1
|
Jansen LAR, Welch MA, Plant LD, Baro DJ. Crosstalk between PKA and PIAS3 regulates cardiac Kv4 channel SUMOylation. Cell Commun Signal 2024; 22:422. [PMID: 39223673 PMCID: PMC11367828 DOI: 10.1186/s12964-024-01795-4] [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: 03/28/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Post-translational SUMOylation of nuclear and cytosolic proteins maintains homeostasis in eukaryotic cells and orchestrates programmed responses to changes in metabolic demand or extracellular stimuli. In excitable cells, SUMOylation tunes the biophysical properties and trafficking of ion channels. Ion channel SUMOylation status is determined by the opposing enzyme activities of SUMO ligases and deconjugases. Phosphorylation also plays a permissive role in SUMOylation. SUMO deconjugases have been identified for several ion channels, but their corresponding E3 ligases remain unknown. This study shows PIAS3, a.k.a. KChAP, is a bona fide SUMO E3 ligase for Kv4.2 and HCN2 channels in HEK cells, and endogenous Kv4.2 and Kv4.3 channels in cardiomyocytes. PIAS3-mediated SUMOylation at Kv4.2-K579 increases channel surface expression through a rab11a-dependent recycling mechanism. PKA phosphorylation at Kv4.2-S552 reduces the current mediated by Kv4 channels in HEK293 cells, cardiomyocytes, and neurons. This study shows PKA mediated phosphorylation blocks Kv4.2-K579 SUMOylation in HEK cells and cardiomyocytes. Together, these data identify PIAS3 as a key downstream mediator in signaling cascades that control ion channel surface expression.
Collapse
Affiliation(s)
| | - Meghyn A Welch
- Department of Biology, Georgia State University, Atlanta, GA, USA
- Present Address: Section on Molecular Neurophysiology and Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Leigh D Plant
- Department of Pharmaceutical Sciences, Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Deborah J Baro
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Kodirov SA. Adam, amigo, brain, and K channel. Biophys Rev 2023; 15:1393-1424. [PMID: 37975011 PMCID: PMC10643815 DOI: 10.1007/s12551-023-01163-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023] Open
Abstract
Voltage-dependent K+ (Kv) channels are diverse, comprising the classical Shab - Kv2, Shaker - Kv1, Shal - Kv4, and Shaw - Kv3 families. The Shaker family alone consists of Kv1.1, Kv1.2, Kv1.3, Kv1.4, Kv1.5, Kv1.6, and Kv1.7. Moreover, the Shab family comprises two functional (Kv2.1 and Kv2.2) and several "silent" alpha subunits (Kv2.3, Kv5, Kv6, Kv8, and Kv9), which do not generate K current. However, e.g., Kv8.1, via heteromerization, inhibits outward currents of the same family or even that of Shaw. This property of Kv8.1 is similar to those of designated beta subunits or non-selective auxiliary elements, including ADAM or AMIGO proteins. Kv channels and, in turn, ADAM may modulate the synaptic long-term potentiation (LTP). Prevailingly, Kv1.1 and Kv1.5 are attributed to respective brain and heart pathologies, some of which may occur simultaneously. The aforementioned channel proteins are apparently involved in several brain pathologies, including schizophrenia and seizures.
Collapse
Affiliation(s)
- Sodikdjon A. Kodirov
- Department of Biological Sciences, University of Texas at Brownsville, Brownsville, TX 78520 USA
- Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia
- Instituto de Medicina Molecular, Universidade de Lisboa, 1649-028 Lisbon, Portugal
- Almazov Federal Heart, Blood and Endocrinology Centre, Saint Petersburg, 197341 Russia
- Institute for Physiology and Pathophysiology, Johannes Kepler University, Linz, Austria
| |
Collapse
|
3
|
Manickam R, Virzi J, Potti A, Cheng F, Russ DW, Tipparaju SM. Genetic deletion of Kvβ2 (AKR6) causes loss of muscle function and increased inflammation in mice. FRONTIERS IN AGING 2023; 4:1175510. [PMID: 37377453 PMCID: PMC10292803 DOI: 10.3389/fragi.2023.1175510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023]
Abstract
The voltage-gated potassium channels (Kv) are complex ion channels with distinct roles in neurotransmission, electrical conductivity of the heart, and smooth and striated muscle functions. Previously, we demonstrated that deletion of Kvβ2 in mice results in decreased Pax7 protein levels, hindlimb muscles and body weights, and fiber type switching. In the present study, we tested the hypothesis that Kvβ2 regulates skeletal muscle function in mice. The young and old Kvβ2 knockout (KO) and wildtype (WT) mice were utilized to test the aging phenotype and skeletal muscle function. Consistent with our previous finding, we found a significant reduction in hindlimb skeletal muscles mass and body weight in young Kvβ2 KO mice, which was also significantly reduced in old Kvβ2 KO mice compared with age-matched WT mice. Forelimb grip strength, and the hindleg extensor digitorum longus (EDL) muscles force-frequency relations were significantly decreased in young and old Kvβ2 KO mice compared to age-matched WT mice. Analysis of transmission electron microscopy images of EDL muscles in young mice revealed a significant reduction in the sarcomere length for Kvβ2 KO vs. WT. Hematoxylin and eosin-stained tibialis anterior muscles cryosections displayed a significant decrease in the number of medium (2,000-4,000 µm2) and largest (>4,000 µm2) myofibers area in young Kvβ2 KO vs. WT mice. We also found a significant increase in fibrotic tissue area in young Kvβ2 KO mice compared with age-matched WT mice. Analysis of RNA Seq data of the gastrocnemius muscles (GAS) identified significant increase in genes involved in skeletal muscle development, proliferation and cell fate determination, atrophy, energy metabolism, muscle plasticity, inflammation, and a decrease in circadian core clock genes in young Kvβ2 KO vs. WT mice. Several genes were significantly upregulated (384 genes) and downregulated (40 genes) in young Kvβ2 KO mice compared to age-matched WT mice. Further, RT-qPCR analysis of the GAS muscles displayed a significant increase in pro-inflammatory marker Il6 expression in young Kvβ2 KO mice compared to age-matched WT mice. Overall, the present study shows that deletion of Kvβ2 leads to decreased muscles strength and increased inflammation.
Collapse
Affiliation(s)
- Ravikumar Manickam
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Jazmine Virzi
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Anish Potti
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Feng Cheng
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - David W. Russ
- School of Physical Therapy and Rehabilitation Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Srinivas M. Tipparaju
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| |
Collapse
|
4
|
Averin AS, Konakov MV, Pimenov OY, Galimova MH, Berezhnov AV, Nenov MN, Dynnik VV. Regulation of Papillary Muscle Contractility by NAD and Ammonia Interplay: Contribution of Ion Channels and Exchangers. MEMBRANES 2022; 12:1239. [PMID: 36557146 PMCID: PMC9785361 DOI: 10.3390/membranes12121239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/04/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
Various models, including stem cells derived and isolated cardiomyocytes with overexpressed channels, are utilized to analyze the functional interplay of diverse ion currents involved in cardiac automaticity and excitation-contraction coupling control. Here, we used β-NAD and ammonia, known hyperpolarizing and depolarizing agents, respectively, and applied inhibitory analysis to reveal the interplay of several ion channels implicated in rat papillary muscle contractility control. We demonstrated that: 4 mM β-NAD, having no strong impact on resting membrane potential (RMP) and action potential duration (APD90) of ventricular cardiomyocytes, evoked significant suppression of isometric force (F) of paced papillary muscle. Reactive blue 2 restored F to control values, suggesting the involvement of P2Y-receptor-dependent signaling in β-NAD effects. Meantime, 5 mM NH4Cl did not show any effect on F of papillary muscle but resulted in significant RMP depolarization, APD90 shortening, and a rightward shift of I-V relationship for total steady state currents in cardiomyocytes. Paradoxically, NH4Cl, being added after β-NAD and having no effect on RMP, APD, and I-V curve, recovered F to the control values, indicating β-NAD/ammonia antagonism. Blocking of HCN, Kir2.x, and L-type calcium channels, Ca2+-activated K+ channels (SK, IK, and BK), or NCX exchanger reverse mode prevented this effect, indicating consistent cooperation of all currents mediated by these channels and NCX. We suggest that the activation of Kir2.x and HCN channels by extracellular K+, that creates positive and negative feedback, and known ammonia and K+ resemblance, may provide conditions required for the activation of all the chain of channels involved in the interplay. Here, we present a mechanistic model describing an interplay of channels and second messengers, which may explain discovered antagonism of β-NAD and ammonia on rat papillary muscle contractile activity.
Collapse
Affiliation(s)
- Alexey S. Averin
- Institute of Theoretical and Experimental Biophysics, the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Maxim V. Konakov
- Institute of Theoretical and Experimental Biophysics, the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Oleg Y. Pimenov
- Institute of Theoretical and Experimental Biophysics, the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Miliausha H. Galimova
- Institute of Theoretical and Experimental Biophysics, the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Alexey V. Berezhnov
- Institute of Cell Biophysics, the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Miroslav N. Nenov
- Institute of Theoretical and Experimental Biophysics, the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Vladimir V. Dynnik
- Institute of Theoretical and Experimental Biophysics, the Russian Academy of Sciences, Pushchino 142290, Russia
| |
Collapse
|
5
|
Xia R, Tomsits P, Loy S, Zhang Z, Pauly V, Schüttler D, Clauss S. Cardiac Macrophages and Their Effects on Arrhythmogenesis. Front Physiol 2022; 13:900094. [PMID: 35812333 PMCID: PMC9257039 DOI: 10.3389/fphys.2022.900094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/30/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiac electrophysiology is a complex system established by a plethora of inward and outward ion currents in cardiomyocytes generating and conducting electrical signals in the heart. However, not only cardiomyocytes but also other cell types can modulate the heart rhythm. Recently, cardiac macrophages were demonstrated as important players in both electrophysiology and arrhythmogenesis. Cardiac macrophages are a heterogeneous group of immune cells including resident macrophages derived from embryonic and fetal precursors and recruited macrophages derived from circulating monocytes from the bone marrow. Recent studies suggest antiarrhythmic as well as proarrhythmic effects of cardiac macrophages. The proposed mechanisms of how cardiac macrophages affect electrophysiology vary and include both direct and indirect interactions with other cardiac cells. In this review, we provide an overview of the different subsets of macrophages in the heart and their possible interactions with cardiomyocytes under both physiologic conditions and heart disease. Furthermore, we elucidate similarities and differences between human, murine and porcine cardiac macrophages, thus providing detailed information for researchers investigating cardiac macrophages in important animal species for electrophysiologic research. Finally, we discuss the pros and cons of mice and pigs to investigate the role of cardiac macrophages in arrhythmogenesis from a translational perspective.
Collapse
Affiliation(s)
- Ruibing Xia
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Philipp Tomsits
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Simone Loy
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Zhihao Zhang
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Valerie Pauly
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Dominik Schüttler
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Sebastian Clauss
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
6
|
Li Y, Duan H, Yi J, Wang G, Cheng W, Feng L, Liu J. Kv4.2 phosphorylation by PKA drives Kv4.2 - KChIP2 dissociation, leading to Kv4.2 out of lipid rafts and internalization. Am J Physiol Cell Physiol 2022; 323:C190-C201. [PMID: 35508186 DOI: 10.1152/ajpcell.00307.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sympathetic regulation of the Kv4.2 transient outward potassium current is critical for the acute electrical and contractile response of the myocardium under physiological and pathological conditions. Previous studies have suggested that KChIP2, the key auxiliary subunit of Kv4 channels, is required for the sympathetic regulation of Kv4.2 current densities. Of interest, Kv4.2 and KChIP2, and key components mediating acute sympathetic signaling transduction are present in lipid rafts, which are profoundly involved in regulation of Ito densities in rat ventricular myocytes. However, little is known about the mechanisms of Kv4.2-raft association and its connection with acute sympathetic regulation. With the aid of high-resolution fluorescent microscope, we demonstrate that KChIP2 assists Kv4.2 localization in lipid rafts in HEK293 cells. Moreover, PKA-mediated Kv4.2 phosphorylation, the downstream signaling event of acute sympathetic stimulation, induced dissociation between Kv4.2 and KChIP2, resulting in Kv4.2 shifting out of lipid rafts in KChIP2-expressed HEK293.The mutation that mimics Kv4.2 phosphorylation by PKA similarly disrupted Kv4.2 interaction with KChIP2 and also decreased the surface stability of Kv4.2. The attenuated Kv4.2-KChIP2 interaction was also observed in native neonatal rat ventricular myocytes (NRVMs) upon acute adrenergic stimulation with phenylephrine (PE). Furthermore, PE accelerated internalization of Kv4.2 in native NRVMs, but disruption of lipid rafts dampens this reaction. In conclusion, KChIP2 contributes to targeting Kv4.2 to lipid rafts. Acute adrenergic stimulation induces Kv4.2 - KChIP2 dissociation, leading to Kv4.2 out of lipid rafts and internalization, reinforcing the critical role of Kv4.2-lipid raft association in the essential physiological response of Ito to acute sympathetic regulation.
Collapse
Affiliation(s)
- Ying Li
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Haixia Duan
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Jing Yi
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Gang Wang
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Wanwen Cheng
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Li Feng
- Department of Cardiology, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Jie Liu
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Changes in ion channel expression and function associated with cardiac arrhythmogenic remodeling by Sorbs2. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166247. [PMID: 34487812 DOI: 10.1016/j.bbadis.2021.166247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022]
Abstract
The Sorbin and SH3 domain-containing protein 2 (Sorbs2) is an important component of cardiomyocyte sarcomere. It has been recently reported that loss of Sorbs2 is causally associated with arrhythmogenic cardiomyopathy in human. However, the ionic mechanisms leading to cardiac arrhythmogenesis by Sorbs2 deficiency are unknown. In this study, we hypothesized that Sorbs2 plays an important role in regulating cardiac ion channel expression and function. Using electrophysiological and molecular biological approaches, we found that the Sorbs2 knockout (KO) mice progressively developed cardiac structural and electrical remodeling as early as 1 to 2 months of age and died prematurely at 5 to 7 months of age. Electrocardiographic recordings showed that Sorbs2 KO mice had conduction delays, spontaneous ventricular extrasystoles and polymorphic ventricular tachyarrhythmia. Intracellular recordings revealed abnormal action potentials with depolarized resting potential, reduced upstroke velocity, prolonged repolarization, and effective refractory period in the ventricular preparations of Sorbs2 KO mice. Patch clamp experiments demonstrated that Sorbs2 KO mice displayed distinct abnormalities in the expression and function of cardiac ion channels, including those of the voltage-gated Na+ channels, L-type Ca2+ channels, the voltage-gated K+ channels and the inward-rectifier K+ channels. Moreover, Sorbs2 physically interacted with the RNAs and/or proteins of important cardiac ion channels and directly regulated their expression in vitro. Our results indicate that Sorbs2 plays a pivotal role in the regulation of cardiac channel physiology. Loss of Sorbs2 promotes cardiac ion channelopathies and life-threatening arrhythmias.
Collapse
|
8
|
Feyen DAM, McKeithan WL, Bruyneel AAN, Spiering S, Hörmann L, Ulmer B, Zhang H, Briganti F, Schweizer M, Hegyi B, Liao Z, Pölönen RP, Ginsburg KS, Lam CK, Serrano R, Wahlquist C, Kreymerman A, Vu M, Amatya PL, Behrens CS, Ranjbarvaziri S, Maas RGC, Greenhaw M, Bernstein D, Wu JC, Bers DM, Eschenhagen T, Metallo CM, Mercola M. Metabolic Maturation Media Improve Physiological Function of Human iPSC-Derived Cardiomyocytes. Cell Rep 2021; 32:107925. [PMID: 32697997 PMCID: PMC7437654 DOI: 10.1016/j.celrep.2020.107925] [Citation(s) in RCA: 202] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/15/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have enormous potential for the study of human cardiac disorders. However, their physiological immaturity severely limits their utility as a model system and their adoption for drug discovery. Here, we describe maturation media designed to provide oxidative substrates adapted to the metabolic needs of human iPSC (hiPSC)-CMs. Compared with conventionally cultured hiPSC-CMs, metabolically matured hiPSC-CMs contract with greater force and show an increased reliance on cardiac sodium (Na+) channels and sarcoplasmic reticulum calcium (Ca2+) cycling. The media enhance the function, long-term survival, and sarcomere structures in engineered heart tissues. Use of the maturation media made it possible to reliably model two genetic cardiac diseases: long QT syndrome type 3 due to a mutation in the cardiac Na+ channel SCN5A and dilated cardiomyopathy due to a mutation in the RNA splicing factor RBM20. The maturation media should increase the fidelity of hiPSC-CMs as disease models. Physiological immaturity of iPSC-derived cardiomyocytes limits their fidelity as disease models. Feyen et al. developed a low glucose, high oxidative substrate media that increase maturation of ventricular-like hiPSC-CMs in 2D and 3D cultures relative to standard protocols. Improved characteristics include a low resting Vm, rapid depolarization, and increased Ca2+ dependence and force generation.
Collapse
Affiliation(s)
- Dries A M Feyen
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wesley L McKeithan
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Arne A N Bruyneel
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sean Spiering
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Larissa Hörmann
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bärbel Ulmer
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hui Zhang
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Francesca Briganti
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michaela Schweizer
- Electron Microscopy Unit, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bence Hegyi
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Zhandi Liao
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | | | - Kenneth S Ginsburg
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Chi Keung Lam
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ricardo Serrano
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Christine Wahlquist
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Alexander Kreymerman
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michelle Vu
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Prashila L Amatya
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Charlotta S Behrens
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sara Ranjbarvaziri
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Renee G C Maas
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Matthew Greenhaw
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Daniel Bernstein
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Joseph C Wu
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Mark Mercola
- Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
9
|
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: 93] [Impact Index Per Article: 23.3] [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
|
10
|
Abstract
Voltage-gated Kv1.1 potassium channel α-subunits are broadly expressed in the nervous system where they act as critical regulators of neuronal excitability. Mutations in the KCNA1 gene, which encodes Kv1.1, are associated with the neurological diseases episodic ataxia and epilepsy. Studies in mouse models have shown that Kv1.1 is important for neural control of the heart and that Kcna1 deletion leads to cardiac dysfunction that appears to be brain-driven. Traditionally, KCNA1 was not believed to be expressed in the heart. However, recent studies have revealed that Kv1.1 subunits are not only present in cardiomyocytes, but that they also make an important heart-intrinsic functional contribution to outward K+ currents and action potential repolarization. This review recounts the winding history of discovery of KCNA1 gene expression and neurocardiac function from fruit flies to mammals and from brain to heart and looks at some of the salient questions that remain to be answered regarding emerging cardiac roles of Kv1.1.
Collapse
Affiliation(s)
- Edward Glasscock
- a Department of Biological Sciences , Southern Methodist University , Dallas , TX , USA
| |
Collapse
|
11
|
Akyuz E, Villa C. A novel role of cardiac inwardly rectifying potassium channels explaining autonomic cardiovascular dysfunctions in a cuprizone-induced mouse model of multiple sclerosis. Auton Neurosci 2020; 225:102647. [PMID: 32087534 DOI: 10.1016/j.autneu.2020.102647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS), believed to have an autoimmune etiology. MS patients showed an increased cardiovascular (CV) risk probably related to an impairment in the autonomic control of CV functions, but the underlying molecular mechanisms are not completely elucidated. Inwardly-rectifying potassium (Kir) channels play a key role in cardiac excitability by contributing to the repolarization phase of action potential and were recently identified as target of the autoantibody response in MS patients. Therefore, we investigated the role of cardiac Kir channels in the CV dysfunctions occurring in MS. Cardiac functions were evaluated by electrocardiographic recordings (ECG) in cuprizone-fed C57BL/6 mice, a classic demyelination animal model. Gene expression profiling of cardiac Kir2.2, Kir4.1 and Kir6.2 channels was performed using real-time PCR in mice. Cuprizone-induced mouse model was confirmed by immunohistochemistry analysis showing demyelination in the corpus callosum. ECG recordings from mice showed a significant decreased duration of the P wave and RR interval as well as an increase of the heart rate in cuprizone-treated mice as compared with the controls. Significant increased relative expression levels of Kcnj11 and Kcnj12, encoding for Kir6.2 and Kir2.2 channels respectively, were observed in mouse heart tissue, whereas no differences were found in mRNA levels of Kir4.1 channel as compared with controls. For the first time, these findings provided valuable insights into the potential role of Kir channels in cardiac problems associated with MS.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey.
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
| |
Collapse
|
12
|
Zhang H, Zhang H, Wang C, Wang Y, Zou R, Shi C, Guan B, Gamper N, Xu Y. Auxiliary subunits control biophysical properties and response to compound NS5806 of the Kv4 potassium channel complex. FASEB J 2019; 34:807-821. [PMID: 31914636 PMCID: PMC6972550 DOI: 10.1096/fj.201902010rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 12/19/2022]
Abstract
Kv4 pore‐forming subunits co‐assemble with β‐subunits including KChIP2 and DPP6 and the resultant complexes conduct cardiac transient outward K+ current (Ito). Compound NS5806 has been shown to potentate Ito in canine cardiomyocytes; however, its effects on Ito in other species yet to be determined. We found that NS5806 inhibited native Ito in a concentration‐dependent manner (0.1~30 μM) in both mouse ventricular cardiomyocytes and human‐induced pluripotent stem cell‐derived cardiomyocytes (hiPSC‐CMs), but potentiated Ito in the canine cardiomyocytes. In HEK293 cells co‐transfected with cloned Kv4.3 (or Kv4.2) and β‐subunit KChIP2, NS5806 significantly increased the peak current amplitude and slowed the inactivation. In contrast, NS5806 suppressed the current and accelerated inactivation of the channels when cells were co‐transfected with Kv4.3 (or Kv4.2), KChIP2 and another β‐subunit, DPP6‐L (long isoform). Western blot analysis showed that DPP6‐L was dominantly expressed in both mouse ventricular myocardium and hiPSC‐CMs, while it was almost undetectable in canine ventricular myocardium. In addition, low level of DPP6‐S expression was found in canine heart, whereas levels of KChIP2 expression were comparable among all three species. siRNA knockdown of DPP6 antagonized the Ito inhibition by NS5806 in hiPSC‐CMs. Molecular docking simulation suggested that DPP6‐L may associate with KChIP2 subunits. Mutations of putative KChIP2‐interacting residues of DPP6‐L reversed the inhibitory effect of NS5806 into potentiation of the current. We conclude that a pharmacological modulator can elicit opposite regulatory effects on Kv4 channel complex among different species, depending on the presence of distinct β‐subunits. These findings provide novel insight into the molecular design and regulation of cardiac Ito. Since Ito is a potential therapeutic target for treatment of multiple cardiovascular diseases, our data will facilitate the development of new therapeutic Ito modulators.
Collapse
Affiliation(s)
- Hongxue Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Hua Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Chanjuan Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Yuhong Wang
- Institute of Masteria Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ruya Zou
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Chenxia Shi
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Bingcai Guan
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| |
Collapse
|
13
|
Metabolic regulation of Kv channels and cardiac repolarization by Kvβ2 subunits. J Mol Cell Cardiol 2019; 137:93-106. [PMID: 31639389 DOI: 10.1016/j.yjmcc.2019.09.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 09/24/2019] [Accepted: 09/28/2019] [Indexed: 11/21/2022]
Abstract
Voltage-gated potassium (Kv) channels control myocardial repolarization. Pore-forming Kvα proteins associate with intracellular Kvβ subunits, which bind pyridine nucleotides with high affinity and differentially regulate channel trafficking, plasmalemmal localization and gating properties. Nevertheless, it is unclear how Kvβ subunits regulate myocardial K+ currents and repolarization. Here, we tested the hypothesis that Kvβ2 subunits regulate the expression of myocardial Kv channels and confer redox sensitivity to Kv current and cardiac repolarization. Co-immunoprecipitation and in situ proximity ligation showed that in cardiac myocytes, Kvβ2 interacts with Kv1.4, Kv1.5, Kv4.2, and Kv4.3. Cardiac myocytes from mice lacking Kcnab2 (Kvβ2-/-) had smaller cross sectional areas, reduced sarcolemmal abundance of Kvα binding partners, reduced Ito, IK,slow1, and IK,slow2 densities, and prolonged action potential duration compared with myocytes from wild type mice. These differences in Kvβ2-/- mice were associated with greater P wave duration and QT interval in electrocardiograms, and lower ejection fraction, fractional shortening, and left ventricular mass in echocardiographic and morphological assessments. Direct intracellular dialysis with a high NAD(P)H:NAD(P)+ accelerated Kv inactivation in wild type, but not Kvβ2-/- myocytes. Furthermore, elevated extracellular levels of lactate increased [NADH]i and prolonged action potential duration in wild type cardiac myocytes and perfused wild type, but not Kvβ2-/-, hearts. Taken together, these results suggest that Kvβ2 regulates myocardial electrical activity by supporting the functional expression of proteins that generate Ito and IK,slow, and imparting redox and metabolic sensitivity to Kv channels, thereby coupling cardiac repolarization to myocyte metabolism.
Collapse
|
14
|
Kodirov SA, Zhuravlev VL, Brachmann J. Prevailing Effects of Ibutilide on Fast Delayed Rectifier K + Channel. J Membr Biol 2019; 252:609-616. [PMID: 31584122 DOI: 10.1007/s00232-019-00098-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 07/29/2019] [Indexed: 11/27/2022]
Abstract
Effects of ibutilide, a class III antiarrhythmic drug, on delayed rectifier potassium currents (IK) in freshly isolated guinea pig ventricular myocytes were studied. Experiments were performed using the whole-cell configuration of patch-clamp technique under blockade of L-type calcium currents (Cav1). Ibutilide at concentrations ranging between 10 nM and 100 µM inhibited IKr in dose-dependent manner with a half maximal effective concentration of 2.03 ± 0.74 µM (n = 5-10). The amplitude of tail currents activated by prepulse to + 20 mV was decreased from 253 ± 52 to 130 ± 25 pA (n = 8, p < 0.01) in the presence of 1 µM ibutilide. The envelope test revealed time-dependent changes in ratio of IK-tail/ΔIK during 0.2-2 s pulse durations in the absence of drug. With ibutilide, regardless of pulse duration, a relatively constant ratio was estimated, indicative of predominant involvement of IKr component. The slow IKs persisted to greater extent even at 100 μM ibutilide revealing a distinguishable selectivity toward the IKr component.
Collapse
Affiliation(s)
- Sodikdjon A Kodirov
- Department of Cardiology, University Hospital Heidelberg, 69120, Heidelberg, Germany. .,Department of Physiology, Saint Petersburg University, Saint Petersburg, Russia, 199034. .,Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteura St., 02-093, Warsaw, Poland. .,Department of Cardiology, Klinikum Coburg, Teaching Hospital of the University of Würzburg, Coburg, Germany.
| | - Vladimir L Zhuravlev
- Department of Cardiology, University Hospital Heidelberg, 69120, Heidelberg, Germany.,Department of Physiology, Saint Petersburg University, Saint Petersburg, Russia, 199034
| | - Johannes Brachmann
- Department of Cardiology, University Hospital Heidelberg, 69120, Heidelberg, Germany.,Department of Cardiology, Klinikum Coburg, Teaching Hospital of the University of Würzburg, Coburg, Germany
| |
Collapse
|
15
|
Zhang Q, Lyu W, Yu M, Niu Y, Meng Z. Investigating the inotropic effect of pyruvic acid on the isolated rat heart and its underlying mechanism. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 71:103206. [PMID: 31212135 DOI: 10.1016/j.etap.2019.103206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 05/09/2019] [Accepted: 06/06/2019] [Indexed: 06/09/2023]
Abstract
Pyruvic acid is important organic chemical intermediates that plays a role in cardiomyocyte pathophysiology and therapy. This study sought to explore the inotropic effects of pyruvic acid on the function of the isolated rat hearts and investigate its underlying mechanism. Pyruvic acid produced a greater negative inotropic effect compared to HCl and sodium pyruvate in a concentration-dependent pattern in the hearts. The role of low dose of pyruvic acid on heart function was regulated by pyruvic acid molecules and high dose pyruvic acid may be influenced by pyruvic acid molecules and pH. Kv channels may be involved in the pyruvic acid-induced negative inotropic effect. Finally, pyruvic acid markedly increased the level of LDH and CK and reduced the level of Ca2+Mg2+-ATPase and Na+K+-ATPase. These results suggest that pyruvic acid may modulate cardiac function at physiological or low doses but can cause damage to cardiomyocytes at high doses.
Collapse
Affiliation(s)
- Quanxi Zhang
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China.
| | - Wenru Lyu
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Mengbin Yu
- Institute of NBC Defence, Beijing 102205, China
| | - Yuxin Niu
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Ziqiang Meng
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
16
|
Autoimmune and inflammatory K+ channelopathies in cardiac arrhythmias: Clinical evidence and molecular mechanisms. Heart Rhythm 2019; 16:1273-1280. [DOI: 10.1016/j.hrthm.2019.02.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Indexed: 12/30/2022]
|
17
|
Kodirov SA. Tale of tail current. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 150:78-97. [PMID: 31238048 DOI: 10.1016/j.pbiomolbio.2019.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/22/2019] [Accepted: 06/20/2019] [Indexed: 02/07/2023]
Abstract
The largest biomass of channel proteins is located in unicellular organisms and bacteria that have no organs. However, orchestrated bidirectional ionic currents across the cell membrane via the channels are important for the functioning of organs of organisms, and equally concern both fauna or flora. Several ion channels are activated in the course of action potentials. One of the hallmarks of voltage-dependent channels is a 'tail current' - deactivation as observed after prior and sufficient activation predominantly at more depolarized potentials e.g. for Kv while upon hyperpolarization for HCN α subunits. Tail current also reflects the timing of channel closure that is initiated upon termination of stimuli. Finally, deactivation of currents during repolarization could be a selective estimate for given channel as in case of HERG, if dedicated long and more depolarized 'tail pulse' is used. Since from a holding potential of e.g. -70 mV are often a family of outward K+ currents comprising IA and IK are simultaneously activated in native cells.
Collapse
Affiliation(s)
- Sodikdjon A Kodirov
- Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, Russia; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Almazov Federal Heart, Blood and Endocrinology Centre, Saint Petersburg, 197341, Russia; Institute of Experimental Medicine, I. P. Pavlov Department of Physiology, Russian Academy of Medical Sciences, Saint Petersburg, Russia; Laboratory of Emotions' Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, 02-093, Poland.
| |
Collapse
|
18
|
Raph SM, Bhatnagar A, Nystoriak MA. Biochemical and physiological properties of K + channel-associated AKR6A (Kvβ) proteins. Chem Biol Interact 2019; 305:21-27. [PMID: 30926318 DOI: 10.1016/j.cbi.2019.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/14/2019] [Accepted: 03/25/2019] [Indexed: 11/16/2022]
Abstract
Voltage-gated potassium (Kv) channels play an essential role in the regulation of membrane excitability and thereby control physiological processes such as cardiac excitability, neural communication, muscle contraction, and hormone secretion. Members of the Kv1 and Kv4 families are known to associate with auxiliary intracellular Kvβ subunits, which belong to the aldo-keto reductase superfamily. Electrophysiological studies have shown that these proteins regulate the gating properties of Kv channels. Although the three gene products encoding Kvβ proteins are functional enzymes in that they catalyze the nicotinamide adenine dinucleotide phosphate (NAD[P]H)-dependent reduction of a wide range of aldehyde and ketone substrates, the physiological role for these proteins and how each subtype may perform unique roles in coupling membrane excitability with cellular metabolic processes remains unclear. Here, we discuss current knowledge of the enzymatic properties of Kvβ proteins from biochemical studies with their described and purported physiological and pathophysiological influences.
Collapse
Affiliation(s)
- Sean M Raph
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Aruni Bhatnagar
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, KY, 40202, USA
| | - Matthew A Nystoriak
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
19
|
Si M, Trosclair K, Hamilton KA, Glasscock E. Genetic ablation or pharmacological inhibition of Kv1.1 potassium channel subunits impairs atrial repolarization in mice. Am J Physiol Cell Physiol 2019; 316:C154-C161. [PMID: 30427720 PMCID: PMC6397341 DOI: 10.1152/ajpcell.00335.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 12/14/2022]
Abstract
Voltage-gated Kv1.1 potassium channel α-subunits, encoded by the Kcna1 gene, have traditionally been regarded as neural-specific with no expression or function in the heart. However, recent data revealed that Kv1.1 subunits are expressed in atria where they may have an overlooked role in controlling repolarization and arrhythmia susceptibility independent of the nervous system. To explore this concept in more detail and to identify functional and molecular effects of Kv1.1 channel impairment in the heart, atrial cardiomyocyte patch-clamp electrophysiology and gene expression analyses were performed using Kcna1 knockout ( Kcna1-/-) mice. Specifically, we hypothesized that Kv1.1 subunits contribute to outward repolarizing K+ currents in mouse atria and that their absence prolongs cardiac action potentials. In voltage-clamp experiments, dendrotoxin-K (DTX-K), a Kv1.1-specific inhibitor, significantly reduced peak outward K+ currents in wild-type (WT) atrial cells but not Kcna1-/- cells, demonstrating an important contribution by Kv1.1-containing channels to mouse atrial repolarizing currents. In current-clamp recordings, Kcna1-/- atrial myocytes exhibited significant action potential prolongation which was exacerbated in right atria, effects that were partially recapitulated in WT cells by application of DTX-K. Quantitative RT-PCR measurements showed mRNA expression remodeling in Kcna1-/- atria for several ion channel genes that contribute to the atrial action potential including the Kcna5, Kcnh2, and Kcnj2 potassium channel genes and the Scn5a sodium channel gene. This study demonstrates a previously undescribed heart-intrinsic role for Kv1.1 subunits in mediating atrial repolarization, thereby adding a new member to the already diverse collection of known K+ channels in the heart.
Collapse
Affiliation(s)
- Man Si
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center , Shreveport, Louisiana
| | - Krystle Trosclair
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center , Shreveport, Louisiana
| | - Kathryn A Hamilton
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center , Shreveport, Louisiana
| | - Edward Glasscock
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center , Shreveport, Louisiana
| |
Collapse
|
20
|
Morrison JL, Botting KJ, Darby JRT, David AL, Dyson RM, Gatford KL, Gray C, Herrera EA, Hirst JJ, Kim B, Kind KL, Krause BJ, Matthews SG, Palliser HK, Regnault TRH, Richardson BS, Sasaki A, Thompson LP, Berry MJ. Guinea pig models for translation of the developmental origins of health and disease hypothesis into the clinic. J Physiol 2018; 596:5535-5569. [PMID: 29633280 PMCID: PMC6265540 DOI: 10.1113/jp274948] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/19/2018] [Indexed: 12/12/2022] Open
Abstract
Over 30 years ago Professor David Barker first proposed the theory that events in early life could explain an individual's risk of non-communicable disease in later life: the developmental origins of health and disease (DOHaD) hypothesis. During the 1990s the validity of the DOHaD hypothesis was extensively tested in a number of human populations and the mechanisms underpinning it characterised in a range of experimental animal models. Over the past decade, researchers have sought to use this mechanistic understanding of DOHaD to develop therapeutic interventions during pregnancy and early life to improve adult health. A variety of animal models have been used to develop and evaluate interventions, each with strengths and limitations. It is becoming apparent that effective translational research requires that the animal paradigm selected mirrors the tempo of human fetal growth and development as closely as possible so that the effect of a perinatal insult and/or therapeutic intervention can be fully assessed. The guinea pig is one such animal model that over the past two decades has demonstrated itself to be a very useful platform for these important reproductive studies. This review highlights similarities in the in utero development between humans and guinea pigs, the strengths and limitations of the guinea pig as an experimental model of DOHaD and the guinea pig's potential to enhance clinical therapeutic innovation to improve human health.
Collapse
Affiliation(s)
- Janna L. Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health ResearchUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Kimberley J. Botting
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health ResearchUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Anna L. David
- Research Department of Maternal Fetal Medicine, Institute for Women's HealthUniversity College LondonLondonUK
| | - Rebecca M. Dyson
- Department of Paediatrics & Child Health and Centre for Translational PhysiologyUniversity of OtagoWellingtonNew Zealand
| | - Kathryn L. Gatford
- Robinson Research Institute and Adelaide Medical SchoolUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Clint Gray
- Department of Paediatrics & Child Health and Centre for Translational PhysiologyUniversity of OtagoWellingtonNew Zealand
| | - Emilio A. Herrera
- Pathophysiology Program, Biomedical Sciences Institute (ICBM), Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jonathan J. Hirst
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Biomedical Sciences and PharmacyUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Bona Kim
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
| | - Karen L. Kind
- School of Animal and Veterinary SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Bernardo J. Krause
- Division of Paediatrics, Faculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | | | - Hannah K. Palliser
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Biomedical Sciences and PharmacyUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Timothy R. H. Regnault
- Departments of Obstetrics and Gynaecology, Physiology and PharmacologyWestern University, and Children's Health Research Institute and Lawson Health Research InstituteLondonOntarioCanada
| | - Bryan S. Richardson
- Departments of Obstetrics and Gynaecology, Physiology and PharmacologyWestern University, and Children's Health Research Institute and Lawson Health Research InstituteLondonOntarioCanada
| | - Aya Sasaki
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
| | - Loren P. Thompson
- Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Mary J. Berry
- Department of Paediatrics & Child Health and Centre for Translational PhysiologyUniversity of OtagoWellingtonNew Zealand
| |
Collapse
|
21
|
Cheng L, Al-Owais M, Covarrubias ML, Koch WJ, Manning DR, Peers C, Riobo-Del Galdo NA. Coupling of Smoothened to inhibitory G proteins reduces voltage-gated K + currents in cardiomyocytes and prolongs cardiac action potential duration. J Biol Chem 2018; 293:11022-11032. [PMID: 29802197 DOI: 10.1074/jbc.ra118.001989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/10/2018] [Indexed: 12/25/2022] Open
Abstract
SMO (Smoothened), the central transducer of Hedgehog signaling, is coupled to heterotrimeric Gi proteins in many cell types, including cardiomyocytes. In this study, we report that activation of SMO with SHH (Sonic Hedgehog) or a small agonist, purmorphamine, rapidly causes a prolongation of the action potential duration that is sensitive to a SMO inhibitor. In contrast, neither of the SMO agonists prolonged the action potential in cardiomyocytes from transgenic GiCT/TTA mice, in which Gi signaling is impaired, suggesting that the effect of SMO is mediated by Gi proteins. Investigation of the mechanism underlying the change in action potential kinetics revealed that activation of SMO selectively reduces outward voltage-gated K+ repolarizing (Kv) currents in isolated cardiomyocytes and that it induces a down-regulation of membrane levels of Kv4.3 in cardiomyocytes and intact hearts from WT but not from GiCT/TTA mice. Moreover, perfusion of intact hearts with Shh or purmorphamine increased the ventricular repolarization time (QT interval) and induced ventricular arrhythmias. Our data constitute the first report that acute, noncanonical Hh signaling mediated by Gi proteins regulates K+ currents density in cardiomyocytes and sensitizes the heart to the development of ventricular arrhythmias.
Collapse
Affiliation(s)
- Lan Cheng
- From the Departments of Biochemistry & Molecular Biology and
| | - Moza Al-Owais
- the Leeds Institute of Cardiovascular and Metabolic Medicine and
| | | | - Walter J Koch
- the Department of Pharmacology and Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania 19140, and
| | - David R Manning
- the Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Chris Peers
- the Leeds Institute of Cardiovascular and Metabolic Medicine and
| | - Natalia A Riobo-Del Galdo
- From the Departments of Biochemistry & Molecular Biology and .,the Leeds Institute of Cancer and Pathology, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
22
|
A novel role of the antitumor agent tricyclodecan-9-yl-xanthogenate as an open channel blocker of KCNQ1/KCNE1. Eur J Pharmacol 2018; 824:99-107. [DOI: 10.1016/j.ejphar.2018.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/26/2022]
|
23
|
Bae H, Choi J, Kim YW, Lee D, Kim JH, Ko JH, Bang H, Kim T, Lim I. Effects of Nitric Oxide on Voltage-Gated K⁺ Currents in Human Cardiac Fibroblasts through the Protein Kinase G and Protein Kinase A Pathways but Not through S-Nitrosylation. Int J Mol Sci 2018. [PMID: 29534509 PMCID: PMC5877675 DOI: 10.3390/ijms19030814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This study investigated the expression of voltage-gated K+ (KV) channels in human cardiac fibroblasts (HCFs), and the effect of nitric oxide (NO) on the KV currents, and the underlying phosphorylation mechanisms. In reverse transcription polymerase chain reaction, two types of KV channels were detected in HCFs: delayed rectifier K+ channel and transient outward K+ channel. In whole-cell patch-clamp technique, delayed rectifier K+ current (IK) exhibited fast activation and slow inactivation, while transient outward K+ current (Ito) showed fast activation and inactivation kinetics. Both currents were blocked by 4-aminopyridine. An NO donor, S-nitroso-N-acetylpenicillamine (SNAP), increased the amplitude of IK in a concentration-dependent manner with an EC50 value of 26.4 µM, but did not affect Ito. The stimulating effect of SNAP on IK was blocked by pretreatment with 1H-(1,2,4)oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) or by KT5823. 8-bromo-cyclic GMP stimulated the IK. The stimulating effect of SNAP on IK was also blocked by pretreatment with KT5720 or by SQ22536. Forskolin and 8-bromo-cyclic AMP each stimulated IK. On the other hand, the stimulating effect of SNAP on IK was not blocked by pretreatment of N-ethylmaleimide or by DL-dithiothreitol. Our data suggest that NO enhances IK, but not Ito, among KV currents of HCFs, and the stimulating effect of NO on IK is through the PKG and PKA pathways, not through S-nitrosylation.
Collapse
Affiliation(s)
- Hyemi Bae
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Jeongyoon Choi
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Young-Won Kim
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Donghee Lee
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Jung-Ha Kim
- Department of Family Medicine, College of Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Seoul 06973, Korea.
| | - Jae-Hong Ko
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Hyoweon Bang
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| | - Taeho Kim
- Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Seoul 06973, Korea.
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Seoul 06974, Korea.
| |
Collapse
|
24
|
Toib A, Zhang C, Borghetti G, Zhang X, Wallner M, Yang Y, Troupes CD, Kubo H, Sharp TE, Feldsott E, Berretta RM, Zalavadia N, Trappanese DM, Harper S, Gross P, Chen X, Mohsin S, Houser SR. Remodeling of repolarization and arrhythmia susceptibility in a myosin-binding protein C knockout mouse model. Am J Physiol Heart Circ Physiol 2017. [PMID: 28646025 DOI: 10.1152/ajpheart.00167.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is one of the most common genetic cardiac diseases and among the leading causes of sudden cardiac death (SCD) in the young. The cellular mechanisms leading to SCD in HCM are not well known. Prolongation of the action potential (AP) duration (APD) is a common feature predisposing hypertrophied hearts to SCD. Previous studies have explored the roles of inward Na+ and Ca2+ in the development of HCM, but the role of repolarizing K+ currents has not been defined. The objective of this study was to characterize the arrhythmogenic phenotype and cellular electrophysiological properties of mice with HCM, induced by myosin-binding protein C (MyBPC) knockout (KO), and to test the hypothesis that remodeling of repolarizing K+ currents causes APD prolongation in MyBPC KO myocytes. We demonstrated that MyBPC KO mice developed severe hypertrophy and cardiac dysfunction compared with wild-type (WT) control mice. Telemetric electrocardiographic recordings of awake mice revealed prolongation of the corrected QT interval in the KO compared with WT control mice, with overt ventricular arrhythmias. Whole cell current- and voltage-clamp experiments comparing KO with WT mice demonstrated ventricular myocyte hypertrophy, AP prolongation, and decreased repolarizing K+ currents. Quantitative RT-PCR analysis revealed decreased mRNA levels of several key K+ channel subunits. In conclusion, decrease in repolarizing K+ currents in MyBPC KO ventricular myocytes contributes to AP and corrected QT interval prolongation and could account for the arrhythmia susceptibility.NEW & NOTEWORTHY Ventricular myocytes isolated from the myosin-binding protein C knockout hypertrophic cardiomyopathy mouse model demonstrate decreased repolarizing K+ currents and action potential and QT interval prolongation, linking cellular repolarization abnormalities with arrhythmia susceptibility and the risk for sudden cardiac death in hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Amir Toib
- Section of Pediatric Cardiology, St. Christopher's Hospital for Children and Department of Pediatrics, Drexel University College of Medicine, Philadelphia, Pennsylvania; and.,Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Chen Zhang
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Giulia Borghetti
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Xiaoxiao Zhang
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Markus Wallner
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Yijun Yang
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Constantine D Troupes
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Hajime Kubo
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Thomas E Sharp
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Eric Feldsott
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Remus M Berretta
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Neil Zalavadia
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Danielle M Trappanese
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Shavonn Harper
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Polina Gross
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Xiongwen Chen
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Sadia Mohsin
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Steven R Houser
- Cardiovascular Research Center and Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Lazzerini PE, Capecchi PL, Laghi-Pasini F, Boutjdir M. Autoimmune channelopathies as a novel mechanism in cardiac arrhythmias. Nat Rev Cardiol 2017; 14:521-535. [PMID: 28470179 DOI: 10.1038/nrcardio.2017.61] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cardiac arrhythmias confer a considerable burden of morbidity and mortality in industrialized countries. Although coronary artery disease and heart failure are the prevalent causes of cardiac arrest, in 5-15% of patients, structural abnormalities at autopsy are absent. In a proportion of these patients, mutations in genes encoding cardiac ion channels are documented (inherited channelopathies), but, to date, the molecular autopsy is negative in nearly 70% of patients. Emerging evidence indicates that autoimmunity is involved in the pathogenesis of cardiac arrhythmias. In particular, several arrhythmogenic autoantibodies targeting specific calcium, potassium, or sodium channels in the heart have been identified. Experimental and clinical studies demonstrate that these autoantibodies can promote conduction disturbances and life-threatening tachyarrhythmias by inducing substantial electrophysiological changes. In this Review, we propose the term 'autoimmune cardiac channelopathies' to define this novel pathogenic mechanism of cardiac arrhythmias, which could be more frequent and clinically relevant than previously appreciated. Indeed, pathogenic autoantibodies against ion channels are detectable not only in patients with manifest autoimmune disease, but also in apparently healthy individuals, which suggests a causal role in some cases of unexplained arrhythmias and cardiac arrest. Considering this possibility and performing specific testing in patients with 'idiopathic' rhythm disturbances could create novel treatment opportunities.
Collapse
Affiliation(s)
- Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Viale Bracci 16, Siena, 53100, Italy
| | - Pier Leopoldo Capecchi
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Viale Bracci 16, Siena, 53100, Italy
| | - Franco Laghi-Pasini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Viale Bracci 16, Siena, 53100, Italy
| | - Mohamed Boutjdir
- VA New York Harbor Healthcare System, 800 Poly Place, Brooklyn, New York 11209, USA.,SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, New York 11203, USA.,NYU School of Medicine, 550 1st Avenue, New York, New York 10016, USA
| |
Collapse
|
26
|
Edwards AG, Louch WE. Species-Dependent Mechanisms of Cardiac Arrhythmia: A Cellular Focus. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2017; 11:1179546816686061. [PMID: 28469490 PMCID: PMC5392019 DOI: 10.1177/1179546816686061] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/20/2016] [Indexed: 12/17/2022]
Abstract
Although ventricular arrhythmia remains a leading cause of morbidity and mortality, available antiarrhythmic drugs have limited efficacy. Disappointing progress in the development of novel, clinically relevant antiarrhythmic agents may partly be attributed to discrepancies between humans and animal models used in preclinical testing. However, such differences are at present difficult to predict, requiring improved understanding of arrhythmia mechanisms across species. To this end, we presently review interspecies similarities and differences in fundamental cardiomyocyte electrophysiology and current understanding of the mechanisms underlying the generation of afterdepolarizations and reentry. We specifically highlight patent shortcomings in small rodents to reproduce cellular and tissue-level arrhythmia substrate believed to be critical in human ventricle. Despite greater ease of translation from larger animal models, discrepancies remain and interpretation can be complicated by incomplete knowledge of human ventricular physiology due to low availability of explanted tissue. We therefore point to the benefits of mathematical modeling as a translational bridge to understanding and treating human arrhythmia.
Collapse
Affiliation(s)
- Andrew G Edwards
- Center for Biomedical Computing, Simula Research Laboratory, Lysaker, Norway.,Center for Cardiological Innovation, Simula Research Laboratory, Lysaker, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| |
Collapse
|