1
|
Neelsen LC, Riel EB, Rinné S, Schmid FR, Jürs BC, Bedoya M, Langer JP, Eymsh B, Kiper AK, Cordeiro S, Decher N, Baukrowitz T, Schewe M. Ion occupancy of the selectivity filter controls opening of a cytoplasmic gate in the K 2P channel TALK-2. Nat Commun 2024; 15:7545. [PMID: 39215031 PMCID: PMC11364775 DOI: 10.1038/s41467-024-51812-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Two-pore domain K+ (K2P) channel activity was previously thought to be controlled primarily via a selectivity filter (SF) gate. However, recent crystal structures of TASK-1 and TASK-2 revealed a lower gate at the cytoplasmic pore entrance. Here, we report functional evidence of such a lower gate in the K2P channel K2P17.1 (TALK-2, TASK-4). We identified compounds (drugs and lipids) and mutations that opened the lower gate allowing the fast modification of pore cysteine residues. Surprisingly, stimuli that directly target the SF gate (i.e., pHe., Rb+ permeation, membrane depolarization) also opened the cytoplasmic gate. Reciprocally, opening of the lower gate reduced the electric work to open the SF via voltage driven ion binding. Therefore, it appears that the SF is so rigidly locked into the TALK-2 protein structure that changes in ion occupancy can pry open a distant lower gate and, vice versa, opening of the lower gate concurrently promote SF gate opening. This concept might extent to other K+ channels that contain two gates (e.g., voltage-gated K+ channels) for which such a positive gate coupling has been suggested, but so far not directly demonstrated.
Collapse
Affiliation(s)
- Lea C Neelsen
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Elena B Riel
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | - Susanne Rinné
- Institute of Physiology and Pathophysiology, Philipps-University of Marburg, Marburg, Germany
| | | | - Björn C Jürs
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
- MSH Medical School Hamburg, University of Applied Sciences and Medical University, Hamburg, Germany
| | - Mauricio Bedoya
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
- Laboratorio de Bioinformática y Química Computacional (LBQC), Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Jan P Langer
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Bisher Eymsh
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Aytug K Kiper
- Institute of Physiology and Pathophysiology, Philipps-University of Marburg, Marburg, Germany
| | - Sönke Cordeiro
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Philipps-University of Marburg, Marburg, Germany.
| | - Thomas Baukrowitz
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany.
| | - Marcus Schewe
- Institute of Physiology, Christian-Albrechts University of Kiel, Kiel, Germany.
| |
Collapse
|
2
|
Müller ME, Petersenn F, Hackbarth J, Pfeiffer J, Gampp H, Frey N, Lugenbiel P, Thomas D, Rahm AK. Electrophysiological Effects of the Sodium-Glucose Co-Transporter-2 (SGLT2) Inhibitor Dapagliflozin on Human Cardiac Potassium Channels. Int J Mol Sci 2024; 25:5701. [PMID: 38891889 PMCID: PMC11172209 DOI: 10.3390/ijms25115701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
The sodium-glucose co-transporter-2 (SGLT2) inhibitor dapagliflozin is increasingly used in the treatment of diabetes and heart failure. Dapagliflozin has been associated with reduced incidence of atrial fibrillation (AF) in clinical trials. We hypothesized that the favorable antiarrhythmic outcome of dapagliflozin use may be caused in part by previously unrecognized effects on atrial repolarizing potassium (K+) channels. This study was designed to assess direct pharmacological effects of dapagliflozin on cloned ion channels Kv11.1, Kv1.5, Kv4.3, Kir2.1, K2P2.1, K2P3.1, and K2P17.1, contributing to IKur, Ito, IKr, IK1, and IK2P K+ currents. Human channels coded by KCNH2, KCNA5, KCND3, KCNJ2, KCNK2, KCNK3, and KCNK17 were heterologously expressed in Xenopus laevis oocytes, and currents were recorded using the voltage clamp technique. Dapagliflozin (100 µM) reduced Kv11.1 and Kv1.5 currents, whereas Kir2.1, K2P2.1, and K2P17.1 currents were enhanced. The drug did not significantly affect peak current amplitudes of Kv4.3 or K2P3.1 K+ channels. Biophysical characterization did not reveal significant effects of dapagliflozin on current-voltage relationships of study channels. In conclusion, dapagliflozin exhibits direct functional interactions with human atrial K+ channels underlying IKur, IKr, IK1, and IK2P currents. Substantial activation of K2P2.1 and K2P17.1 currents could contribute to the beneficial antiarrhythmic outcome associated with the drug. Indirect or chronic effects remain to be investigated in vivo.
Collapse
Affiliation(s)
- Mara Elena Müller
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (M.E.M.); (P.L.); (D.T.)
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Finn Petersenn
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (M.E.M.); (P.L.); (D.T.)
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Juline Hackbarth
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (M.E.M.); (P.L.); (D.T.)
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Julia Pfeiffer
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (M.E.M.); (P.L.); (D.T.)
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Heike Gampp
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (M.E.M.); (P.L.); (D.T.)
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (M.E.M.); (P.L.); (D.T.)
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Patrick Lugenbiel
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (M.E.M.); (P.L.); (D.T.)
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (M.E.M.); (P.L.); (D.T.)
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Ann-Kathrin Rahm
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (M.E.M.); (P.L.); (D.T.)
- HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
3
|
Geng Y, Li P, Butler A, Wang B, Salkoff L, Magleby KL. BK channels of five different subunit combinations underlie the de novo KCNMA1 G375R channelopathy. J Gen Physiol 2023; 155:e202213302. [PMID: 36995317 PMCID: PMC10067970 DOI: 10.1085/jgp.202213302] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/08/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
The molecular basis of a severe developmental and neurological disorder associated with a de novo G375R variant of the tetrameric BK channel is unknown. Here, we address this question by recording from single BK channels expressed to mimic a G375R mutation heterozygous with a WT allele. Five different types of functional BK channels were expressed: 3% were consistent with WT, 12% with homotetrameric mutant, and 85% with three different types of hybrid (heterotetrameric) channels assembled from both mutant and WT subunits. All channel types except WT showed a marked gain-of-function in voltage activation and a smaller decrease-of-function in single-channel conductance, with both changes in function becoming more pronounced as the number of mutant subunits per tetrameric channel increased. The net cellular response from the five different types of channels comprising the molecular phenotype was a shift of -120 mV in the voltage required to activate half of the maximal current through BK channels, giving a net gain-of-function. The WT and homotetrameric mutant channels in the molecular phenotype were consistent with genetic codominance as each displayed properties of a channel arising from only one of the two alleles. The three types of hybrid channels in the molecular phenotype were consistent with partial dominance as their properties were intermediate between those of mutant and WT channels. A model in which BK channels randomly assemble from mutant and WT subunits, with each subunit contributing increments of activation and conductance, approximated the molecular phenotype of the heterozygous G375R mutation.
Collapse
Affiliation(s)
- Yanyan Geng
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Ping Li
- Department of Neuroscience, Washington University St. Louis, St. Louis, MO, USA
| | - Alice Butler
- Department of Neuroscience, Washington University St. Louis, St. Louis, MO, USA
| | - Bill Wang
- Department of Neuroscience, Washington University St. Louis, St. Louis, MO, USA
| | - Lawrence Salkoff
- Department of Neuroscience, Washington University St. Louis, St. Louis, MO, USA
- Department of Genetics, Washington University St. Louis, St. Louis, MO, USA
| | - Karl L. Magleby
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
4
|
Meng W, Reel PS, Nangia C, Rajendrakumar AL, Hebert HL, Guo Q, Adams MJ, Zheng H, Lu ZH, Ray D, Colvin LA, Palmer CNA, McIntosh AM, Smith BH. A Meta-Analysis of the Genome-Wide Association Studies on Two Genetically Correlated Phenotypes Suggests Four New Risk Loci for Headaches. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:64-76. [PMID: 36939796 PMCID: PMC9883337 DOI: 10.1007/s43657-022-00078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/19/2022]
Abstract
Headache is one of the commonest complaints that doctors need to address in clinical settings. The genetic mechanisms of different types of headache are not well understood while it has been suggested that self-reported headache and self-reported migraine were genetically correlated. In this study, we performed a meta-analysis of genome-wide association studies (GWAS) on the self-reported headache phenotype from the UK Biobank and the self-reported migraine phenotype from the 23andMe using the Unified Score-based Association Test (metaUSAT) software for genetically correlated phenotypes (N = 397,385). We identified 38 loci for headaches, of which 34 loci have been reported before and four loci were newly suggested. The LDL receptor related protein 1 (LRP1)-Signal Transducer and Activator of Transcription 6 (STAT6)-S hort chain D ehydrogenase/R eductase family 9C member 7 (SDR9C7) region in chromosome 12 was the most significantly associated locus with a leading p value of 1.24 × 10-62 of rs11172113. The One Cut homeobox 2 (ONECUT2) gene locus in chromosome 18 was the strongest signal among the four new loci with a p value of 1.29 × 10-9 of rs673939. Our study demonstrated that the genetically correlated phenotypes of self-reported headache and self-reported migraine can be meta-analysed together in theory and in practice to boost study power to identify more variants for headaches. This study has paved way for a large GWAS meta-analysis involving cohorts of different while genetically correlated headache phenotypes. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-022-00078-7.
Collapse
Affiliation(s)
- Weihua Meng
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100 China
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF UK
| | - Parminder S. Reel
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF UK
| | - Charvi Nangia
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF UK
| | - Aravind Lathika Rajendrakumar
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF UK
| | - Harry L. Hebert
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF UK
| | - Qian Guo
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100 China
| | - Mark J. Adams
- Division of Psychiatry, Edinburgh Medical School, University of Edinburgh, Edinburgh, EH10 5HF UK
| | - Hua Zheng
- Department of Anaesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Zen Haut Lu
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, BE1410 Brunei Darussalam
| | | | - Debashree Ray
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205 USA
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Lesley A. Colvin
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF UK
| | - Colin N. A. Palmer
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF UK
| | - Andrew M. McIntosh
- Division of Psychiatry, Edinburgh Medical School, University of Edinburgh, Edinburgh, EH10 5HF UK
| | - Blair H. Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF UK
| |
Collapse
|
5
|
Khoubza L, Gilbert N, Kim EJ, Chatelain FC, Feliciangeli S, Abelanet S, Kang D, Lesage F, Bichet D. Alkaline-sensitive two-pore domain potassium channels form functional heteromers in pancreatic β-cells. J Biol Chem 2022; 298:102447. [PMID: 36063992 PMCID: PMC9520024 DOI: 10.1016/j.jbc.2022.102447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/15/2022] Open
Abstract
Two-pore domain K+ channels (K2P channels), active as dimers, produce inhibitory currents regulated by a variety of stimuli. Among them, TWIK1-related alkalinization-activated K+ channel 1 (TALK1), TWIK1-related alkalinization-activated K+ channel 2 (TALK2), and TWIK1-related acid-sensitive K+ channel 2 (TASK2) form a subfamily of structurally related K2P channels stimulated by extracellular alkalosis. The human genes encoding these proteins are clustered at chromosomal region 6p21 and coexpressed in multiple tissues, including the pancreas. The question whether these channels form functional heteromers remained open. By analyzing single-cell transcriptomic data, we show that these channels are coexpressed in insulin-secreting pancreatic β-cells. Using in situ proximity ligation assay and electrophysiology, we show that they form functional heterodimers both upon heterologous expression and under native conditions in human pancreatic β-cells. We demonstrate that heteromerization of TALK2 with TALK1 or with TASK2 endows TALK2 with sensitivity to extracellular alkalosis in the physiological range. We further show that the association of TASK2 with TALK1 and TALK2 increases their unitary conductance. These results provide a new example of heteromerization in the K2P channel family expanding the range of the potential physiological and pathophysiological roles of TALK1/TALK2/TASK2 channels, not only in insulin-secreting cells but also in the many other tissues in which they are coexpressed.
Collapse
Affiliation(s)
- Lamyaa Khoubza
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| | - Nicolas Gilbert
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| | - Eun-Jin Kim
- Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Franck C Chatelain
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| | - Sylvain Feliciangeli
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France; Inserm, Paris, France
| | - Sophie Abelanet
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| | - Dawon Kang
- Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Florian Lesage
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France; Inserm, Paris, France.
| | - Delphine Bichet
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| |
Collapse
|
6
|
Linet MS, Dores GM, Savage SA. Uncovering the Genetic Etiology of the (Posttherapy) Broken Heart. J Natl Cancer Inst 2022; 114:1054-1056. [PMID: 35698269 PMCID: PMC9360454 DOI: 10.1093/jnci/djac116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- Martha S Linet
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and
Genetics, Department of Health and Human Services, National Cancer Institute, National
Institutes of Health, Rockville, MD, USA
| | - Graça M Dores
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and
Genetics, Department of Health and Human Services, National Cancer Institute, National
Institutes of Health, Rockville, MD, USA
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics,
Department of Health and Human Services, National Cancer Institute, National
Institutes of Health, Rockville, MD, USA
| |
Collapse
|
7
|
Sapkota Y, Ehrhardt MJ, Qin N, Wang Z, Liu Q, Qiu W, Shelton K, Shao Y, Plyler E, Mulder HL, Easton J, Michael JR, Burridge PW, Wang X, Wilson CL, Jefferies JL, Chow EJ, Oeffinger KC, Morton LM, Li C, Yang JJ, Zhang J, Bhatia S, Mulrooney DA, Hudson MM, Robison LL, Armstrong GT, Yasui Y. A Novel Locus on 6p21.2 for Cancer Treatment-Induced Cardiac Dysfunction Among Childhood Cancer Survivors. J Natl Cancer Inst 2022; 114:1109-1116. [PMID: 35698272 PMCID: PMC9360468 DOI: 10.1093/jnci/djac115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/31/2022] [Accepted: 06/01/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Adult survivors of childhood cancer are at increased risk of cardiac late effects. METHODS Using whole-genome sequencing data from 1870 survivors of European ancestry in the St. Jude Lifetime Cohort (SJLIFE) study, genetic variants were examined for association with ejection fraction (EF) and clinically assessed cancer therapy-induced cardiac dysfunction (CCD). Statistically significant findings were validated in 301 SJLIFE survivors of African ancestry and 4020 survivors of European ancestry from the Childhood Cancer Survivor Study. All statistical tests were 2-sided. RESULTS A variant near KCNK17 showed genome-wide significant association with EF (rs2815063-A: EF reduction = 1.6%; P = 2.1 × 10-8) in SJLIFE survivors of European ancestry, which replicated in SJLIFE survivors of African ancestry (EF reduction = 1.5%; P = .004). The rs2815063-A also showed a 1.80-fold (P = .008) risk of severe or disabling or life-threatening CCD and replicated in 4020 Childhood Cancer Survivor Study survivors of European ancestry (odds ratio = 1.40; P = .04). Notably, rs2815063-A was specifically associated among survivors exposed to doxorubicin only, with a stronger effect on EF (3.3% EF reduction) and CCD (2.97-fold). Whole blood DNA methylation data in 1651 SJLIFE survivors of European ancestry showed statistically significant correlation of rs2815063-A with dysregulation of KCNK17 enhancers (false discovery rate <5%), which replicated in 263 survivors of African ancestry. Consistently, the rs2815063-A was associated with KCNK17 downregulation based on RNA sequencing of 75 survivors. CONCLUSIONS Leveraging the 2 largest cohorts of childhood cancer survivors in North America and survivor-specific polygenomic functional data, we identified a novel risk locus for CCD, which showed specificity with doxorubicin-induced cardiac dysfunction and highlighted dysregulation of KCNK17 as the likely molecular mechanism underlying this genetic association.
Collapse
Affiliation(s)
- Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Matthew J Ehrhardt
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Na Qin
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Zhaoming Wang
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
- Department of Computational Biology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Qi Liu
- School of Public Health, University of Alberta, Edmonton, AB,
Canada
| | - Weiyu Qiu
- School of Public Health, University of Alberta, Edmonton, AB,
Canada
| | - Kyla Shelton
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Ying Shao
- Department of Computational Biology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Emily Plyler
- Department of Computational Biology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Heather L Mulder
- Department of Computational Biology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - J Robert Michael
- Department of Computational Biology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University, Chicago,
Il, USA
| | - Xuexia Wang
- Department of Mathematics, University of North Texas, Denton,
TX, USA
| | - Carmen L Wilson
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - John L Jefferies
- Division of Cardiovascular Disease, The University of Tennessee Health
Science Center, Memphis, TN, USA
| | - Eric J Chow
- Clinical Research Division, Fred Hutchinson Cancer Research
Center, WA, USA
| | - Kevin C Oeffinger
- Department of Community and Family Medicine, Duke University,
Durham, NC, USA
| | - Lindsay M Morton
- Raditional Oncology Branch, National Cancer Institute,
Bethesda, MD, USA
| | - Chunliang Li
- Department of Tumor Cell Biology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s
Research Hospital, Memphis, TN, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Smita Bhatia
- Institute of Cancer Outcomes and Survivorship, University of Alabama at
Birmingham, Birmingham, AL, USA
| | - Daniel A Mulrooney
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Melissa M Hudson
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
- Department of Oncology, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research
Hospital, Memphis, TN, USA
| |
Collapse
|
8
|
Shen R, Zuo D, Chen K, Yin Y, Tang K, Hou S, Han B, Xu Y, Liu Z, Chen H. K2P1 leak cation channels contribute to ventricular ectopic beats and sudden death under hypokalemia. FASEB J 2022; 36:e22455. [PMID: 35899468 DOI: 10.1096/fj.202200707r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/28/2022] [Accepted: 07/06/2022] [Indexed: 11/11/2022]
Abstract
Hypokalemia causes ectopic heartbeats, but the mechanisms underlying such cardiac arrhythmias are not understood. In reduced serum K+ concentrations that occur under hypokalemia, K2P1 two-pore domain K+ channels change ion selectivity and switch to conduct inward leak cation currents, which cause aberrant depolarization of resting potential and induce spontaneous action potential of human cardiomyocytes. K2P1 is expressed in the human heart but not in mouse hearts. We test the hypothesis that K2P1 leak cation channels contribute to ectopic heartbeats under hypokalemia, by analysis of transgenic mice, which conditionally express induced K2P1 specifically in hearts, mimicking K2P1 channels in the human heart. Conditional expression of induced K2P1 specifically in the heart of hypokalemic mice results in multiple types of ventricular ectopic beats including single and multiple ventricular premature beats as well as ventricular tachycardia and causes sudden death. In isolated mouse hearts that express induced K2P1, sustained ventricular fibrillation occurs rapidly after perfusion with low K+ concentration solutions that mimic hypokalemic conditions. These observed phenotypes occur rarely in control mice or in the hearts that lack K2P1 expression. K2P1-expressing mouse cardiomyocytes of transgenic mice much more frequently fire abnormal single and/or rhythmic spontaneous action potential in hypokalemic conditions, compared to wild type mouse cardiomyocytes without K2P1 expression. These findings confirm that K2P1 leak cation channels induce ventricular ectopic beats and sudden death of transgenic mice with hypokalemia and imply that K2P1 leak cation channels may play a critical role in human ectopic heartbeats under hypokalemia.
Collapse
Affiliation(s)
- Rongrong Shen
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Dongchuan Zuo
- Key Laboratory of Medical Electrophysiology, Institute of Cardiovascular Research, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Southwest Medical University, Luzhou, China.,Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, USA
| | - Kuihao Chen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, USA.,Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
| | - Yiheng Yin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Kai Tang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Shangwei Hou
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Han
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Zheng Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China.,Cryo-Electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Haijun Chen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, USA
| |
Collapse
|
9
|
Backwell L, Marsh JA. Diverse Molecular Mechanisms Underlying Pathogenic Protein Mutations: Beyond the Loss-of-Function Paradigm. Annu Rev Genomics Hum Genet 2022; 23:475-498. [PMID: 35395171 DOI: 10.1146/annurev-genom-111221-103208] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Most known disease-causing mutations occur in protein-coding regions of DNA. While some of these involve a loss of protein function (e.g., through premature stop codons or missense changes that destabilize protein folding), many act via alternative molecular mechanisms and have dominant-negative or gain-of-function effects. In nearly all cases, these non-loss-of-function mutations can be understood by considering interactions of the wild-type and mutant protein with other molecules, such as proteins, nucleic acids, or small ligands and substrates. Here, we review the diverse molecular mechanisms by which pathogenic mutations can have non-loss-of-function effects, including by disrupting interactions, increasing binding affinity, changing binding specificity, causing assembly-mediated dominant-negative and dominant-positive effects, creating novel interactions, and promoting aggregation and phase separation. We believe that increased awareness of these diverse molecular disease mechanisms will lead to improved diagnosis (and ultimately treatment) of human genetic disorders. Expected final online publication date for the Annual Review of Genomics and Human Genetics, Volume 23 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lisa Backwell
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom;
| | - Joseph A Marsh
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom;
| |
Collapse
|
10
|
Two-Pore-Domain Potassium (K 2P-) Channels: Cardiac Expression Patterns and Disease-Specific Remodelling Processes. Cells 2021; 10:cells10112914. [PMID: 34831137 PMCID: PMC8616229 DOI: 10.3390/cells10112914] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 12/23/2022] Open
Abstract
Two-pore-domain potassium (K2P-) channels conduct outward K+ currents that maintain the resting membrane potential and modulate action potential repolarization. Members of the K2P channel family are widely expressed among different human cell types and organs where they were shown to regulate important physiological processes. Their functional activity is controlled by a broad variety of different stimuli, like pH level, temperature, and mechanical stress but also by the presence of lipids or pharmacological agents. In patients suffering from cardiovascular diseases, alterations in K2P-channel expression and function have been observed, suggesting functional significance and a potential therapeutic role of these ion channels. For example, upregulation of atrial specific K2P3.1 (TASK-1) currents in atrial fibrillation (AF) patients was shown to contribute to atrial action potential duration shortening, a key feature of AF-associated atrial electrical remodelling. Therefore, targeting K2P3.1 (TASK-1) channels might constitute an intriguing strategy for AF treatment. Further, mechanoactive K2P2.1 (TREK-1) currents have been implicated in the development of cardiac hypertrophy, cardiac fibrosis and heart failure. Cardiovascular expression of other K2P channels has been described, functional evidence in cardiac tissue however remains sparse. In the present review, expression, function, and regulation of cardiovascular K2P channels are summarized and compared among different species. Remodelling patterns, observed in disease models are discussed and compared to findings from clinical patients to assess the therapeutic potential of K2P channels.
Collapse
|
11
|
Graff SM, Johnson SR, Leo PJ, Dadi PK, Dickerson MT, Nakhe AY, McInerney-Leo AM, Marshall M, Zaborska KE, Schaub CM, Brown MA, Jacobson DA, Duncan EL. A KCNK16 mutation causing TALK-1 gain of function is associated with maturity-onset diabetes of the young. JCI Insight 2021; 6:138057. [PMID: 34032641 PMCID: PMC8410089 DOI: 10.1172/jci.insight.138057] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/12/2021] [Indexed: 11/17/2022] Open
Abstract
Maturity-onset diabetes of the young (MODY) is a heterogeneous group of monogenic disorders of impaired pancreatic β cell function. The mechanisms underlying MODY include β cell KATP channel dysfunction (e.g., KCNJ11 [MODY13] or ABCC8 [MODY12] mutations); however, no other β cell channelopathies have been associated with MODY to date. Here, we have identified a nonsynonymous coding variant in KCNK16 (NM_001135105: c.341T>C, p.Leu114Pro) segregating with MODY. KCNK16 is the most abundant and β cell-restricted K+ channel transcript, encoding the two-pore-domain K+ channel TALK-1. Whole-cell K+ currents demonstrated a large gain of function with TALK-1 Leu114Pro compared with TALK-1 WT, due to greater single-channel activity. Glucose-stimulated membrane potential depolarization and Ca2+ influx were inhibited in mouse islets expressing TALK-1 Leu114Pro with less endoplasmic reticulum Ca2+ storage. TALK-1 Leu114Pro significantly blunted glucose-stimulated insulin secretion compared with TALK-1 WT in mouse and human islets. These data suggest that KCNK16 is a previously unreported gene for MODY.
Collapse
Affiliation(s)
- Sarah M. Graff
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Stephanie R. Johnson
- Department of Endocrinology, Queensland Children’s Hospital, South Brisbane, Queensland, Australia
- Translational Genomics Group, Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
- Faculty of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Paul J. Leo
- Translational Genomics Group, Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Prasanna K. Dadi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Matthew T. Dickerson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Arya Y. Nakhe
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Aideen M. McInerney-Leo
- Dermatology Research Centre, Dermatology Research Centre, The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Mhairi Marshall
- Translational Genomics Group, Institute of Health and Biomedical Innovation, Faculty of Health, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Karolina E. Zaborska
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Charles M. Schaub
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Matthew A. Brown
- Guy’s and St Thomas’ NHS Foundation Trust and King’s College London NIHR Biomedical Research Centre, King’s College London, London, United Kingdom
| | - David A. Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Emma L. Duncan
- Faculty of Medicine, University of Queensland, Herston, Queensland, Australia
- Department of Twin Research & Genetic Epidemiology, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
12
|
Herrera-Pérez S, Campos-Ríos A, Rueda-Ruzafa L, Lamas JA. Contribution of K2P Potassium Channels to Cardiac Physiology and Pathophysiology. Int J Mol Sci 2021; 22:ijms22126635. [PMID: 34205717 PMCID: PMC8234311 DOI: 10.3390/ijms22126635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 12/28/2022] Open
Abstract
Years before the first two-pore domain potassium channel (K2P) was cloned, certain ion channels had already been demonstrated to be present in the heart with characteristics and properties usually attributed to the TREK channels (a subfamily of K2P channels). K2P channels were later detected in cardiac tissue by RT-PCR, although the distribution of the different K2P subfamilies in the heart seems to depend on the species analyzed. In order to collect relevant information in this regard, we focus here on the TWIK, TASK and TREK cardiac channels, their putative roles in cardiac physiology and their implication in coronary pathologies. Most of the RNA expression data and electrophysiological recordings available to date support the presence of these different K2P subfamilies in distinct cardiac cells. Likewise, we show how these channels may be involved in certain pathologies, such as atrial fibrillation, long QT syndrome and Brugada syndrome.
Collapse
|
13
|
Targeting of Potassium Channels in Cardiac Arrhythmias. Trends Pharmacol Sci 2021; 42:491-506. [PMID: 33858691 DOI: 10.1016/j.tips.2021.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Cardiomyocytes are endowed with a complex repertoire of ion channels, responsible for the generation of action potentials (APs), travelling waves of electrical excitation, propagating throughout the heart and leading to cardiac contractions. Cardiac AP waveforms are shaped by a striking diversity of K+ channels. The pivotal role of K+ channels in cardiac health and disease is underscored by the dramatic impact that K+ channel dysfunction has on cardiac arrhythmias. The development of drugs targeted to specific K+ channels is expected to provide an optimized approach to antiarrhythmic therapy. Here, we review the functional roles of cardiac potassium channels under normal and diseased states. We survey current antiarrhythmic drugs (AADs) targeted to voltage-gated and Ca2+-activated K+ channels and highlight future research opportunities.
Collapse
|
14
|
Wague A, Joseph TT, Woll KA, Bu W, Vaidya KA, Bhanu NV, Garcia BA, Nimigean CM, Eckenhoff RG, Riegelhaupt PM. Mechanistic insights into volatile anesthetic modulation of K2P channels. eLife 2020; 9:59839. [PMID: 33345771 PMCID: PMC7781597 DOI: 10.7554/elife.59839] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/19/2020] [Indexed: 01/01/2023] Open
Abstract
K2P potassium channels are known to be modulated by volatile anesthetic (VA) drugs and play important roles in clinically relevant effects that accompany general anesthesia. Here, we utilize a photoaffinity analog of the VA isoflurane to identify a VA-binding site in the TREK1 K2P channel. The functional importance of the identified site was validated by mutagenesis and biochemical modification. Molecular dynamics simulations of TREK1 in the presence of VA found multiple neighboring residues on TREK1 TM2, TM3, and TM4 that contribute to anesthetic binding. The identified VA-binding region contains residues that play roles in the mechanisms by which heat, mechanical stretch, and pharmacological modulators alter TREK1 channel activity and overlaps with positions found to modulate TASK K2P channel VA sensitivity. Our findings define molecular contacts that mediate VA binding to TREK1 channels and suggest a mechanistic basis to explain how K2P channels are modulated by VAs.
Collapse
Affiliation(s)
- Aboubacar Wague
- Department of Anesthesiology, Weill Cornell Medical College, New York City, United States
| | - Thomas T Joseph
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, United States
| | - Kellie A Woll
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, United States
| | - Weiming Bu
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, United States
| | - Kiran A Vaidya
- Department of Anesthesiology, Weill Cornell Medical College, New York City, United States
| | - Natarajan V Bhanu
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Benjamin A Garcia
- Epigenetics Program, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Crina M Nimigean
- Department of Anesthesiology, Weill Cornell Medical College, New York City, United States.,Department of Physiology and Biophysics, Weill Cornell Medical College, New York City, United States.,Department of Biochemistry, Weill Cornell Medical College, New York City, United States
| | - Roderic G Eckenhoff
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, United States
| | - Paul M Riegelhaupt
- Department of Anesthesiology, Weill Cornell Medical College, New York City, United States
| |
Collapse
|
15
|
Varró A, Tomek J, Nagy N, Virág L, Passini E, Rodriguez B, Baczkó I. Cardiac transmembrane ion channels and action potentials: cellular physiology and arrhythmogenic behavior. Physiol Rev 2020; 101:1083-1176. [PMID: 33118864 DOI: 10.1152/physrev.00024.2019] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrhythmias are among the leading causes of mortality. They often arise from alterations in the electrophysiological properties of cardiac cells and their underlying ionic mechanisms. It is therefore critical to further unravel the pathophysiology of the ionic basis of human cardiac electrophysiology in health and disease. In the first part of this review, current knowledge on the differences in ion channel expression and properties of the ionic processes that determine the morphology and properties of cardiac action potentials and calcium dynamics from cardiomyocytes in different regions of the heart are described. Then the cellular mechanisms promoting arrhythmias in congenital or acquired conditions of ion channel function (electrical remodeling) are discussed. The focus is on human-relevant findings obtained with clinical, experimental, and computational studies, given that interspecies differences make the extrapolation from animal experiments to human clinical settings difficult. Deepening the understanding of the diverse pathophysiology of human cellular electrophysiology will help in developing novel and effective antiarrhythmic strategies for specific subpopulations and disease conditions.
Collapse
Affiliation(s)
- András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - Jakub Tomek
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
16
|
Hasdemir C, Gokcay F, Orman MN, Kocabas U, Payzin S, Sahin H, Nyholt DR, Antzelevitch C. Recognition and clinical implications of high prevalence of migraine in patients with Brugada syndrome and drug-induced type 1 Brugada pattern. J Cardiovasc Electrophysiol 2020; 31:3311-3317. [PMID: 33058326 DOI: 10.1111/jce.14778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/11/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION We have previously reported high 1-year prevalence of migraine in patients with atrial arrhythmias associated with DI-type 1 BrP. The present study was designed to determine the lifetime prevalence of migraine in patients with Brugada syndrome (BrS) or drug-induced type 1 Brugada pattern (DI-type 1 BrP) and control group, to investigate the demographic and clinical characteristics, and to identify clinical variables to predict underlying BrS/DI-type 1 BrP among migraineurs. METHODS AND RESULTS Lifetime prevalence of migraine and migraine characteristics were compared between probands with BrS/DI-type 1 BrP (n = 257) and control group (n = 370). Lifetime prevalence of migraine was 60.7% in patients with BrS/DI-type 1 BrP and 30.3% in control group (p = 3.6 × 10-14 ). On stepwise regression analysis, familial migraine (odds ratio [OR] of 4.4; 95% confidence interval [CI]: 2.0-9.8; p = 1.3 × 10-4 ), vestibular migraine (OR of 5.4; 95% CI: 1.4-21.0); p = .013), migraine with visual aura (OR of 1.8; 95% CI: 1.0-3.4); p = .04) and younger age-at-onset of migraine (OR of 0.95; 95% CI: 0.93-0.98); p = .004) were predictors of underlying BrS/DI-type 1 BrP among migraineurs. Use of anti-migraine drugs classified as "to be avoided" or "preferably avoided" in patients with BrS and several other anti-migraine drugs with potential cardiac INa /ICa channel blocking properties was present in 25.6% and 26.9% of migraineurs with BrS/DI-type 1 BrP, respectively. CONCLUSION Migraine comorbidity is common in patients with BrS/DI-type 1 BrP. We identify several clinical variables that point to an underlying type-1 BrP among migraineurs, necessitating cautious use of certain anti-migraine drugs.
Collapse
Affiliation(s)
- Can Hasdemir
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Figen Gokcay
- Department of Neurology, Ege University School of Medicine, Izmir, Turkey
| | - Mehmet N Orman
- Department of Biostatistics and Medical Informatics, Ege University School of Medicine, Izmir, Turkey
| | - Umut Kocabas
- Department of Cardiology, Baskent University Istanbul Hospital, Istanbul, Turkey
| | - Serdar Payzin
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Hatice Sahin
- Department of Cardiology, Ege University School of Medicine, Izmir, Turkey
| | - Dale R Nyholt
- School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, USA.,Lankenau Heart Institute, Wynnewood, Pennsylvania, USA.,Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Mathie A, Veale EL, Cunningham KP, Holden RG, Wright PD. Two-Pore Domain Potassium Channels as Drug Targets: Anesthesia and Beyond. Annu Rev Pharmacol Toxicol 2020; 61:401-420. [PMID: 32679007 DOI: 10.1146/annurev-pharmtox-030920-111536] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Two-pore domain potassium (K2P) channels stabilize the resting membrane potential of both excitable and nonexcitable cells and, as such, are important regulators of cell activity. There are many conditions where pharmacological regulation of K2P channel activity would be of therapeutic benefit, including, but not limited to, atrial fibrillation, respiratory depression, pulmonary hypertension, neuropathic pain, migraine, depression, and some forms of cancer. Up until now, few if any selective pharmacological regulators of K2P channels have been available. However, recent publications of solved structures with small-molecule activators and inhibitors bound to TREK-1, TREK-2, and TASK-1 K2P channels have given insight into the pharmacophore requirements for compound binding to these sites. Together with the increasing availability of a number of novel, active, small-molecule compounds from K2P channel screening programs, these advances have opened up the possibility of rational activator and inhibitor design to selectively target K2P channels.
Collapse
Affiliation(s)
- Alistair Mathie
- Medway School of Pharmacy, University of Greenwich and University of Kent, Kent ME4 4TB, United Kingdom;
| | - Emma L Veale
- Medway School of Pharmacy, University of Greenwich and University of Kent, Kent ME4 4TB, United Kingdom;
| | - Kevin P Cunningham
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom
| | - Robyn G Holden
- Medway School of Pharmacy, University of Greenwich and University of Kent, Kent ME4 4TB, United Kingdom;
| | | |
Collapse
|
18
|
Clozapine, nimodipine and endosulfan differentially suppress behavioral defects caused by gain-of-function mutations in a two-pore domain K + channel (UNC-58). Neurosci Res 2020; 170:41-49. [PMID: 32681854 DOI: 10.1016/j.neures.2020.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 11/20/2022]
Abstract
Two-pore domain K+ channels (K2Ps) regulate the resting membrane potential in excitable cells and determine ease of depolarization. Gain-of-function (gf) mutations in one of these channels (unc-58) in C. elegans switch it to a Na+ conductance channel and cause tremors, paralysis and other defects. We hypothesized that it should be possible to identify drugs that corrected these defects in unc-58(gf) mutant animals by blocking or modulating the over-active channels. We examined dispersal of animals on food because the absence of effective forward locomotion is the most obvious defect. In addition, we quantified egg release over 24 h. Starting with a known inhibitor of mammalian K2Ps and directed structure-based screening, we evaluated numerous drugs in these assays. Loratadine, which inhibits human KCNK18, significantly improved movement as did methiothepin. We confirmed that endosulfan, a GABA-A receptor antagonist, corrected locomotion in the unc-58(gf) strains. Based on structural similarities to other hits, we found that clozapine, loxapine and amoxapine potently suppressed abnormal phenotypes. Curiously, nimodipine, a Ca++-channel blocker, dramatically improved movement and egg laying in unc-58(e665), but not unc-58(n495) animals. Molecular modeling provided initial insights into a possible basis for this difference based on the location of the e665 and n495 mutations. This research may lead to identification of novel K2P modulators and potential leads for drug discovery.
Collapse
|
19
|
POPDC2 a novel susceptibility gene for conduction disorders. J Mol Cell Cardiol 2020; 145:74-83. [PMID: 32535041 DOI: 10.1016/j.yjmcc.2020.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/22/2020] [Accepted: 06/09/2020] [Indexed: 01/25/2023]
Abstract
Despite recent progress in the understanding of cardiac ion channel function and its role in inherited forms of ventricular arrhythmias, the molecular basis of cardiac conduction disorders often remains unresolved. We aimed to elucidate the genetic background of familial atrioventricular block (AVB) using a whole exome sequencing (WES) approach. In monozygotic twins with a third-degree AVB and in another, unrelated family with first-degree AVB, we identified a heterozygous nonsense mutation in the POPDC2 gene causing a premature stop at position 188 (POPDC2W188⁎), deleting parts of its cAMP binding-domain. Popeye-domain containing (POPDC) proteins are predominantly expressed in the skeletal muscle and the heart, with particularly high expression of POPDC2 in the sinoatrial node of the mouse. We now show by quantitative PCR experiments that in the human heart the POPDC-modulated two-pore domain potassium (K2P) channel TREK-1 is preferentially expressed in the atrioventricular node. Co-expression studies in Xenopus oocytes revealed that POPDC2W188⁎ causes a loss-of-function with impaired TREK-1 modulation. Consistent with the high expression level of POPDC2 in the murine sinoatrial node, POPDC2W188⁎ knock-in mice displayed stress-induced sinus bradycardia and pauses, a phenotype that was previously also reported for POPDC2 and TREK-1 knock-out mice. We propose that the POPDC2W188⁎ loss-of-function mutation contributes to AVB pathogenesis by an aberrant modulation of TREK-1, highlighting that POPDC2 represents a novel arrhythmia gene for cardiac conduction disorders.
Collapse
|
20
|
Arazi E, Blecher G, Zilberberg N. Monoterpenes Differently Regulate Acid-Sensitive and Mechano-Gated K 2P Channels. Front Pharmacol 2020; 11:704. [PMID: 32508645 PMCID: PMC7251055 DOI: 10.3389/fphar.2020.00704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/29/2020] [Indexed: 11/13/2022] Open
Abstract
Potassium K2P (“leak”) channels conduct current across the entire physiological voltage range and carry leak or “background” currents that are, in part, time- and voltage-independent. The activity of K2P channels affects numerous physiological processes, such as cardiac function, pain perception, depression, neuroprotection, and cancer development. We have recently established that, when expressed in Xenopus laevis oocytes, K2P2.1 (TREK-1) channels are activated by several monoterpenes (MTs). Here, we show that, within a few minutes of exposure, other mechano-gated K2P channels, K2P4.1 (TRAAK) and K2P10.1 (TREK-2), are opened by monoterpenes as well (up to an eightfold increase in current). Furthermor\e, carvacrol and cinnamaldehyde robustly enhance currents of the alkaline-sensitive K2P5.1 (up to a 17-fold increase in current). Other members of the K2P potassium channels, K2P17.1, K2P18.1, but not K2P16.1, were also activated by various MTs. Conversely, the activity of members of the acid-sensitive (TASK) K2P channels (K2P3.1 and K2P9.1) was rapidly decreased by monoterpenes. We found that MT selectively decreased the voltage-dependent portion of the current and that current inhibition was reduced with the elevation of external K+ concentration. These findings suggest that penetration of MTs into the outer leaflet of the membrane results in immediate changes at the selectivity filter of members of the TASK channel family. Thus, we suggest MTs as promising new tools for the study of K2P channels’ activity in vitro as well as in vivo.
Collapse
Affiliation(s)
- Eden Arazi
- Department of Life Sciences Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Galit Blecher
- Department of Life Sciences Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Noam Zilberberg
- Department of Life Sciences Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
21
|
TASK channels: channelopathies, trafficking, and receptor-mediated inhibition. Pflugers Arch 2020; 472:911-922. [DOI: 10.1007/s00424-020-02403-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 01/06/2023]
|
22
|
Xian W, Hui X, Tian Q, Wang H, Moretti A, Laugwitz KL, Flockerzi V, Ruppenthal S, Lipp P. Aberrant Deactivation-Induced Gain of Function in TRPM4 Mutant Is Associated with Human Cardiac Conduction Block. Cell Rep 2019; 24:724-731. [PMID: 30021168 DOI: 10.1016/j.celrep.2018.06.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/27/2018] [Accepted: 06/07/2018] [Indexed: 02/04/2023] Open
Abstract
A gain-of-function mutation in the Ca2+-activated transient receptor potential melastatin member 4 (TRPM4A432T) is linked to life-threatening cardiac conduction disturbance, but the underlying mechanism is unclear. For deeper insights, we used photolysis of caged Ca2+, quantitative Ca2+, and electrophysiological measurements. TRPM4A432T's 2-fold larger membrane current was associated with 50% decreased plasma membrane expression. Kinetic analysis unveiled 4-fold slower deactivation that was responsible for the augmented membrane current progressively rising during repetitive human cardiac action potentials. Rational mutagenesis of TRPM4 at position 432 revealed that the bulkiness of the amino acid was key to TRPM4A432T's aberrant gating. Charged amino acids rendered the channel non-functional. The slow deactivation caused by an amino acid substitution at position 432 from alanine to the bulkier threonine represents a key contributor to the gain of function in TRPM4A432T. Thus, our results add a mechanism in the etiology of TRP channel-linked human cardiac channelopathies.
Collapse
Affiliation(s)
- Wenying Xian
- Molecular Cell Biology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| | - Xin Hui
- Molecular Cell Biology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| | - Qinghai Tian
- Molecular Cell Biology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| | - Hongmei Wang
- Experimental and Clinical Pharmacology and Toxicology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| | - Alessandra Moretti
- Department of Medicine I (Cardiology and Angiology), Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Karl-Ludwig Laugwitz
- Department of Medicine I (Cardiology and Angiology), Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| | - Sandra Ruppenthal
- Molecular Cell Biology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany
| | - Peter Lipp
- Molecular Cell Biology, Centre for Molecular Signaling (PZMS), Medical Faculty, Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
23
|
Jacobson DA, Shyng SL. Ion Channels of the Islets in Type 2 Diabetes. J Mol Biol 2019; 432:1326-1346. [PMID: 31473158 DOI: 10.1016/j.jmb.2019.08.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
Abstract
Ca2+ is an essential signal for pancreatic β-cell function. Ca2+ plays critical roles in numerous β-cell pathways such as insulin secretion, transcription, metabolism, endoplasmic reticulum function, and the stress response. Therefore, β-cell Ca2+ handling is tightly controlled. At the plasma membrane, Ca2+ entry primarily occurs through voltage-dependent Ca2+ channels. Voltage-dependent Ca2+ channel activity is dependent on orchestrated fluctuations in the plasma membrane potential or voltage, which are mediated via the activity of many ion channels. During the pathogenesis of type 2 diabetes the β-cell is exposed to stressful conditions, which result in alterations of Ca2+ handling. Some of the changes in β-cell Ca2+ handling that occur under stress result from perturbations in ion channel activity, expression or localization. Defective Ca2+ signaling in the diabetic β-cell alters function, limits insulin secretion and exacerbates hyperglycemia. In this review, we focus on the β-cell ion channels that control Ca2+ handling and how they impact β-cell dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- David A Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7415 MRB4 (Langford), 2213 Garland Avenue, Nashville, TN 37232, USA.
| | - Show-Ling Shyng
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, L224, MRB 624, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
24
|
Bedoya M, Rinné S, Kiper AK, Decher N, González W, Ramírez D. TASK Channels Pharmacology: New Challenges in Drug Design. J Med Chem 2019; 62:10044-10058. [PMID: 31260312 DOI: 10.1021/acs.jmedchem.9b00248] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Rational drug design targeting ion channels is an exciting and always evolving research field. New medicinal chemistry strategies are being implemented to explore the wild chemical space and unravel the molecular basis of the ion channels modulators binding mechanisms. TASK channels belong to the two-pore domain potassium channel family and are modulated by extracellular acidosis. They are extensively distributed along the cardiovascular and central nervous systems, and their expression is up- and downregulated in different cancer types, which makes them an attractive therapeutic target. However, TASK channels remain unexplored, and drugs designed to target these channels are poorly selective. Here, we review TASK channels properties and their known blockers and activators, considering the new challenges in ion channels drug design and focusing on the implementation of computational methodologies in the drug discovery process.
Collapse
Affiliation(s)
- Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Aytug K Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud , Universidad Autónoma de Chile , El Llano Subercaseaux 2801, Piso 6 , 8900000 Santiago , Chile
| |
Collapse
|
25
|
Lv J, Liang Y, Zhang S, Lan Q, Xu Z, Wu X, Kang L, Ren J, Cao Y, Wu T, Lin KL, Yung KKL, Cao X, Pang J, Zhou P. DCPIB, an Inhibitor of Volume-Regulated Anion Channels, Distinctly Modulates K2P Channels. ACS Chem Neurosci 2019; 10:2786-2793. [PMID: 30935201 DOI: 10.1021/acschemneuro.9b00010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
K2P potassium channels stabilize the resting membrane potential in nearly all cells and have been implicated in several neuronal, cardiovascular, and immune diseases. DCPIB, a known specific and potent inhibitor of volume-regulated anion channels (VRAC), has been reported to activate TREK1 and TREK2 in astrocytes and in vitro recently. In the present study, we demonstrated DCPIB also voltage dependently activated TRAAK besides TREK1/TREK2, showing DCPIB activated all TREK subfamily members. In contrast, the compound potently inhibited several other K2P channels with no voltage dependence, including TRESK, TASK1, and TASK3. DCPIB displayed superior selectivity toward TRESK with an IC50 of 0.14 μM, demonstrating at least 100-fold higher affinity over TREK1/TRAAK channels. Furthermore, the impaired ion selectivity filter region greatly impaired the activating effect of DCPIB on TREK1 but not the inhibitory effect of DCPIB on TRESK. This indicates distinct molecular determinants underlying the effect of DCPIB on TREK1 or TRESK channels. Our results showed that DCPIB played diverse effects on K2P channels and could be a useful tool for further investigating structure-function studies of K2P channels.
Collapse
Affiliation(s)
- Jinyan Lv
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yemei Liang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shiqing Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region, China
| | - Qunsheng Lan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ziwei Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyan Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lijun Kang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jing Ren
- Key Laboratory of Mental Health of the Ministry of Education, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ying Cao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ting Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ka Li Lin
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region, China
| | - Ken Kin Lam Yung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region, China
| | - Xiong Cao
- Key Laboratory of Mental Health of the Ministry of Education, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Pingzheng Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
26
|
Wiedmann F, Schlund D, Voigt N, Ratte A, Kraft M, Katus HA, Schmidt C. N-glycosylation-dependent regulation of hK 2P17.1 currents. Mol Biol Cell 2019; 30:1425-1436. [PMID: 30969900 PMCID: PMC6724686 DOI: 10.1091/mbc.e18-10-0687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Two pore-domain potassium (K2P) channels mediate potassium background currents that stabilize the resting membrane potential and facilitate action potential repolarization. In the human heart, hK2P17.1 channels are predominantly expressed in the atria and Purkinje cells. Reduced atrial hK2P17.1 protein levels were described in patients with atrial fibrillation or heart failure. Genetic alterations in hK2P17.1 were associated with cardiac conduction disorders. Little is known about posttranslational modifications of hK2P17.1. Here, we characterized glycosylation of hK2P17.1 and investigated how glycosylation alters its surface expression and activity. Wild-type hK2P17.1 channels and channels lacking specific glycosylation sites were expressed in Xenopus laevis oocytes, HEK-293T cells, and HeLa cells. N-glycosylation was disrupted using N-glycosidase F and tunicamycin. hK2P17.1 expression and activity were assessed using immunoblot analysis and a two-electrode voltage clamp technique. Channel subunits of hK2P17.1 harbor two functional N-glycosylation sites at positions N65 and N94. In hemi-glycosylated hK2P17.1 channels, functionality and membrane trafficking remain preserved. Disruption of both N-glycosylation sites results in loss of hK2P17.1 currents, presumably caused by impaired surface expression. This study confirms diglycosylation of hK2P17.1 channel subunits and its pivotal role in cell-surface targeting. Our findings underline the functional relevance of N-glycosylation in biogenesis and membrane trafficking of ion channels.
Collapse
Affiliation(s)
- Felix Wiedmann
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany.,HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, 69120 Heidelberg, Germany
| | - Daniel Schlund
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg-August University of Göttingen, 37073 Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, University of Göttingen, 37073 Göttingen, Germany
| | - Antonius Ratte
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany.,HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, 69120 Heidelberg, Germany
| | - Manuel Kraft
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany.,HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, 69120 Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany.,HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, 69120 Heidelberg, Germany
| | - Constanze Schmidt
- Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69120 Heidelberg, Germany.,HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
27
|
Rinné S, Kiper AK, Vowinkel KS, Ramírez D, Schewe M, Bedoya M, Aser D, Gensler I, Netter MF, Stansfeld PJ, Baukrowitz T, Gonzalez W, Decher N. The molecular basis for an allosteric inhibition of K +-flux gating in K 2P channels. eLife 2019; 8:39476. [PMID: 30803485 PMCID: PMC6391080 DOI: 10.7554/elife.39476] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 02/06/2019] [Indexed: 01/05/2023] Open
Abstract
Two-pore-domain potassium (K2P) channels are key regulators of many physiological and pathophysiological processes and thus emerged as promising drug targets. As for other potassium channels, there is a lack of selective blockers, since drugs preferentially bind to a conserved binding site located in the central cavity. Thus, there is a high medical need to identify novel drug-binding sites outside the conserved lipophilic central cavity and to identify new allosteric mechanisms of channel inhibition. Here, we identified a novel binding site and allosteric inhibition mechanism, disrupting the recently proposed K+-flux gating mechanism of K2P channels, which results in an unusual voltage-dependent block of leak channels belonging to the TASK subfamily. The new binding site and allosteric mechanism of inhibition provide structural and mechanistic insights into the gating of TASK channels and the basis for the drug design of a new class of potent blockers targeting specific types of K2P channels.
Collapse
Affiliation(s)
- Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Marburg, Germany
| | - Aytug K Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Marburg, Germany
| | - Kirsty S Vowinkel
- Institute for Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Marburg, Germany
| | - David Ramírez
- Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca, Chile.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Marcus Schewe
- Institute of Physiology, University of Kiel, Kiel, Germany
| | - Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca, Chile
| | - Diana Aser
- Institute for Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Marburg, Germany
| | - Isabella Gensler
- Institute for Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Marburg, Germany
| | - Michael F Netter
- Institute for Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Marburg, Germany
| | - Phillip J Stansfeld
- Structural Bioinformatics and Computational Biochemistry Unit, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | | | - Wendy Gonzalez
- Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, Talca, Chile
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Marburg, Germany
| |
Collapse
|
28
|
Ben Soussia I, El Mouridi S, Kang D, Leclercq-Blondel A, Khoubza L, Tardy P, Zariohi N, Gendrel M, Lesage F, Kim EJ, Bichet D, Andrini O, Boulin T. Mutation of a single residue promotes gating of vertebrate and invertebrate two-pore domain potassium channels. Nat Commun 2019; 10:787. [PMID: 30770809 PMCID: PMC6377628 DOI: 10.1038/s41467-019-08710-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 01/23/2019] [Indexed: 01/28/2023] Open
Abstract
Mutations that modulate the activity of ion channels are essential tools to understand the biophysical determinants that control their gating. Here, we reveal the conserved role played by a single amino acid position (TM2.6) located in the second transmembrane domain of two-pore domain potassium (K2P) channels. Mutations of TM2.6 to aspartate or asparagine increase channel activity for all vertebrate K2P channels. Using two-electrode voltage-clamp and single-channel recording techniques, we find that mutation of TM2.6 promotes channel gating via the selectivity filter gate and increases single channel open probability. Furthermore, channel gating can be progressively tuned by using different amino acid substitutions. Finally, we show that the role of TM2.6 was conserved during evolution by rationally designing gain-of-function mutations in four Caenorhabditis elegans K2P channels using CRISPR/Cas9 gene editing. This study thus describes a simple and powerful strategy to systematically manipulate the activity of an entire family of potassium channels. Mutations that modulate the activity of ion channels are essential tools to understand the biophysical determinants that control their gating. Here authors reveal the role played by a single residue in the second transmembrane domain of vertebrate and invertebrate two-pore domain potassium channels.
Collapse
Affiliation(s)
- Ismail Ben Soussia
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France
| | - Sonia El Mouridi
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France
| | - Dawon Kang
- Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University, Jinju, 52727, South Korea
| | - Alice Leclercq-Blondel
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France
| | - Lamyaa Khoubza
- Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, CNRS UMR 7275, Université de Nice Sophia Antipolis, Valbonne, 06560, France
| | - Philippe Tardy
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France
| | - Nora Zariohi
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France
| | - Marie Gendrel
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France
| | - Florian Lesage
- Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, CNRS UMR 7275, Université de Nice Sophia Antipolis, Valbonne, 06560, France
| | - Eun-Jin Kim
- Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University, Jinju, 52727, South Korea
| | - Delphine Bichet
- Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, CNRS UMR 7275, Université de Nice Sophia Antipolis, Valbonne, 06560, France
| | - Olga Andrini
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France.
| | - Thomas Boulin
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France.
| |
Collapse
|
29
|
Nedumaran B, Pineda RH, Rudra P, Lee S, Malykhina AP. Association of genetic polymorphisms in the pore domains of mechano-gated TREK-1 channel with overactive lower urinary tract symptoms in humans. Neurourol Urodyn 2018; 38:144-150. [PMID: 30350878 DOI: 10.1002/nau.23862] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022]
Abstract
AIMS Mechanosensitivity of the urinary bladder is regulated by many factors including mechano-gated two-pore domain (K2 P, KCNK) potassium channels. TWIK-related K+ channel, TREK-1, is a predominantly expressed member of K2 P channel family in the human detrusor, and its expression and function are diminished in patients with overactive lower urinary tract symptoms (LUTS). The changes in channel activity may result from spontaneously occurring gene mutations. The aim of this study was to compare single nucleotide polymorphisms (SNPs) in TREK-1 channel between patients with LUTS and healthy donors. METHODS Six SNPs (rs370266806, rs373919966, rs758937019, rs769301539, rs772497750, and rs775158737) in two pore domains of human TREK-1 gene were analyzed using TaqMan SNP genotyping assay with manufacturer-designed primers and allele-specific probes. The screening was done in control bladders and detrusor specimens from patients with overactive LUTS. Statistical analyses were performed using R, Fisher's exact test and Hardy-Weinberg Equilibrium. RESULTS Six SNPs in two pore domains of the human TREK-1 gene were analyzed in human bladder specimens. The frequencies of rs758937019-CT genotype (P = 0.0016) and rs758937019-T allele (P = 0.0022) were significantly higher in the group with overactive LUTS. There was no significant association of rs775158737-GA genotype and rs775158737-A allele with the overactive LUTS, though they were present only in the overactive LUTS group. CONCLUSIONS Our results provide evidence that altered expression and function of TREK-1 channel in patients with overactive LUTS could be due to genetic polymorphisms in the pore domains of TREK-1 channel (rs758937019).
Collapse
Affiliation(s)
- Balachandar Nedumaran
- Division of Urology, Department of Surgery, University of Colorado Denver, Aurora, Colorado.,Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado Denver, Aurora, Colorado
| | - Ricardo H Pineda
- Division of Urology, Department of Surgery, University of Colorado Denver, Aurora, Colorado
| | - Pratyaydipta Rudra
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado
| | - Sanghee Lee
- Department of Urology, University of California San Diego, La Jolla, California
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
30
|
Boyden PA. Purkinje physiology and pathophysiology. J Interv Card Electrophysiol 2018; 52:255-262. [PMID: 30056516 DOI: 10.1007/s10840-018-0414-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/17/2018] [Indexed: 01/08/2023]
Abstract
There has always been an appreciation of the role of Purkinje fibers in the fast conduction of the normal cardiac impulse. Here, we briefly update our knowledge of this important set of cardiac cells. We discuss the anatomy of a Purkinje fiber strand, the importance of longitudinal conduction within a strand, circus movement within a strand, conduction, and excitability properties of Purkinjes. At the cell level, we discuss the important components of the ion channel makeup in the nonremodeled Purkinjes of healthy hearts. Finally, we discuss the role of the Purkinjes in forming the heritable arrhythmogenic substrates such as long QT, heritable conduction slowing, CPVT, sQT, and Brugada syndromes.
Collapse
Affiliation(s)
- Penelope A Boyden
- Department of Pharmacology, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
31
|
Cardiovascular pharmacology of K 2P17.1 (TASK-4, TALK-2) two-pore-domain K + channels. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:1119-1131. [PMID: 30008082 DOI: 10.1007/s00210-018-1535-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/06/2018] [Indexed: 10/28/2022]
Abstract
K2P17.1 (TASK-4, TALK-2) potassium channels are expressed in the heart and represent potential targets for pharmacological management of atrial and ventricular arrhythmias. Reduced K2P17.1 expression was found in atria and ventricles of heart failure (HF) patients. Modulation of K2P17.1 currents by antiarrhythmic compounds has not been comprehensively studied to date. The objective of this study was to investigate acute effects of clinically relevant antiarrhythmic drugs on human K2P17.1 channels to provide a more complete picture of K2P17.1 electropharmacology. Whole-cell patch clamp and two-electrode voltage clamp electrophysiology was employed to study human K2P17.1 channel pharmacology. K2P17.1 channels expressed in Xenopus laevis oocytes were screened for sensitivity to antiarrhythmic drugs, revealing significant activation by propafenone (+ 296%; 100 μM), quinidine (+ 58%; 100 μM), mexiletine (+ 21%; 100 μM), propranolol (+ 139%; 100 μM), and metoprolol (+ 17%; 100 μM) within 60 min. In addition, the currents were inhibited by amiodarone (- 13%; 100 μM), sotalol (- 10%; 100 μM), verapamil (- 21%; 100 μM), and ranolazine (- 8%; 100 μM). K2P17.1 channels were not significantly affected by ajmaline and carvedilol. Concentration-dependent K2P17.1 activation by propafenone was characterized in more detail. The onset of activation was fast, and current-voltage relationships were not modulated by propafenone. K2P17.1 activation was confirmed in mammalian Chinese hamster ovary cells, revealing 7.8-fold current increase by 100 μM propafenone. Human K2P17.1 channels were sensitive to multiple antiarrhythmic drugs. Differential pharmacological regulation of repolarizing K2P17.1 background K+ channels may be employed for personalized antiarrhythmic therapy.
Collapse
|
32
|
Staudacher I, Illg C, Gierten J, Seehausen S, Schweizer PA, Katus HA, Thomas D. Identification and functional characterization of zebrafish K 2P 17.1 (TASK-4, TALK-2) two-pore-domain K + channels. Eur J Pharmacol 2018; 831:94-102. [DOI: 10.1016/j.ejphar.2018.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022]
|
33
|
Wiedmann F, Schulte JS, Gomes B, Zafeiriou MP, Ratte A, Rathjens F, Fehrmann E, Scholz B, Voigt N, Müller FU, Thomas D, Katus HA, Schmidt C. Atrial fibrillation and heart failure-associated remodeling of two-pore-domain potassium (K2P) channels in murine disease models: focus on TASK-1. Basic Res Cardiol 2018; 113:27. [DOI: 10.1007/s00395-018-0687-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/04/2018] [Indexed: 12/27/2022]
|
34
|
Silbernagel N, Walecki M, Schäfer MKH, Kessler M, Zobeiri M, Rinné S, Kiper AK, Komadowski MA, Vowinkel KS, Wemhöner K, Fortmüller L, Schewe M, Dolga AM, Scekic-Zahirovic J, Matschke LA, Culmsee C, Baukrowitz T, Monassier L, Ullrich ND, Dupuis L, Just S, Budde T, Fabritz L, Decher N. The VAMP-associated protein VAPB is required for cardiac and neuronal pacemaker channel function. FASEB J 2018; 32:6159-6173. [PMID: 29879376 PMCID: PMC6629115 DOI: 10.1096/fj.201800246r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels encode neuronal and cardiac pacemaker currents. The composition of pacemaker channel complexes in different tissues is poorly understood, and the presence of additional HCN modulating subunits was speculated. Here we show that vesicle-associated membrane protein-associated protein B (VAPB), previously associated with a familial form of amyotrophic lateral sclerosis 8, is an essential HCN1 and HCN2 modulator. VAPB significantly increases HCN2 currents and surface expression and has a major influence on the dendritic neuronal distribution of HCN2. Severe cardiac bradycardias in VAPB-deficient zebrafish and VAPB-/- mice highlight that VAPB physiologically serves to increase cardiac pacemaker currents. An altered T-wave morphology observed in the ECGs of VAPB-/- mice supports the recently proposed role of HCN channels for ventricular repolarization. The critical function of VAPB in native pacemaker channel complexes will be relevant for our understanding of cardiac arrhythmias and epilepsies, and provides an unexpected link between these diseases and amyotrophic lateral sclerosis.-Silbernagel, N., Walecki, M., Schäfer, M.-K. H., Kessler, M., Zobeiri, M., Rinné, S., Kiper, A. K., Komadowski, M. A., Vowinkel, K. S., Wemhöner, K., Fortmüller, L., Schewe, M., Dolga, A. M., Scekic-Zahirovic, J., Matschke, L. A., Culmsee, C., Baukrowitz, T., Monassier, L., Ullrich, N. D., Dupuis, L., Just, S., Budde, T., Fabritz, L., Decher, N. The VAMP-associated protein VAPB is required for cardiac and neuronal pacemaker channel function.
Collapse
Affiliation(s)
- Nicole Silbernagel
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Phillips University, Marburg, Germany
| | - Magdalena Walecki
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Phillips University, Marburg, Germany
| | - Martin K-H Schäfer
- Institute of Anatomy and Cell Biology, Philipps University, Marburg, Germany
| | - Mirjam Kessler
- Molecular Cardiology, Department of Internal Medicine II, University Hospital Ulm, Ulm, Germany
| | | | - Susanne Rinné
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Phillips University, Marburg, Germany
| | - Aytug K Kiper
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Phillips University, Marburg, Germany
| | - Marlene A Komadowski
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Phillips University, Marburg, Germany.,Institute of Anatomy and Cell Biology, Philipps University, Marburg, Germany
| | - Kirsty S Vowinkel
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Phillips University, Marburg, Germany
| | - Konstantin Wemhöner
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Phillips University, Marburg, Germany
| | - Lisa Fortmüller
- Department of Cardiology II - Electrophysiology, University Hospital Münster, University of Münster, Munster, Germany
| | - Marcus Schewe
- Institute of Physiology, Christian-Albrechts University, Kiel, Germany
| | - Amalia M Dolga
- Institute of Pharmacology and Clinical Pharmacy, Phillips University, Marburg, Germany
| | - Jelena Scekic-Zahirovic
- Laboratoire de Pharmacologie et Toxicologie NeuroCardiovasculaire, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| | - Lina A Matschke
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Phillips University, Marburg, Germany
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, Phillips University, Marburg, Germany
| | - Thomas Baukrowitz
- Institute of Physiology, Christian-Albrechts University, Kiel, Germany
| | - Laurent Monassier
- Laboratoire de Pharmacologie et Toxicologie NeuroCardiovasculaire, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| | - Nina D Ullrich
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Luc Dupuis
- Laboratoire de Neurobiologie et Pharmacologie Cardiovasculaire, Faculté de Médecine, Université de Strasbourg, Strasbourg, France.,INSERM, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, University Hospital Ulm, Ulm, Germany
| | - Thomas Budde
- Institute for Physiology I, University of Münster, Munster, Germany
| | - Larissa Fabritz
- Department of Cardiology II - Electrophysiology, University Hospital Münster, University of Münster, Munster, Germany.,Institute of Cardiovascular Sciences, University Hospital Birmingham, University of Birmingham, Birmingham, United Kingdom.,Department of Cardiology, University Hospital Birmingham, University of Birmingham, Birmingham, United Kingdom.,Division of Rhythmology, Department of Genetic Epidemiology, University Hospital Münster, University of Münster, Munster, Germany.,Institute of Human Genetics, Department of Genetic Epidemiology, University Hospital Münster, University of Münster, Munster, Germany
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Phillips University, Marburg, Germany
| |
Collapse
|
35
|
Chai S, Wan X, Ramirez-Navarro A, Tesar PJ, Kaufman ES, Ficker E, George AL, Deschênes I. Physiological genomics identifies genetic modifiers of long QT syndrome type 2 severity. J Clin Invest 2018; 128:1043-1056. [PMID: 29431731 DOI: 10.1172/jci94996] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 01/02/2018] [Indexed: 12/11/2022] Open
Abstract
Congenital long QT syndrome (LQTS) is an inherited channelopathy associated with life-threatening arrhythmias. LQTS type 2 (LQT2) is caused by mutations in KCNH2, which encodes the potassium channel hERG. We hypothesized that modifier genes are partly responsible for the variable phenotype severity observed in some LQT2 families. Here, we identified contributors to variable expressivity in an LQT2 family by using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and whole exome sequencing in a synergistic manner. We found that iPSC-CMs recapitulated the clinical genotype-phenotype discordance in vitro. Importantly, iPSC-CMs derived from the severely affected LQT2 patients displayed prolonged action potentials compared with cells from mildly affected first-degree relatives. The iPSC-CMs derived from all patients with hERG R752W mutation displayed lower IKr amplitude. Interestingly, iPSC-CMs from severely affected mutation-positive individuals exhibited greater L-type Ca2+ current. Whole exome sequencing identified variants of KCNK17 and the GTP-binding protein REM2, providing biologically plausible explanations for this variable expressivity. Genome editing to correct a REM2 variant reversed the enhanced L-type Ca2+ current and prolonged action potential observed in iPSC-CMs from severely affected individuals. Thus, our findings showcase the power of combining complementary physiological and genomic analyses to identify genetic modifiers and potential therapeutic targets of a monogenic disorder. Furthermore, we propose that this strategy can be deployed to unravel myriad confounding pathologies displaying variable expressivity.
Collapse
Affiliation(s)
- Sam Chai
- Department of Physiology and Biophysics.,Heart and Vascular Research Center, Department of Medicine, and
| | - Xiaoping Wan
- Heart and Vascular Research Center, Department of Medicine, and
| | | | - Paul J Tesar
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Eckhard Ficker
- Heart and Vascular Research Center, Department of Medicine, and
| | - Alfred L George
- Department of Pharmacology, Northwestern University, Chicago, Illinois, USA
| | - Isabelle Deschênes
- Department of Physiology and Biophysics.,Heart and Vascular Research Center, Department of Medicine, and
| |
Collapse
|
36
|
Symonds JD, Zuberi SM. Genetics update: Monogenetics, polygene disorders and the quest for modifying genes. Neuropharmacology 2017; 132:3-19. [PMID: 29037745 DOI: 10.1016/j.neuropharm.2017.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 10/09/2017] [Accepted: 10/11/2017] [Indexed: 12/19/2022]
Abstract
The genetic channelopathies are a broad collection of diseases. Many ion channel genes demonstrate wide phenotypic pleiotropy, but nonetheless concerted efforts have been made to characterise genotype-phenotype relationships. In this review we give an overview of the factors that influence genotype-phenotype relationships across this group of diseases as a whole, using specific individual channelopathies as examples. We suggest reasons for the limitations observed in these relationships. We discuss the role of ion channel variation in polygenic disease and highlight research that has contributed to unravelling the complex aetiological nature of these conditions. We focus specifically on the quest for modifying genes in inherited channelopathies, using the voltage-gated sodium channels as an example. Epilepsy related to genetic channelopathy is one area in which precision medicine is showing promise. We will discuss the successes and limitations of precision medicine in these conditions. This article is part of the Special Issue entitled 'Channelopathies.'
Collapse
Affiliation(s)
- Joseph D Symonds
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Queen Elizabeth University Hospitals, Glasgow, UK; School of Medicine, University of Glasgow, Glasgow, UK
| | - Sameer M Zuberi
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Queen Elizabeth University Hospitals, Glasgow, UK; School of Medicine, University of Glasgow, Glasgow, UK.
| |
Collapse
|
37
|
Heterodimerization of two pore domain K+ channel TASK1 and TALK2 in living heterologous expression systems. PLoS One 2017; 12:e0186252. [PMID: 29016681 PMCID: PMC5634629 DOI: 10.1371/journal.pone.0186252] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/27/2017] [Indexed: 11/19/2022] Open
Abstract
Two-pore-domain K+ (K2P) channels sense a wide variety of stimuli such as mechanical stress, inhalational anesthetics, and changes in extracellular pH or temperature. The K2P channel activity forms a background K+ current and, thereby, contributes to resting membrane potentials. Six subfamilies including fifteen subtypes of K2P channels have been identified. Each K2P channel molecule with two pores consists of a homodimer of each subtype. In addition, a few heterodimers mainly within the same subfamilies have been found recently. In the present study, the possibility of heterodimerization between TASK1 (TWIK-Related Acid-Sensitive K+ channel) and TALK2 (TWIK-Related Alkaline pH-Activated K+ channel) was examined. These channels belong to separate subfamilies and show extremely different channel properties. Surprisingly, single molecular imaging analyses in this study using a total internal reflection microscope suggested the heterodimerization of TASK1 and TALK2 in a pancreatic cell line, QGP-1. This heterodimer was also detected using a bimolecular fluorescence complementation assay in a HEK293 heterologous expression system. Fluorescence resonance energy transfer analyses showed that the affinity between TASK1 and TALK2 appeared to be close to those of homodimers. Whole-cell patch-clamp recordings revealed that TASK1 currents in HEK293 cells were significantly attenuated by co-expression of a dominant-negative form of TALK2 in comparison with that of wild-type TALK2. The sensitivities of TASK1-TALK2 tandem constructs to extracellular pH and halothane were characterized as a unique hybrid of TASK1 and TALK2. These results suggested that heterodimerization of TASK1 and TALK2 provides cells with the ability to make multiple responses to a variety of physiological and pharmacological stimuli.
Collapse
|
38
|
Decher N, Ortiz-Bonnin B, Friedrich C, Schewe M, Kiper AK, Rinné S, Seemann G, Peyronnet R, Zumhagen S, Bustos D, Kockskämper J, Kohl P, Just S, González W, Baukrowitz T, Stallmeyer B, Schulze-Bahr E. Sodium permeable and "hypersensitive" TREK-1 channels cause ventricular tachycardia. EMBO Mol Med 2017; 9:403-414. [PMID: 28242754 PMCID: PMC5376757 DOI: 10.15252/emmm.201606690] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In a patient with right ventricular outflow tract (RVOT) tachycardia, we identified a heterozygous point mutation in the selectivity filter of the stretch-activated K2P potassium channel TREK-1 (KCNK2 or K2P2.1). This mutation introduces abnormal sodium permeability to TREK-1. In addition, mutant channels exhibit a hypersensitivity to stretch-activation, suggesting that the selectivity filter is directly involved in stretch-induced activation and desensitization. Increased sodium permeability and stretch-sensitivity of mutant TREK-1 channels may trigger arrhythmias in areas of the heart with high physical strain such as the RVOT We present a pharmacological strategy to rescue the selectivity defect of the TREK-1 pore. Our findings provide important insights for future studies of K2P channel stretch-activation and the role of TREK-1 in mechano-electrical feedback in the heart.
Collapse
Affiliation(s)
- Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Marburg, Germany
| | - Beatriz Ortiz-Bonnin
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Marburg, Germany
| | - Corinna Friedrich
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Münster, Germany
| | - Marcus Schewe
- Institute of Physiology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Aytug K Kiper
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Marburg, Germany
| | - Susanne Rinné
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Marburg, Germany
| | - Gunnar Seemann
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sven Zumhagen
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Münster, Germany
| | - Daniel Bustos
- Center for Bioinformatics and Molecular Simulation, University of Talca, Talca, Chile
| | - Jens Kockskämper
- Institute of Pharmacology and Clinical Pharmacy, Biochemical and Pharmacological Center (BPC) Philipps-University of Marburg, Marburg, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Steffen Just
- Molecular Cardiology, University Hospital Ulm, Ulm, Germany
| | - Wendy González
- Center for Bioinformatics and Molecular Simulation, University of Talca, Talca, Chile
| | - Thomas Baukrowitz
- Institute of Physiology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Birgit Stallmeyer
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Münster, Germany
| | - Eric Schulze-Bahr
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Münster, Germany
| |
Collapse
|
39
|
Symonds JD, Zuberi SM. WITHDRAWN: Genetics update: Monogenetics, polygene disorders and the quest for modifying genes. Neuropharmacology 2017:S0028-3908(17)30347-7. [PMID: 28757052 DOI: 10.1016/j.neuropharm.2017.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/17/2017] [Indexed: 11/15/2022]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, https://doi.org/10.1016/j.neuropharm.2017.10.013. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Joseph D Symonds
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Queen Elizabeth University Hospitals, Glasgow, UK; School of Medicine, University of Glasgow, Glasgow, UK
| | - Sameer M Zuberi
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Queen Elizabeth University Hospitals, Glasgow, UK; School of Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
40
|
Proost D, Saenen J, Vandeweyer G, Rotthier A, Alaerts M, Van Craenenbroeck EM, Van Crombruggen J, Mortier G, Wuyts W, Vrints C, Del Favero J, Loeys B, Van Laer L. Targeted Next-Generation Sequencing of 51 Genes Involved in Primary Electrical Disease. J Mol Diagn 2017; 19:445-459. [DOI: 10.1016/j.jmoldx.2017.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 01/18/2023] Open
|
41
|
Chai S, Wan X, Nassal DM, Liu H, Moravec CS, Ramirez-Navarro A, Deschênes I. Contribution of two-pore K + channels to cardiac ventricular action potential revealed using human iPSC-derived cardiomyocytes. Am J Physiol Heart Circ Physiol 2017; 312:H1144-H1153. [PMID: 28341634 DOI: 10.1152/ajpheart.00107.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/16/2017] [Accepted: 03/22/2017] [Indexed: 01/12/2023]
Abstract
Two-pore K+ (K2p) channels have been described in modulating background conductance as leak channels in different physiological systems. In the heart, the expression of K2p channels is heterogeneous with equivocation regarding their functional role. Our objective was to determine the K2p expression profile and their physiological and pathophysiological contribution to cardiac electrophysiology. Induced pluripotent stem cells (iPSCs) generated from humans were differentiated into cardiomyocytes (iPSC-CMs). mRNA was isolated from these cells, commercial iPSC-CM (iCells), control human heart ventricular tissue (cHVT), and ischemic (iHF) and nonischemic heart failure tissues (niHF). We detected 10 K2p channels in the heart. Comparing quantitative PCR expression of K2p channels between human heart tissue and iPSC-CMs revealed K2p1.1, K2p2.1, K2p5.1, and K2p17.1 to be higher expressed in cHVT, whereas K2p3.1 and K2p13.1 were higher in iPSC-CMs. Notably, K2p17.1 was significantly lower in niHF tissues compared with cHVT. Action potential recordings in iCells after K2p small interfering RNA knockdown revealed prolongations in action potential depolarization at 90% repolarization for K2p2.1, K2p3.1, K2p6.1, and K2p17.1. Here, we report the expression level of 10 human K2p channels in iPSC-CMs and how they compared with cHVT. Importantly, our functional electrophysiological data in human iPSC-CMs revealed a prominent role in cardiac ventricular repolarization for four of these channels. Finally, we also identified K2p17.1 as significantly reduced in niHF tissues and K2p4.1 as reduced in niHF compared with iHF. Thus, we advance the notion that K2p channels are emerging as novel players in cardiac ventricular electrophysiology that could also be remodeled in cardiac pathology and therefore contribute to arrhythmias.NEW & NOTEWORTHY Two-pore K+ (K2p) channels are traditionally regarded as merely background leak channels in myriad physiological systems. Here, we describe the expression profile of K2p channels in human-induced pluripotent stem cell-derived cardiomyocytes and outline a salient role in cardiac repolarization and pathology for multiple K2p channels.
Collapse
Affiliation(s)
- Sam Chai
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio.,Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Xiaoping Wan
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Drew M Nassal
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio.,Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Haiyan Liu
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | | | - Angelina Ramirez-Navarro
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Isabelle Deschênes
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio; .,Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| |
Collapse
|
42
|
Therapeutic targeting of two-pore-domain potassium (K(2P)) channels in the cardiovascular system. Clin Sci (Lond) 2016; 130:643-50. [PMID: 26993052 DOI: 10.1042/cs20150533] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The improvement of treatment strategies in cardiovascular medicine is an ongoing process that requires constant optimization. The ability of a therapeutic intervention to prevent cardiovascular pathology largely depends on its capacity to suppress the underlying mechanisms. Attenuation or reversal of disease-specific pathways has emerged as a promising paradigm, providing a mechanistic rationale for patient-tailored therapy. Two-pore-domain K(+) (K(2P)) channels conduct outward K(+) currents that stabilize the resting membrane potential and facilitate action potential repolarization. K(2P) expression in the cardiovascular system and polymodal K2P current regulation suggest functional significance and potential therapeutic roles of the channels. Recent work has focused primarily on K(2P)1.1 [tandem of pore domains in a weak inwardly rectifying K(+) channel (TWIK)-1], K(2P)2.1 [TWIK-related K(+) channel (TREK)-1], and K(2P)3.1 [TWIK-related acid-sensitive K(+) channel (TASK)-1] channels and their role in heart and vessels. K(2P) currents have been implicated in atrial and ventricular arrhythmogenesis and in setting the vascular tone. Furthermore, the association of genetic alterations in K(2P)3.1 channels with atrial fibrillation, cardiac conduction disorders and pulmonary arterial hypertension demonstrates the relevance of the channels in cardiovascular disease. The function, regulation and clinical significance of cardiovascular K(2P) channels are summarized in the present review, and therapeutic options are emphasized.
Collapse
|
43
|
Wang J, Ma Y, Sachs F, Li J, Suchyna TM. GsMTx4-D is a cardioprotectant against myocardial infarction during ischemia and reperfusion. J Mol Cell Cardiol 2016; 98:83-94. [PMID: 27423272 DOI: 10.1016/j.yjmcc.2016.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/25/2016] [Accepted: 07/12/2016] [Indexed: 01/01/2023]
Abstract
GsMTx4 is a selective inhibitor of cationic mechanosensitive ion channels (MSCs) and has helped establish the role of MSCs in cardiac physiology. Inhomogeneous local mechanical stresses due to hypercontracture and swelling during ischemic reperfusion injury (IRI) likely induce elevated MSC activity that can contribute to cation imbalance. The aim of this study was to determine if the D enantiomer of GsMTx4 can act as a cardioprotectant in a mouse IRI model. Ischemia and reperfusion involved ligating a coronary artery followed by release of the ligature. GsMTx4-D was tested by either acute intravenous injection during the ischemic event or by two day pretreatment by intraperitoneal injection, both methods achieving similar results. Based on pharmacokinetic studies, GsMTx4-D dosage was set to achieve expected plasma concentrations between 50 and 5000nM and heart tissue concentrations between 1 and 200nM by intravenous injection. Relative to vehicle injected animals, GsMTx4-D reduced infarct area by ~40% for acute and pretreated animals for both 20 and 45min ischemic challenges. Many indicators of cardiac output were indistinguishable from sham-treated control hearts after GsMTx4-D treatment showing improvement at both 4 and 48h post ischemia, and premature ventricular beats immediately following reperfusion were also significantly reduced. To determine if GsMTx4-D cardioprotection could act directly at the level of cardiomyocytes, we tested its effects in vitro on indicators of IRI damage like cation influx and activation of inflammatory kinases in isolated myocytes cultured under hypoxic conditions. Hypoxia challenged cardiomyocytes treated with 10μM GsMTx4-D showed improved contractility and near normal contraction-related Ca(2+) influx. GsMTx4-D inhibited indicators of ischemic damage such as the apoptotic signaling system JNK/c-Jun, but also inhibited the energy response signaling system Akt kinase. We conclude that GsMTx4-D is a potent cardioprotectant in vivo that may act directly on cardiomyocytes and potentially be useful in multidrug strategies to treat IRI.
Collapse
Affiliation(s)
- Jinli Wang
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Yina Ma
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Frederick Sachs
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Ji Li
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Thomas M Suchyna
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14214, United States.
| |
Collapse
|
44
|
Abstract
Approximately 80 genes in the human genome code for pore-forming subunits of potassium (K(+)) channels. Rare variants (mutations) in K(+) channel-encoding genes may cause heritable arrhythmia syndromes. Not all rare variants in K(+) channel-encoding genes are necessarily disease-causing mutations. Common variants in K(+) channel-encoding genes are increasingly recognized as modifiers of phenotype in heritable arrhythmia syndromes and in the general population. Although difficult, distinguishing pathogenic variants from benign variants is of utmost importance to avoid false designations of genetic variants as disease-causing mutations.
Collapse
Affiliation(s)
- Ahmad S Amin
- Department of Clinical and Experimental Cardiology, Heart Centre, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Arthur A M Wilde
- Department of Clinical and Experimental Cardiology, Heart Centre, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands; King Abdulaziz University, Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders, PO Box 80200, Jeddah 21589, Kingdom of Saudi Arabia.
| |
Collapse
|
45
|
Schindler RFR, Scotton C, Zhang J, Passarelli C, Ortiz-Bonnin B, Simrick S, Schwerte T, Poon KL, Fang M, Rinné S, Froese A, Nikolaev VO, Grunert C, Müller T, Tasca G, Sarathchandra P, Drago F, Dallapiccola B, Rapezzi C, Arbustini E, Di Raimo FR, Neri M, Selvatici R, Gualandi F, Fattori F, Pietrangelo A, Li W, Jiang H, Xu X, Bertini E, Decher N, Wang J, Brand T, Ferlini A. POPDC1(S201F) causes muscular dystrophy and arrhythmia by affecting protein trafficking. J Clin Invest 2015; 126:239-53. [PMID: 26642364 DOI: 10.1172/jci79562] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 10/29/2015] [Indexed: 01/16/2023] Open
Abstract
The Popeye domain-containing 1 (POPDC1) gene encodes a plasma membrane-localized cAMP-binding protein that is abundantly expressed in striated muscle. In animal models, POPDC1 is an essential regulator of structure and function of cardiac and skeletal muscle; however, POPDC1 mutations have not been associated with human cardiac and muscular diseases. Here, we have described a homozygous missense variant (c.602C>T, p.S201F) in POPDC1, identified by whole-exome sequencing, in a family of 4 with cardiac arrhythmia and limb-girdle muscular dystrophy (LGMD). This allele was absent in known databases and segregated with the pathological phenotype in this family. We did not find the allele in a further screen of 104 patients with a similar phenotype, suggesting this mutation to be family specific. Compared with WT protein, POPDC1(S201F) displayed a 50% reduction in cAMP affinity, and in skeletal muscle from patients, both POPDC1(S201F) and WT POPDC2 displayed impaired membrane trafficking. Forced expression of POPDC1(S201F) in a murine cardiac muscle cell line (HL-1) increased hyperpolarization and upstroke velocity of the action potential. In zebrafish, expression of the homologous mutation (popdc1(S191F)) caused heart and skeletal muscle phenotypes that resembled those observed in patients. Our study therefore identifies POPDC1 as a disease gene causing a very rare autosomal recessive cardiac arrhythmia and LGMD, expanding the genetic causes of this heterogeneous group of inherited rare diseases.
Collapse
|
46
|
Ishikawa T, Tsuji Y, Makita N. Inherited bradyarrhythmia: A diverse genetic background. J Arrhythm 2015; 32:352-358. [PMID: 27761158 PMCID: PMC5063261 DOI: 10.1016/j.joa.2015.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/03/2015] [Accepted: 09/16/2015] [Indexed: 12/31/2022] Open
Abstract
Bradyarrhythmia is a common heart rhythm abnormality comprising number of diseases and is associated with decreased heart rate due to the failure of action potential generation and propagation at the sinus node. Permanent pacemaker implantation is often used therapeutically to compensate for decreased heart rate and cardiac output. The vast majority of bradyarrhythmia cases are attributable either to aging or to structural abnormalities of the cardiac conduction system, caused by underlying structural heart disease. However, there is a subset of bradyarrhythmia primarily caused by genetic defects in the absence of aging or underlying structural heart disease. These include several genes that play principal roles in cardiac electrophysiology, heart development, cardioprotection, and the structural integrity of the membrane and sarcomere. Recent advances in the functional analysis of mutations using a heterologous expression system and genetically engineered animal models have provided significant insights into the underlying molecular mechanisms responsible for inherited arrhythmia. In this review, current understandings of the genetic and molecular basis of inherited bradyarrhythmia are presented.
Collapse
Affiliation(s)
- Taisuke Ishikawa
- Department of Molecular Physiology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yukiomi Tsuji
- Department of Molecular Physiology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Naomasa Makita
- Department of Molecular Physiology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Progressive cardiac conduction disorder (PCCD) is an inherited cardiac disease that may present as a primary electrical disease or be associated with structural heart disease. In this brief review, we present recent clinical, genetic, and molecular findings relating to PCCD. RECENT FINDINGS Inherited PCCD in structurally normal hearts has been found to be linked to genetic variants in the ion channel genes SCN5A, SCN1B, SCN10A, TRPM4, and KCNK17, as well as in genes coding for cardiac connexin proteins. In addition, several SCN5A mutations lead to 'cardiac sodium channelopathy overlap syndrome'. Other genes coding for cardiac transcription factors, such as NKX2.5 and TBX5, are involved in the development of the cardiac conduction system and in the morphogenesis of the heart. Mutations in these two genes have been shown to cause cardiac conduction disorders associated with various congenital heart defects. SUMMARY PCCD is a hereditary syndrome, and genetic variants in multiple genes have been described to date. Genetic screening and identification of the causal mutation are crucial for risk stratification and family counselling.
Collapse
|
48
|
Rinné S, Kiper AK, Schlichthörl G, Dittmann S, Netter MF, Limberg SH, Silbernagel N, Zuzarte M, Moosdorf R, Wulf H, Schulze-Bahr E, Rolfes C, Decher N. TASK-1 and TASK-3 may form heterodimers in human atrial cardiomyocytes. J Mol Cell Cardiol 2015; 81:71-80. [DOI: 10.1016/j.yjmcc.2015.01.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 12/30/2014] [Accepted: 01/27/2015] [Indexed: 11/29/2022]
|
49
|
Renigunta V, Schlichthörl G, Daut J. Much more than a leak: structure and function of K₂p-channels. Pflugers Arch 2015; 467:867-94. [PMID: 25791628 DOI: 10.1007/s00424-015-1703-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 03/09/2015] [Indexed: 11/27/2022]
Abstract
Over the last decade, we have seen an enormous increase in the number of experimental studies on two-pore-domain potassium channels (K2P-channels). The collection of reviews and original articles compiled for this special issue of Pflügers Archiv aims to give an up-to-date summary of what is known about the physiology and pathophysiology of K2P-channels. This introductory overview briefly describes the structure of K2P-channels and their function in different organs. Its main aim is to provide some background information for the 19 reviews and original articles of this special issue of Pflügers Archiv. It is not intended to be a comprehensive review; instead, this introductory overview focuses on some unresolved questions and controversial issues, such as: Do K2P-channels display voltage-dependent gating? Do K2P-channels contribute to the generation of action potentials? What is the functional role of alternative translation initiation? Do K2P-channels have one or two or more gates? We come to the conclusion that we are just beginning to understand the extremely complex regulation of these fascinating channels, which are often inadequately described as 'leak channels'.
Collapse
Affiliation(s)
- Vijay Renigunta
- Institute of Physiology and Pathophysiology, Marburg University, 35037, Marburg, Germany
| | | | | |
Collapse
|
50
|
Feliciangeli S, Chatelain FC, Bichet D, Lesage F. The family of K2P channels: salient structural and functional properties. J Physiol 2015; 593:2587-603. [PMID: 25530075 DOI: 10.1113/jphysiol.2014.287268] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/10/2014] [Indexed: 12/11/2022] Open
Abstract
Potassium channels participate in many biological functions, from ion homeostasis to generation and modulation of the electrical membrane potential. They are involved in a large variety of diseases. In the human genome, 15 genes code for K(+) channels with two pore domains (K2P ). These channels form dimers of pore-forming subunits that produce background conductances finely regulated by a range of natural and chemical effectors, including signalling lipids, temperature, pressure, pH, antidepressants and volatile anaesthetics. Since the cloning of TWIK1, the prototypical member of this family, a lot of work has been carried out on their structure and biology. These studies are still in progress, but data gathered so far show that K2P channels are central players in many processes, including ion homeostasis, hormone secretion, cell development and excitability. A growing number of studies underline their implication in physiopathological mechanisms, such as vascular and pulmonary hypertension, cardiac arrhythmias, nociception, neuroprotection and depression. This review gives a synthetic view of the most noticeable features of these channels.
Collapse
Affiliation(s)
- Sylvain Feliciangeli
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| | - Frank C Chatelain
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| | - Delphine Bichet
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| | - Florian Lesage
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| |
Collapse
|