1
|
Peng H, Yuan J, Wang Z, Mo B, Wang Y, Wang Y, Wang Q. NR4A3 prevents diabetes induced atrial cardiomyopathy by maintaining mitochondrial energy metabolism and reducing oxidative stress. EBioMedicine 2024; 106:105268. [PMID: 39098108 PMCID: PMC11334830 DOI: 10.1016/j.ebiom.2024.105268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Atrial cardiomyopathy (ACM) is responsible for atrial fibrillation (AF) and thromboembolic events. Diabetes mellitus (DM) is an important risk factor for ACM. However, the potential mechanism between ACM and DM remains elusive. METHODS Atrial tissue samples were obtained from patients diagnosed with AF or sinus rhythm (SR) to assess alterations in NR4A3 expression, and then two distinct animal models were generated by subjecting Nr4a3-/- mice and WT mice to a high-fat diet (HFD) and Streptozotocin (STZ), while db/db mice were administered AAV9-Nr4a3 or AAV9-ctrl. Subsequently, in vivo and in vitro experiments were conducted to assess the impact of NR4A3 on diabetes-induced atrial remodeling through electrophysiological, biological, and histological analyses. RNA sequencing (RNA-seq) and metabolomics analysis were employed to unravel the downstream mechanisms. FINDINGS The expression of NR4A3 was significantly decreased in atrial tissues of both AF patients and diabetic mice compared to their respective control groups. NR4A3 deficiency exacerbated atrial hypertrophy and atrial fibrosis, and increased susceptibility to pacing-induced AF. Conversely, overexpression of NR4A3 alleviated atrial structural remodeling and reduced AF induction rate. Mechanistically, we confirmed that NR4A3 improves mitochondrial energy metabolism and reduces oxidative stress injury by preserving the transcriptional expression of Sdha, thereby exerting a protective influence on atrial remodeling induced by diabetes. INTERPRETATION Our data confirm that NR4A3 plays a protective role in atrial remodeling caused by diabetes, so it may be a new target for treating ACM. FUNDING This study was supported by the major research program of National Natural Science Foundation of China (NSFC) No: 82370316 (to Q-S. W.), No. 81974041 (to Y-P. W.), and No. 82270447 (to Y-P. W.) and Fundation of Shanghai Hospital Development Center (No. SHDC2022CRD044 to Q-S. W.).
Collapse
Affiliation(s)
- Hong Peng
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiali Yuan
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhengshuai Wang
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Binfeng Mo
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yihui Wang
- The Department of Radiology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuepeng Wang
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Qunshan Wang
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
2
|
Duan HY, Barajas-Martinez H, Antzelevitch C, Hu D. The potential anti-arrhythmic effect of SGLT2 inhibitors. Cardiovasc Diabetol 2024; 23:252. [PMID: 39010053 PMCID: PMC11251349 DOI: 10.1186/s12933-024-02312-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/16/2024] [Indexed: 07/17/2024] Open
Abstract
Sodium-glucose cotransporter type 2 inhibitors (SGLT2i) were initially recommended as oral anti-diabetic drugs to treat type 2 diabetes (T2D), by inhibiting SGLT2 in proximal tubule and reduce renal reabsorption of sodium and glucose. While many clinical trials demonstrated the tremendous potential of SGLT2i for cardiovascular diseases. 2022 AHA/ACC/HFSA guideline first emphasized that SGLT2i were the only drug class that can cover the entire management of heart failure (HF) from prevention to treatment. Subsequently, the antiarrhythmic properties of SGLT2i have also attracted attention. Although there are currently no prospective studies specifically on the anti-arrhythmic effects of SGLT2i. We provide clues from clinical and fundamental researches to identify its antiarrhythmic effects, reviewing the evidences and mechanism for the SGLT2i antiarrhythmic effects and establishing a novel paradigm involving intracellular sodium, metabolism and autophagy to investigate the potential mechanisms of SGLT2i in mitigating arrhythmias.
Collapse
Affiliation(s)
- Hong-Yi Duan
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, Hubei, China
| | - Hector Barajas-Martinez
- Lankenau Institute for Medical Research, Lankenau Heart Institute, Wynnewood, PA, 19096, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, 19107, USA
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, Lankenau Heart Institute, Wynnewood, PA, 19096, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, 19107, USA
| | - Dan Hu
- Department of Cardiology and Cardiovascular Research Institute, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, Hubei, China.
| |
Collapse
|
3
|
Sarkar A, Fanous KI, Marei I, Ding H, Ladjimi M, MacDonald R, Hollenberg MD, Anderson TJ, Hill MA, Triggle CR. Repurposing Metformin for the Treatment of Atrial Fibrillation: Current Insights. Vasc Health Risk Manag 2024; 20:255-288. [PMID: 38919471 PMCID: PMC11198029 DOI: 10.2147/vhrm.s391808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Metformin is an orally effective anti-hyperglycemic drug that despite being introduced over 60 years ago is still utilized by an estimated 120 to 150 million people worldwide for the treatment of type 2 diabetes (T2D). Metformin is used off-label for the treatment of polycystic ovary syndrome (PCOS) and for pre-diabetes and weight loss. Metformin is a safe, inexpensive drug with side effects mostly limited to gastrointestinal issues. Prospective clinical data from the United Kingdom Prospective Diabetes Study (UKPDS), completed in 1998, demonstrated that metformin not only has excellent therapeutic efficacy as an anti-diabetes drug but also that good glycemic control reduced the risk of micro- and macro-vascular complications, especially in obese patients and thereby reduced the risk of diabetes-associated cardiovascular disease (CVD). Based on a long history of clinical use and an excellent safety record metformin has been investigated to be repurposed for numerous other diseases including as an anti-aging agent, Alzheimer's disease and other dementias, cancer, COVID-19 and also atrial fibrillation (AF). AF is the most frequently diagnosed cardiac arrythmia and its prevalence is increasing globally as the population ages. The argument for repurposing metformin for AF is based on a combination of retrospective clinical data and in vivo and in vitro pre-clinical laboratory studies. In this review, we critically evaluate the evidence that metformin has cardioprotective actions and assess whether the clinical and pre-clinical evidence support the use of metformin to reduce the risk and treat AF.
Collapse
Affiliation(s)
- Aparajita Sarkar
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Kareem Imad Fanous
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Isra Marei
- Department of Pharmacology & Medical Education, Weill Cornell Medicine- Qatar, Doha, Qatar
| | - Hong Ding
- Department of Pharmacology & Medical Education, Weill Cornell Medicine- Qatar, Doha, Qatar
| | - Moncef Ladjimi
- Department of Biochemistry & Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Ross MacDonald
- Health Sciences Library, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, and Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Todd J Anderson
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael A Hill
- Dalton Cardiovascular Research Center & Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Chris R Triggle
- Department of Pharmacology & Medical Education, Weill Cornell Medicine- Qatar, Doha, Qatar
| |
Collapse
|
4
|
Vogt S, Ramzan R, Cybulski P, Rhiel A, Weber P, Ruppert V, Irqsusi M, Rohrbach S, Niemann B, Mirow N, Rastan AJ. The ratio of cytochrome c oxidase subunit 4 isoform 4I1 and 4I2 mRNA is changed in permanent atrial fibrillation. ESC Heart Fail 2024; 11:1525-1539. [PMID: 38149324 PMCID: PMC11098639 DOI: 10.1002/ehf2.14607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 08/11/2023] [Accepted: 11/16/2023] [Indexed: 12/28/2023] Open
Abstract
AIMS The conditions of hypoxia are suggested to induce permanent atrial fibrillation (AF). The regulation of COX4I2 and COX4I1 depends on oxygen availability in tissues. A role of COX4I2 in the myocardium of AF patients is supposed for pathogenesis of AF and subsequent alterations in the electron transfer chain (ETC) under hypoxia. METHODS AND RESULTS In vitro, influence of hypoxia on HeLa 53 cells was studied and elevated parts of COX 4I2 were confirmed. Myocardial biopsies were taken ex vivo from the patients' Right Atria with SR (n = 31) and AF (n = 11), respectively. RT- PCR for mRNA expresson, mitochondrial respiration by polarography and the protein content of cytochrome c oxidase (CytOx) subunit 4I1 and CytOx subunit 4I2 by ELISA were studied. Clinical data were correlated to the findings of gene expressions in parallel. Patients with permanent AF had a change in isoform 4I2/4I1 expression along with a decrease of isoform COX 4I1 expression. The 4I2/4I1 ratio of mRNA expression was increased from 0.630 to 1.058 in comparison. However, the protein content of CytOx subunit 4 was much lower in the AF group, whereas the respiration/units enzyme activity in both groups remained the same. CONCLUSIONS This study describes a possible molecular correlate for the development of AF. Due to the known functional significance of COX 4I2, mitochondrial dysfunction can be assumed as a part of the pathogenesis of AF.
Collapse
Affiliation(s)
- Sebastian Vogt
- Cardiovascular Research LabPhilipps‐University MarburgMarburgGermany
- Department of Cardiac and Vascular SurgeryUniversity Hospital of Giessen and MarburgMarburgGermany
| | - Rabia Ramzan
- Cardiovascular Research LabPhilipps‐University MarburgMarburgGermany
- Department of Cardiac and Vascular SurgeryUniversity Hospital of Giessen and MarburgMarburgGermany
| | - Pia Cybulski
- Cardiovascular Research LabPhilipps‐University MarburgMarburgGermany
| | - Annika Rhiel
- Cardiovascular Research LabPhilipps‐University MarburgMarburgGermany
| | - Petra Weber
- Cardiovascular Research LabPhilipps‐University MarburgMarburgGermany
| | - Volker Ruppert
- Department of CardiologyUniversity Hospital of Giessen and MarburgMarburgGermany
| | - Marc Irqsusi
- Department of Cardiac and Vascular SurgeryUniversity Hospital of Giessen and MarburgMarburgGermany
| | - Susanne Rohrbach
- Institute of PhysiologyJustus Liebig University GiessenGiessenGermany
| | - Bernd Niemann
- Department of Cardiac and Vascular SurgeryUniversity Hospital of Giessen and MarburgGiessenGermany
| | - Nikolas Mirow
- Department of Cardiac and Vascular SurgeryUniversity Hospital of Giessen and MarburgMarburgGermany
| | - Ardawan J. Rastan
- Department of Cardiac and Vascular SurgeryUniversity Hospital of Giessen and MarburgMarburgGermany
| |
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
|
Zhou D, Zhang Y, Zhu M, Zhang X, Zhang X, Lv J, Tang W, Weng Q, Lin Y, Tong L, Zhong Z, Zhang Y, Zhang M, Lai M, Wang D. mROS‑calcium feedback loop promotes lethal ventricular arrhythmias and sudden cardiac death in early myocardial ischemia. Int J Mol Med 2024; 53:5. [PMID: 37997788 PMCID: PMC10712693 DOI: 10.3892/ijmm.2023.5329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Lethal ventricular arrhythmia‑sudden cardiac death (LVA‑SCD) occurs frequently during the early stage of myocardial ischemia (MI). However, the mechanism underlying higher LVA‑SCD incidence is still poorly understood. The present study aimed to explore the role of mitochondrial reactive oxygen species (mROS) and Ca2+ crosstalk in promoting LVA‑SCD in early MI. RyR2 S2814A mice and their wild‑type littermates were used. MitoTEMPO was applied to scavenge mitochondrial ROS (mROS). Mice were subjected to severe MI and the occurrence of LVA‑SCD was evaluated. Levels of mitochondrial ROS and calcium (mitoCa2+), cytosolic ROS (cytoROS), and calcium (cytoCa2+), RyR2 Ser‑2814 phosphorylation, CaMKII Met‑282 oxidation, mitochondrial membrane potential (MMP), and glutathione/oxidized glutathione (GSH/GSSG) ratio in the myocardia were detected. Dynamic changes in mROS after hypoxia were investigated using H9c2 cells. Moreover, the myocardial phosphoproteome was analyzed to explore the related mechanisms facilitating mROS‑Ca2+ crosstalk and LVA‑SCD. There was a high incidence (~33.9%) of LVA‑SCD in early MI. Mice who underwent SCD displayed notably elevated levels of myocardial ROS and mROS, and the latter was validated in H9c2 cells. These mice also demonstrated overloads of cytoplasmic and mitochondrial Ca2+, decreased MMP and reduced GSH/GSSG ratio, upregulated RyR2‑S2814 phosphorylation and CaMKII‑M282 oxidation and transient hyperphosphorylation of mitochondrial proteomes in the myocardium. mROS‑specific scavenging by a mitochondria‑targeted antioxidant agent (MitoTEMPO) corrected these SCD‑induced alterations. S2814A mice with a genetically inactivated CaMKII phosphorylation site in RyR2 exhibited decreased overloads in cytoplasmic and mitochondrial Ca2+ and demonstrated similar effects as MitoTEMPO to correct SCD‑induced changes and prevent SCD post‑MI. The data confirmed crosstalk between mROS and Ca2+ in promoting LVA‑SCD. Therefore, we provided evidence that there is a higher incidence of LVA‑SCD in early MI, which may be attributed to a positive feedback loop between mROS and Ca2+ imbalance.
Collapse
Affiliation(s)
- Danya Zhou
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- School of Forensic Medicine, Xinxiang Key Laboratory of Forensic Toxicology, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Ye Zhang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Mengting Zhu
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xiaojun Zhang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, Guangdong 515041, P.R. China
| | - Xiaojuan Zhang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Junyao Lv
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Wanting Tang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Qi Weng
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yang Lin
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Lejun Tong
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Zhiwei Zhong
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yanmei Zhang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Mengxuan Zhang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Minchao Lai
- Department of Neurology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Dian Wang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
7
|
Cardiomyocyte-specific loss of plasma membrane calcium ATPase 1 impacts cardiac rhythm and is associated with ventricular repolarisation dysfunction. J Mol Cell Cardiol 2022; 172:41-51. [PMID: 35926724 DOI: 10.1016/j.yjmcc.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 07/09/2022] [Accepted: 07/27/2022] [Indexed: 12/14/2022]
Abstract
Plasma membrane calcium ATPase 1 (PMCA1, Atp2b1) is emerging as a key contributor to cardiac physiology, involved in calcium handling and myocardial signalling. In addition, genome wide association studies have associated PMCA1 in several areas of cardiovascular disease including hypertension and myocardial infarction. Here, we investigated the role of PMCA1 in basal cardiac function and heart rhythm stability. Cardiac structure, heart rhythm and arrhythmia susceptibility were assessed in a cardiomyocyte-specific PMCA1 deletion (PMCA1CKO) mouse model. PMCA1CKO mice developed abnormal heart rhythms related to ventricular repolarisation dysfunction and displayed an increased susceptibility to ventricular arrhythmias. We further assessed the levels of cardiac ion channels using qPCR and found a downregulation of the voltage-dependent potassium channels, Kv4.2, with a corresponding reduction in the transient outward potassium current which underlies ventricular repolarisation in the murine heart. The changes in heart rhythm were found to occur in the absence of any structural cardiomyopathy. To further assess the molecular changes occurring in PMCA1CKO hearts, we performed proteomic analysis. Functional characterisation of differentially expressed proteins suggested changes in pathways related to metabolism, protein-binding, and pathways associated cardiac function including β-adrenergic signalling. Together, these data suggest an important role for PMCA1 in basal cardiac function in relation to heart rhythm control, with reduced cardiac PMCA1 expression resulting in an increased risk of arrhythmia development.
Collapse
|
8
|
Identifying disease-critical cell types and cellular processes by integrating single-cell RNA-sequencing and human genetics. Nat Genet 2022; 54:1479-1492. [PMID: 36175791 PMCID: PMC9910198 DOI: 10.1038/s41588-022-01187-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/18/2022] [Indexed: 12/13/2022]
Abstract
Genome-wide association studies provide a powerful means of identifying loci and genes contributing to disease, but in many cases, the related cell types/states through which genes confer disease risk remain unknown. Deciphering such relationships is important for identifying pathogenic processes and developing therapeutics. In the present study, we introduce sc-linker, a framework for integrating single-cell RNA-sequencing, epigenomic SNP-to-gene maps and genome-wide association study summary statistics to infer the underlying cell types and processes by which genetic variants influence disease. The inferred disease enrichments recapitulated known biology and highlighted notable cell-disease relationships, including γ-aminobutyric acid-ergic neurons in major depressive disorder, a disease-dependent M-cell program in ulcerative colitis and a disease-specific complement cascade process in multiple sclerosis. In autoimmune disease, both healthy and disease-dependent immune cell-type programs were associated, whereas only disease-dependent epithelial cell programs were prominent, suggesting a role in disease response rather than initiation. Our framework provides a powerful approach for identifying the cell types and cellular processes by which genetic variants influence disease.
Collapse
|
9
|
Young WJ, Lahrouchi N, Isaacs A, Duong T, Foco L, Ahmed F, Brody JA, Salman R, Noordam R, Benjamins JW, Haessler J, Lyytikäinen LP, Repetto L, Concas MP, van den Berg ME, Weiss S, Baldassari AR, Bartz TM, Cook JP, Evans DS, Freudling R, Hines O, Isaksen JL, Lin H, Mei H, Moscati A, Müller-Nurasyid M, Nursyifa C, Qian Y, Richmond A, Roselli C, Ryan KA, Tarazona-Santos E, Thériault S, van Duijvenboden S, Warren HR, Yao J, Raza D, Aeschbacher S, Ahlberg G, Alonso A, Andreasen L, Bis JC, Boerwinkle E, Campbell A, Catamo E, Cocca M, Cutler MJ, Darbar D, De Grandi A, De Luca A, Ding J, Ellervik C, Ellinor PT, Felix SB, Froguel P, Fuchsberger C, Gögele M, Graff C, Graff M, Guo X, Hansen T, Heckbert SR, Huang PL, Huikuri HV, Hutri-Kähönen N, Ikram MA, Jackson RD, Junttila J, Kavousi M, Kors JA, Leal TP, Lemaitre RN, Lin HJ, Lind L, Linneberg A, Liu S, MacFarlane PW, Mangino M, Meitinger T, Mezzavilla M, Mishra PP, Mitchell RN, Mononen N, Montasser ME, Morrison AC, Nauck M, Nauffal V, Navarro P, Nikus K, Pare G, Patton KK, Pelliccione G, Pittman A, Porteous DJ, Pramstaller PP, Preuss MH, Raitakari OT, Reiner AP, Ribeiro ALP, Rice KM, Risch L, Schlessinger D, Schotten U, Schurmann C, Shen X, Shoemaker MB, Sinagra G, Sinner MF, Soliman EZ, Stoll M, Strauch K, Tarasov K, Taylor KD, Tinker A, Trompet S, Uitterlinden A, Völker U, Völzke H, Waldenberger M, Weng LC, Whitsel EA, Wilson JG, Avery CL, Conen D, Correa A, Cucca F, Dörr M, Gharib SA, Girotto G, Grarup N, Hayward C, Jamshidi Y, Järvelin MR, Jukema JW, Kääb S, Kähönen M, Kanters JK, Kooperberg C, Lehtimäki T, Lima-Costa MF, Liu Y, Loos RJF, Lubitz SA, Mook-Kanamori DO, Morris AP, O'Connell JR, Olesen MS, Orini M, Padmanabhan S, Pattaro C, Peters A, Psaty BM, Rotter JI, Stricker B, van der Harst P, van Duijn CM, Verweij N, Wilson JF, Arking DE, Ramirez J, Lambiase PD, Sotoodehnia N, Mifsud B, Newton-Cheh C, Munroe PB. Genetic analyses of the electrocardiographic QT interval and its components identify additional loci and pathways. Nat Commun 2022; 13:5144. [PMID: 36050321 PMCID: PMC9436946 DOI: 10.1038/s41467-022-32821-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 08/17/2022] [Indexed: 11/10/2022] Open
Abstract
The QT interval is an electrocardiographic measure representing the sum of ventricular depolarization and repolarization, estimated by QRS duration and JT interval, respectively. QT interval abnormalities are associated with potentially fatal ventricular arrhythmia. Using genome-wide multi-ancestry analyses (>250,000 individuals) we identify 177, 156 and 121 independent loci for QT, JT and QRS, respectively, including a male-specific X-chromosome locus. Using gene-based rare-variant methods, we identify associations with Mendelian disease genes. Enrichments are observed in established pathways for QT and JT, and previously unreported genes indicated in insulin-receptor signalling and cardiac energy metabolism. In contrast for QRS, connective tissue components and processes for cell growth and extracellular matrix interactions are significantly enriched. We demonstrate polygenic risk score associations with atrial fibrillation, conduction disease and sudden cardiac death. Prioritization of druggable genes highlight potential therapeutic targets for arrhythmia. Together, these results substantially advance our understanding of the genetic architecture of ventricular depolarization and repolarization.
Collapse
Affiliation(s)
- William J Young
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS trust, London, UK
| | - Najim Lahrouchi
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Aaron Isaacs
- Deptartment of Physiology, Cardiovascular Research Institute Maastricht CARIM, Maastricht University, Maastricht, The Netherlands
- Maastricht Center for Systems Biology MaCSBio, Maastricht University, Maastricht, The Netherlands
| | - ThuyVy Duong
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luisa Foco
- Eurac Research, Institute for Biomedicine affiliated with the University of Lübeck, Bolzano, Italy
| | - Farah Ahmed
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Reem Salman
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
| | - Raymond Noordam
- Department of Internal Medicine, section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan-Walter Benjamins
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Jeffrey Haessler
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Linda Repetto
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Maria Pina Concas
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Marten E van den Berg
- Department of Epidemiology, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Stefan Weiss
- DZHK German Centre for Cardiovascular Research; partner site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics; Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Antoine R Baldassari
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Departments of Biostatistics and Medicine, University of Washington, Seattle, WA, USA
| | - James P Cook
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Daniel S Evans
- California Pacific Medical Center, Research Institute, San Francisco, CA, USA
| | - Rebecca Freudling
- Department of Cardiology, University Hospital, LMU Munich, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Oliver Hines
- Genetics Research Centre, St George's University of London, London, UK
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Jonas L Isaksen
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Honghuang Lin
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, Jackson, USA
| | - Arden Moscati
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- IBE, Faculty of Medicine, LMU Munich, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics IMBEI, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Casia Nursyifa
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yong Qian
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, US
| | - Anne Richmond
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Carolina Roselli
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
| | - Kathleen A Ryan
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eduardo Tarazona-Santos
- Department of Genetics, Ecology and Evolution, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte/Minas Gerais, Brazil
| | - Sébastien Thériault
- Population Health Research Institute, McMaster University, Hamilton, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec, Canada
| | - Stefan van Duijvenboden
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Helen R Warren
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Dania Raza
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Brighton and Sussex Medical School, Brighton, UK
| | - Stefanie Aeschbacher
- Cardiovascular Research Institute Basel, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Gustav Ahlberg
- Laboratory for Molecular Cardiology, The Heart Centre, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Laura Andreasen
- Laboratory for Molecular Cardiology, The Heart Centre, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Archie Campbell
- Usher Institute, University of Edinburgh, Nine, Edinburgh Bioquarter, 9 Little France Road, Edinburgh, UK
- Health Data Research UK, University of Edinburgh, Nine, Edinburgh Bioquarter, 9 Little France Road, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Eulalia Catamo
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Massimiliano Cocca
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Michael J Cutler
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
| | - Dawood Darbar
- Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Alessandro De Grandi
- Eurac Research, Institute for Biomedicine affiliated with the University of Lübeck, Bolzano, Italy
| | - Antonio De Luca
- Cardiothoracovascular Department, ASUGI, University of Trieste, Trieste, Italy
| | - Jun Ding
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, US
| | - Christina Ellervik
- Department of Data and Data Support, Region Zealand, 4180, Sorø, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
- Demoulas Center for Cardiac Arrhythmias and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Stephan B Felix
- DZHK German Centre for Cardiovascular Research; partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B - Cardiology, Pneumology, Infectious Diseases, Intensive Care Medicine; University Medicine Greifswald, Greifswald, Germany
| | - Philippe Froguel
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- University of Lille Nord de France, Lille, France
- CNRS UMR8199, Institut Pasteur de Lille, Lille, France
| | - Christian Fuchsberger
- Eurac Research, Institute for Biomedicine affiliated with the University of Lübeck, Bolzano, Italy
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, USA
| | - Martin Gögele
- Eurac Research, Institute for Biomedicine affiliated with the University of Lübeck, Bolzano, Italy
| | - Claus Graff
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics/Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics/David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology/University of Washington, Seattle, WA, USA
| | - Paul L Huang
- Cardiology Division and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Heikki V Huikuri
- Research Unit of Internal Medicine, Medical Research Center Oulu, University of Oulu and University Hospital of Oulu, Oulu, Finland
| | - Nina Hutri-Kähönen
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
- Department of Pediatrics, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Centre for Skills Training and Simulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Rebecca D Jackson
- Center for Clinical and Translational Science, Ohio State Medical Center, Columbus, OH, USA
| | - Juhani Junttila
- Research Unit of Internal Medicine, Medical Research Center Oulu, University of Oulu and University Hospital of Oulu, Oulu, Finland
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Jan A Kors
- Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, NL, The Netherlands
| | - Thiago P Leal
- Department of Genetics, Ecology and Evolution, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte/Minas Gerais, Brazil
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Henry J Lin
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics/Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics/David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Lars Lind
- Deptartment of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simin Liu
- Center for Global Cardiometabolic Health, Departments of Epidemiology, Medicine and Surgery, Brown University, Providence, USA
| | - Peter W MacFarlane
- Institute of Health and Wellbeing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, UK
| | - Thomas Meitinger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research, partner site: Munich Heart Alliance, Munich, Germany
| | - Massimo Mezzavilla
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Rebecca N Mitchell
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - May E Montasser
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Matthias Nauck
- DZHK German Centre for Cardiovascular Research; partner site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Victor Nauffal
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, Finland
- Department of Cardiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Guillaume Pare
- Population Health Research Institute, McMaster University, Hamilton, Canada
| | - Kristen K Patton
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Giulia Pelliccione
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Alan Pittman
- Genetics Research Centre, St George's University of London, London, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Peter P Pramstaller
- Eurac Research, Institute for Biomedicine affiliated with the University of Lübeck, Bolzano, Italy
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Alexander P Reiner
- Department of Epidemiology/University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Antonio Luiz P Ribeiro
- Department of Internal Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Brazil, Belo Horizonte, Minas Gerais, Brazil
- Cardiology Service and Telehealth Center, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil, Belo Horizonte, Minas Gerais, Brazil
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Lorenz Risch
- Labormedizinisches zentrum Dr. Risch, Vaduz, Liechtenstein
- Faculty of Medical Sciences, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
- Center of Laboratory Medicine, University Institute of Clinical Chemistry, University of Bern, Inselspital, Bern, Switzerland
| | - David Schlessinger
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institute of Health, Baltimore, US
| | - Ulrich Schotten
- Deptartment of Physiology, Cardiovascular Research Institute Maastricht CARIM, Maastricht University, Maastricht, The Netherlands
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Hasso Plattner Institute, University of Potsdam, Potsdam, Germany
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xia Shen
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Greater Bay Area Institute of Precision Medicine Guangzhou, Fudan University, Nansha District, Guangzhou, China
| | - M Benjamin Shoemaker
- Department of Medicine, Division of Cardiovascular Medicine, Arrhythmia Section, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, ASUGI, University of Trieste, Trieste, Italy
| | - Moritz F Sinner
- Department of Cardiology, University Hospital, LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research, partner site: Munich Heart Alliance, Munich, Germany
| | - Elsayed Z Soliman
- Epidemiological Cardiology Research Center EPICARE, Wake Forest School of Medicine, Winston Salem, USA
| | - Monika Stoll
- Maastricht Center for Systems Biology MaCSBio, Maastricht University, Maastricht, The Netherlands
- Dept. of Biochemistry, Cardiovascular Research Institute Maastricht CARIM, Maastricht University, Maastricht, NL, The Netherlands
- Institute of Human Genetics, Genetic Epidemiology, University of Muenster, Muenster, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- IBE, Faculty of Medicine, LMU Munich, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics IMBEI, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Kirill Tarasov
- Laboratory of Cardiovascular Sciences, National Institute on Aging, National Institute of Health, Baltimore, US
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics/Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics/David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Andrew Tinker
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Stella Trompet
- Department of Internal Medicine, section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Uwe Völker
- DZHK German Centre for Cardiovascular Research; partner site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics; Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- DZHK German Centre for Cardiovascular Research; partner site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Melanie Waldenberger
- DZHK (German Centre for Cardiovascular Research, partner site: Munich Heart Alliance, Munich, Germany
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Lu-Chen Weng
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, USA
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, USA
- Department of Cardiology, Beth Israel Deaconess Medical Center, Boston, USA
| | - Christy L Avery
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Canada
| | - Adolfo Correa
- Departments of Medicine, Pediatrics and Population Health Science, University of Mississippi Medical Center, Jackson, USA
| | - Francesco Cucca
- Institute of Genetic and Biomedical Rsearch, Italian National Research Council, Monserrato, Italy
| | - Marcus Dörr
- DZHK German Centre for Cardiovascular Research; partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B - Cardiology, Pneumology, Infectious Diseases, Intensive Care Medicine; University Medicine Greifswald, Greifswald, Germany
| | - Sina A Gharib
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Giorgia Girotto
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Yalda Jamshidi
- Genetics Research Centre, St George's University of London, London, UK
| | - Marjo-Riitta Järvelin
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Stefan Kääb
- Department of Cardiology, University Hospital, LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research, partner site: Munich Heart Alliance, Munich, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jørgen K Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Yongmei Liu
- Department of Medicine, Duke University, Durham, NC, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven A Lubitz
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
- Demoulas Center for Cardiac Arrhythmias and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew P Morris
- Department of Health Data Science, University of Liverpool, Liverpool, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Jeffrey R O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Michele Orini
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS trust, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Cristian Pattaro
- Eurac Research, Institute for Biomedicine affiliated with the University of Lübeck, Bolzano, Italy
| | - Annette Peters
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research, partner site: Munich Heart Alliance, Munich, Germany
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology/University of Washington, Seattle, WA, USA
- Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics/Harbor-UCLA Medical Center, Torrance, CA, USA
- Departments of Pediatrics and Human Genetics/David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Bruno Stricker
- Department of Epidemiology, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
- Department of Cardiology, Heart and Lung Division, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cornelia M van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Niek Verweij
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julia Ramirez
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Pier D Lambiase
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS trust, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Borbala Mifsud
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Christopher Newton-Cheh
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Cardiovascular Research Center, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Patricia B Munroe
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK.
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
10
|
Samuel TJ, Lai S, Schär M, Wu KC, Steinberg AM, Wei AC, Anderson M, Tomaselli GF, Gerstenblith G, Bottomley PA, Weiss RG. Myocardial ATP depletion detected noninvasively predicts sudden cardiac death risk in heart failure patients. JCI Insight 2022; 7:157557. [PMID: 35579938 PMCID: PMC9309047 DOI: 10.1172/jci.insight.157557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/06/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Sudden cardiac death (SCD) remains a worldwide public health problem in need of better noninvasive predictive tools. Current guidelines for primary preventive SCD therapies, such as implantable cardioverter defibrillators (ICDs), are based on left ventricular ejection fraction (LVEF), but these guidelines are imprecise: fewer than 5% of ICDs deliver lifesaving therapy per year. Impaired cardiac metabolism and ATP depletion cause arrhythmias in experimental models, but to our knowledge a link between arrhythmias and cardiac energetic abnormalities in people has not been explored, nor has the potential for metabolically predicting clinical SCD risk. METHODS We prospectively measured myocardial energy metabolism noninvasively with phosphorus magnetic resonance spectroscopy in patients with no history of significant arrhythmias prior to scheduled ICD implantation for primary prevention in the setting of reduced LVEF (≤35%). RESULTS By 2 different analyses, low myocardial ATP significantly predicted the composite of subsequent appropriate ICD firings for life-threatening arrhythmias and cardiac death over approximately 10 years. Life-threatening arrhythmia risk was approximately 3-fold higher in patients with low ATP and independent of established risk factors, including LVEF. In patients with normal ATP, rates of appropriate ICD firings were several-fold lower than reported rates of ICD complications and inappropriate firings. CONCLUSION To the best of our knowledge, these are the first data linking in vivo myocardial ATP depletion and subsequent significant arrhythmic events in people, suggesting an energetic component to clinical life-threatening ventricular arrhythmogenesis. The findings support investigation of metabolic strategies that limit ATP loss to treat or prevent life-threatening cardiac arrhythmias and herald noninvasive metabolic imaging as a complementary SCD risk stratification tool. TRIAL REGISTRATION ClinicalTrials.gov NCT00181233. FUNDING This work was supported by the DW Reynolds Foundation, the NIH (grants HL61912, HL056882, HL103812, HL132181, HL140034), and Russell H. Morgan and Clarence Doodeman endowments at Johns Hopkins.
Collapse
Affiliation(s)
- T Jake Samuel
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Shenghan Lai
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, United States of America
| | - Michael Schär
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Katherine C Wu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Angela M Steinberg
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - An-Chi Wei
- Department of Electrical Engineering, National Taiwan University, Tapei, Taiwan
| | - Mark Anderson
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, United States of America
| | - Gordon F Tomaselli
- Division of Cardiology, Department of Medicine, The Albert Einstein College of Medicine, Bronx, United States of America
| | - Gary Gerstenblith
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Paul A Bottomley
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Robert G Weiss
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, United States of America
| |
Collapse
|
11
|
Jagadeesh KA, Dey KK, Montoro DT, Mohan R, Gazal S, Engreitz JM, Xavier RJ, Price AL, Regev A. Identifying disease-critical cell types and cellular processes across the human body by integration of single-cell profiles and human genetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.19.436212. [PMID: 34845454 PMCID: PMC8629197 DOI: 10.1101/2021.03.19.436212] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Genome-wide association studies (GWAS) provide a powerful means to identify loci and genes contributing to disease, but in many cases the related cell types/states through which genes confer disease risk remain unknown. Deciphering such relationships is important for identifying pathogenic processes and developing therapeutics. Here, we introduce sc-linker, a framework for integrating single-cell RNA-seq (scRNA-seq), epigenomic maps and GWAS summary statistics to infer the underlying cell types and processes by which genetic variants influence disease. We analyzed 1.6 million scRNA-seq profiles from 209 individuals spanning 11 tissue types and 6 disease conditions, and constructed gene programs capturing cell types, disease progression, and cellular processes both within and across cell types. We evaluated these gene programs for disease enrichment by transforming them to SNP annotations with tissue-specific epigenomic maps and computing enrichment scores across 60 diseases and complex traits (average N= 297K). Cell type, disease progression, and cellular process programs captured distinct heritability signals even within the same cell type, as we show in multiple complex diseases that affect the brain (Alzheimer’s disease, multiple sclerosis), colon (ulcerative colitis) and lung (asthma, idiopathic pulmonary fibrosis, severe COVID-19). The inferred disease enrichments recapitulated known biology and highlighted novel cell-disease relationships, including GABAergic neurons in major depressive disorder (MDD), a disease progression M cell program in ulcerative colitis, and a disease-specific complement cascade process in multiple sclerosis. In autoimmune disease, both healthy and disease progression immune cell type programs were associated, whereas for epithelial cells, disease progression programs were most prominent, perhaps suggesting a role in disease progression over initiation. Our framework provides a powerful approach for identifying the cell types and cellular processes by which genetic variants influence disease.
Collapse
|
12
|
Richter C, Hinkel R. Research('s) Sweet Hearts: Experimental Biomedical Models of Diabetic Cardiomyopathy. Front Cardiovasc Med 2021; 8:703355. [PMID: 34368257 PMCID: PMC8342758 DOI: 10.3389/fcvm.2021.703355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Diabetes and the often accompanying cardiovascular diseases including cardiomyopathy represent a complex disease, that is reluctant to reveal the molecular mechanisms and underlying cellular responses. Current research projects on diabetic cardiomyopathy are predominantly based on animal models, in which there are not only obvious advantages, such as genetics that can be traced over generations and the directly measurable influence of dietary types, but also not despisable disadvantages. Thus, many studies are built up on transgenic rodent models, which are partly comparable to symptoms in humans due to their genetic alterations, but on the other hand are also under discussion regarding their clinical relevance in the translation of biomedical therapeutic approaches. Furthermore, a focus on transgenic rodent models ignores spontaneously occurring diabetes in larger mammals (such as dogs or pigs), which represent with their anatomical similarity to humans regarding their cardiovascular situation appealing models for testing translational approaches. With this in mind, we aim to shed light on the currently most popular animal models for diabetic cardiomyopathy and, by weighing the advantages and disadvantages, provide decision support for future animal experimental work in the field, hence advancing the biomedical translation of promising approaches into clinical application.
Collapse
Affiliation(s)
- Claudia Richter
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany.,Partnersite Goettingen, German Center for Cardiovascular Research (DZHK e.V.), Goettingen, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany.,Partnersite Goettingen, German Center for Cardiovascular Research (DZHK e.V.), Goettingen, Germany.,Stiftung Tierärztliche Hochschule Hannover, University of Veterinary Medicine, Hanover, Germany
| |
Collapse
|
13
|
Li HL, Lip GYH, Feng Q, Fei Y, Tse YK, Wu MZ, Ren QW, Tse HF, Cheung BMY, Yiu KH. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) and cardiac arrhythmias: a systematic review and meta-analysis. Cardiovasc Diabetol 2021; 20:100. [PMID: 33962654 PMCID: PMC8106208 DOI: 10.1186/s12933-021-01293-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
Background Cardiac arrhythmias are associated with poorer outcomes in patients with heart failure (HF), diabetes mellitus (DM), and chronic kidney disease (CKD). Previous studies have shown inconsistent conclusions regarding the association between sodium-glucose cotransporter 2 inhibitors (SGLT2i) and the risk of developing arrhythmias. This study aims to investigate the association of SGLT2i treatment with arrhythmia outcomes in clinical trials of patients with HF, DM, or CKD. Methods MEDLINE, EMBASE, and ClinicalTrials.gov were searched from inception up to 27 August 2020. Randomized controlled trials that randomized patients with DM, CKD, or HF to SGLT2i or placebo were included. The outcomes of interest include atrial fibrillation (AF), embolic stroke, atrial flutter (AFL), AF/AFL, ventricular tachycardia (VT), and cardiac arrest. Relative risks (RRs) and 95% confidence intervals (CI) were pooled using a random-effects model. Results Out of 4,532 citations, 22 trials with altogether 52,115 patients were included (mean age 63.2 years; 33,747 [64.8%] of participants were men). SGLT2i were associated with a lower risk of AF (RR 0.82, 95% CI 0.70–0.96), embolic stroke (RR 0.32, 95% CI 0.12–0.85), AF/AFL (RR 0.82, 95% CI 0.71–0.95), and VT (RR 0.73, 95% CI 0.53–0.99), while the risk reductions in AFL (RR 0.83, 95% CI 0.58–1.17) and cardiac arrest (RR 0.83, 95% CI 0.61–1.14) did not reach statistical significance. The associations appeared to be consistent across different baseline conditions (DM vs CKD vs HF; atherosclerotic cardiovascular disease [ASCVD] vs no ASCVD) and the SGLT2i used. Conclusions SGLT2i reduced the risk of cardiac arrhythmias. Our study provides further evidence for recommending the use of SGLT2i in patients with DM, CKD, and HF. Further research is needed to fully elucidate the mechanism by which SGLT2i protect against arrhythmias. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-021-01293-8.
Collapse
Affiliation(s)
- Hang-Long Li
- Division of Cardiology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Room 1929B/K1931, Block K, Hong Kong, China.,Division of Cardiology, Department of Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Qi Feng
- Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Yue Fei
- Division of Clinical Pharmacology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Yi-Kei Tse
- Division of Cardiology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Room 1929B/K1931, Block K, Hong Kong, China
| | - Mei-Zhen Wu
- Division of Cardiology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Room 1929B/K1931, Block K, Hong Kong, China.,Division of Cardiology, Department of Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Qing-Wen Ren
- Division of Cardiology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Room 1929B/K1931, Block K, Hong Kong, China.,Division of Cardiology, Department of Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Hung-Fat Tse
- Division of Cardiology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Room 1929B/K1931, Block K, Hong Kong, China.,Division of Cardiology, Department of Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Bernard-M Y Cheung
- Division of Clinical Pharmacology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Kai-Hang Yiu
- Division of Cardiology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Room 1929B/K1931, Block K, Hong Kong, China. .,Division of Cardiology, Department of Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
14
|
Sengul C, Sen A, Barutcu S, Cakir C, Sarikaya R. Association of Mild Hyperbilirubinemia with Decreased ECG-Based Ventricular Repolarization Parameters in Young Men. Lab Med 2021; 52:226-231. [PMID: 32885227 DOI: 10.1093/labmed/lmaa063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Hyperbilirubinemia is associated with protection against various oxidative stress-mediated diseases. We aimed to investigate the association between bilirubin and novel electrocardiography (ECG)-based ventricular repolarization parameters. METHODS We enrolled 201 healthy men with mild hyperbilirubinemia (group 1) and 219 healthy men with normal bilirubin levels (group 2). The Tpeak-Tend (Tp-e) interval (defined as the interval from the peak of the T wave to the end of the T wave), corrected (c) Tp-e interval, QT interval, cQT interval, and Tp-e interval/QT interval ratio were measured from leads V5 and V6 with 20 mm/mV amplitude and 50 mm/second rate. RESULTS The Tp-e interval, cTp-e interval, and Tp-e interval/QT interval ratio were significantly lower in group 1 compared with group 2. The cTp-e interval showed a significant negative correlation with total bilirubin, conjugated bilirubin, and unconjugated bilirubin. The cTp-e interval (odds ratio [OR], 0.900; P =.002) and Tp-e interval/QT interval ratio (OR, 0.922; P =.04) were significantly associated with mild hyperbilirubinemia. CONCLUSION We showed the association of mild hyperbilirubinemia with decreased novel ECG-based ventricular repolarization parameters.
Collapse
Affiliation(s)
- Cihan Sengul
- Department of Cardiology, University of Health Sciences, Van Education and Research Hospital, Van, Turkey
| | - Ahmet Sen
- Department of Biochemistry, University of Health Sciences, Van Education and Research Hospital, Van, Turkey
| | - Suleyman Barutcu
- Department of Cardiology, University of Health Sciences, Van Education and Research Hospital, Van, Turkey
| | - Cayan Cakir
- Department of Cardiology, University of Health Sciences, Van Education and Research Hospital, Van, Turkey
| | - Remzi Sarikaya
- Department of Cardiology, University of Health Sciences, Van Education and Research Hospital, Van, Turkey
| |
Collapse
|
15
|
Volani C, Rainer J, Hernandes VV, Meraviglia V, Pramstaller PP, Smárason SV, Pompilio G, Casella M, Sommariva E, Paglia G, Rossini A. Metabolic Signature of Arrhythmogenic Cardiomyopathy. Metabolites 2021; 11:metabo11040195. [PMID: 33805952 PMCID: PMC8064316 DOI: 10.3390/metabo11040195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/06/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic-based cardiac disease accompanied by severe ventricular arrhythmias and a progressive substitution of the myocardium with fibro-fatty tissue. ACM is often associated with sudden cardiac death. Due to the reduced penetrance and variable expressivity, the presence of a genetic defect is not conclusive, thus complicating the diagnosis of ACM. Recent studies on human induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) obtained from ACM individuals showed a dysregulated metabolic status, leading to the hypothesis that ACM pathology is characterized by an impairment in the energy metabolism. However, despite efforts having been made for the identification of ACM specific biomarkers, there is still a substantial lack of information regarding the whole metabolomic profile of ACM patients. The aim of the present study was to investigate the metabolic profiles of ACM patients compared to healthy controls (CTRLs). The targeted Biocrates AbsoluteIDQ® p180 assay was used on plasma samples. Our analysis showed that ACM patients have a different metabolome compared to CTRLs, and that the pathways mainly affected include tryptophan metabolism, arginine and proline metabolism and beta oxidation of fatty acids. Altogether, our data indicated that the plasma metabolomes of arrhythmogenic cardiomyopathy patients show signs of endothelium damage and impaired nitric oxide (NO), fat, and energy metabolism.
Collapse
Affiliation(s)
- Chiara Volani
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
- Correspondence:
| | - Johannes Rainer
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Vinicius Veri Hernandes
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Viviana Meraviglia
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Peter Paul Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Sigurður Vidir Smárason
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138 Milan, Italy; (G.P.); (E.S.)
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20138 Milan, Italy
| | - Michela Casella
- Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy;
- Cardiology and Arrhythmology Clinic, University Hospital Ospedali Riuniti Umberto I-Lancisi-Salesi, 60126 Ancona, Italy
- Department of Clinical, Special and Dental Sciences, Marche Polytechnic University, 60126 Ancona, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138 Milan, Italy; (G.P.); (E.S.)
| | - Giuseppe Paglia
- School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, 20854 Vedano al Lambro, Italy;
| | - Alessandra Rossini
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy; (J.R.); (V.V.H.); (V.M.); (P.P.P.); (S.V.S.); (A.R.)
| |
Collapse
|
16
|
Xie D, Wu J, Wu Q, Zhang X, Zhou D, Dai W, Zhu M, Wang D. Integrating proteomic, lipidomic and metabolomic data to construct a global metabolic network of lethal ventricular tachyarrhythmias (LVTA) induced by aconitine. J Proteomics 2021; 232:104043. [PMID: 33161167 DOI: 10.1016/j.jprot.2020.104043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/10/2020] [Accepted: 11/02/2020] [Indexed: 02/05/2023]
Abstract
Lethal ventricular tachyarrhythmias (LVTA)-related sudden cardiac death (SCD) is one of the major causes of death worldwide. However, the mechanisms underlying LVTA induced by myocardial ion channel diseases (MICDs) are not yet fully understood. Here, we produced an LVTA rat model induced by aconitine, to mimic MICDs-elicited LVTA, and constructed a global pathway network via integrating proteomic and lipidomic data, and our previously published metabolomic data. Results showed that both proteome and lipidome were disturbed during the LVTA process. Most of the differentially expressed proteins and lipid species were correlated. Proteomic data indicated disturbance of energy metabolism (e.g. fatty acid β-oxidation and the tricarboxylic acid cycle) and activation of the protein kinase C and nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase pathway; these alterations led to lowered ATP and elevated ROS, respectively. Altered levels of the Ca2+ handling proteins suggested aberrant intracellular Ca2+ homeostasis, which might also be secondary to the shortage of ATP and oxidative stress. Significantly, the disrupted pathways implied by proteomic data were largely confirmed by lipidomic and metabolomic data. Collectively, we have constructed a metabolic pathway network of aconitine-induced LVTA using a multi-omics strategy, which confers great promise for the deeper interpretation of the mechanisms underlying LVTA. SIGNIFICANCE: In this study, we integrated proteomics, lipidomics and metabolomics to explore the pathophysiological processes of LVTA induced by aconitine. It is innovative to try to integrate these three omics in a study exploring the relative mechanisms. Here, based on the DEPs and differentially abundant lipid species (DALPs) between the LVTA groups and the controls, and the different metabolites discovered previously from the same model, we have successfully constructed a global metabolic network. Taken together, the multi-omics integration strategies used in this study show the potential for a new interpretation of the pathophysiological processes of LVTA induced by different conditions and open the possibility to explore deeper and broader mechanisms of other diseases.
Collapse
Affiliation(s)
- Dezhi Xie
- Department of Forensic Medicine, Shantou University Medical College, Shantou 515041, China
| | - Jiayan Wu
- Department of Forensic Medicine, Shantou University Medical College, Shantou 515041, China
| | - Qian Wu
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China
| | - Xiaojun Zhang
- Central laboratory, Shantou University Medical College, Shantou 515041, China
| | - Danya Zhou
- Department of Forensic Medicine, Shantou University Medical College, Shantou 515041, China
| | - Wentao Dai
- Shanghai Center for Bioinformation Technology, Shanghai 201203, China
| | - Mengting Zhu
- Department of Forensic Medicine, Shantou University Medical College, Shantou 515041, China
| | - Dian Wang
- Department of Forensic Medicine, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
17
|
Mason FE, Pronto JRD, Alhussini K, Maack C, Voigt N. Cellular and mitochondrial mechanisms of atrial fibrillation. Basic Res Cardiol 2020; 115:72. [PMID: 33258071 PMCID: PMC7704501 DOI: 10.1007/s00395-020-00827-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/26/2020] [Indexed: 11/06/2022]
Abstract
The molecular mechanisms underlying atrial fibrillation (AF), the most common form of arrhythmia, are poorly understood and therefore target-specific treatment options remain an unmet clinical need. Excitation–contraction coupling in cardiac myocytes requires high amounts of adenosine triphosphate (ATP), which is replenished by oxidative phosphorylation in mitochondria. Calcium (Ca2+) is a key regulator of mitochondrial function by stimulating the Krebs cycle, which produces nicotinamide adenine dinucleotide for ATP production at the electron transport chain and nicotinamide adenine dinucleotide phosphate for the elimination of reactive oxygen species (ROS). While it is now well established that mitochondrial dysfunction plays an important role in the pathophysiology of heart failure, this has been less investigated in atrial myocytes in AF. Considering the high prevalence of AF, investigating the role of mitochondria in this disease may guide the path towards new therapeutic targets. In this review, we discuss the importance of mitochondrial Ca2+ handling in regulating ATP production and mitochondrial ROS emission and how alterations, particularly in these aspects of mitochondrial activity, may play a role in AF. In addition to describing research advances, we highlight areas in which further studies are required to elucidate the role of mitochondria in AF.
Collapse
Affiliation(s)
- Fleur E Mason
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Julius Ryan D Pronto
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Khaled Alhussini
- Department of Thoracic and Cardiovascular Surgery, University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center Würzburg, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany. .,Department of Internal Medicine I, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany.
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany. .,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
18
|
Chinyere IR, Hutchinson M, Moukabary T, Koevary JW, Juneman E, Goldman S, Lancaster JJ. Modulating the Infarcted Ventricle's Refractoriness with an Epicardial Biomaterial. J Investig Med 2020; 69:364-370. [PMID: 33115956 DOI: 10.1136/jim-2020-001486] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2020] [Indexed: 11/03/2022]
Abstract
Patients diagnosed with heart failure with reduced ejection fraction (HFrEF) are at increased risk of monomorphic ventricular tachycardia (VT) and ventricular fibrillation. The presence of myocardial fibrosis provides both anatomical and functional barriers that promote arrhythmias in these patients. Propagation of VT in a reentrant circuit depends on the presence of excitable myocardium and the refractoriness of the circuit. We hypothesize that myocardial refractoriness can be modulated surgically in a model of HFrEF, leading to decreased susceptibility to VT.Male Sprague-Dawley rats were infarcted via permanent left coronary artery ligation. At 3 weeks post-infarction, engineered grafts composed of human dermal fibroblasts cultured into a polyglactin-910 biomaterial were implanted onto the epicardium to cover the area of infarction. Three weeks post-graft treatment, all rats underwent a terminal electrophysiologic study to compare monophasic action potential electroanatomic maps and susceptibility to inducible monomorphic VT.HFrEF rats (n=29) demonstrated a longer (p=0.0191) ventricular effective refractory period (ERP) and a greater (p=0.0394) VT inducibility compared with sham (n=7). HFrEF rats treated with the graft (n=12) exhibited no change in capture threshold (p=0.3220), but had a longer ventricular ERP (p=0.0029) compared with HFrEF. No statistically significant change in VT incidence was found between HFrEF rats treated with the graft and untreated HFrEF rats (p=0.0834).Surgical deployment of a fibroblast-containing biomaterial in a rodent ischemic cardiomyopathy model prolonged ventricular ERP as measured by programmed electrical stimulation. This hypothesis-generating study warrants additional studies to further characterize the antiarrhythmic or proarrhythmic effects of this novel surgical therapy.
Collapse
Affiliation(s)
| | - Mathew Hutchinson
- Sarver Heart Center, University of Arizona Arizona Health Sciences Center, Tucson, Arizona, USA
| | - Talal Moukabary
- Sarver Heart Center, University of Arizona Arizona Health Sciences Center, Tucson, Arizona, USA
| | - Jen Watson Koevary
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA
| | - Elizabeth Juneman
- Sarver Heart Center, University of Arizona Arizona Health Sciences Center, Tucson, Arizona, USA
| | - Steven Goldman
- Sarver Heart Center, University of Arizona Arizona Health Sciences Center, Tucson, Arizona, USA
| | - Jordan J Lancaster
- Sarver Heart Center, University of Arizona Arizona Health Sciences Center, Tucson, Arizona, USA
| |
Collapse
|
19
|
Yu J, Zhou Y, Li L, Zhang K, Gao L, He X, Dong H. Identification of Biomarkers Related to Atrial Fibrillation With Mitral Regurgitation. Am J Med Sci 2020; 361:319-326. [PMID: 33541709 DOI: 10.1016/j.amjms.2020.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 09/24/2020] [Accepted: 10/07/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND We aimed to explore the biomarkers associated with atrial fibrillation (AF) with mitral regurgitation (MR). METHODS The gene expression profile data GSE115574 were downloaded from Gene Expression Omnibus database, which were obtained from patients with degenerative MR with AF and sinus rhythm (SR). The differentially expressed genes (DEGs) in samples of AF with MR compared with those of SR with MR were selected, followed by functional enrichment analysis, protein-protein interaction (PPI) network analysis, transcription factor (TF) prediction, and drug-gene interaction prediction. RESULTS By comparing the genes' expression profiles between AF with MR and SR with MR, 379 DEGs were obtained. The upregulated genes, such as NMNAT2, LDHB, and hexosaminidase subunit beta (HEXB), were significantly enriched in metabolic pathways. Hub genes, such as amyloid beta precursor protein (APP), CDH2, SPP1, and STC2, were significantly associated with functions related to extracellular matrix organization and vitamin D response. Additionally, two TFs, PRDM3 and LSM6, were predicted for the key module genes. APP predicted the most drug molecules, that is, 22 molecules, and SPP1 predicted 10 drug molecules. CONCLUSIONS Dysregulation of the metabolic pathway may play a critical role in AF with MR. Changes in functions related to the extracellular matrix and vitamin D response may also be associated with AF progression in patients with MR. Furthermore, APP, STC2, and SPP1 may serve as potential therapeutic targets of AF.
Collapse
Affiliation(s)
- Jiajian Yu
- Equipment Division, Foshan Nanhai District Health Care Hospital for Women and Children, Nanhai Children's Hospital, Foshan, China
| | - Yu Zhou
- Division of Vascular Surgery, National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangdong Engineering Laboratory of Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liwen Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Kunyi Zhang
- Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China; Department of Radiation Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Lijuan Gao
- Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China; Department of Radiation Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Xuyu He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Haojian Dong
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
20
|
Ng PCI, Chan JYW, Leung RKK, Li J, Ren Z, Chan AWH, Xu Y, Lee SS, Wang R, Ji X, Zheng J, Chan DPC, Yew WW, Lee SMY. Role of oxidative stress in clofazimine-induced cardiac dysfunction in a zebrafish model. Biomed Pharmacother 2020; 132:110749. [PMID: 33017766 DOI: 10.1016/j.biopha.2020.110749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/19/2020] [Accepted: 09/07/2020] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Clofazimine (CFZ), a riminophenazine, is now commonly used in the treatment of multidrug-resistant tuberculosis. However, its use may be potentially associated with cardiac dysfunction in some individuals. In this study, the zebrafish heart, by merit of its developmental and genetic characteristics being in homology with that of human, was chosen as an animal model for evaluation of such dysfunction. METHODS Morphological and physiological parameters were used to assess cardiac dysfunction. Transcriptome analysis was performed, followed by validation with real-time quantitative PCR, for delineation of the relevant genomics. RESULTS Exposure of 2 dpf zebrafish to 4 mg/L CFZ for 2 days, adversely affected cardiac functions including significant decreases in HR, SV, CO, and FS, with observable pathophysiological developments of pericardial effusion and blood accumulation in the heart, in comparison with the control group. In addition, genes which respond to xenobiotic stimulus, related to oxygen transport, glutathione metabolism and extracellular matrix -receptor interactions, were significantly enriched among the differentially up-regulated genes. Antioxidant response element motif was enriched in the 5000 base pair upstream regions of the differentially expressed genes. Co-administration of N-acetylcysteine was shown to protect zebrafish against the development of CFZ-induced cardiac dysfunction. CONCLUSIONS This study suggests an important role of oxidative stress as a major pathogenetic mechanism of riminophenazine-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Phoebe C I Ng
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Judy Y W Chan
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Ross K K Leung
- School of Public Health, University of Hong Kong, Hong Kong, China; Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China
| | - J Li
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Z Ren
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Anthony W H Chan
- Department of Anatomical & Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China
| | - Y Xu
- Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China
| | - S S Lee
- Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China
| | - R Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xia Ji
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Denise P C Chan
- Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China.
| | - W W Yew
- Stanley Ho Centre for Emerging Infectious Diseases, Chinese University of Hong Kong, Hong Kong, China.
| | - Simon M Y Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
21
|
Chakraborty P, Rose RA, Nair K, Downar E, Nanthakumar K. The rationale for repurposing funny current inhibition for management of ventricular arrhythmia. Heart Rhythm 2020; 18:130-137. [PMID: 32738405 DOI: 10.1016/j.hrthm.2020.07.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/14/2020] [Accepted: 07/25/2020] [Indexed: 11/26/2022]
Abstract
Management of ventricular arrhythmia in structural heart disease is complicated by the toxicity of the limited antiarrhythmic options available. In others, proarrhythmia and deleterious hemodynamic and noncardiac effects prevent practical use. This necessitates new thinking in therapeutic agents for ventricular arrhythmia in structural heart disease. Ivabradine, a funny current (If) inhibitor, has proven safety in heart failure, angina, and inappropriate sinus tachycardia. Although it is commonly known that funny channels are primarily expressed in the sinoatrial node, atrioventricular node, and conducting system of the ventricle, ivabradine is known to exert effects on metabolism, ion homeostasis, and membrane electrophysiology of remodeled ventricular myocardium. This review considers novel concepts and evidence from clinical and experimental studies regarding this paradigm, with a potential role of ivabradine in ventricular arrhythmia.
Collapse
Affiliation(s)
- Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; University Health Network, Toronto, Ontario, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute of Alberta, An entity of the University of Calgary and Alberta Health Services, Calgary, Alberta, Canada
| | - Krishnakumar Nair
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; University Health Network, Toronto, Ontario, Canada
| | - Eugene Downar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; University Health Network, Toronto, Ontario, Canada
| | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
22
|
Tourki B, Kain V, Shaikh SR, Leroy X, Serhan CN, Halade GV. Deficit of resolution receptor magnifies inflammatory leukocyte directed cardiorenal and endothelial dysfunction with signs of cardiomyopathy of obesity. FASEB J 2020; 34:10560-10573. [PMID: 32543720 DOI: 10.1096/fj.202000495rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
Chronic unresolved inflammation is the primary determinant of cardiovascular disease. Precise mechanisms that define the genesis of unresolved inflammation in heart failure with preserved ejection fraction (HFpEF) are of interest due to the obesity epidemic. To examine the obesity phenotype and its direct/indirect consequences, multiple approaches were employed using the lipoxin receptor (abbreviated as ALX) dysfunction mouse model. Indirect calorimetry analyses revealed that the deletion of ALX dysregulated energy metabolism driving toward age-related obesity. Heart function data suggest that obesity-prone ALX deficient mice had impaired myocardium strain. Comprehensive measurement of chemokines, extracellular matrix, and arrhythmogenic arrays confirmed the dysregulation of multiple ion channels gene expression with amplified inflammatory chemokines and cytokines response at the age of 4 months compared with WT counterparts. Quantitative analyses of leukocytes demonstrated an increase of proinflammatory Ly6Chi CCR2+ macrophages in the spleen and heart at a steady-state resulting in an inflamed splenocardiac axis. Signs of subtle inflammation were marked with cardiorenal, endothelial defects with decreased CD31 and eNOS and an increased iNOS and COX2 expression. Thus, ALX receptor deficiency serves as an experimental model that defines multiple cellular and molecular mechanisms in HFpEF that could be a target for the development of HFpEF therapy in cardiovascular medicine.
Collapse
Affiliation(s)
- Bochra Tourki
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, USA
| | - Vasundhara Kain
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, USA
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
23
|
Ning H, Cui Y, Song X, Chen L, Yin Z, Hua L, Ren F, Suo Y, Wang X, Zhang H, Hu D, Ge Y. iTRAQ-based proteomic analysis reveals key proteins affecting cardiac function in broilers that died of sudden death syndrome. Poult Sci 2020; 98:6472-6482. [PMID: 31509194 PMCID: PMC8913949 DOI: 10.3382/ps/pez532] [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: 08/12/2019] [Accepted: 08/31/2019] [Indexed: 12/26/2022] Open
Abstract
Sudden death syndrome (SDS), which is a cardiac-related condition commonly observed in chickens selected for rapid growth, causes significant economic losses to the global poultry industry. Its pathogenesis in broilers is poorly understood, and little is known about the proteome of the heart tissue of SDS broilers. A quantitative proteomic approach using isobaric tags for relative and absolute quantification labeling of peptides was used to characterize the protein expression profiles in the left ventricle of SDS broilers. These proteins were further analyzed by bioinformatics, and two proteins were validated by western blot analysis. We identified 186 differentially expressed proteins (DEPs), of which 72 were upregulated, and 114 were downregulated in the SDS group. Functional annotation suggested that 7 DEPs were related to cardiac muscle contraction, and another 7 DEPs were related to cardiac energy metabolism. Protein interaction network predictions indicated that differences in cardiac muscle contraction between SDS and healthy groups were regulated by troponin T, tropomyosin alpha-1 chain, fast myosin heavy chain HCIII, myosin-1B, coronin, and myoglobin, whereas differences in cardiac energy metabolism and biosynthesis of amino acids were regulated by gamma-enolase, phosphoglycerate mutase, NADH-ubiquinone oxidoreductase chain 2, serine/threonine-protein kinase, myoglobin, and alpha-amylase. Our expression profiles provide useful information and new insights into key proteins to elucidate SDS for further studies.
Collapse
Affiliation(s)
- Hongmei Ning
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Yunli Cui
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Xiaochao Song
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Lingli Chen
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Zhihong Yin
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China.,Postdoctoral Research and Development Base, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Liushuai Hua
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Fei Ren
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Yu Suo
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Xinrui Wang
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Hongli Zhang
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Dongfang Hu
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China.,Postdoctoral Research and Development Base, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Yaming Ge
- College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China
| |
Collapse
|
24
|
Wu Y, Zhang Y, Chen M, Yang Q, Zhuang S, Lv L, Zuo Z, Wang C. Exposure to low-level metalaxyl impacts the cardiac development and function of zebrafish embryos. J Environ Sci (China) 2019; 85:1-8. [PMID: 31471016 DOI: 10.1016/j.jes.2019.03.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 03/21/2019] [Accepted: 03/21/2019] [Indexed: 06/10/2023]
Abstract
Metalaxyl is an anilide pesticide that is widely used to control plant diseases caused by Peronosporales species. In order to study the toxic effects, zebrafish embryos were exposed to metalaxyl at nominal concentrations of 5, 50 and 500 ng/L for 72 hr, and the cardiac development and functioning of larvae were observed. The results showed that metalaxyl exposure resulted in increased rates of pericardial edema, heart hemorrhage and cardiac malformation. The distance between the sinus venosus and bulbus arteriosus, stroke volume, cardiac output and heart rate were significantly increased in larvae exposed to 50 and 500 ng/L metalaxyl compared to solvent control larvae. Significant upregulation in the transcription of tbx5, gata4 and myh6 was observed in the 50 and 500 ng/L treatments, and that of nkx2.5 and myl7 was observed in the 5, 50 and 500 ng/L groups. These disturbances may be related to cardiac developmental and functional defects in the larvae. The activity of Na+/K+-ATPase and Ca2+-ATPase was significantly increased in zebrafish embryos exposed to 500 ng/L metalaxyl, and the mRNA levels of genes related to ATPase (atp2a11, atp1b2b, and atp1a3b) (in the 50 and 500 ng/L groups) and calcium channels (cacna1ab) (in the 500 ng/L group) were significantly downregulated; these changes might be associated with heart arrhythmia and functional failure.
Collapse
Affiliation(s)
- Yuqiong Wu
- Wuyi University, College of Tea and Food Science, Wuyishan 354300, China.
| | - Ying Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China.
| | - Meng Chen
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem Research, Xiamen University, Xiamen 361005, China
| | - Qihong Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Shanshan Zhuang
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem Research, Xiamen University, Xiamen 361005, China
| | - Liangju Lv
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
25
|
Ribeiro AJS, Guth BD, Engwall M, Eldridge S, Foley CM, Guo L, Gintant G, Koerner J, Parish ST, Pierson JB, Brock M, Chaudhary KW, Kanda Y, Berridge B. Considerations for an In Vitro, Cell-Based Testing Platform for Detection of Drug-Induced Inotropic Effects in Early Drug Development. Part 2: Designing and Fabricating Microsystems for Assaying Cardiac Contractility With Physiological Relevance Using Human iPSC-Cardiomyocytes. Front Pharmacol 2019; 10:934. [PMID: 31555128 PMCID: PMC6727630 DOI: 10.3389/fphar.2019.00934] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022] Open
Abstract
Contractility of the myocardium engines the pumping function of the heart and is enabled by the collective contractile activity of its muscle cells: cardiomyocytes. The effects of drugs on the contractility of human cardiomyocytes in vitro can provide mechanistic insight that can support the prediction of clinical cardiac drug effects early in drug development. Cardiomyocytes differentiated from human-induced pluripotent stem cells have high potential for overcoming the current limitations of contractility assays because they attach easily to extracellular materials and last long in culture, while having human- and patient-specific properties. Under these conditions, contractility measurements can be non-destructive and minimally invasive, which allow assaying sub-chronic effects of drugs. For this purpose, the function of cardiomyocytes in vitro must reflect physiological settings, which is not observed in cultured cardiomyocytes derived from induced pluripotent stem cells because of the fetal-like properties of their contractile machinery. Primary cardiomyocytes or tissues of human origin fully represent physiological cellular properties, but are not easily available, do not last long in culture, and do not attach easily to force sensors or mechanical actuators. Microengineered cellular systems with a more mature contractile function have been developed in the last 5 years to overcome this limitation of stem cell-derived cardiomyocytes, while simultaneously measuring contractile endpoints with integrated force sensors/actuators and image-based techniques. Known effects of engineered microenvironments on the maturity of cardiomyocyte contractility have also been discovered in the development of these systems. Based on these discoveries, we review here design criteria of microengineered platforms of cardiomyocytes derived from pluripotent stem cells for measuring contractility with higher physiological relevance. These criteria involve the use of electromechanical, chemical and morphological cues, co-culture of different cell types, and three-dimensional cellular microenvironments. We further discuss the use and the current challenges for developing and improving these novel technologies for predicting clinical effects of drugs based on contractility measurements with cardiomyocytes differentiated from induced pluripotent stem cells. Future research should establish contexts of use in drug development for novel contractility assays with stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Brian D Guth
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany.,PreClinical Drug Development Platform (PCDDP), North-West University, Potchefstroom, South Africa
| | - Michael Engwall
- Safety Pharmacology and Animal Research Center, Amgen Research, Thousand Oaks, CA, United States
| | - Sandy Eldridge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - C Michael Foley
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - Liang Guo
- Laboratory of Investigative Toxicology, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Gary Gintant
- Department of Integrative Pharmacology, Integrated Sciences and Technology, AbbVie, North Chicago, IL, United States
| | - John Koerner
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Stanley T Parish
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Jennifer B Pierson
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Mathew Brock
- Department of Safety Assessment, Genentech, South San Francisco, CA, United States
| | - Khuram W Chaudhary
- Global Safety Pharmacology, GlaxoSmithKline plc, Collegeville, PA, United States
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa, Japan
| | - Brian Berridge
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| |
Collapse
|
26
|
Hu HJ, Zhang C, Tang ZH, Qu SL, Jiang ZS. Regulating the Warburg effect on metabolic stress and myocardial fibrosis remodeling and atrial intracardiac waveform activity induced by atrial fibrillation. Biochem Biophys Res Commun 2019; 516:653-660. [PMID: 31242971 DOI: 10.1016/j.bbrc.2019.06.055] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 10/26/2022]
Abstract
Atrial fibrillation (AF) is associated with metabolic stress and induces myocardial fibrosis reconstruction by increasing glycolysis. One goal in the treatment of paroxysmal AF (p-AF) is to improve myocardial fibrosis reconstruction and myocardial metabolic stress caused by the Warburg effect. Adopted male canine that rapid right atrial pacing (RAP) for 6 days to establish a p-AF model. The canines were pre-treated with phenylephrine (PE) or dichloroacetic acid (DCA) before exposure to p-AF or non-p-AF. P-wave duration (Pmax), minimum P-wave duration (Pmin), P wave dispersion (PWD), atrial effective refractory period (AERP) and AERP dispersion (AERPd) were measured in canine atrial cardiomyocytes. Pyruvate dehydrogenase kinase-1 (PDK-1), PDK-4, lactate dehydrogenase A (LDHA), pyruvate dehydrogenase (PDH), citrate synthase (CS), isocitrate dehydrogenase (IDH), and matrix metalloproteinase 9 (MMP-9) were evaluated by western blotting and reverse transcription polymerase chain reaction (RT-PCR), content of adenosine monophosphate (AMP), adenosine triphosphate (ATP), lactic acid and glycogen, and activity of LDHA, PDK-1 and PDK-4 were evaluated by enzyme-linked immunosorbent assay (ELISA), myocardial tissue glycogen content was evaluated by PAS, myocardial fibrosis remodeling was evaluated by hematoxylin and eosin (H&E) and Masson staining. Our findings demonstrated that p-AF increases the Warburg effect-related metabolic stress and myocardial fibrosis remodeling by increasing the expression and activity of PDK-1, PDK-4, and LDHA, content of AMP and lactic acid, and the ratio of AMP/ATP and decreasing the expression of PDH, CS, and IDH, and glycogen content. In addition, p-AF can induce cardiomyocyte fibrosis remodeling and increase MMP-9 expression, and p-AF also increases atrial intracardiac waveform activity by prolonging Pmax, Pmin, PWD, and AERPd and shortening AERP. PDK isoforms agonists (PE) produce a similar p-AF pathological effect and can produce synergistic effects with p-AF, further increasing Warburg effect-related metabolic stress, myocardial fibrosis remodeling, and atrial intracardiac waveform activity. In contrast, the use of PDK-specific inhibitors (DCA) completely reverses these pathophysiological changes induced by p-AF. We demonstrate that p-AF can induce the Warburg effect in canine atrial cardiomyocytes and significantly improve p-AF-induced metabolic stress, myocardial fibrosis remodeling, and atrial intracardiac waveform activity by inhibiting the Warburg effect.
Collapse
Affiliation(s)
- Heng-Jing Hu
- Department of Cardiology Lab, First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China; Postdoctoral Research Station of Basic Medicine, University of South China, Hengyang, Hunan Province, China
| | - Chi Zhang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan Province, China
| | - Zhi-Han Tang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan Province, China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan Province, China
| | - Zhi-Sheng Jiang
- Department of Cardiology Lab, First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China; Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan Province, China; Postdoctoral Research Station of Basic Medicine, University of South China, Hengyang, Hunan Province, China.
| |
Collapse
|
27
|
Hegyi B, Bers DM, Bossuyt J. CaMKII signaling in heart diseases: Emerging role in diabetic cardiomyopathy. J Mol Cell Cardiol 2019; 127:246-259. [PMID: 30633874 DOI: 10.1016/j.yjmcc.2019.01.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023]
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) is upregulated in diabetes and significantly contributes to cardiac remodeling with increased risk of cardiac arrhythmias. Diabetes is frequently associated with atrial fibrillation, coronary artery disease, and heart failure, which may further enhance CaMKII. Activation of CaMKII occurs downstream of neurohormonal stimulation (e.g. via G-protein coupled receptors) and involve various posttranslational modifications including autophosphorylation, oxidation, S-nitrosylation and O-GlcNAcylation. CaMKII signaling regulates diverse cellular processes in a spatiotemporal manner including excitation-contraction and excitation-transcription coupling, mechanics and energetics in cardiac myocytes. Chronic activation of CaMKII results in cellular remodeling and ultimately arrhythmogenic alterations in Ca2+ handling, ion channels, cell-to-cell coupling and metabolism. This review addresses the detrimental effects of the upregulated CaMKII signaling to enhance the arrhythmogenic substrate and trigger mechanisms in the heart. We also briefly summarize preclinical studies using kinase inhibitors and genetically modified mice targeting CaMKII in diabetes. The mechanistic understanding of CaMKII signaling, cardiac remodeling and arrhythmia mechanisms may reveal new therapeutic targets and ultimately better treatment in diabetes and heart disease in general.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| |
Collapse
|
28
|
Yew WW, Chang KC. Management of adverse reactions to high-dose moxifloxacin used in multidrug-resistant tuberculosis treatment programmes. Respirology 2018; 24:201-203. [PMID: 30520535 DOI: 10.1111/resp.13452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Wing Wai Yew
- Stanley Ho Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Chiu Chang
- Tuberculosis and Chest Service, Centre for Health Protection, Department of Health, Hong Kong, China
| |
Collapse
|
29
|
Grisanti LA. Diabetes and Arrhythmias: Pathophysiology, Mechanisms and Therapeutic Outcomes. Front Physiol 2018; 9:1669. [PMID: 30534081 PMCID: PMC6275303 DOI: 10.3389/fphys.2018.01669] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/06/2018] [Indexed: 12/17/2022] Open
Abstract
The prevalence of diabetes is rapidly increasing and closely associated with cardiovascular morbidity and mortality. While the major cardiovascular complication associated with diabetes is coronary artery disease, it is becoming increasingly apparent that diabetes impacts the electrical conduction system in the heart, resulting in atrial fibrillation, and ventricular arrhythmias. The relationship between diabetes and arrhythmias is complex and multifactorial including autonomic dysfunction, atrial and ventricular remodeling and molecular alterations. This review will provide a comprehensive overview of the link between diabetes and arrhythmias with insight into the common molecular mechanisms, structural alterations and therapeutic outcomes.
Collapse
Affiliation(s)
- Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
30
|
Mačianskienė R, Martišienė I, Navalinskas A, Treinys R, Andriulė I, Jurevičius J. Mechanism of Action Potential Prolongation During Metabolic Inhibition in the Whole Rabbit Heart. Front Physiol 2018; 9:1077. [PMID: 30140239 PMCID: PMC6095129 DOI: 10.3389/fphys.2018.01077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/19/2018] [Indexed: 11/24/2022] Open
Abstract
Myocardial ischemia is associated with significant changes in action potential (AP) duration, which has a biphasic response to metabolic inhibition. Here, we investigated the mechanism of initial AP prolongation in whole Langendorff-perfused rabbit heart. We used glass microelectrodes to record APs transmurally. Simultaneously, optical AP, calcium transient (CaT), intracellular pH, and magnesium concentration changes were recorded using fluorescent dyes. The fluorescence signals were recorded using an EMCCD camera equipped with emission filters; excitation was induced by LEDs. We demonstrated that metabolic inhibition by carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP) resulted in AP shortening preceded by an initial prolongation and that there were no important differences in the response throughout the wall of the heart and in the apical/basal direction. AP prolongation was reduced by blocking the ICaL and transient outward potassium current (Ito) with diltiazem (DTZ) and 4-aminopyridine (4-AP), respectively. FCCP, an uncoupler of oxidative phosphorylation, induced reductions in CaTs and intracellular pH and increased the intracellular Mg2+ concentration. In addition, resting potential depolarization was observed, clearly indicating a decrease in the inward rectifier K+ current (IK1) that can retard AP repolarization. Thus, we suggest that the main currents responsible for AP prolongation during metabolic inhibition are the ICaL, Ito, and IK1, the activities of which are modulated mainly by changes in intracellular ATP, calcium, magnesium, and pH.
Collapse
Affiliation(s)
- Regina Mačianskienė
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Irma Martišienė
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Antanas Navalinskas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rimantas Treinys
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Inga Andriulė
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Jonas Jurevičius
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
31
|
Crook R, Issitt R. Cardiopulmonary Bypass for a Patient With Congenital Hyperinsulinemia. World J Pediatr Congenit Heart Surg 2018; 11:NP117-NP119. [PMID: 29334830 DOI: 10.1177/2150135117742649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Congenital hyperinsulinism is a clinical syndrome of pancreatic β-cell dysfunction characterized by failure to suppress insulin secretion in the presence of hypoglycemia. Here, we describe the concerns, the techniques used to ameliorate these potential problems, and the outcomes for a child with this condition undergoing cardiopulmonary bypass for correction of an atrial septal defect.
Collapse
Affiliation(s)
- Richard Crook
- Department of Perfusion, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Richard Issitt
- Department of Perfusion, Great Ormond Street Hospital for Children, London, United Kingdom
| |
Collapse
|
32
|
Matasic DS, Brenner C, London B. Emerging potential benefits of modulating NAD + metabolism in cardiovascular disease. Am J Physiol Heart Circ Physiol 2017; 314:H839-H852. [PMID: 29351465 DOI: 10.1152/ajpheart.00409.2017] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) and related metabolites are central mediators of fuel oxidation and bioenergetics within cardiomyocytes. Additionally, NAD+ is required for the activity of multifunctional enzymes, including sirtuins and poly(ADP-ribose) polymerases that regulate posttranslational modifications, DNA damage responses, and Ca2+ signaling. Recent research has indicated that NAD+ participates in a multitude of processes dysregulated in cardiovascular diseases. Therefore, supplementation of NAD+ precursors, including nicotinamide riboside that boosts or repletes the NAD+ metabolome, may be cardioprotective. This review examines the molecular physiology and preclinical data with respect to NAD+ precursors in heart failure-related cardiac remodeling, ischemic-reperfusion injury, and arrhythmias. In addition, alternative NAD+-boosting strategies and potential systemic effects of NAD+ supplementation with implications on cardiovascular health and disease are surveyed.
Collapse
Affiliation(s)
- Daniel S Matasic
- Division of Cardiovascular Medicine, Department of Medicine, University of Iowa , Iowa City, Iowa.,Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa , Iowa City, Iowa.,Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
| | - Charles Brenner
- Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa.,Department of Biochemistry, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Barry London
- Division of Cardiovascular Medicine, Department of Medicine, University of Iowa , Iowa City, Iowa.,Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa , Iowa City, Iowa.,Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
| |
Collapse
|
33
|
Rodgers JL, Samal E, Mohapatra S, Panguluri SK. Hyperoxia-induced cardiotoxicity and ventricular remodeling in type-II diabetes mice. Heart Vessels 2017; 33:561-572. [PMID: 29209776 DOI: 10.1007/s00380-017-1100-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/01/2017] [Indexed: 12/14/2022]
Abstract
Hyperoxia, or supplemental oxygen, is regularly used in the clinical setting for critically ill patients in ICU. However, several recent studies have demonstrated the negative impact of this treatment in patients in critical care, including increased rates of lung and cardiac injury, as well as increased mortality. The purpose of this study was to determine the predisposition for arrhythmias and electrical remodeling in a type 2 diabetic mouse model (db/db), as a result of hyperoxia treatment. For this, db/db and their heterozygous controls were treated with hyperoxia (> 90% oxygen) or normoxia (normal air) for 72-h. Immediately following hyperoxia or normoxia treatments, mice underwent surface ECG. Excised left ventricles were used to assess ion channel expression, including for Kv1.4, Kv1.5, Kv4.2, and KChIP2. Serum cardiac markers were also measured, including cardiac troponin I and lactate dehydrogenase. Our results showed that db/db mice have increased sensitivity to arrhythmia. Normoxia-treated db/db mice displayed features of arrhythmia, including QTc and JT prolongation, as well as QRS prolongation. A significant increase in QRS prolongation was also observed in hyperoxia-treated db/db mice, when compared to hyperoxia-treated heterozygous control mice. Db/db mice were also shown to exhibit ion channel dysregulation, as demonstrated by down-regulation in Kv1.5, Kv4.2, and KChIP2 under hyperoxia conditions. From these results, we conclude that: (1) diabetic mice showed distinct pathophysiology, when compared to heterozygous controls, both in normoxia and hyperoxia conditions. (2) Diabetic mice were more susceptible to arrhythmia at normal air conditions; this effect was exacerbated at hyperoxia conditions. (3) Unlike in heterozygous controls, diabetic mice did not demonstrate cardiac hypertrophy as a result of hyperoxia. (4) Ion channel remodeling was also observed in db/db mice under hyperoxia condition similar to its heterozygous controls.
Collapse
Affiliation(s)
- Jennifer Leigh Rodgers
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., MDC-30, Tampa, FL, 33612, USA
| | - Eva Samal
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Subhra Mohapatra
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Siva Kumar Panguluri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., MDC-30, Tampa, FL, 33612, USA.
| |
Collapse
|
34
|
Valli H, Ahmad S, Fraser JA, Jeevaratnam K, Huang CLH. Pro-arrhythmic atrial phenotypes in incrementally paced murine Pgc1β -/- hearts: effects of age. Exp Physiol 2017; 102:1619-1634. [PMID: 28960529 PMCID: PMC5725712 DOI: 10.1113/ep086589] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
New Findings What is the central question of this study? Can we experimentally replicate atrial pro‐arrhythmic phenotypes associated with important chronic clinical conditions, including physical inactivity, obesity, diabetes mellitus and metabolic syndrome, compromising mitochondrial function, and clarify their electrophysiological basis? What is the main finding and its importance? Electrocardiographic and intracellular cardiomyocyte recording at progressively incremented pacing rates demonstrated age‐dependent atrial arrhythmic phenotypes in Langendorff‐perfused murine Pgc1β−/− hearts for the first time. We attributed these to compromised action potential conduction and excitation wavefronts, whilst excluding alterations in recovery properties or temporal electrophysiological instabilities, clarifying these pro‐arrhythmic changes in chronic metabolic disease.
Atrial arrhythmias, most commonly manifesting as atrial fibrillation, represent a major clinical problem. The incidence of atrial fibrillation increases with both age and conditions associated with energetic dysfunction. Atrial arrhythmic phenotypes were compared in young (12–16 week) and aged (>52 week) wild‐type (WT) and peroxisome proliferative activated receptor, gamma, coactivator 1 beta (Ppargc1b)‐deficient (Pgc1β−/−) Langendorff‐perfused hearts, previously used to model mitochondrial energetic disorder. Electrophysiological explorations were performed using simultaneous whole‐heart ECG and intracellular atrial action potential (AP) recordings. Two stimulation protocols were used: an S1S2 protocol, which imposed extrasystolic stimuli at successively decremented intervals following regular pulse trains; and a regular pacing protocol at successively incremented frequencies. Aged Pgc1β−/− hearts showed greater atrial arrhythmogenicity, presenting as atrial tachycardia and ectopic activity. Maximal rates of AP depolarization (dV/dtmax) were reduced in Pgc1β−/− hearts. Action potential latencies were increased by the Pgc1β−/− genotype, with an added interactive effect of age. In contrast, AP durations to 90% recovery (APD90) were shorter in Pgc1β−/− hearts despite similar atrial effective recovery periods amongst the different groups. These findings accompanied paradoxical decreases in the incidence and duration of alternans in the aged and Pgc1β−/− hearts. Limiting slopes of restitution curves of APD90 against diastolic interval were correspondingly reduced interactively by Pgc1β−/− genotype and age. In contrast, reduced AP wavelengths were associated with Pgc1β−/− genotype, both independently and interacting with age, through the basic cycle lengths explored, with the aged Pgc1β−/− hearts showing the shortest wavelengths. These findings thus implicate AP wavelength in possible mechanisms for the atrial arrhythmic changes reported here.
Collapse
Affiliation(s)
- Haseeb Valli
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - James A Fraser
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Kamalan Jeevaratnam
- Physiological Laboratory, University of Cambridge, Cambridge, UK.,PU-RCSI School of Medicine, Perdana University, Serdang, Selangor Darul Ehsan, Malaysia.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Cambridge, UK.,Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
35
|
Cardiotoxic (Arrhythmogenic) Effects of 1,1-Difluoroethane Due to Electrolyte Imbalance and Cardiomyocyte Damage. Am J Forensic Med Pathol 2017; 38:115-125. [PMID: 28263233 DOI: 10.1097/paf.0000000000000262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Inhalant abuse is the intentional inhalation of chemical vapors to attain euphoric effects. Many common household products are abused by inhalation and one is 1,1-difluoroethane (DFE), which is a halogenated hydrocarbon used in refrigeration, dust-off spray, and airbrush painting. Although many human DFE abuse cases have been studied, the etiology and mechanism of sudden death is still unknown. In this study, an animal model was used to simulate the human conditions of DFE inhalation abuse that results in sudden death.Current research targets mechanistic studies involving electrolyte changes and cardiomyocyte damage after DFE administration in vivo. To investigate these changes, Sprague Dawley rats (N = 6) were exposed to 30 seconds of 20 L/min of DFE in multiple doses. Isoflurane acted as a control. Two additional groups, epinephrine and epinephrine + DFE, were included to simulate the clinical condition of DFE abuse. Plasma sodium, potassium, calcium, and magnesium levels were measured, followed by lactate dehydrogenase, creatine kinase, and cardiac troponin I levels. In addition, oxidative stress markers were also evaluated in all animal groups. Electrolyte levels showed a significant rise in plasma potassium and magnesium levels for the treated groups. In addition, lactate dehydrogenase, creatine kinase, and cardiac troponin I levels in DFE and epinephrine + DFE administered rats were significantly elevated as compared with control. Some oxidative stress makers were also elevated significantly in treatment groups. Furthermore, histopathological analysis showed hyperemia/congestion in treated rats.These results support cardiotoxic effects indicating that DFE results in fatal arrhythmias, and the study can be important during clinical cases involving inhalant abuse.
Collapse
|
36
|
Gore-Panter SR, Rennison JH, Van Wagoner DR. Genetic-Genomic Insights Into the Metabolic Determinants of Spontaneous Atrial Fibrillation. Circ Arrhythm Electrophysiol 2017; 10:CIRCEP.117.005636. [PMID: 28784606 DOI: 10.1161/circep.117.005636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
| | - Julie H Rennison
- From the Department of Molecular Cardiology, Cleveland Clinic, OH
| | | |
Collapse
|
37
|
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: 4.7] [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
|
38
|
Steiner H, Geist M, Goldenberg I, Suleiman M, Glikson M, Tenenbaum A, Swissa M, Fisman EZ, Golovchiner G, Strasberg B, Barsheshet A. Characteristics and outcomes of diabetic patients with an implantable cardioverter defibrillator in a real world setting: results from the Israeli ICD registry. Cardiovasc Diabetol 2016; 15:160. [PMID: 27905927 PMCID: PMC5134232 DOI: 10.1186/s12933-016-0478-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/22/2016] [Indexed: 01/24/2023] Open
Abstract
Aims There are limited data regarding the effect of diabetes mellitus (DM) on the risks of both appropriate and inappropriate implantable cardioverter defibrillator (ICD) therapy. The present study was designed to compare the outcome of appropriate and inappropriate ICD therapy in patients with or without DM. Methods and results The risk of a first appropriate ICD therapy for ventricular tachyarrhythmias (including anti tachycardia pacing and shock) was compared between 764 DM and 1346 non-DM patients enrolled in the national Israeli ICD registry. We also compared the risks of inappropriate ICD therapy, and death or cardiac hospitalization between diabetic and non-diabetic patients. Diabetic patients were older, were more likely to have ischemic cardiomyopathy, lower ejection fraction, atrial fibrillation, and other co-morbidities. The 3-year cumulative incidence of appropriate ICD therapy was similar in the DM and non-DM groups (12 and 13%, respectively, p = 0.983). Multivariate analysis showed that DM did not affect the risk of appropriate ICD therapy (HR = 1.07, 95% CI 0.78–1.47, p = 0.694) or inappropriate therapy (HR = 0.72, 95% CI 0.42–1.23, p = 0.232). However, DM was associated with a 31% increased risk for death or cardiac hospitalization (p = 0.005). Results were similar in subgroup analyses including ICD and defibrillators with cardiac resynchronization therapy function recipients, primary or secondary prevention indication for an ICD. Conclusions Despite a significant excess of cardiac hospitalizations and mortality in the diabetic population, there was no difference in the rate of ICD treatments, suggesting that the outcome difference is not related to arrhythmias.
Collapse
Affiliation(s)
- Hillel Steiner
- The Edith Wolfson Medical Center, Holon, affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Department of Cardiology, The Edith Wolfson Medical Center, Holon, Israel.
| | - Michael Geist
- The Edith Wolfson Medical Center, Holon, affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Goldenberg
- The Chaim Sheba Medical Center, Tel Hashomer, affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Michael Glikson
- The Chaim Sheba Medical Center, Tel Hashomer, affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alexander Tenenbaum
- The Chaim Sheba Medical Center, Tel Hashomer, affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Cardiovascular Diabetology Research Foundation, Holon, Israel
| | - Moshe Swissa
- Kaplan Medical Center, Rehovot The Hebrew University, Jerusalem, Israel
| | - Enrique Z Fisman
- The Chaim Sheba Medical Center, Tel Hashomer, affiliated with the Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Cardiovascular Diabetology Research Foundation, Holon, Israel
| | | | | | | | | |
Collapse
|
39
|
Devalla HD, Gélinas R, Aburawi EH, Beqqali A, Goyette P, Freund C, Chaix MA, Tadros R, Jiang H, Le Béchec A, Monshouwer-Kloots JJ, Zwetsloot T, Kosmidis G, Latour F, Alikashani A, Hoekstra M, Schlaepfer J, Mummery CL, Stevenson B, Kutalik Z, de Vries AA, Rivard L, Wilde AA, Talajic M, Verkerk AO, Al-Gazali L, Rioux JD, Bhuiyan ZA, Passier R. TECRL, a new life-threatening inherited arrhythmia gene associated with overlapping clinical features of both LQTS and CPVT. EMBO Mol Med 2016; 8:1390-1408. [PMID: 27861123 PMCID: PMC5167130 DOI: 10.15252/emmm.201505719] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Genetic causes of many familial arrhythmia syndromes remain elusive. In this study, whole‐exome sequencing (WES) was carried out on patients from three different families that presented with life‐threatening arrhythmias and high risk of sudden cardiac death (SCD). Two French Canadian probands carried identical homozygous rare variant in TECRL gene (p.Arg196Gln), which encodes the trans‐2,3‐enoyl‐CoA reductase‐like protein. Both patients had cardiac arrest, stress‐induced atrial and ventricular tachycardia, and QT prolongation on adrenergic stimulation. A third patient from a consanguineous Sudanese family diagnosed with catecholaminergic polymorphic ventricular tachycardia (CPVT) had a homozygous splice site mutation (c.331+1G>A) in TECRL. Analysis of intracellular calcium ([Ca2+]i) dynamics in human induced pluripotent stem cell‐derived cardiomyocytes (hiPSC‐CMs) generated from this individual (TECRLHom‐hiPSCs), his heterozygous but clinically asymptomatic father (TECRLHet‐hiPSCs), and a healthy individual (CTRL‐hiPSCs) from the same Sudanese family, revealed smaller [Ca2+]i transient amplitudes as well as elevated diastolic [Ca2+]i in TECRLHom‐hiPSC‐CMs compared with CTRL‐hiPSC‐CMs. The [Ca2+]i transient also rose markedly slower and contained lower sarcoplasmic reticulum (SR) calcium stores, evidenced by the decreased magnitude of caffeine‐induced [Ca2+]i transients. In addition, the decay phase of the [Ca2+]i transient was slower in TECRLHom‐hiPSC‐CMs due to decreased SERCA and NCX activities. Furthermore, TECRLHom‐hiPSC‐CMs showed prolonged action potentials (APs) compared with CTRL‐hiPSC‐CMs. TECRL knockdown in control human embryonic stem cell‐derived CMs (hESC‐CMs) also resulted in significantly longer APs. Moreover, stimulation by noradrenaline (NA) significantly increased the propensity for triggered activity based on delayed afterdepolarizations (DADs) in TECRLHom‐hiPSC‐CMs and treatment with flecainide, a class Ic antiarrhythmic drug, significantly reduced the triggered activity in these cells. In summary, we report that mutations in TECRL are associated with inherited arrhythmias characterized by clinical features of both LQTS and CPVT. Patient‐specific hiPSC‐CMs recapitulated salient features of the clinical phenotype and provide a platform for drug screening evidenced by initial identification of flecainide as a potential therapeutic. These findings have implications for diagnosis and treatment of inherited cardiac arrhythmias.
Collapse
Affiliation(s)
- Harsha D Devalla
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Roselle Gélinas
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Elhadi H Aburawi
- Department of Pediatrics, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Abdelaziz Beqqali
- Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Christian Freund
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center hiPSC Core Facility, Leiden, The Netherlands
| | - Marie-A Chaix
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Rafik Tadros
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hui Jiang
- Beijing Genomics Institute, Shenzhen, China.,Shenzhen Key Laboratory of Genomics, Shenzhen, China.,The Guangdong Enterprise Key Laboratory of Human Disease Genomics, Shenzhen, China
| | - Antony Le Béchec
- Vital-IT group, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Tom Zwetsloot
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Georgios Kosmidis
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Maaike Hoekstra
- Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jurg Schlaepfer
- Service de Cardiologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Christine L Mummery
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Brian Stevenson
- Vital-IT group, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Zoltan Kutalik
- Vital-IT group, Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Institute of Social and Preventive Medicine, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Antoine Af de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,ICIN-Netherlands Heart Institute, Utrecht, The Netherlands
| | - Léna Rivard
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Arthur Am Wilde
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders, Jeddah, Saudi Arabia
| | - Mario Talajic
- Montreal Heart Institute, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Arie O Verkerk
- Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lihadh Al-Gazali
- Department of Pediatrics, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - John D Rioux
- Montreal Heart Institute, Montreal, QC, Canada .,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Zahurul A Bhuiyan
- Laboratoire Génétiqué Moléculaire, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Robert Passier
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands .,Department of Applied Stem Cell Technologies, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| |
Collapse
|
40
|
Ritchie HE, Ragnerstam C, Gustafsson E, Jonsson JM, Webster WS. Control of the heart rate of rat embryos during the organogenic period. HYPOXIA 2016; 4:147-159. [PMID: 27878135 PMCID: PMC5108485 DOI: 10.2147/hp.s115050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The aim of this study was to gain insight into whether the first trimester embryo could control its own heart rate (HR) in response to hypoxia. The gestational day 13 rat embryo is a good model for the human embryo at 5–6 weeks gestation, as the heart is comparable in development and, like the human embryo, has no functional autonomic nerve supply at this stage. Utilizing a whole-embryo culture technique, we examined the effects of different pharmacological agents on HR under normoxic (95% oxygen) and hypoxic (20% oxygen) conditions. Oxygen concentrations ≤60% caused a concentration-dependent decrease in HR from normal levels of ~210 bpm. An adenosine agonist, AMP-activated protein kinase (AMPK) activator and KATP channel opener all caused bradycardia in normoxic conditions; however, putative antagonists for these systems failed to prevent or ameliorate hypoxia-induced bradycardia. This suggests that the activation of one or more of these systems is not the primary cause of the observed hypoxia-induced bradycardia. Inhibition of oxidative phosphorylation also decreased HR in normoxic conditions, highlighting the importance of ATP levels. The β-blocker metoprolol caused a concentration-dependent reduction in HR supporting reports that β1-adrenergic receptors are present in the early rat embryonic heart. The cAMP inducer colforsin induced a positive chronotropic effect in both normoxic and hypoxic conditions. Overall, the embryonic HR at this stage of development is responsive to the level of oxygenation, probably as a consequence of its influence on ATP production.
Collapse
Affiliation(s)
- Helen E Ritchie
- Discipline of Biomedical Science, Sydney Medical School, University of Sydney, Lidcombe
| | - Carolina Ragnerstam
- Department of Anatomy and Histology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Elin Gustafsson
- Department of Anatomy and Histology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Johanna M Jonsson
- Department of Anatomy and Histology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - William S Webster
- Department of Anatomy and Histology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
41
|
Cardioprotective effects of lipoic acid, quercetin and resveratrol on oxidative stress related to thyroid hormone alterations in long-term obesity. J Nutr Biochem 2016; 33:36-44. [DOI: 10.1016/j.jnutbio.2016.02.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 02/15/2016] [Accepted: 02/23/2016] [Indexed: 12/20/2022]
|
42
|
Abstract
Classical angina refers to typical substernal discomfort triggered by effort or emotions, relieved with rest or nitroglycerin. The well-accepted pathogenesis is an imbalance between oxygen supply and demand. Goals in therapy are improvement in quality of life by limiting the number and severity of attacks, protection against future lethal events, and measures to lower the burden of risk factors to slow disease progression. New pathophysiological data, drugs, as well as conceptual and technological advances have improved patient care over the past decade. Behavioral changes to improve diets, increase physical activity, and encourage adherence to cardiac rehabilitation programs, are difficult to achieve but are effective.
Collapse
Affiliation(s)
- Richard Kones
- The Cardiometabolic Research Institute, 8181 Fannin Street, Unit 314, Houston, TX 77054, USA.
| | - Umme Rumana
- The Cardiometabolic Research Institute, 8181 Fannin Street, Unit 314, Houston, TX 77054, USA
| |
Collapse
|
43
|
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: 7.8] [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
|
44
|
Shahreyar M, Mupiddi V, Choudhuri I, Sra J, Tajik AJ, Jahangir A. Implantable cardioverter defibrillators in diabetics: efficacy and safety in patients at risk of sudden cardiac death. Expert Rev Cardiovasc Ther 2015; 13:897-906. [PMID: 26098816 DOI: 10.1586/14779072.2015.1059276] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Diabetes mellitus is a major risk factor for arrhythmogenesis and is associated with a two-fold increase in all-cause mortality and a four-fold increase in cardiovascular mortality including sudden cardiac death when compared with nondiabetics. Implantable cardioverter defibrillators (ICD) have been shown to effectively reduce arrhythmic death and all-cause mortality in patients with severe myocardial dysfunction. With a high competing risk of nonarrhythmic cardiac and noncardiac death, survival benefit of ICD in patients with diabetes mellitus could be reduced, but the subanalysis of diabetic patients in randomized clinical trials provides reassurance regarding a similar beneficial survival effect of ICD and cardiac resynchronization therapy in diabetics, as observed in the overall population with advanced heart disease. In this article, the authors highlight some of the clinical issues related to diabetes, summarize the data on the efficacy of ICD in diabetics when compared with nondiabetics and discuss concerns related to ICD implantation in patients with diabetes.
Collapse
Affiliation(s)
- Muhammad Shahreyar
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging (CIRCA), Aurora University of Wisconsin Medical Group, Milwaukee, WI, USA
| | | | | | | | | | | |
Collapse
|
45
|
Ivanov SM, Lagunin AA, Pogodin PV, Filimonov DA, Poroikov VV. Identification of Drug Targets Related to the Induction of Ventricular Tachyarrhythmia Through a Systems Chemical Biology Approach. Toxicol Sci 2015; 145:321-36. [DOI: 10.1093/toxsci/kfv054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
46
|
Uysal F, Ozboyaci E, Bostan O, Saglam H, Semizel E, Cil E. Evaluation of electrocardiographic parameters for early diagnosis of autonomic dysfunction in children and adolescents with type-1 diabetes mellitus. Pediatr Int 2014; 56:675-80. [PMID: 24617770 DOI: 10.1111/ped.12329] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 01/14/2014] [Accepted: 02/04/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND The aim of this study was to identify the sensitivity of electrocardiogram (ECG) in early diagnosis of cardiac autonomic function disorder in children with type 1 diabetes mellitus. METHODS A total of 150 children and adolescents with type 1 diabetes mellitus were enrolled between June 2009 and June 2010, as well as 100 age- and sex-matched healthy control children. Twelve-lead ECG was done in all cases and heart rate, QT and QTc interval, dispersion of P wave (Pd), and of QT (QTd) and QTc interval (QTcd) were measured. The clinical and demographic features such as age, gender, duration of follow up and level of HbA1c and fasting glucose were obtained and the effects of these parameters on ECG measurements were investigated. RESULTS The mean age of the patients and controls was 11.61 ± 3.72 years and 10.92 ± 3.2 years, respectively. QT and QTc interval and QTcd interval were significantly higher in diabetic children compared to healthy controls but these ECG findings were not associated with the duration of diabetes or glycemic state. Pd was significantly higher in the diabetic patients with HbA1c >7.5% compared to control, and this was also found in patients that were followed up >1 year. CONCLUSIONS Cardiac autonomic function disorder, which is one of the most important causes of morbidity and mortality, may emerge in the course of type 1 diabetes mellitus. It can be diagnosed on ECG even when the patients are asymptomatic.
Collapse
Affiliation(s)
- Fahrettin Uysal
- Department of Pediatric Cardiology, Medical Faculty, University of Uludag, Gorukle, Bursa, Turkey
| | | | | | | | | | | |
Collapse
|
47
|
Behn C, Dinamarca GA, De Gregorio NF, Lips V, Vivaldi EA, Soza D, Guerra MA, Jiménez RF, Lecannelier EA, Varela H, Silva-Urra JA. Age-Related Arrhythmogenesis on Ascent and Descent: “Autonomic Conflicts” on Hypoxia/Reoxygenation at High Altitude? High Alt Med Biol 2014; 15:356-63. [DOI: 10.1089/ham.2013.1092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Claus Behn
- Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
- Mutual de Seguridad CChC, Chile
| | | | | | - Viviana Lips
- Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Ennio A Vivaldi
- Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | | | | | - Raúl F Jiménez
- Facultad de Ciencias Físicas y Matemáticas, Universidad de Concepción, Concepción, Chile
| | | | - Héctor Varela
- Facultad de Ciencias Básicas, Universidad de Antofagasta, Antofagasta, Chile
| | - Juan A Silva-Urra
- Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| |
Collapse
|
48
|
Ng FS, Holzem KM, Koppel AC, Janks D, Gordon F, Wit AL, Peters NS, Efimov IR. Adverse remodeling of the electrophysiological response to ischemia-reperfusion in human heart failure is associated with remodeling of metabolic gene expression. Circ Arrhythm Electrophysiol 2014; 7:875-82. [PMID: 25114062 DOI: 10.1161/circep.113.001477] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Ventricular arrhythmias occur more frequently in heart failure during episodes of ischemia-reperfusion although the mechanisms underlying this in humans are unclear. We assessed, in explanted human hearts, the remodeled electrophysiological response to acute ischemia-reperfusion in heart failure and its potential causes, including the remodeling of metabolic gene expression. METHODS AND RESULTS We optically mapped coronary-perfused left ventricular wedge preparations from 6 human end-stage failing hearts (F) and 6 donor hearts rejected for transplantation (D). Preparations were subjected to 30 minutes of global ischemia, followed by 30 minutes of reperfusion. Failing hearts had exaggerated electrophysiological responses to ischemia-reperfusion, with greater action potential duration shortening (P<0.001 at 8-minute ischemia; P=0.001 at 12-minute ischemia) and greater conduction slowing during ischemia, delayed recovery of electric excitability after reperfusion (F, 4.8±1.8 versus D, 1.0±0 minutes; P<0.05), and incomplete restoration of action potential duration and conduction velocity early after reperfusion. Expression of 46 metabolic genes was probed using custom-designed TaqMan arrays, using extracted RNA from 15 failing and 9 donor hearts. Ten genes important in cardiac metabolism were downregulated in heart failure, with SLC27A4 and KCNJ11 significantly downregulated at a false discovery rate of 0%. CONCLUSIONS We demonstrate, for the first time in human hearts, that the electrophysiological response to ischemia-reperfusion in heart failure is accelerated during ischemia with slower recovery after reperfusion. This can enhance spatial conduction and repolarization gradients across the ischemic border and increase arrhythmia susceptibility. This adverse response was associated with downregulation of expression of cardiac metabolic genes.
Collapse
Affiliation(s)
- Fu Siong Ng
- From the Department of Biomedical Engineering, Washington University in Saint Louis, MO (F.S.N., K.M.H., A.C.K., D.J., I.R.E.); National Heart & Lung Institute (F.S.N., N.S.P.) and Statistical Advisory Service (F.G.), Imperial College London, London, United Kingdom; and Department of Pharmacology, Columbia University, New York, NY (A.L.W.)
| | - Katherine M Holzem
- From the Department of Biomedical Engineering, Washington University in Saint Louis, MO (F.S.N., K.M.H., A.C.K., D.J., I.R.E.); National Heart & Lung Institute (F.S.N., N.S.P.) and Statistical Advisory Service (F.G.), Imperial College London, London, United Kingdom; and Department of Pharmacology, Columbia University, New York, NY (A.L.W.)
| | - Aaron C Koppel
- From the Department of Biomedical Engineering, Washington University in Saint Louis, MO (F.S.N., K.M.H., A.C.K., D.J., I.R.E.); National Heart & Lung Institute (F.S.N., N.S.P.) and Statistical Advisory Service (F.G.), Imperial College London, London, United Kingdom; and Department of Pharmacology, Columbia University, New York, NY (A.L.W.)
| | - Deborah Janks
- From the Department of Biomedical Engineering, Washington University in Saint Louis, MO (F.S.N., K.M.H., A.C.K., D.J., I.R.E.); National Heart & Lung Institute (F.S.N., N.S.P.) and Statistical Advisory Service (F.G.), Imperial College London, London, United Kingdom; and Department of Pharmacology, Columbia University, New York, NY (A.L.W.)
| | - Fabiana Gordon
- From the Department of Biomedical Engineering, Washington University in Saint Louis, MO (F.S.N., K.M.H., A.C.K., D.J., I.R.E.); National Heart & Lung Institute (F.S.N., N.S.P.) and Statistical Advisory Service (F.G.), Imperial College London, London, United Kingdom; and Department of Pharmacology, Columbia University, New York, NY (A.L.W.)
| | - Andrew L Wit
- From the Department of Biomedical Engineering, Washington University in Saint Louis, MO (F.S.N., K.M.H., A.C.K., D.J., I.R.E.); National Heart & Lung Institute (F.S.N., N.S.P.) and Statistical Advisory Service (F.G.), Imperial College London, London, United Kingdom; and Department of Pharmacology, Columbia University, New York, NY (A.L.W.)
| | - Nicholas S Peters
- From the Department of Biomedical Engineering, Washington University in Saint Louis, MO (F.S.N., K.M.H., A.C.K., D.J., I.R.E.); National Heart & Lung Institute (F.S.N., N.S.P.) and Statistical Advisory Service (F.G.), Imperial College London, London, United Kingdom; and Department of Pharmacology, Columbia University, New York, NY (A.L.W.)
| | - Igor R Efimov
- From the Department of Biomedical Engineering, Washington University in Saint Louis, MO (F.S.N., K.M.H., A.C.K., D.J., I.R.E.); National Heart & Lung Institute (F.S.N., N.S.P.) and Statistical Advisory Service (F.G.), Imperial College London, London, United Kingdom; and Department of Pharmacology, Columbia University, New York, NY (A.L.W.).
| |
Collapse
|
49
|
Stable ischemic heart disease. Cardiol Clin 2014; 32:333-51. [PMID: 25091962 DOI: 10.1016/j.ccl.2014.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Classical angina refers to typical substernal discomfort triggered by effort or emotions, relieved with rest or nitroglycerin. The well-accepted pathogenesis is an imbalance between oxygen supply and demand. Goals in therapy are improvement in quality of life by limiting the number and severity of attacks, protection against future lethal events, and measures to lower the burden of risk factors to slow disease progression. New pathophysiological data, drugs, as well as conceptual and technological advances have improved patient care over the past decade. Behavioral changes to improve diets, increase physical activity, and encourage adherence to cardiac rehabilitation programs, are difficult to achieve but are effective.
Collapse
|
50
|
Zhang Y, Huang L, Zuo Z, Chen Y, Wang C. Phenanthrene exposure causes cardiac arrhythmia in embryonic zebrafish via perturbing calcium handling. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2013; 142-143:26-32. [PMID: 23948075 DOI: 10.1016/j.aquatox.2013.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/18/2013] [Accepted: 07/19/2013] [Indexed: 06/02/2023]
Abstract
Phenanthrene (Phe) is one of the most abundant and ubiquitous polycyclic aromatic hydrocarbons in the aquatic environment. It is known that Phe has cardiotoxic effects, but knowledge concerning the mechanism of cardiac dysfunction caused by Phe is still limited. In this study, zebrafish embryos were exposed to environmentally relevant concentrations of Phe, and an increase of an irregular rhythm was observed in Phe treated embryos. Disordered calcium (Ca(2+)) handling characterized by impaired sarcoplasmic reticulum Ca(2+) uptake, and obvious Ca(2+) accumulation in the cytoplasm was observed in rat embryonic cardiac myoblasts (H9C2) exposed to Phe. The mRNA level as well as protein expression of the SERCA2a Ca(2+) pump in zebrafish hearts or H9C2 cells was significantly decreased by Phe exposure. The activity of Ca(2+)-ATPase in H9C2 cells was inhibited by Phe. Both the mRNA and protein levels of TBX5, a direct regulator of SERCA2a, were significantly decreased by Phe exposure. These results suggested that exposure to Phe could lead to arrhythmia in zebrafish embryos via perturbing the calcium handling pathway.
Collapse
Affiliation(s)
- Youyu Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | | | | | | | | |
Collapse
|