
|
Salazar-Ramírez F, Ramos-Mondragón R, García-Rivas G. Mitochondrial and Sarcoplasmic Reticulum Interconnection in Cardiac Arrhythmia. Front Cell Dev Biol 2021; 8:623381. [PMID: 33585462 PMCID: PMC7876262 DOI: 10.3389/fcell.2020.623381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/30/2020] [Indexed: 12/31/2022] Open
Abstract
Ca2+ plays a pivotal role in mitochondrial energy production, contraction, and apoptosis. Mitochondrial Ca2+-targeted fluorescent probes have demonstrated that mitochondria Ca2+ transients are synchronized with Ca2+ fluxes occurring in the sarcoplasmic reticulum (SR). The presence of specialized proteins tethering SR to mitochondria ensures the local Ca2+ flux between these organelles. Furthermore, communication between SR and mitochondria impacts their functionality in a bidirectional manner. Mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniplex is essential for ATP production and controlled reactive oxygen species levels for proper cellular signaling. Conversely, mitochondrial ATP ensures the proper functioning of SR Ca2+-handling proteins, which ensures that mitochondria receive an adequate supply of Ca2+. Recent evidence suggests that altered SR Ca2+ proteins, such as ryanodine receptors and the sarco/endoplasmic reticulum Ca2+ ATPase pump, play an important role in maintaining proper cardiac membrane excitability, which may be initiated and potentiated when mitochondria are dysfunctional. This recognized mitochondrial role offers the opportunity to develop new therapeutic approaches aimed at preventing cardiac arrhythmias in cardiac disease.
Collapse
Affiliation(s)
- Felipe Salazar-Ramírez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Cardiovascular, Monterrey, Mexico
| | - Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Cardiovascular, Monterrey, Mexico.,TecSalud, Centro de Investigación Biomédica, Hospital Zambrano-Hellion, San Pedro Garza García, Mexico.,TecSalud, Centro de Medicina Funcional, Hospital Zambrano-Hellion, San Pedro Garza García, Mexico
| |
Collapse
|
18
|
Brown SM, Larsen NK, Thankam FG, Agrawal DK. Regulatory role of cardiomyocyte metabolism via AMPK activation in modulating atrial structural, contractile, and electrical properties following atrial fibrillation. Can J Physiol Pharmacol 2020; 99:36-41. [PMID: 33049144 DOI: 10.1139/cjpp-2020-0313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The number of patients diagnosed with atrial fibrillation (AF) has been rising due to increased incidence, enhanced detection methods, and greater survival rates following diagnosis. Due to this increase, AF is now the most commonly diagnosed arrhythmia in clinical practice. AF is characterized by irregular, high-frequency contractions of atrial myocytes that lead to turbulent blood flow and the potential for thrombus formation, stroke, or heart failure. These high-frequency contractions of the atrial myocytes cause an imbalance between metabolic supply and demand. Although advances have been made in understanding the pathophysiology of AF, the etiology and underlying pathogenic mechanism remain unknown. However, recent evidence suggests that cardiomyocyte metabolism involving 5' AMP-activated protein kinase (AMPK) activation is altered in patients with AF. Here, we critically reviewed the current understanding of AMPK activation in AF and how it could affect structural, contractile, and electrophysiological cellular properties in the pathogenesis of AF.
Collapse
Affiliation(s)
- Sean M Brown
- Creighton University School of Medicine, Omaha, NE 68178, USA
| | | | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
19
|
Li B, Po SS, Zhang B, Bai F, Li J, Qin F, Liu N, Sun C, Xiao Y, Tu T, Zhou S, Liu Q. Metformin regulates adiponectin signalling in epicardial adipose tissue and reduces atrial fibrillation vulnerability. J Cell Mol Med 2020; 24:7751-7766. [PMID: 32441464 PMCID: PMC7348162 DOI: 10.1111/jcmm.15407] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 03/31/2020] [Accepted: 04/22/2020] [Indexed: 12/26/2022] Open
Abstract
Epicardial adipose tissue (EAT) remodelling is closely related to the pathogenesis of atrial fibrillation (AF). We investigated whether metformin (MET) prevents AF‐dependent EAT remodelling and AF vulnerability in dogs. A canine AF model was developed by 6‐week rapid atrial pacing (RAP), and electrophysiological parameters were measured. Effective refractory periods (ERP) were decreased in the left and right atrial appendages as well as in the left atrium (LA) and right atrium (RA). MET attenuated the RAP‐induced increase in ERP dispersion, cumulative window of vulnerability, AF inducibility and AF duration. RAP increased reactive oxygen species (ROS) production and nuclear factor kappa‐B (NF‐κB) phosphorylation; up‐regulated interleukin‐6 (IL‐6), tumour necrosis factor‐α (TNF‐α) and transforming growth factor‐β1 (TGF‐β1) levels in LA and EAT; decreased peroxisome proliferator‐activated receptor gamma (PPARγ) and adiponectin (APN) expression in EAT and was accompanied by atrial fibrosis and adipose infiltration. MET reversed these alterations. In vitro, lipopolysaccharide (LPS) exposure increased IL‐6, TNF‐α and TGF‐β1 expression and decreased PPARγ/APN expression in 3T3‐L1 adipocytes, which were all reversed after MET administration. Indirect coculture of HL‐1 cells with LPS‐stimulated 3T3‐L1 conditioned medium (CM) significantly increased IL‐6, TNF‐α and TGF‐β1 expression and decreased SERCA2a and p‐PLN expression, while LPS + MET CM and APN treatment alleviated the inflammatory response and sarcoplasmic reticulum Ca2+ handling dysfunction. MET attenuated the RAP‐induced increase in AF vulnerability, remodelling of atria and EAT adipokines production profiles. APN may play a key role in the prevention of AF‐dependent EAT remodelling and AF vulnerability by MET.
Collapse
Affiliation(s)
- Biao Li
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Sunny S Po
- Heart Rhythm Institute and Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Baojian Zhang
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China.,Department of Cardiology, the Affiliated Chinese Medicine Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Province, China
| | - Fan Bai
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Jiayi Li
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Fen Qin
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Na Liu
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Chao Sun
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Yichao Xiao
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Tao Tu
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Shenghua Zhou
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Qiming Liu
- Department of Cardiology/Cardiac Catheterization Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| |
Collapse
|
20
|
Linking cellular energy state to atrial fibrillation pathogenesis: Potential role of adenosine monophosphate-activated protein kinase. Heart Rhythm 2020; 17:1398-1404. [PMID: 32268208 DOI: 10.1016/j.hrthm.2020.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/28/2020] [Indexed: 01/01/2023]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is the cellular stress-sensing molecule. Apart from maintaining cellular energy balance, AMPK controls expression and regulation of ion channels and ion transporters, including cytosolic Ca2+ handling proteins. Emerging evidence suggests that metabolic impairment plays a crucial role in the pathogenesis of atrial fibrillation. AMPK activation is thought to be protective by preventing metabolic stress, favorably modulating membrane electrophysiology including cytosolic Ca2+ dynamics; preventing cellular growth; and hypertrophic remodeling. This review considers current concepts and evidence from clinical and experimental studies regarding the role of AMPK in atrial fibrillation.
Collapse
|
21
|
Scholz B, Schulte JS, Hamer S, Himmler K, Pluteanu F, Seidl MD, Stein J, Wardelmann E, Hammer E, Völker U, Müller FU. HDAC (Histone Deacetylase) Inhibitor Valproic Acid Attenuates Atrial Remodeling and Delays the Onset of Atrial Fibrillation in Mice. Circ Arrhythm Electrophysiol 2019; 12:e007071. [PMID: 30879335 PMCID: PMC6426346 DOI: 10.1161/circep.118.007071] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is available in the text. Background: A structural, electrical and metabolic atrial remodeling is central in the development of atrial fibrillation (AF) contributing to its initiation and perpetuation. In the heart, HDACs (histone deacetylases) control remodeling associated processes like hypertrophy, fibrosis, and energy metabolism. Here, we analyzed, whether the HDAC class I/IIa inhibitor valproic acid (VPA) is able to attenuate atrial remodeling in CREM-IbΔC-X (cAMP responsive element modulator isoform IbΔC-X) transgenic mice, a mouse model of extensive atrial remodeling with age-dependent progression from spontaneous atrial ectopy to paroxysmal and finally long-lasting AF. Methods: VPA was administered for 7 or 25 weeks to transgenic and control mice. Atria were analyzed macroscopically and using widefield and electron microscopy. Action potentials were recorded from atrial cardiomyocytes using patch-clamp technique. ECG recordings documented the onset of AF. A proteome analysis with consecutive pathway mapping identified VPA-mediated proteomic changes and related pathways. Results: VPA attenuated many components of atrial remodeling that are present in transgenic mice, animal AF models, and human AF. VPA significantly (P<0.05) reduced atrial dilatation, cardiomyocyte enlargement, atrial fibrosis, and the disorganization of myocyte’s ultrastructure. It significantly reduced the occurrence of atrial thrombi, reversed action potential alterations, and finally delayed the onset of AF by 4 to 8 weeks. Increased histone H4-acetylation in atria from VPA-treated transgenic mice verified effective in vivo HDAC inhibition. Cardiomyocyte-specific genetic inactivation of HDAC2 in transgenic mice attenuated the ultrastructural disorganization of myocytes comparable to VPA. Finally, VPA restrained dysregulation of proteins in transgenic mice that are involved in a multitude of AF relevant pathways like oxidative phosphorylation or RhoA (Ras homolog gene family, member A) signaling and disease functions like cardiac fibrosis and apoptosis of muscle cells. Conclusions: Our results suggest that VPA, clinically available, well-tolerated, and prescribed to many patients for years, has the therapeutic potential to delay the development of atrial remodeling and the onset of AF in patients at risk.
Collapse
Affiliation(s)
- Beatrix Scholz
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Jan Sebastian Schulte
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Sabine Hamer
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Kirsten Himmler
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Florentina Pluteanu
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Matthias Dodo Seidl
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Juliane Stein
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Germany (E.W.)
| | - Elke Hammer
- Interfaculty Institute of Genetics und Functional Genomics, University Medicine Greifswald, Germany (E.H., U.V.).,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany (E.H., U.V.)
| | - Uwe Völker
- Interfaculty Institute of Genetics und Functional Genomics, University Medicine Greifswald, Germany (E.H., U.V.).,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany (E.H., U.V.)
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| |
Collapse
|
22
|
Bai F, Liu Y, Tu T, Li B, Xiao Y, Ma Y, Qin F, Xie J, Zhou S, Liu Q. Metformin regulates lipid metabolism in a canine model of atrial fibrillation through AMPK/PPAR-α/VLCAD pathway. Lipids Health Dis 2019; 18:109. [PMID: 31077199 PMCID: PMC6511207 DOI: 10.1186/s12944-019-1059-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Background Atrial lipid metabolic remodeling is critical for the process of atrial fibrillation (AF). Abnormal Fatty acid (FA) metabolism in cardiomyocytes is involved in the pathogenesis of AF. MET (Metformin), an AMPK (AMP-activated protein kinase) activator, has been found to be associated with a decreased risk of AF in patients with type 2 diabetes. However, the specific mechanism remains unknown. Methods Fifteen mongrel dogs were divided into three groups: SR, ARP (pacing with 800 beats/min for 6 h), ARP plus MET (treated with MET (100 mg/kg/day) for two weeks before pacing). We assessed metabolic factors, speed limiting enzymes circulating biochemical metabolites (substrates and products), atrial electrophysiology and accumulation of lipid droplets. Results The expression of AMPK increased in the ARP group and significantly increased in the MET+ARP group comparing to the SR group. In the ARP group, the expressions of PPARα、PGC-1α and VLCAD were down-regulated, while the concentration of free fatty acid and triglyceride and the lipid deposition in LAA (left atrial appendage) increased. Moreover, AERP and AERPd have also been found abnormally in this process. Pretreatment with MET before receiving ARP reversed the alterations aforementioned. Conclusions The FA metabolism in LAA is altered in the ARP group, mainly characterized by the abnormal expression of the rate-limiting enzyme. Metformin reduces lipid accumulation and promotes β-oxidation of FA in AF models partially through AMPK/PPAR-α/VLCAD pathway. Our study indicates that MET may inhibit the FA lipid metabolic remodeling in AF.
Collapse
Affiliation(s)
- Fan Bai
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yaozhong Liu
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Tao Tu
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Biao Li
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yichao Xiao
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yingxu Ma
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Fen Qin
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Jing Xie
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Shenghua Zhou
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Qiming Liu
- Department of Cardiology/Cardiac Catheterization Lab, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China.
| |
Collapse
|
23
|
Lin ZH, Li YC, Wu SJ, Zheng C, Lin YZ, Lian H, Lin WQ, Lin JF. Eliciting α7-nAChR exerts cardioprotective effects on ischemic cardiomyopathy via activation of AMPK signalling. J Cell Mol Med 2019; 23:4746-4758. [PMID: 31062470 PMCID: PMC6584557 DOI: 10.1111/jcmm.14363] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/10/2019] [Accepted: 04/14/2019] [Indexed: 01/23/2023] Open
Abstract
Our previous studies have reported that agonist of α7 nicotinic acetylcholine receptors prevented electrophysiological dysfunction of rats with ischaemic cardiomyopathy (ICM) by eliciting the cholinergic anti‐inflammatory pathway (CAP). Adenosine monophosphate‐activated protein kinase (AMPK) signalling is widely recognized exerting cardioprotective effect in various cardiomyopathy. Here, we aimed to investigate whether the protective effects of the CAP are associated with AMPK signalling in ICM. In vivo, coronary artery of rats was ligated for 4 weeks to induce the ICM and then treated with PNU‐282987 (CAP agonist) and BML‐275 dihydrochloride (AMPK antagonist) for 4 weeks. In vitro, primary macrophages harvested from rats were induced inflammation by Lipopolysaccharide (LPS) treatment and then treated with PNU‐282987 and BML‐275 dihydrochloride. In vivo, exciting CAP by PUN‐282987 elicited an activation of AMPK signalling, alleviated ventricular remodeling, modified the cardiac electrophysiological function, reduced the cardiac expression of collagens and inflammatory cytokines and maintained the integrity of ultrastructure in the ischemic heart. However, the benefits of CAP excitation were blunted by AMPK signaling antagonization. In vitro, excitation of the CAP was observed inhibiting the nuclear transfer of NF‐κB p65 of macrophages and promoting the transformation of Ly‐6Chigh macrophages into Ly‐6Clow macrophages. However, inhibiting AMPK signalling by BML‐275 dihydrochloride reversed the CAP effect on LPS‐treated macrophages. Finally, our findings suggest that eliciting the CAP modulates the inflammatory response in ICM through regulating AMPK signalling.
Collapse
Affiliation(s)
- Zhong-Hao Lin
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue-Chun Li
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shu-Jie Wu
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cheng Zheng
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuan-Zheng Lin
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hao Lian
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei-Qian Lin
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia-Feng Lin
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
24
|
Qian C, Li H, Chang D, Wei B, Wang Y. Identification of functional lncRNAs in atrial fibrillation by integrative analysis of the lncRNA-mRNA network based on competing endogenous RNAs hypothesis. J Cell Physiol 2018; 234:11620-11630. [PMID: 30478836 DOI: 10.1002/jcp.27819] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/06/2018] [Indexed: 12/14/2022]
Abstract
A mounting body of evidence has suggested that long noncoding RNAs (lncRNAs) play critical roles in human diseases by acting as competing endogenous RNAs (ceRNAs). However, the functions and ceRNA mechanisms of lncRNAs in atrial fibrillation (AF) remain to date unclear. In this study, we constructed an AF-related lncRNA-mRNA network (AFLMN) based on ceRNA theory, by integrating probe reannotation pipeline and microRNA (miRNA)-target regulatory interactions. Two lncRNAs with central topological properties in the AFLMN were first obtained. By using bidirectional hierarchical clustering, we identified two modules containing four lncRNAs, which were significantly enriched in many known pathways of AF. To elucidate the ceRNA interactions in certain disease or normal condition, the dysregulated lncRNA-mRNA crosstalks in AF were further analyzed, and six hub lncRNAs were obtained from the network. Furthermore, random walk analysis of the AFLMN suggested that lncRNA RP11-296O14.3 may function importantly in the pathological process of AF. All these eight lncRNAs that were identified from previous steps (RP11-363E7.4, GAS5, RP11-410L14.2, HAGLR, RP11-421L21.3, RP11-111K18.2, HOTAIRM1, and RP11-296O14.3) exhibited a strong diagnostic power for AF. The results of our study provide new insights into the functional roles and regulatory mechanisms of lncRNAs in AF, and facilitate the discovery of novel diagnostic biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hang Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Danqi Chang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Baozhu Wei
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yanggan Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.,Medical Research Institute of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
25
|
Zhang Y, Zhang S, Liu Z, Zhao X, Yuan Y, Sheng L, Li Y. Resveratrol prevents atrial fibrillation by inhibiting atrial structural and metabolic remodeling in collagen-induced arthritis rats. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:1179-1190. [PMID: 30135998 DOI: 10.1007/s00210-018-1554-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 08/09/2018] [Indexed: 12/13/2022]
Abstract
Rheumatoid arthritis (RA) causes atrial remodeling that induces the occurrence and maintenance of atrial fibrillation (AF). In this study, we explored the influence of RA on atrial fibrillation and the potential therapeutic effects of resveratrol in a rat model. The following three groups of female Wistar rats (8 weeks old) were used in this study: control, collagen-induced arthritis (CIA), and resveratrol. Rats in the CIA and resveratrol groups were injected twice with type II collagen in Freund's incomplete adjuvant. Three weeks after the second injection, resveratrol (10 mg kg-1 day-1) was administered for 4 weeks. Subsequently, atrial electrophysiological parameters were measured. Levels of inflammatory factors in the atria and serum were measured. Atrial histopathological changes were assessed using microscopy, and cardiomyocyte apoptosis and fibrosis were assessed using TUNEL and Masson's staining. Apoptosis-related and fibrosis-related proteins were assessed using Western blotting. Atrial adenosine triphosphate (ATP) and free fatty acid (FFA) levels were tested using ELISA. Glycogen accumulation and metabolism-related protein expression were assessed. AF inducibility and duration were markedly increased in CIA rats and were reduced by resveratrol. CIA also increased the atrial and serum IL-6 and TNF-a levels and induced atrial apoptosis and fibrosis, which were attenuated by resveratrol. Moreover, CIA induced the impairment of atrial energy metabolism by inhibiting the AMPK/PGC-1α pathway, which was reversed by resveratrol. Resveratrol protects against RA-induced atrial structural and metabolic remodeling, which may provide a new potential therapeutic treatment for RA-related AF.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Song Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Zonghong Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Xinbo Zhao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yue Yuan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Li Sheng
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yue Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China.
- Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
26
|
Li J, Bai F, Li B, Liu Q. AMPK: An Ambiguous Position for Atrial Fibrillation. Trends Endocrinol Metab 2018; 29:369. [PMID: 29573948 DOI: 10.1016/j.tem.2018.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 02/03/2018] [Accepted: 02/20/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Jiayi Li
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Fan Bai
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Biao Li
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qiming Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Li X, Yu L, Gao J, Bi X, Zhang J, Xu S, Wang M, Chen M, Qiu F, Fu G. Apelin Ameliorates High Glucose-Induced Downregulation of Connexin 43 via AMPK-Dependent Pathway in Neonatal Rat Cardiomyocytes. Aging Dis 2018; 9:66-76. [PMID: 29392082 PMCID: PMC5772859 DOI: 10.14336/ad.2017.0426] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/26/2017] [Indexed: 11/10/2022] Open
Abstract
Diabetes Mellitus is a common disorder, with increasing risk of cardiac arrhythmias. Studies have shown that altered connexin expression and gap junction remodeling under hyperglycemia contribute to the high prevalence of cardiac arrhythmias and even sudden death. Connexin 43 (Cx43), a major protein that assembles to form cardiac gap junctions, has been found to be downregulated under high glucose conditions, along with inhibition of gap junctional intercellular communication (GJIC). While, apelin, a beneficial adipokine, increases Cx43 protein expression in mouse and human embryonic stem cells during cardiac differentiation. However, it remains unknown whether apelin influences GJIC capacity in cardiomyocytes. Here, using Western blotting and dye transfer assays, we found that Cx43 protein expression was reduced and GJIC was impaired after treatment with high glucose, which, however, could be abrogated after apelin treatment for 48 h. We also found that apelin increased Cx43 expression under normal glucose. Real-time PCR showed that the Cx43 mRNA was not significantly affected under high glucose conditions in the presence of apelin or high glucose and apelin. High glucose decreased the phosphorylation of AMPKα; however, apelin activated AMPKα. Interestingly, we found that Cx43 expression was increased after treatment with AICAR, an activator of AMPK signaling. AMPKα inhibition mediated with transfection of siRNA-AMPKα1 and siRNA-AMPKα2 abolished the protective effect of apelin on Cx43 expression. Our data suggest that apelin attenuates high glucose-induced Cx43 downregulation and improves the loss of functional gap junctions partly through the AMPK pathway.
Collapse
Affiliation(s)
- Xiaoting Li
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lu Yu
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Gao
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xukun Bi
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juhong Zhang
- 2Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shiming Xu
- 3Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Meihui Wang
- 4Biomedical Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengmeng Chen
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fuyu Qiu
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guosheng Fu
- 1Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Kim N, Jung Y, Nam M, Sun Kang M, Lee MK, Cho Y, Choi EK, Hwang GS, Soo Kim H. Angiotensin II affects inflammation mechanisms via AMPK-related signalling pathways in HL-1 atrial myocytes. Sci Rep 2017; 7:10328. [PMID: 28871102 PMCID: PMC5583339 DOI: 10.1038/s41598-017-09675-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 07/27/2017] [Indexed: 11/30/2022] Open
Abstract
Inflammation is a common cause of cardiac arrhythmia. Angiotensin ІІ (Ang ІІ) is a major contributing factor in the pathogenesis of cardiac inflammation; however, its underlying molecular mechanism remains unclear. Here, we explored the effect of Ang ІІ on inflammatory mechanisms and oxidative stress using HL-1 atrial myocytes. We showed that Ang ІІ activated c-Jun N-terminal kinase (JNK) phosphorylation and other inflammatory markers, such as transforming growth factor-β1 (TGF-β1) and tumor necrosis factor-α (TNF-α). Ang ІІ decreased oxygen consumption rate, which resulted in reactive oxygen species (ROS) generation and inhibition of ROS blocked Ang II-mediated JNK phosphorylation and TGF-β1 induction. Ang ІІ induced the expression of its specific receptor, AT1R. Ang II-induced intracellular calcium production associated with Ang ІІ-mediated signalling pathways. In addition, the generated ROS and calcium stimulated AMPK phosphorylation. Inhibiting AMPK blocked Ang II-mediated JNK and TGF-β signalling pathways. Ang ІІ concentration, along with TGF-β1 and tumor necrosis factor-α levels, was slightly increased in plasma of patients with atrial fibrillation. Taken together, these results suggest that Ang ІІ induces inflammation mechanisms through an AMPK-related signalling pathway. Our results provide new molecular targets for the development of therapeutics for inflammation-related conditions, such as atrial fibrillation.
Collapse
Affiliation(s)
- Nami Kim
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Youngae Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Miso Nam
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Mi Sun Kang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Min Kyung Lee
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea
| | - Youngjin Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Eue-Keun Choi
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 120-140, Republic of Korea. .,Chemistry & Nanoscience, Ewha Womans University, Seoul, Republic of Korea.
| | - Hyeon Soo Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| |
Collapse
|
29
|
Harada M, Melka J, Sobue Y, Nattel S. Metabolic Considerations in Atrial Fibrillation ― Mechanistic Insights and Therapeutic Opportunities ―. Circ J 2017; 81:1749-1757. [DOI: 10.1253/circj.cj-17-1058] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - Jonathan Melka
- Department of Medicine and Research Center, Montreal Heart Institute
- Université de Montréal
- Department of Pharmacology and Therapeutics, McGill University
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen
| | - Yoshihiro Sobue
- Department of Medicine and Research Center, Montreal Heart Institute
- Université de Montréal
- Department of Pharmacology and Therapeutics, McGill University
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen
| | - Stanley Nattel
- Department of Medicine and Research Center, Montreal Heart Institute
- Université de Montréal
- Department of Pharmacology and Therapeutics, McGill University
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen
| |
Collapse
|
30
|
Jankyova S, Rubintova D, Janosikova L, Panek P, Foltanova T, Kralova E. The Effects of Pycnogenol® as Add-on Drug to Metformin Therapy in Diabetic Rats. Phytother Res 2016; 30:1354-1361. [PMID: 27170051 DOI: 10.1002/ptr.5639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 01/03/2025]
Abstract
The progression of diabetes mellitus leads in time to the development of serious cardiovascular complications. Pycnogenol® (PYC) belongs to strong antioxidants that may interfere with different pathways playing an important role in diseases associated with oxidative stress. Metformin (MET), commonly used antidiabetic drug, has cardio-protective effects via activation of AMP kinase (AMPK). In our study, we examined the effects of PYC as add-on drug to metformin therapy in streptozotocin (STZ)-induced diabetic rats. Our results revealed that both used agents, PYC and MET, showed improvement of blood glucose levels, vascular reactivity, left ventricular hypertrophy, expression of AMPK, glucose transporter 4 (GLUT4) and calcium/calmodulin-dependent protein kinase II (CaMKII) in left ventricle of the hearts. However, the combination of these interventions has failed to possess higher efficacy. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Stanislava Jankyova
- Department of Pharmacology and Toxicology, Comenius University in Bratislava, Faculty of Pharmacy, Slovak Republic
| | - Dominika Rubintova
- Department of Pharmacology and Toxicology, Comenius University in Bratislava, Faculty of Pharmacy, Slovak Republic
| | - Lenka Janosikova
- Department of Pharmacology and Toxicology, Comenius University in Bratislava, Faculty of Pharmacy, Slovak Republic
| | - Peter Panek
- Department of Pharmacology and Toxicology, Comenius University in Bratislava, Faculty of Pharmacy, Slovak Republic
| | - Tatiana Foltanova
- Department of Pharmacology and Toxicology, Comenius University in Bratislava, Faculty of Pharmacy, Slovak Republic
| | - Eva Kralova
- Department of Pharmacology and Toxicology, Comenius University in Bratislava, Faculty of Pharmacy, Slovak Republic
| |
Collapse
|
31
|
Energy metabolic alterations in the progression of atrial fibrillation: Potential role of AMP-activated protein kinase as a critical regulator. Int J Cardiol 2016; 212:14-5. [DOI: 10.1016/j.ijcard.2016.03.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/19/2015] [Accepted: 03/12/2016] [Indexed: 01/16/2023]
|
32
|
Qiu J, Zhou S, Liu Q. Phosphorylated AMP-activated protein kinase slows down the atrial fibrillation progression by activating Connexin43. Int J Cardiol 2016; 208:56-7. [PMID: 26828380 DOI: 10.1016/j.ijcard.2016.01.201] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/15/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Jie Qiu
- Department of Cardiology/Cardiac Catheterisation Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province 410011, China
| | - Shenghua Zhou
- Department of Cardiology/Cardiac Catheterisation Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province 410011, China
| | - Qiming Liu
- Department of Cardiology/Cardiac Catheterisation Lab, Second Xiangya Hospital, Central South University, Changsha City, Hunan Province 410011, China.
| |
Collapse
|
33
|
Kim GE, Ross JL, Xie C, Su KN, Zaha VG, Wu X, Palmeri M, Ashraf M, Akar JG, Russell KS, Akar FG, Young LH. LKB1 deletion causes early changes in atrial channel expression and electrophysiology prior to atrial fibrillation. Cardiovasc Res 2015; 108:197-208. [PMID: 26378152 PMCID: PMC4571838 DOI: 10.1093/cvr/cvv212] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 07/09/2015] [Accepted: 07/14/2015] [Indexed: 12/19/2022] Open
Abstract
AIMS Liver kinase B1 (LKB1) is a protein kinase that activates the metabolic regulator AMP-activated protein kinase (AMPK) and other related kinases. Deletion of LKB1 in mice leads to cardiomyopathy and atrial fibrillation (AF). However, the specific role of the LKB1 pathway in early atrial biology remains unknown. Thus, we investigated whether LKB1 deletion altered atrial channel expression and electrophysiological function in a cardiomyocyte-specific knockout mouse model. METHODS AND RESULTS We performed a systematic comparison of αMHC-Cre LKB1(fl/fl) and littermate LKB1(fl/fl) male mice. This included analysis of gene expression, histology, and echocardiography, as well as cellular and tissue-level electrophysiology using patch-clamp recordings in vitro, optical mapping ex vivo, and ECG recordings in vivo. At postnatal day 1, atrial depolarization was prolonged, and Nav1.5 and Cx40 expression were markedly down-regulated in MHC-Cre LKB1(fl/fl) mice. Inward sodium current density was significantly decreased in MHC-Cre LKB1(fl/fl) neonatal atrial myocytes. Subsequently, additional alterations in atrial channel expression, atrial fibrosis, and spontaneous onset of AF developed by 2 weeks of age. In adult mice, abnormalities of interatrial conduction and bi-atrial electrical coupling were observed, likely promoting the perpetuation of AF. Mice with AMPK-inactivated hearts demonstrated modest overlap in channel expression with MHC-Cre LKB1(fl/fl) hearts, but retained normal structure, electrophysiological function and contractility. CONCLUSIONS Deletion of LKB1 causes early defects in atrial channel expression, action potential generation and conduction, which precede widespread atrial remodelling, fibrosis and AF. LKB1 is critical for normal atrial growth and electrophysiological function.
Collapse
Affiliation(s)
- Grace E Kim
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Jenna L Ross
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Chaoqin Xie
- Cardiovascular Research Center, Mt. Sinai School of Medicine, New York, NY 10029, USA
| | - Kevin N Su
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Vlad G Zaha
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Xiaohong Wu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Monica Palmeri
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Mohammed Ashraf
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Joseph G Akar
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Kerry S Russell
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Fadi G Akar
- Cardiovascular Research Center, Mt. Sinai School of Medicine, New York, NY 10029, USA
| | - Lawrence H Young
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
34
|
Harada M, Tadevosyan A, Qi X, Xiao J, Liu T, Voigt N, Karck M, Kamler M, Kodama I, Murohara T, Dobrev D, Nattel S. Atrial Fibrillation Activates AMP-Dependent Protein Kinase and its Regulation of Cellular Calcium Handling: Potential Role in Metabolic Adaptation and Prevention of Progression. J Am Coll Cardiol 2015; 66:47-58. [PMID: 26139058 DOI: 10.1016/j.jacc.2015.04.056] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 04/05/2015] [Accepted: 04/23/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is associated with metabolic stress, which activates adenosine monophosphate-regulated protein kinase (AMPK). OBJECTIVES This study sought to examine AMPK response to AF and associated metabolic stress, along with consequences for atrial cardiomyocyte Ca(2+) handling. METHODS Calcium ion (Ca(2+)) transients (CaTs) and cell shortening (CS) were measured in dog and human atrial cardiomyocytes. AMPK phosphorylation and AMPK association with Ca(2+)-handling proteins were evaluated by immunoblotting and immunoprecipitation. RESULTS CaT amplitude and CS decreased at 4-min glycolysis inhibition (GI) but returned to baseline at 8 min, suggesting cellular adaptation to metabolic stress, potentially due to AMPK activation. GI increased AMPK-activating phosphorylation, and an AMPK inhibitor, compound C (CompC), abolished the adaptation of CaT and CS to GI. The AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) increased CaT amplitude and CS, restoring CompC-induced CaT and CS decreases. CompC decreased L-type calcium channel current (ICa,L), along with ICa,L-triggered CaT amplitude and sarcoplasmic reticulum (SR) Ca(2+) content under voltage clamp conditions in dog cells and suppressed CaT and ICa,L in human cardiomyocytes. Small interfering ribonucleic acid-based AMPK knockdown decreased CaT amplitude in neonatal rat cardiomyocytes. L-type Ca(2+) channel α subunits coimmunoprecipitated with AMPKα. Atrial AMPK-activating phosphorylation was enhanced by 1 week of electrically maintained AF in dogs; fractional AMPK phosphorylation was increased in paroxysmal AF and reduced in longstanding persistent AF patients. CONCLUSIONS AMPK is activated by metabolic stress and AF, and helps maintain the intactness of atrial ICa,L, Ca(2+) handling, and cell contractility. AMPK contributes to the atrial compensatory response to AF-related metabolic stress; AF-related metabolic responses may be an interesting new therapeutic target.
Collapse
Affiliation(s)
- Masahide Harada
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada; Department of Cardiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Artavazd Tadevosyan
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada
| | - Xiaoyan Qi
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada
| | - Jiening Xiao
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada
| | - Tao Liu
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Niels Voigt
- Institute of Pharmacology, West German Cardiac and Vascular Center, School of Medicine, University Duisburg-Essen, Essen, Germany
| | - Matthias Karck
- Department of Cardiac Surgery, Heidelberg University, Heidelberg, Germany
| | - Markus Kamler
- Department of Cardiac Surgery, Huttrop Heart Center, Essen, Germany
| | | | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Cardiac and Vascular Center, School of Medicine, University Duisburg-Essen, Essen, Germany
| | - Stanley Nattel
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
35
|
Irregular rhythm and atrial metabolism are key for the evolution of proarrhythmic atrial remodeling in atrial fibrillation. Basic Res Cardiol 2015; 110:41. [PMID: 26018792 DOI: 10.1007/s00395-015-0498-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 05/22/2015] [Indexed: 12/18/2022]
|
36
|
Arrhythmia causes lipid accumulation and reduced glucose uptake. Basic Res Cardiol 2015; 110:40. [DOI: 10.1007/s00395-015-0497-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 04/16/2015] [Accepted: 05/22/2015] [Indexed: 12/17/2022]
|
37
|
Ghezelbash S, Molina CE, Dobrev D. Altered atrial metabolism: an underappreciated contributor to the initiation and progression of atrial fibrillation. J Am Heart Assoc 2015; 4:e001808. [PMID: 25773300 PMCID: PMC4392451 DOI: 10.1161/jaha.115.001808] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Shokoufeh Ghezelbash
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany (S.G., C.E.M., D.D.)
| | - Cristina E Molina
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany (S.G., C.E.M., D.D.)
| | - Dobromir Dobrev
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany (S.G., C.E.M., D.D.) DZHK, associated site UniversityDuisburg-Essen, German Centre for Cardiovascular Research, Germany (D.D.)
| |
Collapse
|
38
|
Saks V, Schlattner U, Tokarska-Schlattner M, Wallimann T, Bagur R, Zorman S, Pelosse M, Santos PD, Boucher F, Kaambre T, Guzun R. Systems Level Regulation of Cardiac Energy Fluxes Via Metabolic Cycles: Role of Creatine, Phosphotransfer Pathways, and AMPK Signaling. SYSTEMS BIOLOGY OF METABOLIC AND SIGNALING NETWORKS 2014. [DOI: 10.1007/978-3-642-38505-6_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Corradi F, Paolini L, De Caterina R. Ranolazine in the prevention of anthracycline cardiotoxicity. Pharmacol Res 2014; 79:88-102. [DOI: 10.1016/j.phrs.2013.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 11/06/2013] [Accepted: 11/06/2013] [Indexed: 12/19/2022]
|