1
|
Romero-Becerra R, Cruz FM, Mora A, Lopez JA, Ponce-Balbuena D, Allan A, Ramos-Mondragón R, González-Terán B, León M, Rodríguez ME, Leiva-Vega L, Guerrero-Serna G, Jimenez-Vazquez EN, Filgueiras-Rama D, Vázquez J, Jalife J, Sabio G. p38γ/δ activation alters cardiac electrical activity and predisposes to ventricular arrhythmia. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1204-1220. [PMID: 39196141 DOI: 10.1038/s44161-023-00368-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 10/19/2023] [Indexed: 08/29/2024]
Abstract
Ventricular fibrillation (VF) is a leading immediate cause of sudden cardiac death. There is a strong association between aging and VF, although the mechanisms are unclear, limiting the availability of targeted therapeutic interventions. Here we found that the stress kinases p38γ and p38δ are activated in the ventricles of old mice and mice with genetic or drug-induced arrhythmogenic conditions. We discovered that, upon activation, p38γ and p38δ cooperatively increase the susceptibility to stress-induced VF. Mechanistically, our data indicate that activated p38γ and p38δ phosphorylate ryanodine receptor 2 (RyR2) disrupt Kv4.3 channel localization, promoting sarcoplasmic reticulum calcium leak, Ito current reduction and action potential duration prolongation. In turn, this led to aberrant intracellular calcium handling, premature ventricular complexes and enhanced susceptibility to VF. Blocking this pathway protected genetically modified animals from VF development and reduced the VF duration in aged animals. These results indicate that p38γ and p38δ are a potential therapeutic target for sustained VF prevention.
Collapse
Affiliation(s)
| | - Francisco M Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Juan Antonio Lopez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Daniela Ponce-Balbuena
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrew Allan
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Roberto Ramos-Mondragón
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Bárbara González-Terán
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Gladstone Institutes, San Francisco, CA, USA
| | - Marta León
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Guadalupe Guerrero-Serna
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Eric N Jimenez-Vazquez
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - David Filgueiras-Rama
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
2
|
Nayak A, S B A, Bardhan M, Rashmi R, Arunachal G, Prathyusha P, Nalini A, Sathyaprabha T, Udupa K. Evaluation of Cardiac, Autonomic Functions in Ambulant Patients with Duchenne Muscular Dystrophy. SN COMPREHENSIVE CLINICAL MEDICINE 2023; 5:138. [PMID: 37193318 PMCID: PMC10160717 DOI: 10.1007/s42399-023-01473-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 05/18/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder caused by dystrophin gene mutation resulting in muscle weakness, motor delays, difficulty in standing, and inability to walk by 12 years. As disease progresses, it leads to cardiac and respiratory failure. Evaluation of cardiac autonomic status and echocardiography in DMD patients at a young age can be a potential biomarker to assess disease progression. This study aimed to investigate the younger DMD population of 5-11years of age with mild to moderate cardiac involvement for early detection using non-invasive and cost-effective tools. Genetically confirmed male DMD patients, aged 5-11 years (n = 47), screened from the outpatient department of a tertiary neuroscience institution were subjected to heart rate variability and echocardiographic analysis, and values were correlated with their clinical variables. DMD patients showed a significantly higher difference in HR, interventricular septum, E m/s, and E-wave to A-wave (E/A) ratio than normal values (p < 0.001). Significantly higher HR indicates initial sinus tachycardia and decreased IVD (d), and increased E m/s and E/A ratio mark the onset of cardiac symptoms in DMD patients even though its chamber dimension remains normal and are associated with cardiac muscle fibrosis.
Collapse
Affiliation(s)
- Amritharekha Nayak
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| | - Apoorva S B
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| | - Mainak Bardhan
- Department of Neurology, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, 560029 India
| | - R. Rashmi
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| | - G. Arunachal
- Department of Human Genetics, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, 560029 India
| | - P.V. Prathyusha
- Department of Biostatistics, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, 560029 India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, 560029 India
| | - T.N. Sathyaprabha
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| | - Kaviraja Udupa
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| |
Collapse
|
3
|
Lazzeroni D, Villatore A, Souryal G, Pili G, Peretto G. The Aging Heart: A Molecular and Clinical Challenge. Int J Mol Sci 2022; 23:16033. [PMID: 36555671 PMCID: PMC9783309 DOI: 10.3390/ijms232416033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Aging is associated with an increasing burden of morbidity, especially for cardiovascular diseases (CVDs). General cardiovascular risk factors, ischemic heart diseases, heart failure, arrhythmias, and cardiomyopathies present a significant prevalence in older people, and are characterized by peculiar clinical manifestations that have distinct features compared with the same conditions in a younger population. Remarkably, the aging heart phenotype in both healthy individuals and patients with CVD reflects modifications at the cellular level. An improvement in the knowledge of the physiological and pathological molecular mechanisms underlying cardiac aging could improve clinical management of older patients and offer new therapeutic targets.
Collapse
Affiliation(s)
| | - Andrea Villatore
- School of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Department of Arrhythmology and Cardiac Electrophysiology, Ospedale San Raffaele, 20132 Milan, Italy
| | - Gaia Souryal
- School of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Gianluca Pili
- School of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Giovanni Peretto
- School of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Department of Arrhythmology and Cardiac Electrophysiology, Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
4
|
Nagy N, Tóth N, Nánási PP. Antiarrhythmic and Inotropic Effects of Selective Na +/Ca 2+ Exchanger Inhibition: What Can We Learn from the Pharmacological Studies? Int J Mol Sci 2022; 23:ijms232314651. [PMID: 36498977 PMCID: PMC9736231 DOI: 10.3390/ijms232314651] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
Life-long stable heart function requires a critical balance of intracellular Ca2+. Several ion channels and pumps cooperate in a complex machinery that controls the influx, release, and efflux of Ca2+. Probably one of the most interesting and most complex players of this crosstalk is the Na+/Ca2+ exchanger, which represents the main Ca2+ efflux mechanism; however, under some circumstances, it can also bring Ca2+ into the cell. Therefore, the inhibition of the Na+/Ca2+ exchanger has emerged as one of the most promising possible pharmacological targets to increase Ca2+ levels, to decrease arrhythmogenic depolarizations, and to reduce excessive Ca2+ influx. In line with this, as a response to increasing demand, several more or less selective Na+/Ca2+ exchanger inhibitor compounds have been developed. In the past 20 years, several results have been published regarding the effect of Na+/Ca2+ exchanger inhibition under various circumstances, e.g., species, inhibitor compounds, and experimental conditions; however, the results are often controversial. Does selective Na+/Ca2+ exchanger inhibition have any future in clinical pharmacological practice? In this review, the experimental results of Na+/Ca2+ exchanger inhibition are summarized focusing on the data obtained by novel highly selective inhibitors.
Collapse
Affiliation(s)
- Norbert Nagy
- ELKH-SZTE Research Group of Cardiovascular Pharmacology, 6720 Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-682; Fax: +36-62-545-680
| | - Noémi Tóth
- ELKH-SZTE Research Group of Cardiovascular Pharmacology, 6720 Szeged, Hungary
| | - Péter P. Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
5
|
Angelini M, Pezhouman A, Savalli N, Chang MG, Steccanella F, Scranton K, Calmettes G, Ottolia M, Pantazis A, Karagueuzian HS, Weiss JN, Olcese R. Suppression of ventricular arrhythmias by targeting late L-type Ca2+ current. J Gen Physiol 2021; 153:212725. [PMID: 34698805 PMCID: PMC8552156 DOI: 10.1085/jgp.202012584] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/15/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022] Open
Abstract
Ventricular arrhythmias, a leading cause of sudden cardiac death, can be triggered by cardiomyocyte early afterdepolarizations (EADs). EADs can result from an abnormal late activation of L-type Ca2+ channels (LTCCs). Current LTCC blockers (class IV antiarrhythmics), while effective at suppressing EADs, block both early and late components of ICa,L, compromising inotropy. However, computational studies have recently demonstrated that selective reduction of late ICa,L (Ca2+ influx during late phases of the action potential) is sufficient to potently suppress EADs, suggesting that effective antiarrhythmic action can be achieved without blocking the early peak ICa,L, which is essential for proper excitation–contraction coupling. We tested this new strategy using a purine analogue, roscovitine, which reduces late ICa,L with minimal effect on peak current. Scaling our investigation from a human CaV1.2 channel clone to rabbit ventricular myocytes and rat and rabbit perfused hearts, we demonstrate that (1) roscovitine selectively reduces ICa,L noninactivating component in a human CaV1.2 channel clone and in ventricular myocytes native current, (2) the pharmacological reduction of late ICa,L suppresses EADs and EATs (early after Ca2+ transients) induced by oxidative stress and hypokalemia in isolated myocytes, largely preserving cell shortening and normal Ca2+ transient, and (3) late ICa,L reduction prevents/suppresses ventricular tachycardia/fibrillation in ex vivo rabbit and rat hearts subjected to hypokalemia and/or oxidative stress. These results support the value of an antiarrhythmic strategy based on the selective reduction of late ICa,L to suppress EAD-mediated arrhythmias. Antiarrhythmic therapies based on this idea would modify the gating properties of CaV1.2 channels rather than blocking their pore, largely preserving contractility.
Collapse
Affiliation(s)
- Marina Angelini
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Arash Pezhouman
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Nicoletta Savalli
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Marvin G Chang
- Department of Anesthesia and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Federica Steccanella
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Kyle Scranton
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Guillaume Calmettes
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Michela Ottolia
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,University of California, Los Angeles Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Antonios Pantazis
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Hrayr S Karagueuzian
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - James N Weiss
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Riccardo Olcese
- Division of Molecular Medicine, Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,University of California, Los Angeles Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
6
|
Verheule S, Schotten U. Electrophysiological Consequences of Cardiac Fibrosis. Cells 2021; 10:cells10113220. [PMID: 34831442 PMCID: PMC8625398 DOI: 10.3390/cells10113220] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/27/2022] Open
Abstract
For both the atria and ventricles, fibrosis is generally recognized as one of the key determinants of conduction disturbances. By definition, fibrosis refers to an increased amount of fibrous tissue. However, fibrosis is not a singular entity. Various forms can be distinguished, that differ in distribution: replacement fibrosis, endomysial and perimysial fibrosis, and perivascular, endocardial, and epicardial fibrosis. These different forms typically result from diverging pathophysiological mechanisms and can have different consequences for conduction. The impact of fibrosis on propagation depends on exactly how the patterns of electrical connections between myocytes are altered. We will therefore first consider the normal patterns of electrical connections and their regional diversity as determinants of propagation. Subsequently, we will summarize current knowledge on how different forms of fibrosis lead to a loss of electrical connectivity in order to explain their effects on propagation and mechanisms of arrhythmogenesis, including ectopy, reentry, and alternans. Finally, we will discuss a histological quantification of fibrosis. Because of the different forms of fibrosis and their diverging effects on electrical propagation, the total amount of fibrosis is a poor indicator for the effect on conduction. Ideally, an assessment of cardiac fibrosis should exclude fibrous tissue that does not affect conduction and differentiate between the various types that do; in this article, we highlight practical solutions for histological analysis that meet these requirements.
Collapse
|
7
|
Sadredini M, Haugsten Hansen M, Frisk M, Louch WE, Lehnart SE, Sjaastad I, Stokke MK. CaMKII inhibition has dual effects on spontaneous Ca 2+ release and Ca 2+ alternans in ventricular cardiomyocytes from mice with a gain-of-function RyR2 mutation. Am J Physiol Heart Circ Physiol 2021; 321:H446-H460. [PMID: 34270372 DOI: 10.1152/ajpheart.00011.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In conditions with abnormally increased activity of the cardiac ryanodine receptor (RyR2), Ca2+/calmodulin-dependent protein kinase II (CaMKII) can contribute to a further destabilization of RyR2 that results in triggered arrhythmias. Therefore, inhibition of CaMKII in such conditions has been suggested as a strategy to suppress RyR2 activity and arrhythmias. However, suppression of RyR2 activity can lead to the development of arrhythmogenic Ca2+ alternans. The aim of this study was to test whether the suppression of RyR2 activity caused by inhibition of CaMKII increases propensity for Ca2+ alternans. We studied spontaneous Ca2+ release events and Ca2+ alternans in isolated left ventricular cardiomyocytes from mice carrying the gain-of-function RyR2 mutation RyR2-R2474S and from wild-type mice. CaMKII inhibition by KN-93 effectively decreased the frequency of spontaneous Ca2+ release events in RyR2-R2474S cardiomyocytes exposed to the β-adrenoceptor agonist isoprenaline. However, KN-93-treated RyR2-R2474S cardiomyocytes also showed increased propensity for Ca2+ alternans and increased Ca2+ alternans ratio compared with both an inactive analog of KN-93 and with vehicle-treated controls. This increased propensity for Ca2+ alternans was explained by prolongation of Ca2+ release refractoriness. Importantly, the increased propensity for Ca2+ alternans in KN-93-treated RyR2-R2474S cardiomyocytes did not surpass that of wild type. In conclusion, inhibition of CaMKII efficiently reduces spontaneous Ca2+ release but promotes Ca2+ alternans in RyR2-R2474S cardiomyocytes with a gain-of-function RyR2 mutation. The dominant effect in RyR2-R2474S is to reduce spontaneous Ca2+ release, which supports this intervention as a therapeutic strategy in this specific condition. However, future studies on CaMKII inhibition in conditions with increased propensity for Ca2+ alternans should include investigation of both phenomena.NEW & NOTEWORTHY Genetically increased RyR2 activity promotes arrhythmogenic Ca2+ release. Inhibition of CaMKII suppresses RyR2 activity and arrhythmogenic Ca2+ release. Suppression of RyR2 activity prolongs refractoriness of Ca2+ release. Prolonged refractoriness of Ca2+ release leads to arrhythmogenic Ca2+ alternans. CaMKII inhibition promotes Ca2+ alternans by prolonging Ca2+ release refractoriness.
Collapse
Affiliation(s)
- Mani Sadredini
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Cardiac Research Centre, University of Oslo, Oslo, Norway
| | - Marie Haugsten Hansen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Cardiac Research Centre, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Cardiac Research Centre, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Cardiac Research Centre, University of Oslo, Oslo, Norway
| | - Stephan E Lehnart
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Cardiac Research Centre, University of Oslo, Oslo, Norway
| | - Mathis Korseberg Stokke
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Cardiac Research Centre, University of Oslo, Oslo, Norway.,Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
8
|
Imran H, Nester W, Elgendy IY, Saad M. Role of sodium glucose co-transporter 2 inhibitors in patients with heart failure: an elusive mechanism. Ann Med 2020; 52:178-190. [PMID: 32393068 PMCID: PMC7877993 DOI: 10.1080/07853890.2020.1767298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a major cause of morbidity and mortality worldwide, and the burden of HF continues to rise. There has been an interest in sodium-glucose co-transporter-2 (SGLT2) inhibitors for their role in reducing HF hospitalizations in pivotal trials. Since these agents were approved by the Food and Drug Administration for the management of diabetes mellitus, multiple small trials and analyses have tried to explain the underlying beneficial mechanisms in HF . In this review, we discuss different mechanisms by which SGLT2 inhibitors play hemodynamic, metabolic, and cellular roles in different HF phenotypes. We also address issues pertaining to the safety of these relatively newer agents.KEY MESSAGESSGLT2 inhibitors are associated with a reduction in HF hospitalizations in both diabetics and non-diabetics.The beneficial role of SGLT2 inhibitors in reducing HF hospitalization is observed among participants with established cardiovascular disease/HF and at-risk population.SGLT2 inhibitors pose an important role in renal protection, another mechanism by which these medications can be helpful in HF patients.
Collapse
Affiliation(s)
- Hafiz Imran
- Cardiovascular Institute, Warren Alpert Medical School at Brown University, Providence, RI, USA
| | - William Nester
- Cardiovascular Institute, Warren Alpert Medical School at Brown University, Providence, RI, USA
| | - Islam Y. Elgendy
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Marwan Saad
- Cardiovascular Institute, Warren Alpert Medical School at Brown University, Providence, RI, USA
| |
Collapse
|
9
|
Cardiac function and incidence of unexplained myocardial scarring in patients with primary carnitine deficiency - a cardiac magnetic resonance study. Sci Rep 2019; 9:13909. [PMID: 31558765 PMCID: PMC6763485 DOI: 10.1038/s41598-019-50458-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/11/2019] [Indexed: 11/15/2022] Open
Abstract
Primary carnitine deficiency (PCD) not treated with L-Carnitine can lead to sudden cardiac death. To our knowledge, it is unknown if asymptomatic patients treated with L-Carnitine suffer from myocardial scarring and thus be at greater risk of potentially serious arrhythmia. Cardiac evaluation of function and myocardial scarring is non-invasively best supported by cardiac magnetic resonance imaging (CMR) with late gadolinium enhancement (LGE). The study included 36 PCD patients, 17 carriers and 17 healthy subjects. A CMR cine stack in the short-axis plane were acquired to evaluate left ventricle (LV) systolic and diastolic function and a similar LGE stack to evaluate myocardial scarring and replacement fibrosis. LV volumes and ejection fraction were not different between PCD patients, carriers and healthy subjects. However, LV mass was higher in PCD patients with the severe homozygous mutation, c.95 A > G (p = 0.037; n = 17). Among homozygous PCD patients there were two cases of unexplained myocardial scarring and this is in contrast to no myocardial scarring in any of the other study participants (p = 0.10). LV mass was increased in PCD patients. L-carnitine supplementation is essential in order to prevent potentially lethal cardiac arrhythmia and serious adverse cardiac remodeling.
Collapse
|
10
|
Wang Y, Tang Y, Zou Y, Wang D, Zhu L, Tian T, Wang J, Bao J, Hui R, Kang L, Song L, Wang J. Plasma level of big endothelin-1 predicts the prognosis in patients with hypertrophic cardiomyopathy. Int J Cardiol 2017; 243:283-289. [PMID: 28587741 DOI: 10.1016/j.ijcard.2017.03.162] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/24/2017] [Accepted: 03/31/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cardiac remodeling is one of major pathological process in hypertrophic cardiomyopathy (HCM). Endothelin-1 has been linked to cardiac remodeling. Big endothelin-1 is the precursor of endothelin-1. METHODS A total of 245 patients with HCM were enrolled from 1999 to 2011 and partitioned to low, middle and high level groups according to their plasma big endothelin-1 levels. RESULTS At baseline, significant associations were found between high level of big endothelin-1 and left atrium size, heart function and atrial fibrillation. Big endothelin-1 was positively correlated with N-terminal B-type natriuretic peptide (r=0.291, p<0.001) and late gadolinium enhancement (LGE) on magnetic resonance imaging (r=0.222, p=0.016). During a follow-up of 3 (range, 2-5) years, big endothelin-1 level was positively associated with the risks of all-cause mortality, cardiovascular death and progression to NYHA class 3 or 4 (p=0.020, 0.044 and 0.032, respectively). The rate of above events in the highest tertile were 18.1%, 15.7%, 24.2%, respectively. After adjusting for multiple factors related to survival and cardiac function, the significance remained in the association of big endothelin-1 with the risk of all-cause mortality (hazard ratio (HR)=4.94, 95% confidence interval (CI) 1.07-22.88; p=0.041) and progression to NYHA class 3 or 4 (HR=4.10, 95%CI 1.32-12.75, p=0.015). CONCLUSION Our study showed that high level of plasma big endothelin-1 predicted prognosis for patients with HCM and it can be added to the marker panel in stratifying HCM patients for giving treatment priority to those at high risk.
Collapse
Affiliation(s)
- Yilu Wang
- Department of ICU, China Meitan General Hospital, Beijing, China
| | - Yida Tang
- State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yubao Zou
- State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Dong Wang
- State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ling Zhu
- Department of Cardiology, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Tao Tian
- State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jizheng Wang
- Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jingru Bao
- Center for Cardiovascular Diseases, PLA Navy General Hospital, Beijing, China
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lianming Kang
- State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lei Song
- State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Sino-German Laboratory for Molecular Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| | - Ji Wang
- Department of ICU, China Meitan General Hospital, Beijing, China.
| |
Collapse
|
11
|
Karagueuzian HS, Pezhouman A, Angelini M, Olcese R. Enhanced Late Na and Ca Currents as Effective Antiarrhythmic Drug Targets. Front Pharmacol 2017; 8:36. [PMID: 28220073 PMCID: PMC5292429 DOI: 10.3389/fphar.2017.00036] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/18/2017] [Indexed: 11/30/2022] Open
Abstract
While recent advances clarified the molecular and cellular modes of action of antiarrhythmic drugs (AADs), their link to suppression of dynamical arrhythmia mechanisms remains only partially understood. The current classifications of AADs (Classes I, III, and IV) rely on blocking peak Na, K and L-type calcium currents (ICa,L), with Class II with dominant beta receptor blocking activity and Class V including drugs with diverse classes of actions. The discovery that the calcium and redox sensor, cardiac Ca/calmodulin-dependent protein kinase II (CaMKII) enhances both the late Na (INa-L) and the late ICa,L in patients at high risk of VT/VF provided a new and a rational AAD target. Pathological rise of either or both of INa-L and late ICa,L are demonstrated to promote cellular early afterdepolarizations (EADs) and EAD-mediated triggered activity that can initiate VT/VF in remodeled hearts. Selective inhibition of the INa-L without affecting their peak transients with the highly specific prototype drug, GS-967 suppresses these EAD-mediated VT/VFs. As in the case of INa-L, selective inhibition of the late ICa,L without affecting its peak with the prototype drug, roscovitine suppressed oxidative EAD-mediated VT/VF. These findings indicate that specific blockers of the late inward currents without affecting their peaks (gating modifiers), offer a new and effective AAD class action i.e., “Class VI.” The development of safe drugs with selective Class VI actions provides a rational and effective approach to treat VT/VF particularly in cardiac conditions associated with enhanced CaMKII activity such as heart failure.
Collapse
Affiliation(s)
- Hrayr S Karagueuzian
- Translational Arrhythmia Section, David Geffen School of Medicine, University of California, Los AngelesLos Angeles, CA, USA; Cardiovascular Research Laboratory, Departments of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los AngelesLos Angeles, CA, USA
| | - Arash Pezhouman
- Translational Arrhythmia Section, David Geffen School of Medicine, University of California, Los AngelesLos Angeles, CA, USA; Cardiovascular Research Laboratory, Departments of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los AngelesLos Angeles, CA, USA
| | - Marina Angelini
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles Los Angeles, CA, USA
| | - Riccardo Olcese
- Cardiovascular Research Laboratory, Departments of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los AngelesLos Angeles, CA, USA; Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los AngelesLos Angeles, CA, USA; Department of Physiology, David Geffen School of Medicine, University of California, Los AngelesLos Angeles, CA, USA
| |
Collapse
|
12
|
Sridhar S, Vandersickel N, Panfilov AV. Effect of myocyte-fibroblast coupling on the onset of pathological dynamics in a model of ventricular tissue. Sci Rep 2017; 7:40985. [PMID: 28106124 PMCID: PMC5247688 DOI: 10.1038/srep40985] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/13/2016] [Indexed: 12/23/2022] Open
Abstract
Managing lethal cardiac arrhythmias is one of the biggest challenges in modern cardiology, and hence it is very important to understand the factors underlying such arrhythmias. While early afterdepolarizations (EAD) of cardiac cells is known to be one such arrhythmogenic factor, the mechanisms underlying the emergence of tissue level arrhythmias from cellular level EADs is not fully understood. Another known arrhythmogenic condition is fibrosis of cardiac tissue that occurs both due to aging and in many types of heart diseases. In this paper we describe the results of a systematic in-silico study, using the TNNP model of human cardiac cells and MacCannell model for (myo)fibroblasts, on the possible effects of diffuse fibrosis on arrhythmias occurring via EADs. We find that depending on the resting potential of fibroblasts (VFR), M-F coupling can either increase or decrease the region of parameters showing EADs. Fibrosis increases the probability of occurrence of arrhythmias after a single focal stimulation and this effect increases with the strength of the M-F coupling. While in our simulations, arrhythmias occur due to fibrosis induced ectopic activity, we do not observe any specific fibrotic pattern that promotes the occurrence of these ectopic sources.
Collapse
Affiliation(s)
- S. Sridhar
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Nele Vandersickel
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Alexander V. Panfilov
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
13
|
Kohajda Z, Farkas-Morvay N, Jost N, Nagy N, Geramipour A, Horváth A, Varga RS, Hornyik T, Corici C, Acsai K, Horváth B, Prorok J, Ördög B, Déri S, Tóth D, Levijoki J, Pollesello P, Koskelainen T, Otsomaa L, Tóth A, Baczkó I, Leprán I, Nánási PP, Papp JG, Varró A, Virág L. The Effect of a Novel Highly Selective Inhibitor of the Sodium/Calcium Exchanger (NCX) on Cardiac Arrhythmias in In Vitro and In Vivo Experiments. PLoS One 2016; 11:e0166041. [PMID: 27832106 PMCID: PMC5104402 DOI: 10.1371/journal.pone.0166041] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 10/21/2016] [Indexed: 11/18/2022] Open
Abstract
Background In this study the effects of a new, highly selective sodium-calcium exchanger (NCX) inhibitor, ORM-10962 were investigated on cardiac NCX current, Ca2+ transients, cell shortening and in experimental arrhythmias. The level of selectivity of the novel inhibitor on several major transmembrane ion currents (L-type Ca2+ current, major repolarizing K+ currents, late Na+ current, Na+/K+ pump current) was also determined. Methods Ion currents in single dog ventricular cells (cardiac myocytes; CM), and action potentials in dog cardiac multicellular preparations were recorded utilizing the whole-cell patch clamp and standard microelectrode techniques, respectively. Ca2+ transients and cell shortening were measured in fluorescent dye loaded isolated dog myocytes. Antiarrhythmic effects of ORM-10962 were studied in anesthetized ouabain (10 μg/kg/min i.v.) pretreated guinea pigs and in ischemia-reperfusion models (I/R) of anesthetized coronary artery occluded rats and Langendorff perfused guinea pigs hearts. Results ORM-10962 significantly reduced the inward/outward NCX currents with estimated EC50 values of 55/67 nM, respectively. The compound, even at a high concentration of 1 μM, did not modify significantly the magnitude of ICaL in CMs, neither had any apparent influence on the inward rectifier, transient outward, the rapid and slow components of the delayed rectifier potassium currents, the late and peak sodium and Na+/K+ pump currents. NCX inhibition exerted moderate positive inotropic effect under normal condition, negative inotropy when reverse, and further positive inotropic effect when forward mode was facilitated. In dog Purkinje fibres 1 μM ORM-10962 decreased the amplitude of digoxin induced delayed afterdepolarizations (DADs). Pre-treatment with 0.3 mg/kg ORM-10962 (i.v.) 10 min before starting ouabain infusion significantly delayed the development and recurrence of ventricular extrasystoles (by about 50%) or ventricular tachycardia (by about 30%) in anesthetized guinea pigs. On the contrary, ORM-10962 pre-treatment had no apparent influence on the time of onset or the severity of I/R induced arrhythmias in anesthetized rats and in Langendorff perfused guinea-pig hearts. Conclusions The present study provides strong evidence for a high efficacy and selectivity of the NCX-inhibitory effect of ORM-10962. Selective NCX inhibition can exert positive as well as negative inotropic effect depending on the actual operation mode of NCX. Selective NCX blockade may contribute to the prevention of DAD based arrhythmogenesis, in vivo, however, its effect on I/R induced arrhythmias is still uncertain.
Collapse
Affiliation(s)
- Zsófia Kohajda
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Nikolett Farkas-Morvay
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Norbert Jost
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
- Department of Pathophysiology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Norbert Nagy
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Amir Geramipour
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - András Horváth
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Richárd S. Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Tibor Hornyik
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Claudia Corici
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Károly Acsai
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Prorok
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Balázs Ördög
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Szilvia Déri
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Dániel Tóth
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | | | | | | | | | - András Tóth
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
- Department of Pathophysiology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - István Leprán
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Péter P. Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Julius Gy Papp
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
- * E-mail:
| |
Collapse
|
14
|
Yang HQ, Subbotina E, Ramasamy R, Coetzee WA. Cardiovascular K ATP channels and advanced aging. PATHOBIOLOGY OF AGING & AGE RELATED DISEASES 2016; 6:32517. [PMID: 27733235 PMCID: PMC5061878 DOI: 10.3402/pba.v6.32517] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/12/2016] [Accepted: 09/14/2016] [Indexed: 12/20/2022]
Abstract
With advanced aging, there is a decline in innate cardiovascular function. This decline is not general in nature. Instead, specific changes occur that impact the basic cardiovascular function, which include alterations in biochemical pathways and ion channel function. This review focuses on a particular ion channel that couple the latter two processes, namely the KATP channel, which opening is promoted by alterations in intracellular energy metabolism. We show that the intrinsic properties of the KATP channel changes with advanced aging and argue that the channel can be further modulated by biochemical changes. The importance is widespread, given the ubiquitous nature of the KATP channel in the cardiovascular system where it can regulate processes as diverse as cardiac function, blood flow and protection mechanisms against superimposed stress, such as cardiac ischemia. We highlight questions that remain to be answered before the KATP channel can be considered as a viable target for therapeutic intervention.
Collapse
Affiliation(s)
- Hua-Qian Yang
- Department of Pediatrics, NYU School of Medicine, New York, NY, USA
| | | | - Ravichandran Ramasamy
- Department of Medicine, NYU School of Medicine, New York, NY, USA.,Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY, USA
| | - William A Coetzee
- Department of Pediatrics, NYU School of Medicine, New York, NY, USA.,Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY, USA.,Department of Physiology & Neuroscience, NYU School of Medicine, New York, NY, USA;
| |
Collapse
|
15
|
Henry BL, Gabris B, Li Q, Martin B, Giannini M, Parikh A, Patel D, Haney J, Schwartzman DS, Shroff SG, Salama G. Relaxin suppresses atrial fibrillation in aged rats by reversing fibrosis and upregulating Na+ channels. Heart Rhythm 2016; 13:983-91. [PMID: 26711798 PMCID: PMC4801709 DOI: 10.1016/j.hrthm.2015.12.030] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) contributes significantly to morbidity and mortality in elderly patients and has been correlated with enhanced age-dependent atrial fibrosis. Reversal of atrial fibrosis has been proposed as therapeutic strategy to suppress AF. OBJECTIVE To test the ability of relaxin to reverse age-dependent atrial fibrosis and suppress AF. METHODS Aged F-344 rats (24 months old) were treated with subcutaneous infusion of vehicle or relaxin (0.4 mg/kg/day) for 2 weeks. Rat hearts were excised, perfused on a Langendorff apparatus, and stained with voltage and Ca(2+) indicator dyes. Optical mapping and programmed electrical stimulation was used to test arrhythmia vulnerability and changes in electrophysiological characteristics. Changes in protein expression and Na(+) current density (INa) were measured by tissue immunofluorescence and whole-cell patch clamp technique. RESULTS In aged rats, sustained AF was readily induced with a premature pulse (n = 7/8) and relaxin treatment suppressed sustained AF by a premature impulse or burst pacing (n = 1/6) (P < .01). Relaxin significantly increased atrial action potential conduction velocity and decreased atrial fibrosis. Relaxin treatment increased Nav1.5 expression (n = 6; 36% ± 10%) and decreased total collagen and collagen I (n = 5-6; 55%-66% ± 15%) in aged atria (P < .05) and decreased collagen I and III and TGF-β1 mRNA (P < .05). Voltage-clamp experiments demonstrated that relaxin treatment (100 nM for 2 days) increased atrial INa by 46% ± 4% (n = 12-13/group, P < .02). CONCLUSION Relaxin suppresses AF through an increase in atrial conduction velocity by decreasing atrial fibrosis and increasing INa. These data provide compelling evidence that relaxin may serve as an effective therapy to manage AF in geriatric patients by reversing fibrosis and modulating cardiac ionic currents.
Collapse
Affiliation(s)
- Brian L Henry
- University of Pittsburgh School of Medicine, Heart and Vascular Institute, Pittsburgh, Pennsylvania
| | - Beth Gabris
- University of Pittsburgh School of Medicine, Heart and Vascular Institute, Pittsburgh, Pennsylvania
| | - Qiao Li
- University of Pittsburgh School of Medicine, Heart and Vascular Institute, Pittsburgh, Pennsylvania
| | - Brian Martin
- University of Pittsburgh Department of Bioengineering, and the McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| | - Marianna Giannini
- University of Pittsburgh School of Medicine, Heart and Vascular Institute, Pittsburgh, Pennsylvania
| | - Ashish Parikh
- University of Pittsburgh Department of Bioengineering, and the McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| | - Divyang Patel
- University of Pittsburgh School of Medicine, Heart and Vascular Institute, Pittsburgh, Pennsylvania
| | - Jamie Haney
- University of Pittsburgh Department of Bioengineering, and the McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| | - David S Schwartzman
- University of Pittsburgh School of Medicine, Heart and Vascular Institute, Pittsburgh, Pennsylvania
| | - Sanjeev G Shroff
- University of Pittsburgh Department of Bioengineering, and the McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| | - Guy Salama
- University of Pittsburgh School of Medicine, Heart and Vascular Institute, Pittsburgh, Pennsylvania,; University of Pittsburgh Department of Bioengineering, and the McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
16
|
Abstract
KATP channels are integral to the functions of many cells and tissues. The use of electrophysiological methods has allowed for a detailed characterization of KATP channels in terms of their biophysical properties, nucleotide sensitivities, and modification by pharmacological compounds. However, even though they were first described almost 25 years ago (Noma 1983, Trube and Hescheler 1984), the physiological and pathophysiological roles of these channels, and their regulation by complex biological systems, are only now emerging for many tissues. Even in tissues where their roles have been best defined, there are still many unanswered questions. This review aims to summarize the properties, molecular composition, and pharmacology of KATP channels in various cardiovascular components (atria, specialized conduction system, ventricles, smooth muscle, endothelium, and mitochondria). We will summarize the lessons learned from available genetic mouse models and address the known roles of KATP channels in cardiovascular pathologies and how genetic variation in KATP channel genes contribute to human disease.
Collapse
Affiliation(s)
- Monique N Foster
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - William A Coetzee
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| |
Collapse
|
17
|
Stary V, Puppala D, Scherrer-Crosbie M, Dillmann WH, Armoundas AA. SERCA2a upregulation ameliorates cellular alternans induced by metabolic inhibition. J Appl Physiol (1985) 2016; 120:865-75. [PMID: 26846549 DOI: 10.1152/japplphysiol.00588.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 01/27/2016] [Indexed: 12/26/2022] Open
Abstract
Cardiac alternans has been associated with the incidence of ventricular tachyarrhythmias and sudden cardiac death. The aim of this study was to investigate the effect of impaired mitochondrial function in the genesis of cellular alternans and to examine whether modulating the sarcoplasmic reticulum (SR) Ca(2+)ameliorates the level of alternans. Cardiomyocytes isolated from control and doxycyline-induced sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a (SERCA2a)-upregulated mice were loaded with two different Ca(2+)indicators to selectively measure mitochondrial and cytosolic Ca(2+)using a custom-made fluorescence photometry system. The degree of alternans was defined as the alternans ratio (AR) [1 - (small Ca(2+)intensity)/(large Ca(2+)intensity)]. Blocking of complex I and II, cytochrome-coxidase, F0F1synthase, α-ketoglutarate dehydrogenase of the electron transport chain, increased alternans in both control and SERCA2a mice (P< 0.01). Changes in AR in SERCA2a-upregulated mice were significantly less pronounced than those observed in control in seven of nine tested conditions (P< 0.04).N-acetyl-l-cysteine (NAC), rescued alternans in myocytes that were previously exposed to an oxidizing agent (P< 0.001). CGP, an antagonist of the mitochondrial Na(+)-Ca(2+)exchanger, had the most severe effect on AR. Exposure to cyclosporin A, a blocker of the mitochondrial permeability transition pore reduced CGP-induced alternans (P< 0.0001). The major findings of this study are that impairment of mitochondrial Ca(2+)cycling and energy production leads to a higher amplitude of alternans in both control and SERCA2a-upregulated mice, but changes in SERCA2a-upregulated mice are less severe, indicating that SERCA2a mice are more capable of sustaining electrical stability during stress. This suggests a relationship between sarcoplasmic Ca(2+)content and mitochondrial dysfunction during alternans, which may potentially help to understand changes in Ca(2+)signaling in myocytes from diseased hearts, leading to new therapeutic targets.
Collapse
Affiliation(s)
- Victoria Stary
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts; Department of Cardiology and Pulmonology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany; and
| | - Dheeraj Puppala
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Wolfgang H Dillmann
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Antonis A Armoundas
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts;
| |
Collapse
|
18
|
Nguyen TP, Sovari AA, Pezhouman A, Iyer S, Cao H, Ko CY, Bapat A, Vahdani N, Ghanim M, Fishbein MC, Karagueuzian HS. Increased susceptibility of spontaneously hypertensive rats to ventricular tachyarrhythmias in early hypertension. J Physiol 2016; 594:1689-707. [PMID: 26775607 DOI: 10.1113/jp271318] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 12/07/2015] [Indexed: 01/19/2023] Open
Abstract
Hypertension is a risk factor for sudden cardiac death caused by ventricular tachycardia and fibrillation (VT/VF). We hypothesized that, in early hypertension, the susceptibility to stress-induced VT/VF increases. We compared the susceptibility of 5- to 6-month-old male spontaneously hypertensive rats (SHR) and age/sex-matched normotensive rats (NR) to VT/VF during challenge with oxidative stress (H2 O2 ; 0.15 mmol l(-1) ). We found that only SHR hearts exhibited left ventricular fibrosis and hypertrophy. H2 O2 promoted VT in all 30 SHR but none of the NR hearts. In 33% of SHR cases, focal VT degenerated to VF within 3 s. Simultaneous voltage-calcium optical mapping of Langendorff-perfused SHR hearts revealed that H2 O2 -induced VT/VF arose spontaneously from focal activations at the base and mid left ventricular epicardium. Microelectrode recording of SHR hearts showed that VT was initiated by early afterdepolarization (EAD)-mediated triggered activity. However, despite the increased susceptibility of SHR hearts to VT/VF, patch clamped isolated SHR ventricular myocytes developed EADs and triggered activity to the same extent as NR ventricular myocytes, except with larger EAD amplitude. During the early stages of hypertension, when challenged with oxidative stress, SHR hearts showed an increased ventricular arrhythmogenicity that stems primarily from tissue remodelling (hypertrophy, fibrosis) rather than cellular electrophysiological changes. Our findings highlight the need for early hypertension treatment to minimize myocardial fibrosis, ventricular hypertrophy, and arrhythmias.
Collapse
Affiliation(s)
- Thao P Nguyen
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine
| | - Ali A Sovari
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine.,Present address: Department of Medicine, Cedars-Sinai Medical Centre, Los Angeles, CA, USA
| | - Arash Pezhouman
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine
| | - Shankar Iyer
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine
| | - Hong Cao
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine.,Present address: Department of Physiology, Wuhan University, Wuhan, China
| | - Christopher Y Ko
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine
| | - Aneesh Bapat
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine.,Present address: Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nooshin Vahdani
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine.,Present address: School of Pharmacy, West Coast University, Los Angeles, CA, USA
| | - Mostafa Ghanim
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine
| | - Michael C Fishbein
- Department of Pathology, UCLA David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Hrayr S Karagueuzian
- UCLA Cardiovascular Research Laboratory, Division of Cardiology, Department of Medicine
| |
Collapse
|
19
|
Karagueuzian HS. Synergism between Enhanced Late Inward Currents and Tissue Fibrosis in the Initiation of Spontaneous Ventricular Tachyarrhythmias. ACTA ACUST UNITED AC 2016; 2. [PMID: 27722204 DOI: 10.16966/2379-769x.120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- H S Karagueuzian
- UCLA Cardiovascular Research Laboratory, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
20
|
Pezhouman A, Singh N, Song Z, Nivala M, Eskandari A, Cao H, Bapat A, Ko CY, Nguyen T, Qu Z, Karagueuzian HS, Weiss JN. Molecular Basis of Hypokalemia-Induced Ventricular Fibrillation. Circulation 2015; 132:1528-1537. [PMID: 26269574 DOI: 10.1161/circulationaha.115.016217] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 08/05/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Hypokalemia is known to promote ventricular arrhythmias, especially in combination with class III antiarrhythmic drugs like dofetilide. Here, we evaluated the underlying molecular mechanisms. METHODS AND RESULTS Arrhythmias were recorded in isolated rabbit and rat hearts or patch-clamped ventricular myocytes exposed to hypokalemia (1.0-3.5 mmol/L) in the absence or presence of dofetilide (1 μmol/L). Spontaneous early afterdepolarizations (EADs) and ventricular tachycardia/fibrillation occurred in 50% of hearts at 2.7 mmol/L [K] in the absence of dofetilide and 3.3 mmol/L [K] in its presence. Pretreatment with the Ca-calmodulin kinase II (CaMKII) inhibitor KN-93, but not its inactive analogue KN-92, abolished EADs and hypokalemia-induced ventricular tachycardia/fibrillation, as did the selective late Na current (INa) blocker GS-967. In intact hearts, moderate hypokalemia (2.7 mmol/L) significantly increased tissue CaMKII activity. Computer modeling revealed that EAD generation by hypokalemia (with or without dofetilide) required Na-K pump inhibition to induce intracellular Na and Ca overload with consequent CaMKII activation enhancing late INa and the L-type Ca current. K current suppression by hypokalemia and dofetilide alone in the absence of CaMKII activation were ineffective at causing EADs. CONCLUSIONS We conclude that Na-K pump inhibition by even moderate hypokalemia plays a critical role in promoting EAD-mediated arrhythmias by inducing a positive feedback cycle activating CaMKII and enhancing late INa. Class III antiarrhythmic drugs like dofetilide sensitize the heart to this positive feedback loop.
Collapse
Affiliation(s)
- Arash Pezhouman
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Neha Singh
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Zhen Song
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Michael Nivala
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Anahita Eskandari
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Hong Cao
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Aneesh Bapat
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Christopher Y Ko
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Thao Nguyen
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Zhilin Qu
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Hrayr S Karagueuzian
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - James N Weiss
- UCLA Cardiovascular Research Laboratory, Departments of Medicine (Cardiology) and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
21
|
Nagy N, Kormos A, Kohajda Z, Szebeni Á, Szepesi J, Pollesello P, Levijoki J, Acsai K, Virág L, Nánási PP, Papp JG, Varró A, Tóth A. Selective Na(+) /Ca(2+) exchanger inhibition prevents Ca(2+) overload-induced triggered arrhythmias. Br J Pharmacol 2015; 171:5665-81. [PMID: 25073832 DOI: 10.1111/bph.12867] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 07/03/2014] [Accepted: 07/25/2014] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND AND PURPOSE Augmented Na(+) /Ca(2+) exchanger (NCX) activity may play a crucial role in cardiac arrhythmogenesis; however, data regarding the anti-arrhythmic efficacy of NCX inhibition are debatable. Feasible explanations could be the unsatisfactory selectivity of NCX inhibitors and/or the dependence of the experimental model on the degree of Ca(2+) i overload. Hence, we used NCX inhibitors SEA0400 and the more selective ORM10103 to evaluate the efficacy of NCX inhibition against arrhythmogenic Ca(2+) i rise in conditions when [Ca(2+) ]i was augmented via activation of the late sodium current (INaL ) or inhibition of the Na(+) /K(+) pump. EXPERIMENTAL APPROACH Action potentials (APs) were recorded from canine papillary muscles and Purkinje fibres by microelectrodes. NCX current (INCX ) was determined in ventricular cardiomyocytes utilizing the whole-cell patch clamp technique. Ca(2+) i transients (CaTs) were monitored with a Ca(2+) -sensitive fluorescent dye, Fluo-4. KEY RESULTS Enhanced INaL increased the Ca(2+) load and AP duration (APD). SEA0400 and ORM10103 suppressed INCX and prevented/reversed the anemone toxin II (ATX-II)-induced [Ca(2+) ]i rise without influencing APD, CaT or cell shortening, or affecting the ATX-II-induced increased APD. ORM10103 significantly decreased the number of strophanthidin-induced spontaneous diastolic Ca(2+) release events; however, SEA0400 failed to restrict the veratridine-induced augmentation in Purkinje-ventricle APD dispersion. CONCLUSIONS AND IMPLICATIONS Selective NCX inhibition - presumably by blocking rev INCX (reverse mode NCX current) - is effective against arrhythmogenesis caused by [Na(+) ]i -induced [Ca(2+) ]i elevation, without influencing the AP waveform. Therefore, selective INCX inhibition, by significantly reducing the arrhythmogenic trigger activity caused by the perturbed Ca(2+) i handling, should be considered as a promising anti-arrhythmic therapeutic strategy.
Collapse
Affiliation(s)
- Norbert Nagy
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Morita N, Mandel WJ, Kobayashi Y, Karagueuzian HS. Cardiac fibrosis as a determinant of ventricular tachyarrhythmias. J Arrhythm 2014; 30:389-394. [PMID: 25642299 DOI: 10.1016/j.joa.2013.12.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Animal and emerging clinical studies have demonstrated that increased ventricular fibrosis in a setting of reduced repolarization reserve promotes early afterdepolarizations (EADs) and triggered activity that can initiate ventricular tachycardia and ventricular fibrillation (VT/VF). Increased ventricular fibrosis plays a key facilitatory role in allowing oxidative and metabolic stress-induced EADs to manifest as triggered activity causing VT/VF. The lack of such an arrhythmogenic effect by the same stressors in normal non-fibrotic hearts highlights the importance of fibrosis in the initiation of VT/VF. These findings suggest that antifibrotic therapy combined with therapy designed to increase ventricular repolarization reserve may act synergistically to reduce the risk of sudden cardiac death.
Collapse
Affiliation(s)
- Norishige Morita
- Division of Cardiology, Department of Medicine, Tokai University Hachioji Hospital, Tokyo, Japan
| | - William J Mandel
- Translational Arrhythmia Research Section, Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Yoshinori Kobayashi
- Division of Cardiology, Department of Medicine, Tokai University Hachioji Hospital, Tokyo, Japan
| | - Hrayr S Karagueuzian
- Division of Cardiology, Department of Medicine, Tokai University Hachioji Hospital, Tokyo, Japan
| |
Collapse
|
23
|
Behn C, Dinamarca GA, De Gregorio NF, Lips V, Vivaldi EA, Soza D, Guerra MA, Jiménez RF, Lecannelier EA, Varela H, Silva-Urra JA. Age-Related Arrhythmogenesis on Ascent and Descent: “Autonomic Conflicts” on Hypoxia/Reoxygenation at High Altitude? High Alt Med Biol 2014; 15:356-63. [DOI: 10.1089/ham.2013.1092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Claus Behn
- Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
- Mutual de Seguridad CChC, Chile
| | | | | | - Viviana Lips
- Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Ennio A Vivaldi
- Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | | | | | - Raúl F Jiménez
- Facultad de Ciencias Físicas y Matemáticas, Universidad de Concepción, Concepción, Chile
| | | | - Héctor Varela
- Facultad de Ciencias Básicas, Universidad de Antofagasta, Antofagasta, Chile
| | - Juan A Silva-Urra
- Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| |
Collapse
|
24
|
Driessen HE, Bourgonje VJA, van Veen TAB, Vos MA. New antiarrhythmic targets to control intracellular calcium handling. Neth Heart J 2014; 22:198-213. [PMID: 24733689 PMCID: PMC4016334 DOI: 10.1007/s12471-014-0549-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sudden cardiac death due to ventricular arrhythmias is a major problem. Drug therapies to prevent SCD do not provide satisfying results, leading to the demand for new antiarrhythmic strategies. New targets include Ca2+/Calmodulin-dependent protein kinase II (CaMKII), the Na/Ca exchanger (NCX), the Ryanodine receptor (RyR, and its associated protein FKBP12.6 (Calstabin)) and the late component of the sodium current (INa-Late), all related to intracellular calcium (Ca2+) handling. In this review, drugs interfering with these targets (SEA-0400, K201, KN-93, W7, ranolazine, sophocarpine, and GS-967) are evaluated and their future as clinical compounds is considered. These new targets prove to be interesting; however more insight into long-term drug effects is necessary before clinical applicability becomes reality.
Collapse
Affiliation(s)
- H E Driessen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM, Utrecht, the Netherlands,
| | | | | | | |
Collapse
|
25
|
Lee HL, Lu CH, Chang PC, Chou CC, Wo HT, Wen MS. Contrasting effects of HMR1098 on arrhythmogenicity in a Langendorff-perfused phase-2 myocardial infarction rabbit model. Pacing Clin Electrophysiol 2014; 37:1058-66. [PMID: 24645834 DOI: 10.1111/pace.12381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/05/2014] [Accepted: 01/28/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND The stability of dynamic factors has been reported to play a role in the antiarrhythmic actions of adenosine triphosphate (ATP)-sensitive potassium channel (KATP) opener in phase-2 myocardial infarction (MI) hearts. In the situation of the downregulation of KATP, the effects of KATP blocker (HMR1098) on the dynamic factors and electrophysiological changes during phase-2 MI remain unclear. METHODS Dual voltage and intracellular Ca(2+) (Cai) optical mapping was performed in nine Langendorff-perfused hearts 4-5 hours after coronary artery ligation and five control hearts. Electrophysiology studies, including action potential duration (APD) restitution, conduction velocity (CV), inducibility of ventricular fibrillation (VF), VF dominant frequency, APD and Cai alternans, and Cai decay, were performed. The same protocol was repeated in the presence of HMR1098 (10 μm) after the baseline studies. RESULTS HMR1098 significantly prolonged APD and effective refractory period to prevent sustained VF in five of nine MI hearts and two of five control hearts compared to none at baseline in both groups. On the other hand, HMR1098 steepened APD restitution slope to enhance spatially concordant alternans in both groups. In the phase-2 MI group, HMR1098 steepened CV restitution slope and enhanced spatially discordant alternans (SDA), which might account for a decreased pacing threshold of VF induction during HMR1098 infusion in phase-2 MI hearts. CONCLUSIONS In phase-2 MI hearts, HMR1098 has contrasting effects on arrhythmogenesis, suppressing reentry and VF persistence but facilitating VF inducibility. The mechanism is the intensified induction of SDA because of the steepened APD and CV restitution slopes.
Collapse
Affiliation(s)
- Hui-Ling Lee
- Department of Anesthesia, Chang Gung Memorial Hospital, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
26
|
Pezhouman A, Madahian S, Stepanyan H, Ghukasyan H, Qu Z, Belardinelli L, Karagueuzian HS. Selective inhibition of late sodium current suppresses ventricular tachycardia and fibrillation in intact rat hearts. Heart Rhythm 2013; 11:492-501. [PMID: 24291413 DOI: 10.1016/j.hrthm.2013.11.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Indexed: 01/02/2023]
Abstract
BACKGROUND Enhanced late inward Na current (INa-L) modulates action potential duration (APD) and plays a key role in the genesis of early afterdepolarizations (EADs) and delayed afterdepolarizations (DADs) and triggered activity. OBJECTIVE The purpose of this study was to define the influence of selective block of INa-L on EAD- and DAD-mediated triggered ventricular tachycardia (VT) and ventricular fibrillation (VF) in intact hearts using (GS967), a selective and potent (IC50 = 0.13 ± 0.01 μM) blocker of INa-L. METHODS VT/VF were induced either by local aconitine injection (50 μg) in the left ventricular muscle of adult (3-4 months) male rats (N = 21) or by arterial perfusion of 0.1 mM hydrogen peroxide (H2O2) in aged male rats (24-26 months, N = 16). The left ventricular epicardial surface of the isolated-perfused hearts was optically mapped using fluorescent voltage-sensitive dye, and microelectrode recordings of action potentials were made adjacent to the aconitine injection site. The suppressive and preventive effects of GS967 (1 μM) against EAD/DAD-mediated VT/VF were then determined. RESULTS Aconitine induced VT in all 13 hearts studied. Activation map (N = 6) showed that the VT was initiated by a focal activity arising from the aconitine injection site (cycle length [CL] 84 ± 12) that degenerated to VF (CL 52 ± 8 ms) within a few seconds. VF was maintained by multifocal activity with occasional incomplete reentrant wavefronts. Administration of GS967 suppressed the VT/VF in 10 of 13 hearts (P < .001). Preexposure to GS967 for 15 minutes before aconitine injection prevented initiation of VT/VF in 5 of 8 additional hearts (P < .02). VF reoccurred within 10 minutes on washout of GS967. Microelectrode recordings (N = 7) showed that VT/VF was initiated by EAD- and DAD-mediated triggered activity at CL of 86 ± 14 ms (NS from VT CL) that preceded the VF. GS967 shortened APD, flattened the slope of the dynamic APD restitution curve, and reduced APD dispersion from 42 ± 12 ms to 8 ± 3 ms (P < .01). H2O2 perfusion in eight fibrotic aged hearts promoted EAD-mediated focal VT/VF, which was suppressed by GS967 in five hearts (P < .02). CONCLUSION The selective INa-L blocker GS967 effectively suppresses and prevents aconitine and oxidative stress-induced EADs, DADs, and focal VT/VF. Suppression of EADs, DADs, and reduction of APD dispersion make GS967 a potentially useful antiarrhythmic drug in conditions of enhanced INa-L.
Collapse
Affiliation(s)
- Arash Pezhouman
- Translational Arrhythmia Research Section, UCLA Cardiovascular Research Laboratory, Department of Medicine Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Sepideh Madahian
- Translational Arrhythmia Research Section, UCLA Cardiovascular Research Laboratory, Department of Medicine Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Hayk Stepanyan
- Translational Arrhythmia Research Section, UCLA Cardiovascular Research Laboratory, Department of Medicine Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Hayk Ghukasyan
- Translational Arrhythmia Research Section, UCLA Cardiovascular Research Laboratory, Department of Medicine Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Zhilin Qu
- Translational Arrhythmia Research Section, UCLA Cardiovascular Research Laboratory, Department of Medicine Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Hrayr S Karagueuzian
- Translational Arrhythmia Research Section, UCLA Cardiovascular Research Laboratory, Department of Medicine Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California.
| |
Collapse
|
27
|
Karagueuzian HS. Letter to the editor: “The role of short QT interval and elevated LV end-diastolic pressure in the genesis of ventricular tachycardia and fibrillation”. Am J Physiol Heart Circ Physiol 2013; 305:H1405. [DOI: 10.1152/ajpheart.00581.2013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
28
|
Pizzuto MF, Suzuki G, Banas MD, Heavey B, Fallavollita JA, Canty JM. Reply to “Letter to the editor: ‘The role of short QT interval and elevated LV end-diastolic pressure in the genesis of ventricular tachycardia and fibrillation’”. Am J Physiol Heart Circ Physiol 2013; 305:H1406. [DOI: 10.1152/ajpheart.00659.2013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Matthew F. Pizzuto
- Center for Research in Cardiovascular Medicine, University at Buffalo, Buffalo, New York
| | - Gen Suzuki
- Center for Research in Cardiovascular Medicine, University at Buffalo, Buffalo, New York
- Department of Medicine, University at Buffalo, Buffalo, New York
| | - Michael D. Banas
- Center for Research in Cardiovascular Medicine, University at Buffalo, Buffalo, New York
- Department of Medicine, University at Buffalo, Buffalo, New York
| | - Brendan Heavey
- Center for Research in Cardiovascular Medicine, University at Buffalo, Buffalo, New York
| | - James A. Fallavollita
- Veterans Affairs Western New York Health Care System, Buffalo, New York
- Center for Research in Cardiovascular Medicine, University at Buffalo, Buffalo, New York
- Department of Medicine, University at Buffalo, Buffalo, New York
| | - John M. Canty
- Veterans Affairs Western New York Health Care System, Buffalo, New York
- Center for Research in Cardiovascular Medicine, University at Buffalo, Buffalo, New York
- Department of Medicine, University at Buffalo, Buffalo, New York
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, New York; and
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| |
Collapse
|
29
|
Qu Z, Xie LH, Olcese R, Karagueuzian HS, Chen PS, Garfinkel A, Weiss JN. Early afterdepolarizations in cardiac myocytes: beyond reduced repolarization reserve. Cardiovasc Res 2013; 99:6-15. [PMID: 23619423 DOI: 10.1093/cvr/cvt104] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Early afterdepolarizations (EADs) are secondary voltage depolarizations during the repolarizing phase of the action potential, which can cause lethal cardiac arrhythmias. The occurrence of EADs requires a reduction in outward current and/or an increase in inward current, a condition called reduced repolarization reserve. However, this generalized condition is not sufficient for EAD genesis and does not explain the voltage oscillations manifesting as EADs. Here, we summarize recent progress that uses dynamical theory to build on and advance our understanding of EADs beyond the concept of repolarization reserve, towards the goal of developing a holistic and integrative view of EADs and their role in arrhythmogenesis. We first introduce concepts from nonlinear dynamics that are relevant to EADs, namely, Hopf bifurcation leading to oscillations and basin of attraction of an equilibrium or oscillatory state. We then present a theory of phase-2 EADs in nonlinear dynamics, which includes the formation of quasi-equilibrium states at the plateau voltage, their stabilities, and the bifurcations leading to and terminating the oscillations. This theory shows that the L-type calcium channel plays a unique role in causing the nonlinear dynamical behaviours necessary for EADs. We also summarize different mechanisms of phase-3 EADs. Based on the dynamical theory, we discuss the roles of each of the major ionic currents in the genesis of EADs, and potential therapeutic targets.
Collapse
Affiliation(s)
- Zhilin Qu
- Department of Medicine , David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Sovari AA, Rutledge CA, Jeong EM, Dolmatova E, Arasu D, Liu H, Vahdani N, Gu L, Zandieh S, Xiao L, Bonini MG, Duffy HS, Dudley SC. Mitochondria oxidative stress, connexin43 remodeling, and sudden arrhythmic death. Circ Arrhythm Electrophysiol 2013; 6:623-31. [PMID: 23559673 DOI: 10.1161/circep.112.976787] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Previously, we showed that a mouse model (ACE8/8) of cardiac renin-angiotensin system activation has a high rate of spontaneous ventricular tachycardia and sudden cardiac death secondary to a reduction in connexin43 level. Angiotensin-II activation increases reactive oxygen species (ROS) production, and ACE8/8 mice show increased cardiac ROS. We sought to determine the source of ROS and whether ROS played a role in the arrhythmogenesis. METHODS AND RESULTS Wild-type and ACE8/8 mice with and without 2 weeks of treatment with L-NIO (NO synthase inhibitor), sepiapterin (precursor of tetrahydrobiopterin), MitoTEMPO (mitochondria-targeted antioxidant), TEMPOL (a general antioxidant), apocynin (nicotinamide adenine dinucleotide phosphate oxidase inhibitor), allopurinol (xanthine oxidase inhibitor), and ACE8/8 crossed with P67 dominant negative mice to inhibit the nicotinamide adenine dinucleotide phosphate oxidase were studied. Western blotting, detection of mitochondrial ROS by MitoSOX Red, electron microscopy, immunohistochemistry, fluorescent dye diffusion technique for functional assessment of connexin43, telemetry monitoring, and in vivo electrophysiology studies were performed. Treatment with MitoTEMPO reduced sudden cardiac death in ACE8/8 mice (from 74% to 18%; P<0.005), decreased spontaneous ventricular premature beats, decreased ventricular tachycardia inducibility (from 90% to 17%; P<0.05), diminished elevated mitochondrial ROS to the control level, prevented structural damage to mitochondria, resulted in 2.6-fold increase in connexin43 level at the gap junctions, and corrected gap junction conduction. None of the other antioxidant therapies prevented ventricular tachycardia and sudden cardiac death in ACE8/8 mice. CONCLUSIONS Mitochondrial oxidative stress plays a central role in angiotensin II-induced gap junction remodeling and arrhythmia. Mitochondria-targeted antioxidants may be effective antiarrhythmic drugs in cases of renin-angiotensin system activation.
Collapse
Affiliation(s)
- Ali A Sovari
- Section of Cardiology and Center for Cardiovascular Research, University of Illinois at Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Karagueuzian HS, Stepanyan H, Mandel WJ. Bifurcation theory and cardiac arrhythmias. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2013; 3:1-16. [PMID: 23459417 PMCID: PMC3584649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 01/16/2013] [Indexed: 06/01/2023]
Abstract
In this paper we review two types of dynamic behaviors defined by the bifurcation theory that are found to be particularly useful in describing two forms of cardiac electrical instabilities that are of considerable importance in cardiac arrhythmogenesis. The first is action potential duration (APD) alternans with an underlying dynamics consistent with the period doubling bifurcation theory. This form of electrical instability could lead to spatially discordant APD alternans leading to wavebreak and reentrant form of tachyarrhythmias. Factors that modulate the APD alternans are discussed. The second form of bifurcation of importance to cardiac arrhythmogenesis is the Hopf-homoclinic bifurcation that adequately describes the dynamics of the onset of early afterdepolarization (EAD)-mediated triggered activity (Hopf) that may cause ventricular tachycardia and ventricular fibrillation (VT/VF respectively). The self-termination of the triggered activity is compatible with the homoclinic bifurcation. Ionic and intracellular calcium dynamics underlying these dynamics are discussed using available experimental and simulation data. The dynamic analysis provides novel insights into the mechanisms of VT/VF, a major cause of sudden cardiac death in the US.
Collapse
Affiliation(s)
- Hrayr S Karagueuzian
- Translational Arrhythmia Research Section, UCLA Cardiovascular Research Laboratory and the Division of Cardiology, Departments of Medicine David Geffen School of Medicine at UCLA Los Angeles, California
| | | | | |
Collapse
|
32
|
Karagueuzian HS, Nguyen TP, Qu Z, Weiss JN. Oxidative stress, fibrosis, and early afterdepolarization-mediated cardiac arrhythmias. Front Physiol 2013; 4:19. [PMID: 23423152 PMCID: PMC3573324 DOI: 10.3389/fphys.2013.00019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 01/25/2013] [Indexed: 01/06/2023] Open
Abstract
Animal and clinical studies have demonstrated that oxidative stress, a common pathophysiological factor in cardiac disease, reduces repolarization reserve by enhancing the L-type calcium current, the late Na, and the Na-Ca exchanger, promoting early afterdepolarizations (EADs) that can initiate ventricular tachycardia and ventricular fibrillation (VT/VF) in structurally remodeled hearts. Increased ventricular fibrosis plays a key facilitatory role in allowing oxidative-stress induced EADs to manifest as triggered activity and VT/VF, since normal non-fibrotic hearts are resistant to arrhythmias when challenged with similar or higher levels of oxidative stress. The findings imply that antifibrotic therapy, in addition to therapies designed to suppress EAD formation at the cellular level, may be synergistic in reducing the risk of sudden cardiac death.
Collapse
Affiliation(s)
- Hrayr S Karagueuzian
- Cardiovascular Research Laboratory, Translational Arrhythmia Research Section, David Geffen School of Medicine at UCLA Los Angeles, CA, USA
| | | | | | | |
Collapse
|
33
|
Mirza M, Strunets A, Shen WK, Jahangir A. Mechanisms of arrhythmias and conduction disorders in older adults. Clin Geriatr Med 2013; 28:555-73. [PMID: 23101571 DOI: 10.1016/j.cger.2012.08.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Aging is associated with an increased prevalence of cardiac arrhythmias, which contribute to higher morbidity and mortality in the elderly. The frequency of cardiac arrhythmias, particularly atrial fibrillation and ventricular tachyarrhythmia, is projected to increase as the population ages, greatly impacting health care resource utilization. Several clinical factors associated with the risk of arrhythmias have been identified in the population, yet the molecular bases for the increased predisposition to arrhythmogenesis in the elderly are not fully understood. This review highlights the epidemiology of cardiac dysrhythmias, changes in cardiac structure and function associated with aging, and the basis for arrhythmogenesis in the elderly.
Collapse
Affiliation(s)
- Mahek Mirza
- Center for Integrative Research on Cardiovascular Aging (CIRCA), Aurora University of Wisconsin Medical Group, Aurora Health Care, 3033 South 27th Street, Milwaukee, WI 53215, USA
| | | | | | | |
Collapse
|
34
|
Bao L, Taskin E, Foster M, Ray B, Rosario R, Ananthakrishnan R, Howlett SE, Schmidt AM, Ramasamy R, Coetzee WA. Alterations in ventricular K(ATP) channel properties during aging. Aging Cell 2013; 12:167-76. [PMID: 23173756 DOI: 10.1111/acel.12033] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2012] [Indexed: 12/27/2022] Open
Abstract
Coronary heart disease remains the principle cause of mortality in the United States. During aging, the efficiency of the cardiovascular system is decreased and the aged heart is less tolerant to ischemic injury. ATP-sensitive K(+) (K(ATP)) channels protect the myocardium against ischemic damage. We investigated how aging affects cardiac K(ATP) channels in the Fischer 344 rat model. Expression of K(ATP) channel subunit mRNA and protein levels was unchanged in hearts from 26-month-old vs. 4-month-old rats. Interestingly, the mRNA expression of several other ion channels (> 80) was also largely unchanged, suggesting that posttranscriptional regulatory mechanisms occur during aging. The whole-cell K(ATP) channel current density was strongly diminished in ventricular myocytes from aged male rat hearts (also observed in aged C57BL/6 mouse myocytes). Experiments with isolated patches (inside-out configuration) demonstrated that the K(ATP) channel unitary conductance was unchanged, but that the inhibitory effect of cytosolic ATP on channel activity was enhanced in the aged heart. The mean patch current was diminished, consistent with the whole-cell data. We incorporated these findings into an empirical model of the K(ATP) channel and numerically simulated the effects of decreased cytosolic ATP levels on the human action potential. This analysis predicts lesser activation of K(ATP) channels by metabolic impairment in the aged heart and a diminished action potential shortening. This study provides insights into the changes in K(ATP) channels during aging and suggests that the protective role of these channels during ischemia is significantly compromised in the aged individual.
Collapse
Affiliation(s)
- Li Bao
- Pediatrics; NYU School of Medicine; New York; NY; USA
| | - Eylem Taskin
- Pediatrics; NYU School of Medicine; New York; NY; USA
| | | | - Beevash Ray
- Medicine; NYU School of Medicine; New York; NY; USA
| | - Rosa Rosario
- Medicine; NYU School of Medicine; New York; NY; USA
| | | | - Susan E. Howlett
- Department of Pharmacology; Dalhousie University; 5850 College Street; PO Box 15000; Sir Charles Tupper Medical Building; Halifax; Nova Scotia; Canada; B3H 4R2
| | | | | | | |
Collapse
|
35
|
|
36
|
Swaminathan PD, Purohit A, Hund TJ, Anderson ME. Calmodulin-dependent protein kinase II: linking heart failure and arrhythmias. Circ Res 2012; 110:1661-77. [PMID: 22679140 DOI: 10.1161/circresaha.111.243956] [Citation(s) in RCA: 226] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Understanding relationships between heart failure and arrhythmias, important causes of suffering and sudden death, remains an unmet goal for biomedical researchers and physicians. Evidence assembled over the past decade supports a view that activation of the multifunctional Ca(2+) and calmodulin-dependent protein kinase II (CaMKII) favors myocardial dysfunction and cell membrane electrical instability. CaMKII activation follows increases in intracellular Ca(2+) or oxidation, upstream signals with the capacity to transition CaMKII into a Ca(2+) and calmodulin-independent constitutively active enzyme. Constitutively active CaMKII appears poised to participate in disease pathways by catalyzing the phosphorylation of classes of protein targets important for excitation-contraction coupling and cell survival, including ion channels and Ca(2+) homeostatic proteins, and transcription factors that drive hypertrophic and inflammatory gene expression. This rich diversity of downstream targets helps to explain the potential for CaMKII to simultaneously affect mechanical and electrical properties of heart muscle cells. Proof-of-concept studies from a growing number of investigators show that CaMKII inhibition is beneficial for improving myocardial performance and for reducing arrhythmias. We review the molecular physiology of CaMKII and discuss CaMKII actions at key cellular targets and results of animal models of myocardial hypertrophy, dysfunction, and arrhythmias that suggest CaMKII inhibition may benefit myocardial function while reducing arrhythmias.
Collapse
Affiliation(s)
- Paari Dominic Swaminathan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
37
|
Rodrigo R. Prevention of postoperative atrial fibrillation: novel and safe strategy based on the modulation of the antioxidant system. Front Physiol 2012; 3:93. [PMID: 22518106 PMCID: PMC3325031 DOI: 10.3389/fphys.2012.00093] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Accepted: 03/26/2012] [Indexed: 01/15/2023] Open
Abstract
Postoperative atrial fibrillation (AF) is the most common arrhythmia following cardiac surgery with extracorporeal circulation. The pathogenesis of postoperative AF is multifactorial. Oxidative stress, caused by the unavoidable ischemia-reperfusion event occurring in this setting, is a major contributory factor. Reactive oxygen species (ROS)-derived effects could result in lipid peroxidation, protein carbonylation, or DNA oxidation of cardiac tissue, thus leading to functional and structural myocardial remodeling. The vulnerability of myocardial tissue to the oxidative challenge is also dependent on the activity of the antioxidant system. High ROS levels, overwhelming this system, should result in deleterious cellular effects, such as the induction of necrosis, apoptosis, or autophagy. Nevertheless, tissue exposure to low to moderate ROS levels could trigger a survival response with a trend to reinforce the antioxidant defense system. Administration of n-3 polyunsaturated fatty acids (PUFA), known to involve a moderate ROS production, is consistent with a diminished vulnerability to the development of postoperative AF. Accordingly, supplementation of n-3 PUFA successfully reduced the incidence of postoperative AF after coronary bypass grafting. This response is due to an up-regulation of antioxidant enzymes, as shown in experimental models. In turn, non-enzymatic antioxidant reinforcement through vitamin C administration prior to cardiac surgery has also reduced the postoperative AF incidence. Therefore, it should be expected that a mixed therapy result in an improvement of the cardioprotective effect by modulating both components of the antioxidant system. We present novel available evidence supporting the hypothesis of an effective prevention of postoperative AF including a two-step therapeutic strategy: n-3 PUFA followed by vitamin C supplementation to patients scheduled for cardiac surgery with extracorporeal circulation. The present study should encourage the design of clinical trials aimed to test the efficacy of this strategy to offer new therapeutic opportunities to patients challenged by ischemia-reperfusion events not solely in heart, but also in other organs such as kidney or liver in transplantation surgeries.
Collapse
Affiliation(s)
- Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of ChileSantiago, Chile
| |
Collapse
|
38
|
Bapat A, Nguyen TP, Lee JH, Sovari AA, Fishbein MC, Weiss JN, Karagueuzian HS. Enhanced sensitivity of aged fibrotic hearts to angiotensin II- and hypokalemia-induced early afterdepolarization-mediated ventricular arrhythmias. Am J Physiol Heart Circ Physiol 2012; 302:H2331-40. [PMID: 22467308 DOI: 10.1152/ajpheart.00094.2012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Unlike young hearts, aged hearts are highly susceptible to early afterdepolarization (EAD)-mediated ventricular fibrillation (VF). This differential may result from age-related structural remodeling (fibrosis) or electrical remodeling of ventricular myocytes or both. We used optical mapping and microelectrode recordings in Langendorff-perfused hearts and patch-clamp recordings in isolated ventricular myocytes from aged (24-26 mo) and young (3-4 mo) rats to assess susceptibility to EADs and VF during either oxidative stress with ANG II (2 μM) or ionic stress with hypokalemia (2.7 mM). ANG II caused EAD-mediated VF in 16 of 19 aged hearts (83%) after 32 ± 7 min but in 0 of 9 young hearts (0%). ANG II-mediated VF was suppressed with KN-93 (Ca(2+)/calmodulin-dependent kinase inhibitor) and the reducing agent N-acetylcysteine. Hypokalemia caused EAD-mediated VF in 11 of 11 aged hearts (100%) after 7.4 ± 0.4 min. In 14 young hearts, however, VF did not occur in 6 hearts (43%) or was delayed in onset (31 ± 22 min, P < 0.05) in 8 hearts (57%). In patch-clamped myocytes, ANG II and hypokalemia (n = 6) induced EADs and triggered activity in both age groups (P = not significant) at a cycle length of >0.5 s. When myocytes of either age group were coupled to a virtual fibroblast using the dynamic patch-clamp technique, EADs arose in both groups at a cycle length of <0.5 s. Aged ventricles had significantly greater fibrosis and reduced connexin43 gap junction density compared with young hearts. The lack of differential age-related sensitivity at the single cell level in EAD susceptibility indicates that increased ventricular fibrosis in the aged heart plays a key role in increasing vulnerability to VF induced by oxidative and ionic stress.
Collapse
Affiliation(s)
- Aneesh Bapat
- Translational Arrhythmia Research Section, University of California-Los Angeles Cardiovascular Research Laboratory, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Sovari AA, Karagueuzian HS. Myocardial fibrosis as a risk stratifier for sudden arrhythmic death. Expert Rev Cardiovasc Ther 2012; 9:951-3. [PMID: 21878039 DOI: 10.1586/erc.11.103] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Alternative strategies in arrhythmia therapy: evaluation of Na/Ca exchange as an anti-arrhythmic target. Pharmacol Ther 2011; 134:26-42. [PMID: 22197992 DOI: 10.1016/j.pharmthera.2011.12.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 11/22/2011] [Accepted: 11/22/2011] [Indexed: 01/08/2023]
Abstract
The search for alternative anti-arrhythmic strategies is fueled by an unmet medical need as well as by the opportunities arising from identification of novel targets and novel drugs. Na/Ca exchange is a potential target involved in several types of arrhythmias, such as those related to ischemia-reperfusion, heart failure and also some forms of genetic arrhythmias. Inhibition of Na/Ca exchange is theoretically not only anti-arrhythmic but also increases cellular Ca(2+) content. This could be an advantage in conditions of low inotropy, such as in heart failure, but may also worsen conditions such as the recovery from ischemia or relaxation abnormalities. With the available drugs such as KB-R7943 and SEA-0400 these theories have now been tested in a number of cellular and in vivo models. Experience is overall rather positive and seems less hampered by the potential drawbacks than expected. This may be because the currently available drugs are not highly selective, with additional benefit derived from concurrent effects. While this precludes a definite answer regarding the benefit of a pure NCX inhibitor, they indicate that Na/Ca exchange inhibition as part of a multi-target strategy is an avenue to be considered. Such studies will need further 'bench' work and testing in relevant preclinical models, including chronic disease.
Collapse
|
41
|
Numata A, Miyauchi Y, Ono N, Fishbein MC, Mandel WJ, Lin SF, Weiss JN, Chen PS, Karagueuzian HS. Spontaneous atrial fibrillation initiated by tyramine in canine atria with increased sympathetic nerve sprouting. J Cardiovasc Electrophysiol 2011; 23:415-22. [PMID: 22034958 DOI: 10.1111/j.1540-8167.2011.02197.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Chronic left ventricular myocardial infarction (LVMI) promotes atrial and pulmonary veins (PV) sympathetic nerve sprouting. OBJECTIVES To test the hypothesis that sympathetic stimulation with tyramine initiates atrial fibrillation (AF) by early after depolarization (EAD)-mediated triggered activity at the left atrial PV (LAPV) junction. METHODS LVMI was created in 6 dogs and 6 dogs served as controls. Six to 8 weeks later the activation pattern of the isolated LAPV was optically mapped using dual voltage and intracellular Ca(+2) (Ca(i) (2+) )-sensitive epifluorescent dyes before and after tyramine (5 μM) perfusion. RESULTS Tyramine initiated spontaneous AF in 5 of 6 atria but none in the control group (P < 0.01). The AF was initiated by late phase 3 EAD-mediated triggered activity that arose from the LAPV junction causing functional conduction block in LA, reentry, and AF. The AF was subsequently maintained by mixed reentrant and focal mechanisms. The EADs arose during the late phase 3, when the Ca(i) (2+) level was 64 ± 12% of the peak systolic Ca(i) (2+) transient amplitude, a property caused by tyramine's simultaneous shortening of the action potential duration and lengthening of the Ca(i) (2+) transient duration in the LVMI group but not in the control. Tyrosine hydroxylase and growth associated protein 43 positive nerve sprouts were significantly increased in the sinus node, LAA, and the LSPV in the LVMI group compared to control (P < 0.01). CONCLUSIONS Increased atrial sympathetic nerve sprouts after LVMI makes the LAPV junction susceptible to late phase 3 EAD-mediated triggered and AF during sympathetic stimulation with tyramine.
Collapse
Affiliation(s)
- Ayaka Numata
- Division of Cardiovascular Medicine, Ohashi Medical Center, Toho University, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Karagueuzian HS. Targeting cardiac fibrosis: a new frontier in antiarrhythmic therapy? AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2011; 1:101-109. [PMID: 22254191 PMCID: PMC3253497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 05/29/2011] [Indexed: 05/31/2023]
Abstract
Cardiac fibrosis is known to alter cardiac conduction and promote reentry. Recent evidence indicates that fibrosis characterized by increased interstitial collagen accumulation and increased myofibroblast proliferation also promotes enhanced automaticity and early afterdepolarizations (EADs) causing triggered activity. Fibrosis then becomes an effective therapeutic target for the management of lethal cardiac arrhythmias. While oxidative stress with hydrogen peroxide (H(2)O(2)) is shown to readily promote EADs and triggered activity in isolated rat and rabbit ventricular myocytes however, this same stress fails to cause EADs in well-coupled, non-fibrotic hearts due to source-to-sink mismatches arising from cell-to-cell coupling. The triggered activity in the aged fibrotic hearts causes focal ventricular tachycardia (VT) that degenerates within seconds to ventricular fibrillation (VF) after the emergence of spatially discordant action potential duration alternans leading to wavebreak, reentry and VF. Computer simulations in 2D tissue incorporating variable degrees of fibrosis showed that intermediate (but not mild or very severe) fibrosis promoted EADs and TA. Human studies have shown that myocardial fibrosis was an independent predictor for arrhythmias including sustained VT and VF. A variety of drug classes including, torsemide, a loop diuretic, that inhibits the enzyme involved in the myocardial extracellular generation of collagen type I molecules and the inhibitors of the renin-angiotensin-aldosterone system (RAAS), the mineralocorticoid receptors and endothelin receptors reduce cardiac fibrosis with reduction of myocardial stiffness and improved ventricular function. It is hoped that in the near future effective antifibrotic drug regimen would be developed to reduce the risk of fibrosis related VT and VF.
Collapse
Affiliation(s)
- Hrayr S Karagueuzian
- Translational Arrhythmia Research Section, UCLA Cardiovascular Research Laboratory and the Division of Cardiology, Departments of Medicine, David Geffen School of Medicine at UCLA Los Angeles, California, USA
| |
Collapse
|