1
|
Gomaa AA, Zeid AM, Nagy IM. The role of genetic polymorphisms in KCNN2 in cardiovascular complications in patients with renal failure. Gene 2025; 944:149269. [PMID: 39884404 DOI: 10.1016/j.gene.2025.149269] [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: 11/09/2024] [Revised: 12/30/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Patients with end-stage renal disease (ESRD) are at a higher risk of cardiovascular (CV) complications and mortality compared to the general population. This study aimed to investigate the genetic polymorphisms of KCNN2, a key gene encoding a subtype of small-conductance calcium-activated potassium (SK) channels, which regulate an important SK current pathway potentially involved in the development of CV complications, particularly arrhythmias, in ESRD patients. A total of 169 ESRD patients were enrolled in this study. The patients were divided into two groups based on the presence of CV complications: Group I, consisting of 84 patients without CV complications, and Group II, comprising 85 patients with CV complications. Twelve tagging single nucleotide polymorphisms (tSNPs) in KCNN2 were examined. Polymerase chain reaction (PCR) was performed, and genotyping was correlated with CV complications in each group. The TC and CC genotypes of rs10076582, and the GT and TT genotypes of rs11738819 in the KCNN2 gene, were associated with an increased risk of CV complications in ESRD patients. After adjusting for potential risk factors, these associations remained significant. Additionally, KCNN2 haplotypes with the allele combinations GGCCCTCCGAG and AGTCCTCCGGT were significantly associated with a higher risk of CV complications in ESRD patients. In conclusion, our findings report that specific genetic polymorphisms in the KCNN2 gene, particularly the rs10076582 and rs11738819 variants, as well as GGCCCTCCGAG and AGTCCTCCGGT haplotypes, are significantly associated with an increased risk of cardiovascular complications in ESRD patients. These genetic markers may serve as potential biomarkers for identifying individuals at high risk of cardiovascular complications in this vulnerable population.
Collapse
Affiliation(s)
- Azza A Gomaa
- Internal Medicine Department Menofia University Menofia Egypt.
| | - Amany M Zeid
- Clinical Pathology Department Menofia University Menofia Egypt.
| | - Ibrahim M Nagy
- Medicinal Chemistry Department Menofia University Menofia Egypt.
| |
Collapse
|
2
|
Lin LC, Liu ZY, Tu B, Song K, Sun H, Zhou Y, Sha JM, Zhang Y, Yang JJ, Zhao JY, Tao H. Epigenetic signatures in cardiac fibrosis: Focusing on noncoding RNA regulators as the gatekeepers of cardiac fibroblast identity. Int J Biol Macromol 2024; 254:127593. [PMID: 37898244 DOI: 10.1016/j.ijbiomac.2023.127593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/13/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023]
Abstract
Cardiac fibroblasts play a pivotal role in cardiac fibrosis by transformation of fibroblasts into myofibroblasts, which synthesis and secrete a large number of extracellular matrix proteins. Ultimately, this will lead to cardiac wall stiffness and impaired cardiac performance. The epigenetic regulation and fate reprogramming of cardiac fibroblasts has been advanced considerably in recent decades. Non coding RNAs (microRNAs, lncRNAs, circRNAs) regulate the functions and behaviors of cardiac fibroblasts, including proliferation, migration, phenotypic transformation, inflammation, pyroptosis, apoptosis, autophagy, which can provide the basis for novel targeted therapeutic treatments that abrogate activation and inflammation of cardiac fibroblasts, induce different death pathways in cardiac fibroblasts, or make it sensitive to established pathogenic cells targeted cytotoxic agents and biotherapy. This review summarizes our current knowledge in this field of ncRNAs function in epigenetic regulation and fate determination of cardiac fibroblasts as well as the details of signaling pathways contribute to cardiac fibrosis. Moreover, we will comment on the emerging landscape of lncRNAs and circRNAs function in regulating signal transduction pathways, gene translation processes and post-translational regulation of gene expression in cardiac fibroblast. In the end, the prospect of cardiac fibroblasts targeted therapy for cardiac fibrosis based on ncRNAs is discussed.
Collapse
Affiliation(s)
- Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Bin Tu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Kai Song
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - He Sun
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Yang Zhou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Ji-Ming Sha
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Jian-Yuan Zhao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| |
Collapse
|
3
|
Wang X, Chen X, Xu H, Zhou S, Zheng Y, Keller BB, Cai L. Emerging roles of microRNA-208a in cardiology and reverse cardio-oncology. Med Res Rev 2021; 41:2172-2194. [PMID: 33533026 DOI: 10.1002/med.21790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/22/2020] [Accepted: 01/20/2021] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases (CVDs) and cancer, which are the leading causes of mortality globally, have been viewed as two distinct diseases. However, the fact that cancer and CVDs may coincide has been noted by cardiologists when taking care of patients with CVDs caused by cancer chemotherapy; this entity is designated cardio-oncology. More recently, patients with CVDs have also been found to have increased risk of cancers, termed reverse cardio-oncology. Although reverse cardio-oncology has been highlighted as an important disease state in recent studies, how the diseased heart affects cancer and the potential mediators of the crosstalk between CVDs and cancer are largely unknown. Here, we focus on the roles of cardiac-specific microRNA-208a (miR-208a) in cardiac and cancer biology and explore its essential roles in reverse cardio-oncology. Accumulating evidence has shown that within the heart, increased miR-208a promotes myocardial injury, arrhythmia, cardiac remodeling, and dysfunction and that secreted miR-208a in the circulation may have novel roles in promoting tumor proliferation and invasion. This review, therefore, provides insights into the novel roles of miR-208a in reverse cardio-oncology and strategies to prevent secondary carcinogenesis in patients with early- or late-stage heart failure.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Xinxin Chen
- Department of Burn Surgery, First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Hui Xu
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Shanshan Zhou
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Yang Zheng
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Bradley B Keller
- Cincinnati Children's Heart Institute, Greater Louisville and Western Kentucky Practice, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
4
|
Wu N, Li C, Xu B, Xiang Y, Jia X, Yuan Z, Wu L, Zhong L, Li Y. Circular RNA mmu_circ_0005019 inhibits fibrosis of cardiac fibroblasts and reverses electrical remodeling of cardiomyocytes. BMC Cardiovasc Disord 2021; 21:308. [PMID: 34154526 PMCID: PMC8215745 DOI: 10.1186/s12872-021-02128-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Circular RNA (circRNA) have been reported to play important roles in cardiovascular diseases including myocardial infarction and heart failure. However, the role of circRNA in atrial fibrillation (AF) has rarely been investigated. We recently found a circRNA hsa_circ_0099734 was significantly differentially expressed in the AF patients atrial tissues compared to paired control. We aim to investigate the functional role and molecular mechanisms of mmu_circ_0005019 which is the homologous circRNA in mice of hsa_circ_0099734 in AF. METHODS In order to investigate the effect of mmu_circ_0005019 on the proliferation, migration, differentiation into myofibroblasts and expression of collagen of cardiac fibroblasts, and the effect of mmu_circ_0005019 on the apoptosis and expression of Ito, INA and SK3 of cardiomyocytes, gain- and loss-of-function of cell models were established in mice cardiac fibroblasts and HL-1 atrial myocytes. Dual-luciferase reporter assays and RIP were performed to verify the binding effects between mmu_circ_0005019 and its target microRNA (miRNA). RESULTS In cardiac fibroblasts, mmu_circ_0005019 showed inhibitory effects on cell proliferation and migration. In cardiomyocytes, overexpression of mmu_circ_0005019 promoted Kcnd1, Scn5a and Kcnn3 expression. Knockdown of mmu_circ_0005019 inhibited the expression of Kcnd1, Kcnd3, Scn5a and Kcnn3. Mechanistically, mmu_circ_0005019 exerted biological functions by acting as a miR-499-5p sponge to regulate the expression of its target gene Kcnn3. CONCLUSIONS Our findings highlight mmu_circ_0005019 played a protective role in AF development and might serve as an attractive candidate target for AF treatment.
Collapse
Affiliation(s)
- Na Wu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Chengying Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Bin Xu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Ying Xiang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Xiaoyue Jia
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Zhiquan Yuan
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Long Wu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China
| | - Li Zhong
- Cardiovascular Disease Center, Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, People's Republic of China
| | - Yafei Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), NO. 30 Gaotanyan Street, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
5
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia, largely associated to morbidity and mortality. Over the past decades, research in appearance and progression of this arrhythmia have turned into significant advances in its management. However, the incidence of AF continues to increase with the aging of the population and many important fundamental and translational underlaying mechanisms remain elusive. Here, we review recent advances in molecular and cellular basis for AF initiation, maintenance and progression. We first provide an overview of the basic molecular and electrophysiological mechanisms that lead and characterize AF. Next, we discuss the upstream regulatory factors conducting the underlying mechanisms which drive electrical and structural AF-associated remodeling, including genetic factors (risk variants associated to AF as transcriptional regulators and genetic changes associated to AF), neurohormonal regulation (i.e., cAMP) and oxidative stress imbalance (cGMP and mitochondrial dysfunction). Finally, we discuss the potential therapeutic implications of those findings, the knowledge gaps and consider future approaches to improve clinical management.
Collapse
|
6
|
The regulation of the small-conductance calcium-activated potassium current and the mechanisms of sex dimorphism in J wave syndrome. Pflugers Arch 2021; 473:491-506. [PMID: 33411079 DOI: 10.1007/s00424-020-02500-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/20/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022]
Abstract
Apamin-sensitive small-conductance calcium-activated potassium (SK) current (IKAS) plays an important role in cardiac repolarization under a variety of physiological and pathological conditions. The regulation of cardiac IKAS relies on SK channel expression, intracellular Ca2+, and interaction between SK channel and intracellular Ca2+. IKAS activation participates in multiple types of arrhythmias, including atrial fibrillation, ventricular tachyarrhythmias, and automaticity and conduction abnormality. Recently, sex dimorphisms in autonomic control have been noticed in IKAS activation, resulting in sex-differentiated action potential morphology and arrhythmogenesis. This review provides an update on the Ca2+-dependent regulation of cardiac IKAS and the role of IKAS on arrhythmias, with a special focus on sex differences in IKAS activation. We propose that sex dimorphism in autonomic control of IKAS may play a role in J wave syndrome.
Collapse
|
7
|
Rahm AK, Wieder T, Gramlich D, Müller ME, Wunsch MN, El Tahry FA, Heimberger T, Weis T, Most P, Katus HA, Thomas D, Lugenbiel P. HDAC2-dependent remodeling of K Ca2.2 (KCNN2) and K Ca2.3 (KCNN3) K + channels in atrial fibrillation with concomitant heart failure. Life Sci 2020; 266:118892. [PMID: 33310041 DOI: 10.1016/j.lfs.2020.118892] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
AIMS Atrial fibrillation (AF) with concomitant heart failure (HF) is associated with prolonged atrial refractoriness. Small-conductance, calcium-activated K+ (KCa, KCNN) channels promote action potential (AP) repolarization. KCNN2 and KCNN3 variants are associated with AF risk. In addition, histone deacetylase (HDAC)-related epigenetic mechanisms have been implicated in AP regulation. We hypothesized that HDAC2-dependent remodeling of KCNN2 and KCNN3 expression contributes to atrial arrhythmogenesis in AF complicated by HF. The objectives were to assess HDAC2 and KCNN2/3 transcript levels in AF/HF patients and in a pig model, and to investigate cellular epigenetic effects of HDAC2 inactivation on KCNN expression. MATERIALS AND METHODS HDAC2 and KCNN2/3 transcript levels were quantified in patients with AF and HF, and in a porcine model of atrial tachypacing-induced AF and reduced left ventricular function. Tachypacing and anti-Hdac2 siRNA treatment were employed in HL-1 atrial myocytes to study effects on KCNN2/3 mRNA and KCa protein abundance. KEY FINDINGS Atrial KCNN2 and KCNN3 expression was reduced in AF/HF patients and in a corresponding pig model. HDAC2 displayed significant downregulation in humans and a tendency towards reduced expression in right atrial tissue of pigs. Tachypacing recapitulated downregulation of Kcnn2/KCa2.2, Kcnn3/KCa2.3 and Hdac2/HDAC2, indicating that high atrial rates trigger epigenetic remodeling mechanisms. Finally, knock-down of Hdac2 in vitro reduced Kcnn3/KCa2.3 expression. SIGNIFICANCE KCNN2/3 and HDAC2 expression is suppressed in AF complicated by HF. Hdac2 directly regulates Kcnn3 mRNA levels in atrial cells. The mechanistic and therapeutic significance of epigenetic electrophysiological effects in AF requires further validation.
Collapse
Affiliation(s)
- Ann-Kathrin Rahm
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Teresa Wieder
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Dominik Gramlich
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Mara Elena Müller
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Maximilian N Wunsch
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Fadwa A El Tahry
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Tanja Heimberger
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Tanja Weis
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Patrick Most
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | - Patrick Lugenbiel
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
8
|
Brown BM, Shim H, Christophersen P, Wulff H. Pharmacology of Small- and Intermediate-Conductance Calcium-Activated Potassium Channels. Annu Rev Pharmacol Toxicol 2019; 60:219-240. [PMID: 31337271 DOI: 10.1146/annurev-pharmtox-010919-023420] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The three small-conductance calcium-activated potassium (KCa2) channels and the related intermediate-conductance KCa3.1 channel are voltage-independent K+ channels that mediate calcium-induced membrane hyperpolarization. When intracellular calcium increases in the channel vicinity, it calcifies the flexible N lobe of the channel-bound calmodulin, which then swings over to the S4-S5 linker and opens the channel. KCa2 and KCa3.1 channels are highly druggable and offer multiple binding sites for venom peptides and small-molecule blockers as well as for positive- and negative-gating modulators. In this review, we briefly summarize the physiological role of KCa channels and then discuss the pharmacophores and the mechanism of action of the most commonly used peptidic and small-molecule KCa2 and KCa3.1 modulators. Finally, we describe the progress that has been made in advancing KCa3.1 blockers and KCa2.2 negative- and positive-gating modulators toward the clinic for neurological and cardiovascular diseases and discuss the remaining challenges.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Heesung Shim
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | | | - Heike Wulff
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| |
Collapse
|
9
|
Seifert MB, Olesen MS, Christophersen IE, Nielsen JB, Carlson J, Holmqvist F, Tveit A, Haunsø S, Svendsen JH, Platonov PG. Genetic variants on chromosomes 7p31 and 12p12 are associated with abnormal atrial electrical activation in patients with early-onset lone atrial fibrillation. Ann Noninvasive Electrocardiol 2019; 24:e12661. [PMID: 31152482 DOI: 10.1111/anec.12661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/04/2019] [Accepted: 05/05/2019] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Abnormal P-wave morphology (PWM) has been associated with a history of atrial fibrillation (AF) in earlier studies. Although lone AF is believed to have substantial genetic basis, studies on associations between single nucleotide polymorphisms (SNP) linked to lone AF and PWM have not been reported. We aimed to assess whether SNPs previously associated with lone AF (rs2200733, rs13376333, rs3807989, and rs11047543) are also linked to P-wave abnormalities. METHODS Four SNPs were studied in 176 unrelated individuals with early-onset lone AF (age at onset <50 years), median age 38 years (19-63 years), 149 men. Using sinus rhythm ECG, orthogonal PWM was classified as Type 1-positive in leads X and Y and negative in lead Z, Type 2-positive in leads X and Y and biphasic (-/+) in lead Z, Type 3-positive in lead X and biphasic in lead Y (+/-), and the remaining as atypical. RESULTS Two SNPs were found to be significantly associated with altered P-wave morphology distribution: rs3807989 near the gene CAV1/CAV2 and rs11047543 near the gene SOX5. Both SNPs were associated with a higher risk of non-Type 1 P-wave morphology (rs3807989: OR = 4.8, 95% CI = 2.3-10.2, p < 0.001; rs11047543: OR = 4.7, 95% CI = 1.1-20.5, p = 0.04). No association was observed for rs2200733 and rs13376333. CONCLUSION In this study, the two variants rs3807989 and rs11047543, previously associated with PR interval and lone AF, were associated with altered P-wave morphology distribution in patients with early-onset lone AF. These findings suggest that common genetic variants may modify atrial conduction properties.
Collapse
Affiliation(s)
- Mariam B Seifert
- The Center for Integrative Electrocardiology, Arrhythmia Clinic Skåne University Hospital, Lund University (CIEL), Lund, Sweden.,Department of Cardiology, Frederiksberg Hospital, Copenhagen, Denmark
| | - Morten S Olesen
- Danish National Research Foundation Center for Cardiac Arrhythmia, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ingrid E Christophersen
- The Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.,Department of Medical Research, Baerum Hospital, Vestre Viken Hospital Trust, Rud, Norway
| | - Jonas B Nielsen
- Danish National Research Foundation Center for Cardiac Arrhythmia, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Carlson
- The Center for Integrative Electrocardiology, Arrhythmia Clinic Skåne University Hospital, Lund University (CIEL), Lund, Sweden
| | - Fredrik Holmqvist
- The Center for Integrative Electrocardiology, Arrhythmia Clinic Skåne University Hospital, Lund University (CIEL), Lund, Sweden
| | - Arnljot Tveit
- Department of Medical Research, Baerum Hospital, Vestre Viken Hospital Trust, Rud, Norway
| | - Stig Haunsø
- Danish National Research Foundation Center for Cardiac Arrhythmia, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper H Svendsen
- Danish National Research Foundation Center for Cardiac Arrhythmia, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pyotr G Platonov
- The Center for Integrative Electrocardiology, Arrhythmia Clinic Skåne University Hospital, Lund University (CIEL), Lund, Sweden
| |
Collapse
|
10
|
Vagos M, van Herck IGM, Sundnes J, Arevalo HJ, Edwards AG, Koivumäki JT. Computational Modeling of Electrophysiology and Pharmacotherapy of Atrial Fibrillation: Recent Advances and Future Challenges. Front Physiol 2018; 9:1221. [PMID: 30233399 PMCID: PMC6131668 DOI: 10.3389/fphys.2018.01221] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022] Open
Abstract
The pathophysiology of atrial fibrillation (AF) is broad, with components related to the unique and diverse cellular electrophysiology of atrial myocytes, structural complexity, and heterogeneity of atrial tissue, and pronounced disease-associated remodeling of both cells and tissue. A major challenge for rational design of AF therapy, particularly pharmacotherapy, is integrating these multiscale characteristics to identify approaches that are both efficacious and independent of ventricular contraindications. Computational modeling has long been touted as a basis for achieving such integration in a rapid, economical, and scalable manner. However, computational pipelines for AF-specific drug screening are in their infancy, and while the field is progressing quite rapidly, major challenges remain before computational approaches can fill the role of workhorse in rational design of AF pharmacotherapies. In this review, we briefly detail the unique aspects of AF pathophysiology that determine requirements for compounds targeting AF rhythm control, with emphasis on delimiting mechanisms that promote AF triggers from those providing substrate or supporting reentry. We then describe modeling approaches that have been used to assess the outcomes of drugs acting on established AF targets, as well as on novel promising targets including the ultra-rapidly activating delayed rectifier potassium current, the acetylcholine-activated potassium current and the small conductance calcium-activated potassium channel. Finally, we describe how heterogeneity and variability are being incorporated into AF-specific models, and how these approaches are yielding novel insights into the basic physiology of disease, as well as aiding identification of the important molecular players in the complex AF etiology.
Collapse
Affiliation(s)
- Márcia Vagos
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Ilsbeth G. M. van Herck
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Joakim Sundnes
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Hermenegild J. Arevalo
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Andrew G. Edwards
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Jussi T. Koivumäki
- BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
11
|
Value of multilocus genetic risk score for atrial fibrillation in end-stage kidney disease patients in a Polish population. Sci Rep 2018; 8:9284. [PMID: 29915175 PMCID: PMC6006310 DOI: 10.1038/s41598-018-27382-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 05/25/2018] [Indexed: 12/11/2022] Open
Abstract
Genetic factors play a key role in the pathogenesis of atrial fibrillation (AF). We would like to establish an association between previously described single-nucleotide polymorphisms (SNPs) and AF in haemodialysed patients with end-stage kidney disease (ESKD-HD) as well as to assess the cumulative effect of all genotyped SNPs on AF risk. Sixteen SNPs were genotyped in 113 patients with AF-ESKD-HD and in 157 controls: without AF (NAF) and with ESKD-HD. The distribution of the risk alleles was compared in both groups and between different sub-phenotypes. The multilocus genetic risk score (GRS) was calculated to estimate the cumulative risk conferred by all SNPs. Several loci showed a trend toward an association with permanent AF (perm-AF): CAV1, Cx40 and PITX2. However, GRS was significantly higher in the AF and perm-AF groups, as compared to NAF. Three of the tested variables were independently associated with AF: male sex, history of myocardial infarction (MI) and GRS. The GRS, which combined 13 previously described SNPs, showed a significant and independent association with AF in a Polish population of patients with ESKD-HD and concomitant AF. Further studies on larger groups of patients are needed to confirm the associations.
Collapse
|
12
|
Abstract
Although the mechanism of sudden cardiac death (SCD) in heart failure is not completely known, genetic variations are known to play key roles in this process. Increasing numbers of mutations and variants are being discovered through genome-wide association studies. The genetic variations involved in the mechanisms of SCD have aroused widespread concern. Comprehensive understanding of the genetic variations involved in SCD may help prevent it. To this end, we briefly reviewed the genetic variations involved in SCD and their associations and interactions, and observed that cardiac ion channels are the core molecules involved in this process. Genetic variations involved in cardiac structure, cardiogenesis and development, cell division and differentiation, and DNA replication and transcription are all speculated to be loci involved in SCD. Additionally, the systems involved in neurohumoral regulation as well as substance and energy metabolism are also potentially responsible for susceptibility to SCD. They form an elaborate network and mutually interact with each other to govern the fate of SCD-susceptible individuals.
Collapse
|
13
|
Yu CC, Chia-Ti T, Chen PL, Wu CK, Chiu FC, Chiang FT, Chen PS, Chen CL, Lin LY, Juang JM, Ho LT, Lai LP, Yang WS, Lin JL. KCNN2 polymorphisms and cardiac tachyarrhythmias. Medicine (Baltimore) 2016; 95:e4312. [PMID: 27442679 PMCID: PMC5265796 DOI: 10.1097/md.0000000000004312] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Potassium calcium-activated channel subfamily N member 2 (KCNN2) encodes an integral membrane protein that forms small-conductance calcium-activated potassium (SK) channels. Recent studies in animal models show that SK channels are important in atrial and ventricular repolarization and arrhythmogenesis. However, the importance of SK channels in human arrhythmia remains unclear. The purpose of the present study was to test the association between genetic polymorphism of the SK2 channel and the occurrence of cardiac tachyarrhythmias in humans. We enrolled 327 Han Chinese, including 72 with clinically significant ventricular tachyarrhythmias (VTa) who had a history of aborted sudden cardiac death (SCD) or unexplained syncope, 98 with a history of atrial fibrillation (AF), and 144 normal controls. We genotyped 12 representative tag single nucleotide polymorphisms (SNPs) across a 141-kb genetic region containing the KCNN2 gene; these captured the full haplotype information. The rs13184658 and rs10076582 variants of KCNN2 were associated with VTa in both the additive and dominant models (odds ratio [OR] 2.89, 95% confidence interval [CI] = 1.505-5.545, P = 0.001; and OR 2.55, 95% CI = 1.428-4.566, P = 0.002, respectively). After adjustment for potential risk factors, the association remained significant. The population attributable risks of these 2 variants of VTa were 17.3% and 10.6%, respectively. One variant (rs13184658) showed weak but significant association with AF in a dominant model (OR 1.91, CI = 1.025-3.570], P = 0.042). There was a significant association between the KCNN2 variants and clinically significant VTa. These findings suggest an association between KCNN2 and VTa; it also appears that KCNN2 variants may be adjunctive markers for risk stratification in patients susceptible to SCD.
Collapse
Affiliation(s)
- Chih-Chieh Yu
- Department of Internal Medicine, National Taiwan University Hospital
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University
| | - Tsai Chia-Ti
- Department of Internal Medicine, National Taiwan University Hospital
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University
| | - Pei-Lung Chen
- Department of Internal Medicine, National Taiwan University Hospital
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University
- Department of Medical Genetics, National Taiwan University Hospital
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei
| | - Cho-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital
| | - Fu-Chun Chiu
- Department of Internal Medicine, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin, Taiwan
| | - Fu-Tien Chiang
- Department of Internal Medicine, National Taiwan University Hospital
| | - Peng-Sheng Chen
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chi-Ling Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University
- Graduate Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Lian-Yu Lin
- Department of Internal Medicine, National Taiwan University Hospital
| | - Jyh-Ming Juang
- Department of Internal Medicine, National Taiwan University Hospital
| | - Li-Ting Ho
- Department of Internal Medicine, National Taiwan University Hospital
| | - Ling-Ping Lai
- Department of Internal Medicine, National Taiwan University Hospital
| | - Wei-Shiung Yang
- Department of Internal Medicine, National Taiwan University Hospital
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei
- Correspondence: Jiunn-Lee Lin, Wei-Shiung Yang, Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung San South Road, Taipei City 100, Taiwan (R.O.C.) (e-mail: , )
| | - Jiunn-Lee Lin
- Department of Internal Medicine, National Taiwan University Hospital
- Correspondence: Jiunn-Lee Lin, Wei-Shiung Yang, Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung San South Road, Taipei City 100, Taiwan (R.O.C.) (e-mail: , )
| |
Collapse
|
14
|
Abstract
INTRODUCTION SK channels have functional importance in the cardiac atrium of many species, including humans. Pharmacological blockage of SK channels has been reported to be antiarrhythmic in animal models of atrial fibrillation; however, the exact antiarrhythmic mechanism of SK channel inhibition remains unclear. OBJECTIVES We speculated that together with a direct inhibition of repolarizing SK current, the previously observed depolarization of the atrial resting membrane potential (RMP) after SK channel inhibition reduces sodium channel availability, thereby prolonging the effective refractory period and slowing the conduction velocity (CV). We therefore aimed at elucidating these properties of SK channel inhibition and the underlying antiarrhythmic mechanisms using microelectrode action potential (AP) recordings and CV measurements in isolated rat atrium. Automated patch clamping and two-electrode voltage clamp were used to access INa and IK,ACh, respectively. RESULTS The SK channel inhibitor N-(pyridin-2-yl)-4-(pyridin-2-yl)thiazol-2-amine (ICA) exhibited antiarrhythmic effects. ICA prevented electrically induced runs of atrial fibrillation in the isolated right atrium and induced atrial postrepolarization refractoriness and depolarized RMP. Moreover, ICA (1-10 μM) was found to slow CV; however, because of a marked prolongation of effective refractory period, the calculated wavelength was increased. Furthermore, at increased pacing frequencies, SK channel inhibition by ICA (10-30 μM) demonstrated prominent depression of other sodium channel-dependent parameters. ICA did not inhibit IK,ACh, but at concentrations above 10 μM, ICA use dependently inhibited INa. CONCLUSIONS SK channel inhibition modulates multiple parameters of AP. It prolongs the AP duration and shifts the RMP towards more depolarized potentials through direct ISK block. This indirectly leads to sodium channel inhibition through accumulation of state dependently inactivated channels, which ultimately slows conduction and decreases excitability. However, a contribution from a direct sodium channel inhibition cannot be ruled. We here propose that the primary antiarrhythmic mechanism of SK channel inhibition is through direct potassium channel block and through indirect sodium channel inhibition.
Collapse
|
15
|
Jabbari J, Olesen MS, Yuan L, Nielsen JB, Liang B, Macri V, Christophersen IE, Nielsen N, Sajadieh A, Ellinor PT, Grunnet M, Haunsø S, Holst AG, Svendsen JH, Jespersen T. Common and rare variants in SCN10A modulate the risk of atrial fibrillation. ACTA ACUST UNITED AC 2015; 8:64-73. [PMID: 25691686 DOI: 10.1161/hcg.0000000000000022] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Genome-wide association studies have shown that the common single nucleotide polymorphism rs6800541 located in SCN10A, encoding the voltage-gated Nav1.8 sodium channel, is associated with PR-interval prolongation and atrial fibrillation (AF). Single nucleotide polymorphism rs6800541 is in high linkage disequilibrium with the nonsynonymous variant in SCN10A, rs6795970 (V1073A, r(2)=0.933). We therefore sought to determine whether common and rare SCN10A variants are associated with early onset AF. METHODS AND RESULTS SCN10A was sequenced in 225 AF patients in whom there was no evidence of other cardiovascular disease or dysfunction (lone AF). In an association study of the rs6795970 single nucleotide polymorphism variant, we included 515 AF patients and 2 control cohorts of 730 individuals free of AF and 6161 randomly sampled individuals. Functional characterization of SCN10A variants was performed by whole-cell patch-clamping. In the lone AF cohort, 9 rare missense variants and 1 splice site donor variant were detected. Interestingly, AF patients were found to have higher G allele frequency of rs6795970, which encodes the alanine variant at position 1073 (described from here on as A1073, odds ratio =1.35 [1.16-1.54]; P=2.3×10(-5)). Both of the common variants, A1073 and P1092, induced a gain-of-channel function, whereas the rare missense variants, V94G and R1588Q, resulted in a loss-of-channel function. CONCLUSIONS The common variant A1073 is associated with increased susceptibility to AF. Both rare and common variants have effect on the function of the channel, indicating that these variants influence susceptibility to AF. Hence, our study suggests that SCN10A variations are involved in the genesis of AF.
Collapse
Affiliation(s)
- Javad Jabbari
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Morten S Olesen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Lei Yuan
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Jonas B Nielsen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Bo Liang
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Vincenzo Macri
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Ingrid E Christophersen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Nikolaj Nielsen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Ahmad Sajadieh
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Patrick T Ellinor
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Morten Grunnet
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Stig Haunsø
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Anders G Holst
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Jesper H Svendsen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.)
| | - Thomas Jespersen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA (M.V., P.T.E) and Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA (P.T.E); Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway (I.E.C.); Department of Cardiology, Copenhagen University Hospital of Bispebjerg, Bispebjerg, Denmark (A.S.); and LuCamp, The Lundbeck Foundation Centre for Applied Medical Genomics in Personalized Disease Prediction, Prevention and Care, Copenhagen, Denmark (S.H.).
| |
Collapse
|
16
|
Gundlund A, Christiansen MN, Hansen ML, Olesen JB, Zahir D, Køber L, Gislason GH, Piccini JP, Peterson ED, Torp-Pedersen C, Fosbøl EL. Familial clustering and subsequent incidence of atrial fibrillation among first-degree relatives in Denmark. Europace 2015; 18:658-64. [DOI: 10.1093/europace/euv274] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 07/17/2015] [Indexed: 11/14/2022] Open
|
17
|
|
18
|
Yao JL, Zhou YF, Yang XJ, Qian XD, Jiang WP. KCNN3 SNP rs13376333 on Chromosome 1q21 Confers Increased Risk of Atrial Fibrillation. Int Heart J 2015; 56:511-5. [PMID: 26370375 DOI: 10.1536/ihj.15-133] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To investigate the relationship between KCNN3 SNP (single-nucleotide polymorphism) rs13376333 and risk of atrial fibrillation (AF) and to provide evidence for prevention and treatment for AF.The PubMed, Embase, OVID, Cochrane library, CNKI, and Wan Fang databases were searched to identify studies on the relationship between KCNN3 SNP rs13376333 polymorphism and atrial fibrillation. Two authors performed independent article reviews and study quality assessment using the Newcastle-Ottawa Scale (NOS) checklist.Seven studies involving 24,339 individuals were included in the meta-analysis. The overall combined OR of rs13376333 polymorphism was observed for both lone AF (OR: 1.58 [95%CI: 1.37 to 1.82]; P < 0.001; I(2) = 47.0%) and total AF (OR: 1.33 [95%CI: 1.14 to 1.54]; P < 0.001; I(2) = 0). Further, when stratified by ethnicity, control sources, sample sizes, and genotyping method, similar results were observed in both subgroups. Sensitivity analysis revealed that the source of control was the source of the heterogeneity for lone AF. Omission of any single study had little effect on the combined risk estimate. No evidence of publication bias was found.This meta-analysis suggests that KCNN3 SNP rs13376333 polymorphism significantly increases the risk of lone AF and total AF, which suggests the rs13376333 polymorphism of the KCNN3 gene may play an important role in the pathogenesis of AF.
Collapse
Affiliation(s)
- Jia-Lu Yao
- Department of Cardiology, the First Affiliated Hospital of Soochow University
| | | | | | | | | |
Collapse
|
19
|
Zhang XD, Lieu DK, Chiamvimonvat N. Small-conductance Ca2+ -activated K+ channels and cardiac arrhythmias. Heart Rhythm 2015; 12:1845-51. [PMID: 25956967 PMCID: PMC4662728 DOI: 10.1016/j.hrthm.2015.04.046] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Indexed: 01/04/2023]
Abstract
Small-conductance Ca2+ -activated K+ (SK, KCa2) channels are unique in that they are gated solely by changes in intracellular Ca2+ and, hence, function to integrate intracellular Ca2+ and membrane potentials on a beat-to-beat basis. Recent studies have provided evidence for the existence and functional significance of SK channels in the heart. Indeed, our knowledge of cardiac SK channels has been greatly expanded over the past decade. Interests in cardiac SK channels are further driven by recent studies suggesting the critical roles of SK channels in human atrial fibrillation, the SK channel as a possible novel therapeutic target in atrial arrhythmias, and upregulation of SK channels in heart failure in animal models and in human heart failure. However, there remain critical gaps in our knowledge. Specifically, blockade of SK channels in cardiac arrhythmias has been shown to be both antiarrhythmic and proarrhythmic. This contemporary review provides an overview of the literature on the role of cardiac SK channels in cardiac arrhythmias and serves as a discussion platform for the current clinical perspectives. At the translational level, development of SK channel blockers as a new therapeutic strategy in the treatment of atrial fibrillation and the possible proarrhythmic effects merit further considerations and investigations.
Collapse
Affiliation(s)
- Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California.
| | - Deborah K Lieu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, California; Department of Veterans Affairs, Northern California Health Care System, Mather, California.
| |
Collapse
|
20
|
Luo Z, Yan C, Zhang W, Shen X, Zheng W, Chen F, Cao X, Yang Y, Lin X, Wang Z, Huang M. Association between SNP rs13376333 and rs1131820 in the KCNN3 gene and atrial fibrillation in the Chinese Han population. Clin Chem Lab Med 2015; 52:1867-73. [PMID: 24978901 DOI: 10.1515/cclm-2014-0491] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/10/2014] [Indexed: 11/15/2022]
Abstract
BACKGROUND The small conductance calcium-activated potassium, subfamily N, member 3 (KCNN3) gene rs13376333 and rs1131820 have been shown to be strongly associated with lone atrial fibrillation (AF), while replication association studies between rs13376333 in KCNN3 gene and risk of AF showed conflicting results. The current study tried to validate the impact of SNP rs13376333 and rs1131820 of KCNN3 gene on the risk of AF in the Chinese Han population. METHODS A total of 889 AF patients and 1015 controls were enrolled. Two hundred and seventy-eight cases of AF were lone AF. KCNN3 gene SNP rs13376333 and rs1131820 were genotyped by allele-specific MALDI-TOF mass spectrometry. RESULTS The genotype distribution and allele frequency of rs13376333 polymorphism were not different between total AF patients and controls. However, the genotype distribution of rs13376333 polymorphism was significantly different between lone AF and control group (p<0.001); and T allele frequency was significantly higher in lone AF group than that in controls (7.6% vs 3.6%, p<0.001). Multivariable logistic regression analysis showed that T allele carriers of rs13376333 was significantly associated with lone AF (OR=2.31, 95% CI 1.41-3.78, p=0.001). No relationship between rs1131820 polymorphism and total AF or lone AF was found in this study. CONCLUSIONS KCNN3 rs13376333 polymorphism was associated with lone AF in the Chinese Han population and the T allele carriers may be an independent predictive factor for lone AF.
Collapse
|
21
|
Steffensen AB, Refsgaard L, Andersen MN, Vallet C, Mujezinovic A, Haunsø S, Svendsen JH, Olesen SP, Olesen MS, Schmitt N. IKs Gain- and Loss-of-Function in Early-Onset Lone Atrial Fibrillation. J Cardiovasc Electrophysiol 2015; 26:715-23. [PMID: 25786344 DOI: 10.1111/jce.12666] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/09/2015] [Accepted: 03/13/2015] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Atrial fibrillation (AF) is the most frequent cardiac arrhythmia. The potassium current IKs is essential for cardiac repolarization. Gain-of-function mutation in KCNQ1, the gene encoding the pore-forming α-subunit of the IKs channel (KV 7.1), was the first ion channel dysfunction to be associated with familial AF. We hypothesized that early-onset lone AF is associated with a high prevalence of mutations in KCNQ1. METHODS AND RESULTS We bidirectionally sequenced the entire coding sequence of KCNQ1 in 209 unrelated patients with early-onset lone AF (<40 years) and investigated the identified mutations functionally in a heterologous expression system. We found 4 nonsynonymous KCNQ1 mutations (A46T, R195W, A302V, and R670K) in 4 unrelated patients (38, 31, 39, and 36 years, respectively). None of the mutations were present in the control group (n = 416 alleles). No other mutations were found in genes previously associated with AF. The mutations A46T, R195W, and A302V have previously been associated with long-QT syndrome. In line with previous reports, we found A302V to display a pronounced loss-of-function of the IKs current, while the other mutants exhibited a gain-of-function phenotype. CONCLUSIONS Mutations in the IKs channel leading to gain-of-function have previously been described in familial AF, yet this is the first time a loss-of-function mutation in KCNQ1 is associated with early-onset lone AF. These findings suggest that both gain-of-function and loss-of-function of cardiac potassium currents enhance the susceptibility to AF.
Collapse
Affiliation(s)
- Annette Buur Steffensen
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lena Refsgaard
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Laboratory for Molecular Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Nybo Andersen
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cecilia Vallet
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amer Mujezinovic
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stig Haunsø
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Laboratory for Molecular Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Hastrup Svendsen
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Laboratory for Molecular Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren-Peter Olesen
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Salling Olesen
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Laboratory for Molecular Cardiology, The Heart Centre, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicole Schmitt
- Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Poudel P, Xu Y, Cui Z, Sharma D, Tian B, Paudel S. Atrial fibrillation: recent advances in understanding the role of microRNAs in atrial remodeling with an electrophysiological overview. Cardiology 2015; 131:58-67. [PMID: 25871909 DOI: 10.1159/000375403] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 01/17/2015] [Indexed: 01/08/2023]
Abstract
Atrial fibrillation (AF) is a highly prevalent condition associated with pronounced cardiovascular-related morbidity, mortality and socioeconomic burden. It accounts for more hospitalization days than does any other arrhythmia. This article reviews the basic electrophysiology of AF, electrical and structural remodeling in AF and recent advances in understanding the molecular mechanisms of AF in relation to specific microRNAs. This paper also reviews the potential role of microRNAs as novel therapeutic targets as well as biomarkers in the management of AF. AF shows characteristics typical of altered electrophysiology that promote ectopic activity and facilitate reentry, thereby contributing to the progression from short paroxysmal AF to a persistent, permanent form via atrial remodeling, even in the absence of progressive underlying heart disease. MicroRNAs have been suggested to influence the development of AF by regulating gene expression at the post-transcriptional level. Increasing evidence has identified various microRNA modifications and their impacts on AF initiation and maintenance through electrical and structural remodeling. The discovery of specific microRNAs as novel therapeutic targets and some experimental evidence implicating microRNAs as potential molecular diagnostic markers have had a significant impact on the diagnosis and management of AF and demand further research.
Collapse
Affiliation(s)
- Pradeep Poudel
- International College of Tianjin Medical University, Tianjin, PR China
| | | | | | | | | | | |
Collapse
|
23
|
Huang Y, Li J. MicroRNA208 family in cardiovascular diseases: therapeutic implication and potential biomarker. J Physiol Biochem 2015; 71:479-86. [DOI: 10.1007/s13105-015-0409-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/26/2015] [Indexed: 01/05/2023]
|
24
|
Jabbari J, Olesen MS, Yuan L, Nielsen JB, Liang B, Macri V, Christophersen IE, Nielsen N, Sajadieh A, Ellinor PT, Grunnet M, Haunsø S, Holst AG, Svendsen JH, Jespersen T. Common and Rare Variants in
SCN10A
Modulate the Risk of Atrial Fibrillation. ACTA ACUST UNITED AC 2015. [DOI: 10.1161/circgenetics.113.000442] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background—
Genome-wide association studies have shown that the common single nucleotide polymorphism rs6800541 located in
SCN10A
, encoding the voltage-gated Na
v
1.8 sodium channel, is associated with PR-interval prolongation and atrial fibrillation (AF). Single nucleotide polymorphism rs6800541 is in high linkage disequilibrium with the nonsynonymous variant in
SCN10A
, rs6795970 (V1073A,
r
2
=0.933). We therefore sought to determine whether common and rare
SCN10A
variants are associated with early onset AF.
Methods and Results—
SCN10A
was sequenced in 225 AF patients in whom there was no evidence of other cardiovascular disease or dysfunction (lone AF). In an association study of the rs6795970 single nucleotide polymorphism variant, we included 515 AF patients and 2 control cohorts of 730 individuals free of AF and 6161 randomly sampled individuals. Functional characterization of
SCN10A
variants was performed by whole-cell patch-clamping. In the lone AF cohort, 9 rare missense variants and 1 splice site donor variant were detected. Interestingly, AF patients were found to have higher G allele frequency of rs6795970, which encodes the alanine variant at position 1073 (described from here on as A1073, odds ratio =1.35 [1.16−1.54];
P
=2.3×10
−5
). Both of the common variants, A1073 and P1092, induced a gain-of-channel function, whereas the rare missense variants, V94G and R1588Q, resulted in a loss-of-channel function.
Conclusions—
The common variant A1073 is associated with increased susceptibility to AF. Both rare and common variants have effect on the function of the channel, indicating that these variants influence susceptibility to AF. Hence, our study suggests that
SCN10A
variations are involved in the genesis of AF.
Collapse
Affiliation(s)
- Javad Jabbari
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Morten S. Olesen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Lei Yuan
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Jonas B. Nielsen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Bo Liang
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Vincenzo Macri
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Ingrid E. Christophersen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Nikolaj Nielsen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Ahmad Sajadieh
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Patrick T. Ellinor
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Morten Grunnet
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Stig Haunsø
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Anders G. Holst
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Jesper H. Svendsen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | - Thomas Jespersen
- From the The Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences (J.J., M.S.O., L.Y., J.B.N., B.L., N.N., M.G., S.H., A.G.H., J.H.S., T.J.), Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet (J.J., M.S.O., J.B.N., S.H., A.G.H., J.H.S.), and Department of Clinical Medicine (S.H., J.H.S.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Cardiovascular Research Center,
| | | |
Collapse
|
25
|
Schmitt N, Grunnet M, Olesen SP. Cardiac potassium channel subtypes: new roles in repolarization and arrhythmia. Physiol Rev 2014; 94:609-53. [PMID: 24692356 DOI: 10.1152/physrev.00022.2013] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
About 10 distinct potassium channels in the heart are involved in shaping the action potential. Some of the K+ channels are primarily responsible for early repolarization, whereas others drive late repolarization and still others are open throughout the cardiac cycle. Three main K+ channels drive the late repolarization of the ventricle with some redundancy, and in atria this repolarization reserve is supplemented by the fairly atrial-specific KV1.5, Kir3, KCa, and K2P channels. The role of the latter two subtypes in atria is currently being clarified, and several findings indicate that they could constitute targets for new pharmacological treatment of atrial fibrillation. The interplay between the different K+ channel subtypes in both atria and ventricle is dynamic, and a significant up- and downregulation occurs in disease states such as atrial fibrillation or heart failure. The underlying posttranscriptional and posttranslational remodeling of the individual K+ channels changes their activity and significance relative to each other, and they must be viewed together to understand their role in keeping a stable heart rhythm, also under menacing conditions like attacks of reentry arrhythmia.
Collapse
|
26
|
Single-nucleotide variations in cardiac arrhythmias: prospects for genomics and proteomics based biomarker discovery and diagnostics. Genes (Basel) 2014; 5:254-69. [PMID: 24705329 PMCID: PMC4094932 DOI: 10.3390/genes5020254] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/19/2014] [Accepted: 02/19/2014] [Indexed: 02/08/2023] Open
Abstract
Cardiovascular diseases are a large contributor to causes of early death in developed countries. Some of these conditions, such as sudden cardiac death and atrial fibrillation, stem from arrhythmias—a spectrum of conditions with abnormal electrical activity in the heart. Genome-wide association studies can identify single nucleotide variations (SNVs) that may predispose individuals to developing acquired forms of arrhythmias. Through manual curation of published genome-wide association studies, we have collected a comprehensive list of 75 SNVs associated with cardiac arrhythmias. Ten of the SNVs result in amino acid changes and can be used in proteomic-based detection methods. In an effort to identify additional non-synonymous mutations that affect the proteome, we analyzed the post-translational modification S-nitrosylation, which is known to affect cardiac arrhythmias. We identified loss of seven known S-nitrosylation sites due to non-synonymous single nucleotide variations (nsSNVs). For predicted nitrosylation sites we found 1429 proteins where the sites are modified due to nsSNV. Analysis of the predicted S-nitrosylation dataset for over- or under-representation (compared to the complete human proteome) of pathways and functional elements shows significant statistical over-representation of the blood coagulation pathway. Gene Ontology (GO) analysis displays statistically over-represented terms related to muscle contraction, receptor activity, motor activity, cystoskeleton components, and microtubule activity. Through the genomic and proteomic context of SNVs and S-nitrosylation sites presented in this study, researchers can look for variation that can predispose individuals to cardiac arrhythmias. Such attempts to elucidate mechanisms of arrhythmia thereby add yet another useful parameter in predicting susceptibility for cardiac diseases.
Collapse
|
27
|
Andreasen L, Nielsen JB, Darkner S, Christophersen IE, Jabbari J, Refsgaard L, Thiis JJ, Sajadieh A, Tveit A, Haunsø S, Svendsen JH, Schmitt N, Olesen MS. Brugada syndrome risk loci seem protective against atrial fibrillation. Eur J Hum Genet 2014; 22:1357-61. [PMID: 24667784 DOI: 10.1038/ejhg.2014.46] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 12/12/2013] [Accepted: 12/26/2013] [Indexed: 12/19/2022] Open
Abstract
Several studies have shown an overlap between genes involved in the pathophysiological mechanisms of atrial fibrillation (AF) and Brugada Syndrome (BrS). We investigated whether three single-nucleotide polymorphisms (SNPs) (rs11708996; G>C located intronic to SCN5A, rs10428132; T>G located in SCN10A, and rs9388451; T>C located downstream to HEY2) at loci associated with BrS in a recent genome-wide association study (GWAS) also were associated with AF. A total of 657 patients diagnosed with AF and a control group comprising 741 individuals free of AF were included. The three SNPs were genotyped using TaqMan assays. The frequencies of risk alleles in the AF population and the control population were compared in two-by-two models. One variant, rs10428132 at SCN10A, was associated with a statistically significant decreased risk of AF (odds ratio (OR)=0.77, P=0.001). A meta-analysis was performed by enriching the control population with allele frequencies from controls in the recently published BrS GWAS (2230 alleles). In this meta-analysis, both rs10428132 at SCN10A (OR=0.73, P=5.7 × 10(-6)) and rs11708996 at SCN5A (OR=0.80, P=0.02) showed a statistically significant decreased risk of AF. When assessing the additive effect of the three loci, we found that the risk of AF decreased in a dose-responsive manner with increasing numbers of risk alleles (OR=0.50, P=0.001 for individuals carrying ≥4 risk alleles vs ≤1 allele). In conclusion, the prevalence of three risk alleles previously associated with BrS was lower in AF patients than in patients free of AF, suggesting a protective role of these loci in developing AF.
Collapse
Affiliation(s)
- Laura Andreasen
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark [3] The Ion Channel Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas B Nielsen
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Stine Darkner
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ingrid E Christophersen
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark [3] Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway
| | - Javad Jabbari
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lena Refsgaard
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Thiis
- Departments of Cardiothoracic Surgery and Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ahmad Sajadieh
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Arnljot Tveit
- Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Rud, Norway
| | - Stig Haunsø
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark [3] Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jesper H Svendsen
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark [3] Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nicole Schmitt
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] The Ion Channel Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten S Olesen
- 1] Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark [2] Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Voudris KV, Apostolakis S, Karyofillis P, Doukas K, Zaravinos A, Androutsopoulos VP, Michalis A, Voudris V, Spandidos DA. Genetic diversity of the KCNE1 gene and susceptibility to postoperative atrial fibrillation. Am Heart J 2014; 167:274-280.e1. [PMID: 24439990 DOI: 10.1016/j.ahj.2013.09.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 09/30/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND The human KCNE1 protein forms the β-subunit of the IKs potassium channel and is important in the regulation of the atrial action potential duration. The purpose of this study was to investigate the association between the nonsynonymous 112G>A mutation of the KCNE1 gene and postcardiac surgery atrial fibrillation (AF). METHODS AND RESULTS A cohort of patients scheduled for cardiac surgery was prospectively recruited. The genotype of 112G>A polymorphism was determined using polymerase chain reaction/restriction fragment analysis and confirmed with direct sequencing of the polymerase chain reaction product. In total, 509 patients were recruited in the study, of whom 203 (39.9%) had at least 1 qualifying episode of postoperative AF. An increased frequency of the G allele was observed in the postoperative AF group compared with the group without postoperative AF (0.628 vs 0.552, respectively, P = .016). The individual's relative risk of postoperative AF increased as the number of G alleles increased from 1.36 (95% CI 0.89-2.08) for G allele heterozygotes to 1.62 (95% CI 1.08-2.43) for G allele homozygotes (P = .04 for trend). The multivariate analysis revealed the abnormal ejection fraction (odds ratio [OR] 1.585, 95% CI 1.076-2.331, P = .020), age (OR 1.043, 95% CI 1.022-1.064, P < .001), type of surgery (aortic valve replacement) (OR 1.869, 95% CI 1.094-3.194, P = .022), and the 112G>A genotype (OR 1.401 [in additive model], 95% CI 1.052-1.865, P = .021) to be independent predictors of postoperative AF. CONCLUSION This study confirmed the association of the 112G>A polymorphism and postoperative AF in a cohort of patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Konstantinos V Voudris
- Department of Clinical Virology Faculty of medicine, University of Crete, Heraklion, Crete, Greece
| | - Stavros Apostolakis
- Thrombosis Haemostasis and Vascular Biology Unit, University of Birmingham, Birmingham, United Kingdom; Cardiology Department, Democritus University of Thrace, Alexandroupolis, Greece
| | | | | | - Apostolos Zaravinos
- Department of Clinical Virology Faculty of medicine, University of Crete, Heraklion, Crete, Greece
| | | | - Alkis Michalis
- Department of Cardiac Surgery, Onassis Cardiac Surgery Center, Athens, Greece
| | - Vassilis Voudris
- Department of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Demetrios A Spandidos
- Department of Clinical Virology Faculty of medicine, University of Crete, Heraklion, Crete, Greece.
| |
Collapse
|
29
|
Andreasen L, Nielsen JB, Christophersen IE, Holst AG, Sajadieh A, Tveit A, Haunsø S, Svendsen JH, Schmitt N, Olesen MS. Genetic modifier of the QTc interval associated with early-onset atrial fibrillation. Can J Cardiol 2013; 29:1234-40. [PMID: 24074973 DOI: 10.1016/j.cjca.2013.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 06/14/2013] [Accepted: 06/14/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Both shortening and prolongation of the QTc interval have been associated with atrial fibrillation (AF). We investigated whether 8 single nucleotide polymorphisms (SNPs) at loci previously shown to affect QTc interval duration were associated with lone AF. METHODS We included 358 patients diagnosed with lone AF (defined as onset of AF at < 50 years of age in the absence of traditional cardiovascular risk factors) and a control group consisting of 751 individuals free of AF. The 8 loci were genotyped using TaqMan assays. Genotype frequencies in lone AF cases and controls were compared using an additive logistic regression model. RESULTS Risk of the development of early-onset lone AF in individuals homozygous for the variant rs2968863 (7q36.1) was higher than in individuals with no copies of the risk allele (odds ratio [OR], 2.40; P = 0.001). The association was also significant after Bonferroni correction (P = 0.016). This polymorphism has been shown to decrease the QTc interval by 1.4 ms in genome-wide association studies (GWAS). The genetic variant is situated close to the long QT syndrome (LQTS) type 2 gene KCNH2 that encodes the potassium channel Kv11.1 (hERG). Sanger sequencing of KCNH2 confirmed the known high linkage disequilibrium between rs2968863 and the nonsynonymous variant K897T in KCNH2. No novel mutations were found in the gene. CONCLUSIONS The variant rs2968863 (7q36.1), reported in GWAS to shorten the QTc interval, was found to be associated with early-onset lone AF. This may have implications for the pathophysiological understanding of AF.
Collapse
Affiliation(s)
- Laura Andreasen
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark; Laboratory for Molecular Cardiology, Rigshospitalet, Copenhagen, Denmark; The Ion Channel Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Shi KH, Tao H, Yang JJ, Wu JX, Xu SS, Zhan HY. Role of microRNAs in atrial fibrillation: New insights and perspectives. Cell Signal 2013; 25:2079-84. [DOI: 10.1016/j.cellsig.2013.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/08/2013] [Accepted: 06/14/2013] [Indexed: 01/15/2023]
|
31
|
Ling TY, Wang XL, Chai Q, Lau TW, Koestler CM, Park SJ, Daly RC, Greason KL, Jen J, Wu LQ, Shen WF, Shen WK, Cha YM, Lee HC. Regulation of the SK3 channel by microRNA-499--potential role in atrial fibrillation. Heart Rhythm 2013; 10:1001-9. [PMID: 23499625 PMCID: PMC3710704 DOI: 10.1016/j.hrthm.2013.03.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Indexed: 01/27/2023]
Abstract
BACKGROUND MicroRNAs are important regulators of gene expression, including those involving electrical remodeling in atrial fibrillation (AF). Recently, KCNN3, the gene that encodes the small-conductance calcium-activated potassium channel 3 (SK3), was found to be strongly associated with AF. OBJECTIVES To evaluate the changes in atrial myocardial microRNAs in patients with permanent AF and to determine the role of microRNA on the regulation of cardiac SK3 expression. METHODS Atrial tissue obtained during cardiac surgery from patients (4 sinus rhythm and 4 permanent AF) was analyzed by using microRNA arrays. Potential targets of microRNAs were predicted by using software programs. The effects of specific microRNAs on target gene expression were evaluated in HL-1 cells from a continuously proliferating mouse hyperplastic atrial cardiomyocyte cell line. Interactions between microRNAs and targets were further evaluated by using luciferase reporter assay and by using Argonaute pull-down assay. RESULTS Twenty-one microRNAs showed significant (>2-fold) changes in AF. MicroRNA 499 (miR-499) was upregulated by 2.33-fold (P < .01) in AF atria, whereas SK3 protein expression was downregulated by 46% (P < .05). Transfection of miR-499 mimic in HL-1 cells resulted in the downregulation of SK3 protein expression, while that of miR-499 inhibitor upregulated SK3 expression. Binding of miR-499 to the 3' untranslated region of KCNN3 was confirmed by luciferase reporter assay and by the increased presence of SK3 mRNA in Argonaute pulled-down microRNA-induced silencing complexes after transfection with miR-499. CONCLUSION Atrial miR-499 is significantly upregulated in AF, leading to SK3 downregulation and possibly contributing to the electrical remodeling in AF.
Collapse
Affiliation(s)
- Tian-You Ling
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Internal Medicine, Mayo Clinic Rochester, Minnesota
| | - Xiao-Li Wang
- Department of Internal Medicine, Mayo Clinic Rochester, Minnesota
| | - Qiang Chai
- Department of Internal Medicine, Mayo Clinic Rochester, Minnesota
- The Department of Physiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Tin-Wah Lau
- Department of Internal Medicine, Mayo Clinic Rochester, Minnesota
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Soon J. Park
- Department of Surgery, Mayo Clinic Rochester, Minnesota
| | | | | | - Jin Jen
- Advanced Genomics Technology Center, Mayo Clinic Rochester, Minnesota
| | - Li-Qun Wu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Feng Shen
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Win-Kuang Shen
- Department of Internal Medicine, Mayo Clinic Scottsdale, Arizona
| | - Yong-Mei Cha
- Department of Internal Medicine, Mayo Clinic Rochester, Minnesota
| | - Hon-Chi Lee
- Department of Internal Medicine, Mayo Clinic Rochester, Minnesota
| |
Collapse
|
32
|
Hsueh CH, Chang PC, Hsieh YC, Reher T, Chen PS, Lin SF. Proarrhythmic effect of blocking the small conductance calcium activated potassium channel in isolated canine left atrium. Heart Rhythm 2013; 10:891-8. [PMID: 23376397 PMCID: PMC3663880 DOI: 10.1016/j.hrthm.2013.01.033] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Small conductance calcium activated potassium (SKCa) channels are voltage insensitive and are activated by intracellular calcium. Genome-wide association studies revealed that a variant of SKCa is associated with lone atrial fibrillation in humans. Roles of SKCa in atrial arrhythmias remain unclear. OBJECTIVE To determine roles of SKCa in atrial arrhythmias. METHODS Optical mapping using the isolated canine left atrium was performed. The optical action potential duration (APD) and induction of arrhythmia were evaluated before and after the addition of specific SKCa blockers-apamin or UCL-1684. RESULTS SKCa blockade significantly increased APD₈₀ (188 ± 19 ms vs 147 ± 11 ms; P<.001). The pacing cycle length thresholds to induce 2:2 alternans, and wave breaks were prolonged by SKCa blockade. Increased APD heterogeneity was observed after the SKCa blockade, as measured by the difference between the maximum and the minimum APD (39 ± 4 ms vs 26 ± 5 ms; P<.05), by standard deviation (12.43 ± 2.36 ms vs 7.49 ± 1.47 ms; P<.001), or by coefficient of variation (6.68% ± 0.97% vs 4.90% ± 0.84%; P<.05). No arrhythmia was induced at baseline by an S1-S2 protocol. After SKCa blockade, 4 of 6 atria developed arrhythmia. CONCLUSIONS SKCa blockade promotes arrhythmia and prolongs the pacing cycle length threshold of 2:2 alternans and wave breaks in the canine left atrium. The proarrhythmic effect could be attributed to increased APD heterogeneity in the canine left atrium. This study provides supportive evidence of genome-wide association studies showing association of KCNN3 and lone atrial fibrillation.
Collapse
Affiliation(s)
- Chia-Hsiang Hsueh
- Krannert Institute of Cardiology and Division of Cardiology, Indiana University, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | |
Collapse
|
33
|
Olesen MS, Yuan L, Liang B, Holst AG, Nielsen N, Nielsen JB, Hedley PL, Christiansen M, Olesen SP, Haunsø S, Schmitt N, Jespersen T, Svendsen JH. High prevalence of long QT syndrome-associated SCN5A variants in patients with early-onset lone atrial fibrillation. ACTA ACUST UNITED AC 2012; 5:450-9. [PMID: 22685113 DOI: 10.1161/circgenetics.111.962597] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common cardiac arrhythmia. The cardiac sodium channel, Na(V)1.5, plays a pivotal role in setting the conduction velocity and the initial depolarization of the cardiac myocytes. We hypothesized that early-onset lone AF was associated with genetic variation in SCN5A. METHODS AND RESULTS The coding sequence of SCN5A was sequenced in 192 patients with early-onset lone AF. Eight nonsynonymous mutations (T220I, R340Q, T1304M, F1596I, R1626H, D1819N, R1897W, and V1951M) and 2 rare variants (S216L in 2 patients and F2004L) were identified. Of 11 genopositive probands, 6 (3.2% of the total population) had a variant previously associated with long QT syndrome type 3 (LQTS3). The prevalence of LQTS3-associated variants in the patients with lone AF was much higher than expected, compared with the prevalence in recent exome data (minor allele frequency, 1.6% versus 0.3%; P=0.003), mainly representing the general population. The functional effects of the mutations were analyzed by whole cell patch clamp in HEK293 cells; for 5 of the mutations previously associated with LQTS3, patch-clamp experiments showed an increased sustained sodium current, suggesting a mechanistic overlap between LQTS3 and early-onset lone AF. In 9 of 10 identified mutations and rare variants, we observed compromised biophysical properties affecting the transient peak current. CONCLUSIONS In a cohort of patients with early-onset lone AF, we identified a high prevalence of SCN5A mutations previously associated with LQTS3. Functional investigations of the mutations revealed both compromised transient peak current and increased sustained current.
Collapse
Affiliation(s)
- Morten S Olesen
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Olesen MS, Bentzen BH, Nielsen JB, Steffensen AB, David JP, Jabbari J, Jensen HK, Haunsø S, Svendsen JH, Schmitt N. Mutations in the potassium channel subunit KCNE1 are associated with early-onset familial atrial fibrillation. BMC MEDICAL GENETICS 2012; 13:24. [PMID: 22471742 PMCID: PMC3359244 DOI: 10.1186/1471-2350-13-24] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 04/03/2012] [Indexed: 11/10/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common arrhythmia. The potassium current IKs is essential for cardiac repolarization. Gain-of-function mutations in KV7.1, the pore-forming α-subunit of the IKs channel, have been associated with AF. We hypothesized that early-onset lone AF is associated with mutations in the IKs channel regulatory subunit KCNE1. METHODS In 209 unrelated early-onset lone AF patients (< 40 years) the entire coding sequence of KCNE1 was bidirectionally sequenced. We analyzed the identified KCNE1 mutants electrophysiologically in heterologous expression systems. RESULTS Two non-synonymous mutations G25V and G60D were found in KCNE1 that were not present in the control group (n = 432 alleles) and that have not previously been reported in any publicly available databases or in the exom variant server holding exom data from more than 10.000 alleles. Proband 1 (female, age 45, G25V) had onset of paroxysmal AF at the age of 39 years. Proband 2 (G60D) was diagnosed with lone AF at the age of 33 years. The patient has inherited the mutation from his mother, who also has AF. Both probands had no mutations in genes previously associated with AF. In heterologous expression systems, both mutants showed significant gain-of-function for IKs both with respect to steady-state current levels, kinetic parameters, and heart rate-dependent modulation. CONCLUSIONS Mutations in KV7.1 leading to gain-of-function of IKs current have previously been described in lone AF, yet this is the first time a mutation in the beta-subunit KCNE1 is associated with the disease. This finding further supports the hypothesis that increased potassium current enhances AF susceptibility.
Collapse
Affiliation(s)
- Morten S Olesen
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Olesen MS, Holst AG, Jabbari J, Nielsen JB, Christophersen IE, Sajadieh A, Haunsø S, Svendsen JH. Genetic loci on chromosomes 4q25, 7p31, and 12p12 are associated with onset of lone atrial fibrillation before the age of 40 years. Can J Cardiol 2012; 28:191-5. [PMID: 22336519 DOI: 10.1016/j.cjca.2011.11.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 11/25/2011] [Accepted: 11/25/2011] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Three distinct genetic loci on chromosomes 1q21, 4q25, and 16q22 have been associated with atrial fibrillation (AF) in genome-wide association studies (GWAS). Five additional loci have been associated primarily with the PR interval and subsequently with AF. We aimed to investigate if 8 single nucleotide polymorphisms (SNPs), representing the 8 genomic loci previously linked with AF in genome-wide association studies, were associated with early-onset lone AF. METHODS We included 209 patients with early-onset lone AF, and a control group consisting of 534 individuals free of AF. The 8 SNPs were genotyped using TaqMan assays (Applied Biosystems, Foster City, CA). RESULTS Three SNPs were found to be significantly associated with early-onset lone AF: rs2200733 closest to PITX2 (odds ratio [OR], 1.62; 95% confidence interval [CI], 1.16-2.27; P = 0.004), rs3807989 near to CAV1 (OR 1.35; 95% CI, 1.06-1.72; P = 0.015), and rs11047543 near to SOX5 (OR 1.70; 95% CI, 1.18-2.44; P = 0.004). When correcting for multiple testing, rs2200733 and rs11047543 were still significantly associated with AF. CONCLUSIONS Three SNPs, rs2200733 (4q25), rs3807989 (7p31), and rs11047543 (12p12), were associated with early-onset lone AF. All 3 SNPs are positioned close to genes that in previous studies have been demonstrated to be important for cardiac morphology/development, thereby suggesting a link between these SNPs and structural heart disease. Our results however, indicate that variants in these 3 loci are associated with AF through mechanisms that do not involve major structural abnormalities in the heart.
Collapse
Affiliation(s)
- Morten S Olesen
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Barc J, Koopmann TT. Genome-wide association studies: providers of candidate genes for identification of rare variants? Europace 2011; 13:911-2. [DOI: 10.1093/europace/eur050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
37
|
Olesen MS, Jensen NF, Holst AG, Nielsen JB, Tfelt-Hansen J, Jespersen T, Sajadieh A, Haunsø S, Lund JT, Calloe K, Schmitt N, Svendsen JH. A novel nonsense variant in Nav1.5 cofactor MOG1 eliminates its sodium current increasing effect and may increase the risk of arrhythmias. Can J Cardiol 2011; 27:523.e17-23. [PMID: 21621375 DOI: 10.1016/j.cjca.2011.01.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Accepted: 12/10/2010] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The protein MOG1 is a cofactor of the cardiac sodium channel, Nav1.5. Overexpression of MOG1 in Nav1.5-expressing cells increases sodium current markedly. Mutations in the genes encoding Nav1.5 and its accessory proteins have been associated with cardiac arrhythmias of significant clinical impact. We sought to investigate whether MOG1 is implicated in cardiac arrhythmias. METHODS We performed a genetic screening of the MOG1-encoding gene (gene symbol RANGRF, alias MOG1) in 220 Danish patients with cardiac arrhythmia. Of the 220, 197 were young patients with lone atrial fibrillation and 23 were patients with Brugada syndrome. The effect of one variant was investigated functionally by patch-clamping CHO-K1 cells coexpressing Nav1.5 with MOG1. RESULTS We uncovered a novel heterozygous nonsense variant, c.181G>T (p.E61X), that, however, was also present in control subjects, albeit at a lower frequency (1.8% vs 0.4%, P = 0.078). Electrophysiological investigation showed that the p.E61X variant completely eliminates the sodium current-increasing effect of MOG1 and thereby causes loss of function in the sodium current. When mimicking heterozygosity by coexpression of Nav1.5 with wild-type MOG1 and p.E61X-MOG1, no current decrease was seen. CONCLUSIONS Our screening of Nav1.5 cofactor MOG1 uncovered a novel nonsense variant that appeared to be present at a higher frequency among patients than control subjects. This variant causes MOG1 loss of function and therefore might be disease causing or modifying under certain conditions.
Collapse
Affiliation(s)
- Morten S Olesen
- Department of Cardiology, Laboratory for Molecular Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen Ø, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|