1
|
van Bavel JJA, Pham C, Beekman HDM, Houtman MJC, Bossu A, Sparidans RW, van der Heyden MAG, Vos MA. PI3K/mTOR inhibitor omipalisib prolongs cardiac repolarization along with a mild proarrhythmic outcome in the AV block dog model. Front Cardiovasc Med 2022; 9:956538. [PMID: 35990966 PMCID: PMC9381882 DOI: 10.3389/fcvm.2022.956538] [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: 05/30/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background The phosphoinositide 3-kinase (PI3K) signaling pathway is an interesting target in cancer treatment. The awareness of the proarrhythmic risk of PI3K inhibitors was raised because PI3K is also involved in regulating signaling toward cardiac ion channels. Canine cardiomyocytes treated with PI3K inhibitors show an increased action potential duration and reduced cardiac repolarizing currents. Now, the potential proarrhythmic effect of chronic treatment of PI3K/mTOR inhibitor GSK2126458 (omipalisib) was investigated in the atrioventricular (AV) block dog model. Methods Purpose-bred Mongrel dogs received complete AV block by ablation of the bundle of His and their hearts were paced in the right ventricular apex at VDD-mode (RVA-VDD). In this way, sinus rhythm was maintained for 15 ± 1 days and thereby bradycardia-induced cardiac remodeling was prevented. Dogs received 1 mg/kg omipalisib once (n = 3) or twice (n = 10) a day via oral administration for 7 days. Under standardized conditions (anesthesia, bradycardia at 60 beats/min, and a dofetilide challenge), potential proarrhythmic effects of omipalisib were investigated. Results Twice daily dosing of omipalisib increased accumulative plasma levels compared to once daily dosing accompanied with adverse events. Omipalisib prolonged the QT interval at baseline and more strongly after the dofetilide challenge (490 ± 37 to 607 ± 48 ms). The arrhythmic outcome after omipalisib resulted in single ectopic beats in 30% of dogs perpetuating in multiple ectopic beats and TdP arrhythmia in 20% of dogs. Isolated ventricular cardiomyocytes from omipalisib-treated dogs showed a diminished IKs current density. Conclusion Chronic treatment of PI3K/mTOR inhibitor omipalisib prolonged the QT interval in a preclinical model under standardized proarrhythmic conditions. Furthermore, this study showed that electrical remodeling induced by omipalisib had a mild proarrhythmic outcome.
Collapse
Affiliation(s)
- J. J. A. van Bavel
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - C. Pham
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - H. D. M. Beekman
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - M. J. C. Houtman
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - A. Bossu
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - R. W. Sparidans
- Division Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - M. A. G. van der Heyden
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
- *Correspondence: M. A. G. van der Heyden
| | - M. A. Vos
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
2
|
Effect of hyperglycaemia in combination with moxifloxacin on cardiac repolarization in male and female patients with type I diabetes. Clin Res Cardiol 2022; 111:1147-1160. [PMID: 35596784 PMCID: PMC9525410 DOI: 10.1007/s00392-022-02037-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/05/2022] [Indexed: 11/20/2022]
Abstract
Background Patients with Type 1 diabetes mellitus have been shown to be at a two to ten-fold higher risk of sudden cardiac death (SCD) (Svane et al., Curr Cardiol 2020; 22:112) than the general population, but the underlying mechanism is unclear. Hyperglycaemia is a recognised cause of QTc prolongation; a state patients with type 1 diabetes are more prone to, potentially increasing their risk of ventricular arrhythmia. Understanding the QTc prolongation effect of both hyperglycaemia and the concomitant additive risk of commonly prescribed QTc-prolonging drugs such as Moxifloxacin may help to elucidate the mechanism of sudden cardiac death in this cohort. This single-blinded, placebo-controlled study investigated the extent to which hyperglycaemia prolongs the QTc in controlled conditions, and the potential additive risk of QTc-prolonging medications. Methods 21 patients with type 1 diabetes mellitus were enrolled to a placebo-controlled crossover study at a single clinical trials unit. Patients underwent thorough QTc assessment throughout the study. A ‘hyperglycaemic clamp’ of oral and intravenous glucose was administered with a target blood glucose of > 25 mM and maintained for 2 h on day 1 and day 3, alongside placebo on day 1 and moxifloxacin on day 3. Day 2 served as a control day between the two active treatment days. Thorough QTc assessment was conducted at matched time points over 3 days, and regular blood sampling was undertaken at matched time intervals for glucose levels and moxifloxacin exposure. Results Concentration-effect modelling showed that acute hyperglycaemia prolonged the QTc interval in female and male volunteers with type 1 diabetes by a peak mean increase of 13 ms at 2 h. Peak mean QTc intervals after the administration of intravenous Moxifloxacin during the hyperglycaemic state were increased by a further 9 ms at 2 h, to 22 ms across the entire study population. Regression analysis suggested this additional increase was additive, not exponential. Hyperglycaemia was associated with a significantly greater mean QTc-prolonging effect in females, but the mean peak increase with the addition of moxifloxacin was the same for males and females. This apparent sex difference was likely due to the exclusive use of basal insulin in the male patients, which provided a low level of exogenous insulin during the study assessments thereby mitigating the effects of hyperglycaemia on QTc. This effect was partially overcome by Moxifloxacin administration, suggesting both hyperglycaemia and moxifloxacin prolong QTc by different mechanisms, based on subinterval analysis. Conclusions Hyperglycaemia was found to be a significant cause of QTc prolongation and the additional effect of a QTc-prolonging positive control (moxifloxacin) was found to be additive. Given the high risk of sudden cardiac death in type 1 diabetes mellitus, extra caution should be exercised when prescribing any medication in this cohort for QTc effects, and further research needs to be undertaken to elucidate the exact mechanism underlying this finding and explore the potential prescribing risk in diabetes. Trial Registration NCT number: NCT01984827. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s00392-022-02037-8.
Collapse
|
3
|
Attachaipanich T, Chattipakorn SC, Chattipakorn N. Potential roles of sodium-glucose co-transporter 2 inhibitors in attenuating cardiac arrhythmias in diabetes and heart failure. J Cell Physiol 2022; 237:2404-2419. [PMID: 35324001 DOI: 10.1002/jcp.30727] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/02/2022] [Accepted: 03/12/2022] [Indexed: 12/12/2022]
Abstract
Sodium-glucose co-transporter 2 (SGLT-2) inhibitors are antidiabetic drugs that have been shown to exert cardiovascular benefits. Their benefits including a reduction of cardiovascular events and worsening heart failure have been extended to nondiabetic patients with high-risk. Although both heart failure and diabetes are known to increase risk of cardiac arrhythmias, the effects of SGLT-2 inhibitors on arrhythmia reduction and their underlying mechanisms are still not fully understood. This review aims to summarize the current available evidence ranging from basic research to clinical reports regarding the potential benefits of SGLT-2 inhibitors against cardiac arrhythmias. Previous in vitro and in vivo studies using various models including heart failure and diabetes are comprehensively summarized to examine the evidence of how SGLT-2 inhibitors affect cardiac action potential, cellular ion currents, calcium ion homeostasis, and cardiac mitochondrial function. Clinical reports investigating the association between SGLT-2 inhibitors and arrhythmias including atrial fibrillation and ventricular arrhythmias are also comprehensively summarized. Valuable information obtained from this review can be used to encourage further clinical investigations to warrant the potential use of SGLT-2 inhibitors against cardiac arrhythmias in both diabetic and heart failure settings.
Collapse
Affiliation(s)
- Tanawat Attachaipanich
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
4
|
Jaquenod De Giusti C, Palomeque J, Mattiazzi A. Ca 2+ mishandling and mitochondrial dysfunction: a converging road to prediabetic and diabetic cardiomyopathy. Pflugers Arch 2022; 474:33-61. [PMID: 34978597 PMCID: PMC8721633 DOI: 10.1007/s00424-021-02650-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/16/2022]
Abstract
Diabetic cardiomyopathy is defined as the myocardial dysfunction that suffers patients with diabetes mellitus (DM) in the absence of hypertension and structural heart diseases such as valvular or coronary artery dysfunctions. Since the impact of DM on cardiac function is rather silent and slow, early stages of diabetic cardiomyopathy, known as prediabetes, are poorly recognized, and, on many occasions, cardiac illness is diagnosed only after a severe degree of dysfunction was reached. Therefore, exploration and recognition of the initial pathophysiological mechanisms that lead to cardiac dysfunction in diabetic cardiomyopathy are of vital importance for an on-time diagnosis and treatment of the malady. Among the complex and intricate mechanisms involved in diabetic cardiomyopathy, Ca2+ mishandling and mitochondrial dysfunction have been described as pivotal early processes. In the present review, we will focus on these two processes and the molecular pathway that relates these two alterations to the earlier stages and the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Carolina Jaquenod De Giusti
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Médicas, UNLP, La Plata, Argentina
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Médicas, UNLP, La Plata, Argentina
| | - Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, CCT-La Plata-CONICET, Facultad de Cs. Médicas, UNLP, La Plata, Argentina.
| |
Collapse
|
5
|
Al Kury LT, Chacar S, Alefishat E, Khraibi AA, Nader M. Structural and Electrical Remodeling of the Sinoatrial Node in Diabetes: New Dimensions and Perspectives. Front Endocrinol (Lausanne) 2022; 13:946313. [PMID: 35872997 PMCID: PMC9302195 DOI: 10.3389/fendo.2022.946313] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/14/2022] [Indexed: 11/14/2022] Open
Abstract
The sinoatrial node (SAN) is composed of highly specialized cells that mandate the spontaneous beating of the heart through self-generation of an action potential (AP). Despite this automaticity, the SAN is under the modulation of the autonomic nervous system (ANS). In diabetes mellitus (DM), heart rate variability (HRV) manifests as a hallmark of diabetic cardiomyopathy. This is paralleled by an impaired regulation of the ANS, and by a pathological remodeling of the pacemaker structure and function. The direct effect of diabetes on the molecular signatures underscoring this pathology remains ill-defined. The recent focus on the electrical currents of the SAN in diabetes revealed a repressed firing rate of the AP and an elongation of its tracing, along with conduction abnormalities and contractile failure. These changes are blamed on the decreased expression of ion transporters and cell-cell communication ports at the SAN (i.e., HCN4, calcium and potassium channels, connexins 40, 45, and 46) which further promotes arrhythmias. Molecular analysis crystallized the RGS4 (regulator of potassium currents), mitochondrial thioredoxin-2 (reactive oxygen species; ROS scavenger), and the calcium-dependent calmodulin kinase II (CaMKII) as metabolic culprits of relaying the pathological remodeling of the SAN cells (SANCs) structure and function. A special attention is given to the oxidation of CaMKII and the generation of ROS that induce cell damage and apoptosis of diabetic SANCs. Consequently, the diabetic SAN contains a reduced number of cells with significant infiltration of fibrotic tissues that further delay the conduction of the AP between the SANCs. Failure of a genuine generation of AP and conduction of their derivative waves to the neighboring atrial myocardium may also occur as a result of the anti-diabetic regiment (both acute and/or chronic treatments). All together, these changes pose a challenge in the field of cardiology and call for further investigations to understand the etiology of the structural/functional remodeling of the SANCs in diabetes. Such an understanding may lead to more adequate therapies that can optimize glycemic control and improve health-related outcomes in patients with diabetes.
Collapse
Affiliation(s)
- Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
- *Correspondence: Lina T. Al Kury, ; Moni Nader,
| | - Stephanie Chacar
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Eman Alefishat
- Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ali A. Khraibi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Moni Nader
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- *Correspondence: Lina T. Al Kury, ; Moni Nader,
| |
Collapse
|
6
|
Hegyi B, Ko CY, Bossuyt J, Bers DM. Two-hit mechanism of cardiac arrhythmias in diabetic hyperglycaemia: reduced repolarization reserve, neurohormonal stimulation, and heart failure exacerbate susceptibility. Cardiovasc Res 2021; 117:2781-2793. [PMID: 33483728 PMCID: PMC8683706 DOI: 10.1093/cvr/cvab006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/10/2021] [Indexed: 12/11/2022] Open
Abstract
AIMS Diabetic hyperglycaemia is associated with increased arrhythmia risk. We aimed to investigate whether hyperglycaemia alone can be accountable for arrhythmias or whether it requires the presence of additional pathological factors. METHODS AND RESULTS Action potentials (APs) and arrhythmogenic spontaneous diastolic activities were measured in isolated murine ventricular, rabbit atrial, and ventricular myocytes acutely exposed to high glucose. Acute hyperglycaemia increased the short-term variability (STV) of action potential duration (APD), enhanced delayed afterdepolarizations, and the inducibility of APD alternans during tachypacing in both murine and rabbit atrial and ventricular myocytes. Hyperglycaemia also prolonged APD in mice and rabbit atrial cells but not in rabbit ventricular myocytes. However, rabbit ventricular APD was more strongly depressed by block of late Na+ current (INaL) during hyperglycaemia, consistent with elevated INaL in hyperglycaemia. All the above proarrhythmic glucose effects were Ca2+-dependent and abolished by CaMKII inhibition. Importantly, when the repolarization reserve was reduced by pharmacological inhibition of K+ channels (either Ito, IKr, IKs, or IK1) or hypokalaemia, acute hyperglycaemia further prolonged APD and further increased STV and alternans in rabbit ventricular myocytes. Likewise, when rabbit ventricular myocytes were pretreated with isoproterenol or angiotensin II, hyperglycaemia significantly prolonged APD, increased STV and promoted alternans. Moreover, acute hyperglycaemia markedly prolonged APD and further enhanced STV in failing rabbit ventricular myocytes. CONCLUSION We conclude that even though hyperglycaemia alone can enhance cellular proarrhythmic mechanisms, a second hit which reduces the repolarization reserve or stimulates G protein-coupled receptor signalling greatly exacerbates cardiac arrhythmogenesis in diabetic hyperglycaemia.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, CA 95616, USA
| | - Christopher Y Ko
- Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, CA 95616, USA
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, CA 95616, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, CA 95616, USA
| |
Collapse
|
7
|
Gallego M, Zayas-Arrabal J, Alquiza A, Apellaniz B, Casis O. Electrical Features of the Diabetic Myocardium. Arrhythmic and Cardiovascular Safety Considerations in Diabetes. Front Pharmacol 2021; 12:687256. [PMID: 34305599 PMCID: PMC8295895 DOI: 10.3389/fphar.2021.687256] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
Diabetes is a chronic metabolic disease characterized by hyperglycemia in the absence of treatment. Among the diabetes-associated complications, cardiovascular disease is the major cause of mortality and morbidity in diabetic patients. Diabetes causes a complex myocardial dysfunction, referred as diabetic cardiomyopathy, which even in the absence of other cardiac risk factors results in abnormal diastolic and systolic function. Besides mechanical abnormalities, altered electrical function is another major feature of the diabetic myocardium. Both type 1 and type 2 diabetic patients often show cardiac electrical remodeling, mainly a prolonged ventricular repolarization visible in the electrocardiogram as a lengthening of the QT interval duration. The underlying mechanisms at the cellular level involve alterations on the expression and activity of several cardiac ion channels and their associated regulatory proteins. Consequent changes in sodium, calcium and potassium currents collectively lead to a delay in repolarization that can increase the risk of developing life-threatening ventricular arrhythmias and sudden death. QT duration correlates strongly with the risk of developing torsade de pointes, a form of ventricular tachycardia that can degenerate into ventricular fibrillation. Therefore, QT prolongation is a qualitative marker of proarrhythmic risk, and analysis of ventricular repolarization is therefore required for the approval of new drugs. To that end, the Thorough QT/QTc analysis evaluates QT interval prolongation to assess potential proarrhythmic effects. In addition, since diabetic patients have a higher risk to die from cardiovascular causes than individuals without diabetes, cardiovascular safety of the new antidiabetic drugs must be carefully evaluated in type 2 diabetic patients. These cardiovascular outcome trials reveal that some glucose-lowering drugs actually reduce cardiovascular risk. The mechanism of cardioprotection might involve a reduction of the risk of developing arrhythmia.
Collapse
Affiliation(s)
- Mónica Gallego
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Julián Zayas-Arrabal
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Amaia Alquiza
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Beatriz Apellaniz
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Oscar Casis
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| |
Collapse
|
8
|
Zayas-Arrabal J, Alquiza A, Tuncay E, Turan B, Gallego M, Casis O. Molecular and Electrophysiological Role of Diabetes-Associated Circulating Inflammatory Factors in Cardiac Arrhythmia Remodeling in a Metabolic-Induced Model of Type 2 Diabetic Rat. Int J Mol Sci 2021; 22:ijms22136827. [PMID: 34202017 PMCID: PMC8268936 DOI: 10.3390/ijms22136827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Diabetic patients have prolonged cardiac repolarization and higher risk of arrhythmia. Besides, diabetes activates the innate immune system, resulting in higher levels of plasmatic cytokines, which are described to prolong ventricular repolarization. Methods: We characterize a metabolic model of type 2 diabetes (T2D) with prolonged cardiac repolarization. Sprague-Dawley rats were fed on a high-fat diet (45% Kcal from fat) for 6 weeks, and a low dose of streptozotozin intraperitoneally injected at week 2. Body weight and fasting blood glucose were measured and electrocardiograms of conscious animals were recorded weekly. Plasmatic lipid profile, insulin, cytokines, and arrhythmia susceptibility were determined at the end of the experimental period. Outward K+ currents and action potentials were recorded in isolated ventricular myocytes by patch-clamp. Results: T2D animals showed insulin resistance, hyperglycemia, and elevated levels of plasma cholesterol, triglycerides, TNFα, and IL-1b. They also developed bradycardia and prolonged QTc-interval duration that resulted in increased susceptibility to severe ventricular tachycardia under cardiac challenge. Action potential duration (APD) was prolonged in control cardiomyocytes incubated 24 h with plasma isolated from diabetic rats. However, adding TNFα and IL-1b receptor blockers to the serum of diabetic animals prevented the increased APD. Conclusions: The elevation of the circulating levels of TNFα and IL-1b are responsible for impaired ventricular repolarization and higher susceptibility to cardiac arrhythmia in our metabolic model of T2D.
Collapse
Affiliation(s)
- Julian Zayas-Arrabal
- Departament of Physiology, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, 01006 Vitoria-Gasteiz, Spain; (J.Z.-A.); (A.A.); (M.G.)
| | - Amaia Alquiza
- Departament of Physiology, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, 01006 Vitoria-Gasteiz, Spain; (J.Z.-A.); (A.A.); (M.G.)
| | - Erkan Tuncay
- Department of Biophysics, Faculty of Medicine, Ankara University, 06100 Ankara, Turkey;
| | - Belma Turan
- Department of Biophysics, Faculty of Medicine, Lokman Hekim University, 06510 Ankara, Turkey;
| | - Monica Gallego
- Departament of Physiology, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, 01006 Vitoria-Gasteiz, Spain; (J.Z.-A.); (A.A.); (M.G.)
| | - Oscar Casis
- Departament of Physiology, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, 01006 Vitoria-Gasteiz, Spain; (J.Z.-A.); (A.A.); (M.G.)
- Correspondence: ; Tel.: +34-945013033
| |
Collapse
|
9
|
Ozturk N, Uslu S, Ozdemir S. Diabetes-induced changes in cardiac voltage-gated ion channels. World J Diabetes 2021; 12:1-18. [PMID: 33520105 PMCID: PMC7807254 DOI: 10.4239/wjd.v12.i1.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/05/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus affects the heart through various mechanisms such as microvascular defects, metabolic abnormalities, autonomic dysfunction and incompatible immune response. Furthermore, it can also cause functional and structural changes in the myocardium by a disease known as diabetic cardiomyopathy (DCM) in the absence of coronary artery disease. As DCM progresses it causes electrical remodeling of the heart, left ventricular dysfunction and heart failure. Electrophysiological changes in the diabetic heart contribute significantly to the incidence of arrhythmias and sudden cardiac death in diabetes mellitus patients. In recent studies, significant changes in repolarizing K+ currents, Na+ currents and L-type Ca2+ currents along with impaired Ca2+ homeostasis and defective contractile function have been identified in the diabetic heart. In addition, insulin levels and other trophic factors change significantly to maintain the ionic channel expression in diabetic patients. There are many diagnostic tools and management options for DCM, but it is difficult to detect its development and to effectively prevent its progress. In this review, diabetes-associated alterations in voltage-sensitive cardiac ion channels are comprehensively assessed to understand their potential role in the pathophysiology and pathogenesis of DCM.
Collapse
Affiliation(s)
- Nihal Ozturk
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| | - Serkan Uslu
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| | - Semir Ozdemir
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| |
Collapse
|
10
|
Kobayashi S, Nagao M, Fukuda I, Oikawa S, Sugihara H. Multiple daily insulin injections ameliorate QT interval by lowering blood glucose levels in patients with type 2 diabetes. Ther Adv Endocrinol Metab 2021; 12:20420188211010057. [PMID: 34104393 PMCID: PMC8072833 DOI: 10.1177/20420188211010057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/24/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND A prolonged QT interval plays a causal role in fatal arrhythmia and is known to be a risk factor for sudden cardiac death. Although diabetic patients with microvascular complications tend to have a longer QT interval, the therapeutic effect of diabetes on the QT interval remains unclear. Here, we assessed the changes in QT interval in patients with type 2 diabetes (T2D) who received multiple daily insulin injections. MATERIALS AND METHODS Patients with T2D (n = 34) who were admitted to our hospital and initiated multiple daily insulin injections for glycemic control were enrolled in this study. Clinical measurements, including electrocardiogram, were taken on admission and discharge. The QT interval was measured manually in lead II on the electrocardiogram, and corrected QT interval (QTc) was calculated using Bazett's formula. The change in QTc (ΔQTc) during hospitalization (median, 15 days) and clinical parameters affecting ΔQTc were investigated. RESULTS QTc was shortened from 439 ± 24 to 427 ± 26 ms during hospitalization (p < 0.0001). ΔQTc was positively correlated with the changes in fasting plasma glucose (ΔFPG, r = 0.55, p = 0.0008) and glycated albumin (r = 0.38, p = 0.026) following insulin therapy, but not with the final dose of insulin (r = -0.20, p = 0.26). The multiple regression analyses revealed that ΔFPG was independently associated with ΔQTc. CONCLUSIONS Multiple daily insulin injections can ameliorate QT interval by lowering the blood glucose levels in T2D, suggesting that glycemic control is important for preventing patients with T2D from sudden cardiac death.
Collapse
Affiliation(s)
- Shunsuke Kobayashi
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | | | - Izumi Fukuda
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Shinichi Oikawa
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Hitoshi Sugihara
- Department of Endocrinology, Diabetes and Metabolism, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
11
|
Hyperglycemia regulates cardiac K + channels via O-GlcNAc-CaMKII and NOX2-ROS-PKC pathways. Basic Res Cardiol 2020; 115:71. [PMID: 33237428 DOI: 10.1007/s00395-020-00834-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022]
Abstract
Chronic hyperglycemia and diabetes lead to impaired cardiac repolarization, K+ channel remodeling and increased arrhythmia risk. However, the exact signaling mechanism by which diabetic hyperglycemia regulates cardiac K+ channels remains elusive. Here, we show that acute hyperglycemia increases inward rectifier K+ current (IK1), but reduces the amplitude and inactivation recovery time of the transient outward K+ current (Ito) in mouse, rat, and rabbit myocytes. These changes were all critically dependent on intracellular O-GlcNAcylation. Additionally, IK1 amplitude and Ito recovery effects (but not Ito amplitude) were prevented by the Ca2+/calmodulin-dependent kinase II (CaMKII) inhibitor autocamtide-2-related inhibitory peptide, CaMKIIδ-knockout, and O-GlcNAc-resistant CaMKIIδ-S280A knock-in. Ito reduction was prevented by inhibition of protein kinase C (PKC) and NADPH oxidase 2 (NOX2)-derived reactive oxygen species (ROS). In mouse models of chronic diabetes (streptozotocin, db/db, and high-fat diet), heart failure, and CaMKIIδ overexpression, both Ito and IK1 were reduced in line with the downregulated K+ channel expression. However, IK1 downregulation in diabetes was markedly attenuated in CaMKIIδ-S280A. We conclude that acute hyperglycemia enhances IK1 and Ito recovery via CaMKIIδ-S280 O-GlcNAcylation, but reduces Ito amplitude via a NOX2-ROS-PKC pathway. Moreover, chronic hyperglycemia during diabetes and CaMKII activation downregulate K+ channel expression and function, which may further increase arrhythmia susceptibility.
Collapse
|
12
|
Varró A, Tomek J, Nagy N, Virág L, Passini E, Rodriguez B, Baczkó I. Cardiac transmembrane ion channels and action potentials: cellular physiology and arrhythmogenic behavior. Physiol Rev 2020; 101:1083-1176. [PMID: 33118864 DOI: 10.1152/physrev.00024.2019] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrhythmias are among the leading causes of mortality. They often arise from alterations in the electrophysiological properties of cardiac cells and their underlying ionic mechanisms. It is therefore critical to further unravel the pathophysiology of the ionic basis of human cardiac electrophysiology in health and disease. In the first part of this review, current knowledge on the differences in ion channel expression and properties of the ionic processes that determine the morphology and properties of cardiac action potentials and calcium dynamics from cardiomyocytes in different regions of the heart are described. Then the cellular mechanisms promoting arrhythmias in congenital or acquired conditions of ion channel function (electrical remodeling) are discussed. The focus is on human-relevant findings obtained with clinical, experimental, and computational studies, given that interspecies differences make the extrapolation from animal experiments to human clinical settings difficult. Deepening the understanding of the diverse pathophysiology of human cellular electrophysiology will help in developing novel and effective antiarrhythmic strategies for specific subpopulations and disease conditions.
Collapse
Affiliation(s)
- András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - Jakub Tomek
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
13
|
Xu Parks X, Qudsi H, Braun C, Lopes CMB. The auxiliary subunit KCNE1 regulates KCNQ1 channel response to sustained calcium-dependent PKC activation. PLoS One 2020; 15:e0237591. [PMID: 32833978 PMCID: PMC7446858 DOI: 10.1371/journal.pone.0237591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/29/2020] [Indexed: 11/18/2022] Open
Abstract
The slow cardiac delayed rectifier current (IKs) is formed by KCNQ1 and KCNE1 subunits and is one of the major repolarizing currents in the heart. Decrease of IKs currents either due to inherited mutations or pathological remodeling is associated with increased risk for cardiac arrhythmias and sudden death. Ca2+-dependent PKC isoforms (cPKC) are chronically activated in heart disease and diabetes. Recently, we found that sustained stimulation of the calcium-dependent PKCβII isoform leads to decrease in KCNQ1 subunit membrane localization and KCNQ1/KCNE1 channel activity, although the role of KCNE1 in this regulation was not explored. Here, we show that the auxiliary KCNE1 subunit expression is necessary for channel internalization. A mutation in a KCNE1 phosphorylation site (KCNE1(S102A)) abolished channel internalization in both heterologous expression systems and cardiomyocytes. Altogether, our results suggest that KCNE1(S102) phosphorylation by PKCβII leads to KCNQ1/KCNE1 channel internalization in response to sustained PKC stimulus, while leaving KCNQ1 homomeric channels in the membrane. This preferential internalization is expected to have strong impact on cardiac repolarization. Our results suggest that KCNE1(S102) is an important anti-arrhythmic drug target to prevent IKs pathological remodeling leading to cardiac arrhythmias.
Collapse
Affiliation(s)
- Xiaorong Xu Parks
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - Haani Qudsi
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - Chen Braun
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - Coeli M. B. Lopes
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| |
Collapse
|
14
|
Abstract
Kv7 channels (Kv7.1-7.5) are voltage-gated K+ channels that can be modulated by five β-subunits (KCNE1-5). Kv7.1-KCNE1 channels produce the slow-delayed rectifying K+ current, IKs, which is important during the repolarization phase of the cardiac action potential. Kv7.2-7.5 are predominantly neuronally expressed and constitute the muscarinic M-current and control the resting membrane potential in neurons. Kv7.1 produces drastically different currents as a result of modulation by KCNE subunits. This flexibility allows the Kv7.1 channel to have many roles depending on location and assembly partners. The pharmacological sensitivity of Kv7.1 channels differs from that of Kv7.2-7.5 and is largely dependent upon the number of β-subunits present in the channel complex. As a result, the development of pharmaceuticals targeting Kv7.1 is problematic. This review discusses the roles and the mechanisms by which different signaling pathways affect Kv7.1 and KCNE channels and could potentially provide different ways of targeting the channel.
Collapse
Affiliation(s)
- Emely Thompson
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada;
| | - Jodene Eldstrom
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada;
| | - David Fedida
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada;
| |
Collapse
|
15
|
Zhabyeyev P, Chen X, Vanhaesebroeck B, Oudit GY. PI3Kα in cardioprotection: Cytoskeleton, late Na + current, and mechanism of arrhythmias. Channels (Austin) 2020; 13:520-532. [PMID: 31790629 PMCID: PMC6930018 DOI: 10.1080/19336950.2019.1697127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PI 3-kinase α (PI3Kα) is a lipid kinase that converts phosphatidylinositol-4,5-bisphosphate (PIP2) to phosphatidylinositol-3,4,5-triphosphate (PIP3). PI3Kα regulates a variety of cellular processes such as nutrient sensing, cell cycle, migration, and others. Heightened activity of PI3Kα in many types of cancer made it a prime oncology drug target, but also raises concerns of possible adverse effects on the heart. Indeed, recent advances in preclinical models demonstrate an important role of PI3Kα in the control of cytoskeletal integrity, Na+ channel activity, cardioprotection, and prevention of arrhythmias.
Collapse
Affiliation(s)
- Pavel Zhabyeyev
- Department of Medicine, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Xueyi Chen
- Department of Medicine, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | | | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
16
|
Baczkó I, Hornyik T, Brunner M, Koren G, Odening KE. Transgenic Rabbit Models in Proarrhythmia Research. Front Pharmacol 2020; 11:853. [PMID: 32581808 PMCID: PMC7291951 DOI: 10.3389/fphar.2020.00853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/22/2020] [Indexed: 12/23/2022] Open
Abstract
Drug-induced proarrhythmia constitutes a potentially lethal side effect of various drugs. Most often, this proarrhythmia is mechanistically linked to the drug's potential to interact with repolarizing cardiac ion channels causing a prolongation of the QT interval in the ECG. Despite sophisticated screening approaches during drug development, reliable prediction of proarrhythmia remains very challenging. Although drug-induced long-QT-related proarrhythmia is often favored by conditions or diseases that impair the individual's repolarization reserve, most cellular, tissue, and whole animal model systems used for drug safety screening are based on normal, healthy models. In recent years, several transgenic rabbit models for different types of long QT syndromes (LQTS) with differences in the extent of impairment in repolarization reserve have been generated. These might be useful for screening/prediction of a drug's potential for long-QT-related proarrhythmia, particularly as different repolarizing cardiac ion channels are impaired in the different models. In this review, we summarize the electrophysiological characteristics of the available transgenic LQTS rabbit models, and the pharmacological proof-of-principle studies that have been performed with these models—highlighting the advantages and disadvantages of LQTS models for proarrhythmia research. In the end, we give an outlook on potential future directions and novel models.
Collapse
Affiliation(s)
- István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Tibor Hornyik
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary.,Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Brunner
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Cardiology and Medical Intensive Care, St. Josefskrankenhaus, Freiburg, Germany
| | - Gideon Koren
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, United States
| | - Katja E Odening
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland.,Institute of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
17
|
Loss of insulin signaling may contribute to atrial fibrillation and atrial electrical remodeling in type 1 diabetes. Proc Natl Acad Sci U S A 2020; 117:7990-8000. [PMID: 32198206 DOI: 10.1073/pnas.1914853117] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Atrial fibrillation (AF) is prevalent in diabetes mellitus (DM); however, the basis for this is unknown. This study investigated AF susceptibility and atrial electrophysiology in type 1 diabetic Akita mice using in vivo intracardiac electrophysiology, high-resolution optical mapping in atrial preparations, and patch clamping in isolated atrial myocytes. qPCR and western blotting were used to assess ion channel expression. Akita mice were highly susceptible to AF in association with increased P-wave duration and slowed atrial conduction velocity. In a second model of type 1 DM, mice treated with streptozotocin (STZ) showed a similar increase in susceptibility to AF. Chronic insulin treatment reduced susceptibility and duration of AF and shortened P-wave duration in Akita mice. Atrial action potential (AP) morphology was altered in Akita mice due to a reduction in upstroke velocity and increases in AP duration. In Akita mice, atrial Na+ current (INa) and repolarizing K+ current (IK) carried by voltage gated K+ (Kv1.5) channels were reduced. The reduction in INa occurred in association with reduced expression of SCN5a and voltage gated Na+ (NaV1.5) channels as well as a shift in INa activation kinetics. Insulin potently and selectively increased INa in Akita mice without affecting IK Chronic insulin treatment increased INa in association with increased expression of NaV1.5. Acute insulin also increased INa, although to a smaller extent, due to enhanced insulin signaling via phosphatidylinositol 3,4,5-triphosphate (PIP3). Our study reveals a critical, selective role for insulin in regulating atrial INa, which impacts susceptibility to AF in type 1 DM.
Collapse
|
18
|
Bertero E, Sequeira V, Heymans S, Maack C. Response to 'The possible role of insulin and glucagon in patients with heart failure and Type 2 diabetes'. Eur Heart J 2020; 41:326-327. [PMID: 31329851 DOI: 10.1093/eurheartj/ehz243] [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: 11/12/2022] Open
Affiliation(s)
- Edoardo Bertero
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
| | - Vasco Sequeira
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands.,Netherlands Heart Institute, Moreelsepark 1, 3511EP Utrecht, The Netherlands.,Department of Cardiovascular Sciences, Leuven University, Herestraat 49, 3001 Leuven, Belgium
| | - Christoph Maack
- Department of Translational Science, Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, 97078 Würzburg, Germany
| |
Collapse
|
19
|
Jin X, Jiang Y, Xue G, Yuan Y, Zhu H, Zhan L, Zhuang Y, Huang Q, Shi L, Zhao Y, Li P, Sun Y, Su W, Zhang Y, Yang B, Lu Y, Wang Z, Pan Z. Increase of late sodium current contributes to enhanced susceptibility to atrial fibrillation in diabetic mice. Eur J Pharmacol 2019; 857:172444. [PMID: 31185218 DOI: 10.1016/j.ejphar.2019.172444] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/02/2019] [Accepted: 06/07/2019] [Indexed: 01/10/2023]
Abstract
Studies demonstrated that the incidence of atrial fibrillation is significantly increased in patients with diabetes mellitus. Increase of late sodium current (INaL) has been associated with atrial arrhythmias. However, the role of INaL in the setting of atrial fibrillation in diabetes mellitus remained unknown. In this study, we investigated the alteration of INaL in the atria of diabetic mice and the therapeutic effect of its inhibitor (GS967) on the susceptibility of atrial fibrillation. The whole-cell patch-clamp technique was used to detect single cell electrical activities. The results showed that the density of INaL in diabetic cardiomyocytes was larger than that of the control cells at the holding potential of -100 mV. The action potential duration at both 50% and 90% repolarization, APD50 and APD90, respectively, was markedly increased in diabetic mice than in controls. GS967 application inhibited INaL and shortened APD of diabetic mice. High-frequency electrical stimuli were used to induce atrial arrhythmias. We found that the occurrence rate of atrial fibrillation was significantly increased in diabetic mice, which was alleviated by the administration of GS967. In GS967-treated diabetic mice, the INaL current density was reduced and APD was shortened. In conclusion, the susceptibility to atrial fibrillation was increased in diabetic mice, which is associated with the increased late sodium current and the consequent prolongation of action potential. Inhibition of INaL by GS967 is beneficial against the occurrence of atrial fibrillation in diabetic mice.
Collapse
Affiliation(s)
- Xuexin Jin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Yuan Jiang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Genlong Xue
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Yin Yuan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Haixia Zhu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Linfeng Zhan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Yuting Zhuang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Qihe Huang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Ling Shi
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Yue Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Penghui Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Yilin Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Wanzhen Su
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Yang Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China.
| | - Yanjie Lu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China.
| | - Zhiguo Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China.
| | - Zhenwei Pan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, PR China.
| |
Collapse
|
20
|
Zhabyeyev P, McLean B, Chen X, Vanhaesebroeck B, Oudit GY. Inhibition of PI3Kinase-α is pro-arrhythmic and associated with enhanced late Na + current, contractility, and Ca 2+ release in murine hearts. J Mol Cell Cardiol 2019; 132:98-109. [PMID: 31095940 DOI: 10.1016/j.yjmcc.2019.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/11/2019] [Accepted: 05/09/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Phosphoinositide 3-kinase α (PI3Kα) is a proto-oncogene with high activity in the heart. BYL719 (BYL) is a PI3Kα-selective small molecule inhibitor and a prospective drug for advanced solid tumors. We investigated whether acute pharmacological inhibition of PI3Kα has pro-arrhythmic effects. METHODS & RESULTS In isolated wild-type (WT) cardiomyocytes, pharmacological inhibition of PI3Kα (BYL719) increased contractility by 28%, Ca2+ release by 20%, and prolonged action potential (AP) repolarization by 10-15%. These effects of BYL719 were abolished by inhibition of reverse-mode Na+/Ca2+ exchanger (NCX) (KB-R7943) or by inhibition of late Na+ current (INa-L) (ranolazine). BYL719 had no effect on PI3Kα-deficient cardiomyocytes, suggesting BYL719 effects were PI3Kα-dependent and mediated via NCX and INa-L. INa-L was suppressed by activation of PI3Kα, application of exogenous intracellular PIP3, or ranolazine. Investigation of AP and Ca2+ release in whole heart preparations using epicardial optical mapping showed that inhibition of PI3Kα similarly led to prolongation of AP and enhancement of Ca2+ release. In hearts of PI3Kα-deficient mice, β-adrenergic stimulation in the presence of high Ca2+ concentrations and 12-Hz burst pacing led to delayed afterdepolarizations and ventricular fibrillation. In vivo, administration of BYL719 prolonged QT interval [QTcF (Fridericia) increased by 15%] in WT, but not in PI3Kα-deficient mice. CONCLUSIONS Pharmacological inhibition of PI3Kα is arrhythmogenic due to activation of INa-L leading to increased sarcoplasmic reticulum Ca2+ load and prolonged QT interval. Therefore, monitoring of cardiac electrical activity in patients receiving PI3K inhibitors may provide further insights into the arrhythmogenic potential of PI3Ka inhibition.
Collapse
Affiliation(s)
- Pavel Zhabyeyev
- Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Brent McLean
- Department of Medicine, University of Alberta, Edmonton, Canada; Department of Physiology, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Xueyi Chen
- Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | | | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada.
| |
Collapse
|
21
|
Parks XX, Ronzier E, O-Uchi J, Lopes CM. Fluvastatin inhibits Rab5-mediated IKs internalization caused by chronic Ca 2+-dependent PKC activation. J Mol Cell Cardiol 2019; 129:314-325. [PMID: 30898664 DOI: 10.1016/j.yjmcc.2019.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/26/2019] [Accepted: 03/16/2019] [Indexed: 10/27/2022]
Abstract
Statins, in addition to their cholesterol lowering effects, can prevent isoprenylation of Rab GTPase proteins, a key protein family for the regulation of protein trafficking. Rab-GTPases have been shown to be involved in the control of membrane expression level of ion channels, including one of the major cardiac repolarizing channels, IKs. Decreased IKs function has been observed in a number of disease states and associated with increased propensity for arrhythmias, but the mechanism underlying IKs decrease remains elusive. Ca2+-dependent PKC isoforms (cPKC) are chronically activated in variety of human diseases and have been suggested to acutely regulate IKs function. We hypothesize that chronic cPKC stimulation leads to Rab-mediated decrease in IKs membrane expression, and that can be prevented by statins. In this study we show that chronic cPKC stimulation caused a dramatic Rab5 GTPase-dependent decrease in plasma membrane localization of the IKs pore forming subunit KCNQ1, reducing IKs function. Our data indicates fluvastatin inhibition of Rab5 restores channel localization and function after cPKC-mediated channel internalization. Our results indicate a novel statin anti-arrhythmic effect that would be expected to inhibit pathological electrical remodeling in a number of disease states associated with high cPKC activation. Because Rab-GTPases are important regulators of membrane trafficking they may underlie other statin pleiotropic effects.
Collapse
Affiliation(s)
- Xiaorong Xu Parks
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Elsa Ronzier
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Jin O-Uchi
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America; Lillehei Heart Institute, University of Minnesota, 2231 6th Street SE, Minneapolis, MN 55455, United States of America
| | - Coeli M Lopes
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America.
| |
Collapse
|
22
|
Hegyi B, Bers DM, Bossuyt J. CaMKII signaling in heart diseases: Emerging role in diabetic cardiomyopathy. J Mol Cell Cardiol 2019; 127:246-259. [PMID: 30633874 DOI: 10.1016/j.yjmcc.2019.01.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023]
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) is upregulated in diabetes and significantly contributes to cardiac remodeling with increased risk of cardiac arrhythmias. Diabetes is frequently associated with atrial fibrillation, coronary artery disease, and heart failure, which may further enhance CaMKII. Activation of CaMKII occurs downstream of neurohormonal stimulation (e.g. via G-protein coupled receptors) and involve various posttranslational modifications including autophosphorylation, oxidation, S-nitrosylation and O-GlcNAcylation. CaMKII signaling regulates diverse cellular processes in a spatiotemporal manner including excitation-contraction and excitation-transcription coupling, mechanics and energetics in cardiac myocytes. Chronic activation of CaMKII results in cellular remodeling and ultimately arrhythmogenic alterations in Ca2+ handling, ion channels, cell-to-cell coupling and metabolism. This review addresses the detrimental effects of the upregulated CaMKII signaling to enhance the arrhythmogenic substrate and trigger mechanisms in the heart. We also briefly summarize preclinical studies using kinase inhibitors and genetically modified mice targeting CaMKII in diabetes. The mechanistic understanding of CaMKII signaling, cardiac remodeling and arrhythmia mechanisms may reveal new therapeutic targets and ultimately better treatment in diabetes and heart disease in general.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| |
Collapse
|
23
|
Grisanti LA. Diabetes and Arrhythmias: Pathophysiology, Mechanisms and Therapeutic Outcomes. Front Physiol 2018; 9:1669. [PMID: 30534081 PMCID: PMC6275303 DOI: 10.3389/fphys.2018.01669] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/06/2018] [Indexed: 12/17/2022] Open
Abstract
The prevalence of diabetes is rapidly increasing and closely associated with cardiovascular morbidity and mortality. While the major cardiovascular complication associated with diabetes is coronary artery disease, it is becoming increasingly apparent that diabetes impacts the electrical conduction system in the heart, resulting in atrial fibrillation, and ventricular arrhythmias. The relationship between diabetes and arrhythmias is complex and multifactorial including autonomic dysfunction, atrial and ventricular remodeling and molecular alterations. This review will provide a comprehensive overview of the link between diabetes and arrhythmias with insight into the common molecular mechanisms, structural alterations and therapeutic outcomes.
Collapse
Affiliation(s)
- Laurel A Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
24
|
Type 1 diabetes is associated with T-wave morphology changes. The Thousand & 1 Study. J Electrocardiol 2018; 51:S72-S77. [DOI: 10.1016/j.jelectrocard.2018.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/03/2018] [Accepted: 05/24/2018] [Indexed: 11/19/2022]
|
25
|
Ferdinandy P, Baczkó I, Bencsik P, Giricz Z, Görbe A, Pacher P, Varga ZV, Varró A, Schulz R. Definition of hidden drug cardiotoxicity: paradigm change in cardiac safety testing and its clinical implications. Eur Heart J 2018; 40:1771-1777. [PMID: 29982507 PMCID: PMC6554653 DOI: 10.1093/eurheartj/ehy365] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/12/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022] Open
Abstract
Unexpected cardiac adverse effects are the leading causes of discontinuation of clinical trials and withdrawal of drugs from the market. Since the original observations in the mid-90s, it has been well established that cardiovascular risk factors and comorbidities (such as ageing, hyperlipidaemia, and diabetes) and their medications (e.g. nitrate tolerance, adenosine triphosphate-dependent potassium inhibitor antidiabetic drugs, statins, etc.) may interfere with cardiac ischaemic tolerance and endogenous cardioprotective signalling pathways. Indeed drugs may exert unwanted effects on the diseased and treated heart that is hidden in the healthy myocardium. Hidden cardiotoxic effects may be due to (i) drug-induced enhancement of deleterious signalling due to ischaemia/reperfusion injury and/or the presence of risk factors and/or (ii) inhibition of cardioprotective survival signalling pathways, both of which may lead to ischaemia-related cell death and/or pro-arrhythmic effects. This led to a novel concept of ‘hidden cardiotoxicity’, defined as cardiotoxity of a drug that manifests only in the diseased heart with e.g. ischaemia/reperfusion injury and/or in the presence of its major comorbidities. Little is known on the mechanism of hidden cardiotoxocity, moreover, hidden cardiotoxicity cannot be revealed by the routinely used non-clinical cardiac safety testing methods on healthy animals or tissues. Therefore, here, we emphasize the need for development of novel cardiac safety testing platform involving combined experimental models of cardiac diseases (especially myocardial ischaemia/reperfusion and ischaemic conditioning) in the presence and absence of major cardiovascular comorbidities and/or cotreatments.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, Szeged, Hungary
| | | | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Fishers Lane, Bethesda, MD, USA
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Fishers Lane, Bethesda, MD, USA
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Aulweg 129, Giessen, Germany
| |
Collapse
|
26
|
Yoshimatsu Y, Ishizaka T, Chiba K, Mori K. Usefulness of simultaneous and sequential monitoring of glucose level and electrocardiogram in monkeys treated with gatifloxacin under conscious and nonrestricted conditions. Exp Anim 2018; 67:281-290. [PMID: 29311442 PMCID: PMC5955759 DOI: 10.1538/expanim.17-0136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Drug-induced cardiac electrophysiological abnormalities accompanied by hypoglycemia or
hyperglycemia increase the risk for life-threatening arrhythmia. To assess the
drug-induced cardiotoxic potential associated with extraordinary blood glucose (GLU)
levels, the effect of gatifloxacin (GFLX) which was frequently associated with GLU
abnormality and QT/QTc prolongations in the clinic on blood GLU and electrocardiogram
(ECG) parameters was investigated in cynomolgus monkeys (n=4) given GFLX orally in an
ascending dose regimen (10, 30, 60 and 100 mg/kg). Simultaneous and sequential GLU and ECG
monitoring with a continuous GLU monitoring system and Holter ECG, respectively, were
conducted for 24 h under free-moving conditions. Consequently, GFLX at 30 and 60 mg/kg
dose-dependently induced a transient decrease in GLU without any ECG abnormality 2–4 h
postdose. Highest dose of 100 mg/kg caused severe hypoglycemia with a mean GLU of <30
mg/dL, accompanied by remarkable QT/QTc prolongations by 20–30% in all animals. In
contrast, hyperglycemia without QT/QTc prolongations was noted 24 h after dosing in one
animal. A close correlation between GLU and QTc values was observed in animals treated
with 100 mg/kg, suggesting that GFLX-induced hypoglycemia enhanced QT/QTc prolongations.
Furthermore, the 24-h sequential GLU monitoring data clearly distinguished between
GFLX-induced GLU abnormality and physiological GLU changes influenced by feeding
throughout the day. In conclusion, the combined assessment of continuous GLU and ECG
monitoring is valuable in predicting the drug-induced cardio-electrophysiological risk
associated with both GLU and ECG abnormalities.
Collapse
Affiliation(s)
- Yu Yoshimatsu
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Tomomichi Ishizaka
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Katsuyoshi Chiba
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Kazuhiko Mori
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| |
Collapse
|
27
|
Russell J, Du Toit EF, Peart JN, Patel HH, Headrick JP. Myocyte membrane and microdomain modifications in diabetes: determinants of ischemic tolerance and cardioprotection. Cardiovasc Diabetol 2017; 16:155. [PMID: 29202762 PMCID: PMC5716308 DOI: 10.1186/s12933-017-0638-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/22/2017] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease, predominantly ischemic heart disease (IHD), is the leading cause of death in diabetes mellitus (DM). In addition to eliciting cardiomyopathy, DM induces a ‘wicked triumvirate’: (i) increasing the risk and incidence of IHD and myocardial ischemia; (ii) decreasing myocardial tolerance to ischemia–reperfusion (I–R) injury; and (iii) inhibiting or eliminating responses to cardioprotective stimuli. Changes in ischemic tolerance and cardioprotective signaling may contribute to substantially higher mortality and morbidity following ischemic insult in DM patients. Among the diverse mechanisms implicated in diabetic impairment of ischemic tolerance and cardioprotection, changes in sarcolemmal makeup may play an overarching role and are considered in detail in the current review. Observations predominantly in animal models reveal DM-dependent changes in membrane lipid composition (cholesterol and triglyceride accumulation, fatty acid saturation vs. reduced desaturation, phospholipid remodeling) that contribute to modulation of caveolar domains, gap junctions and T-tubules. These modifications influence sarcolemmal biophysical properties, receptor and phospholipid signaling, ion channel and transporter functions, contributing to contractile and electrophysiological dysfunction, cardiomyopathy, ischemic intolerance and suppression of protective signaling. A better understanding of these sarcolemmal abnormalities in types I and II DM (T1DM, T2DM) can inform approaches to limiting cardiomyopathy, associated IHD and their consequences. Key knowledge gaps include details of sarcolemmal changes in models of T2DM, temporal patterns of lipid, microdomain and T-tubule changes during disease development, and the precise impacts of these diverse sarcolemmal modifications. Importantly, exercise, dietary, pharmacological and gene approaches have potential for improving sarcolemmal makeup, and thus myocyte function and stress-resistance in this ubiquitous metabolic disorder.
Collapse
Affiliation(s)
- Jake Russell
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Eugene F Du Toit
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Hemal H Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California San Diego, San Diego, USA
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia. .,School of Medical Science, Griffith University, Southport, QLD, 4217, Australia.
| |
Collapse
|
28
|
Wu M, Obara Y, Ohshima S, Nagasawa Y, Ishii K. Insulin treatment augments KCNQ1/KCNE1 currents but not KCNQ1 currents, which is associated with an increase in KCNE1 expression. Biochem Biophys Res Commun 2017; 493:409-415. [PMID: 28882596 DOI: 10.1016/j.bbrc.2017.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 09/03/2017] [Indexed: 11/25/2022]
Abstract
Diabetes mellitus affects ion channel physiology. We have previously reported that acute application of insulin suppresses the KCNQ1/KCNE1 currents that play an important role in terminating ventricular action potential. In this study, we investigated the effect of long-term insulin treatment on KCNQ1/KCNE1 currents using the Xenopus oocyte expression system. Insulin treatment with a duration longer than 6 h had an opposite effect to acute insulin application, that is, it augmented the KCNQ1/KCNE1 currents. Inhibitors of PI3K, wortmannin and LY294002, and a MEK inhibitor, U0126, abolished the potentiating effect of long-term insulin treatment. The long-term treatment with insulin had no effect on KCNQ1 currents indicating an essential role of KCNE1 in the insulin effect, which is similar to the acute insulin effect. Cycloheximide, an inhibitor of protein synthesis, and brefeldin A, an inhibitor of protein transport from endoplasmic reticulum, suppressed the long-term insulin effect. Western blotting analysis combined with these pharmacological data suggest that long-term insulin treatment augments KCNQ1/KCNE1 currents by increasing KCNE1 protein expression.
Collapse
Affiliation(s)
- Minghua Wu
- Department of Pharmacology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| | - Yutaro Obara
- Department of Pharmacology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| | - Shingo Ohshima
- Department of Pharmacology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| | - Yoshinobu Nagasawa
- Department of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Toho University, Chiba 274-8510, Japan
| | - Kuniaki Ishii
- Department of Pharmacology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan.
| |
Collapse
|
29
|
Orosz A, Baczkó I, Nyiraty S, Körei AE, Putz Z, Takács R, Nemes A, Várkonyi TT, Balogh L, Ábrahám G, Kempler P, Papp JG, Varró A, Lengyel C. Increased Short-Term Beat-to-Beat QT Interval Variability in Patients with Impaired Glucose Tolerance. Front Endocrinol (Lausanne) 2017; 8:129. [PMID: 28659867 PMCID: PMC5468431 DOI: 10.3389/fendo.2017.00129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 05/29/2017] [Indexed: 01/02/2023] Open
Abstract
Prediabetic states and diabetes are important risk factors for cardiovascular morbidity and mortality. Determination of short-term QT interval variability (STVQT) is a non-invasive method for assessment of proarrhythmic risk. The aim of the study was to evaluate the STVQT in patients with impaired glucose tolerance (IGT). 18 IGT patients [age: 63 ± 11 years, body mass index (BMI): 31 ± 6 kg/m2, fasting glucose: 6.0 ± 0.4 mmol/l, 120 min postload glucose: 9.0 ± 1.0 mmol/l, hemoglobin A1c (HbA1c): 5.9 ± 0.4%; mean ± SD] and 18 healthy controls (age: 56 ± 9 years, BMI: 27 ± 5 kg/m2, fasting glucose: 5.2 ± 0.4 mmol/l, 120 min postload glucose: 5.5 ± 1.3 mmol/l, HbA1c: 5.4 ± 0.3%) were enrolled into the study. ECGs were recorded, processed, and analyzed off-line. The RR and QT intervals were expressed as the average of 30 consecutive beats, the temporal instability of beat-to-beat repolarization was characterized by calculating STVQT as follows: STVQT = Σ|QTn + 1 - QTn| (30x√2)-1. Autonomic function was assessed by means of standard cardiovascular reflex tests. There were no differences between IGT and control groups in QT (411 ± 43 vs 402 ± 39 ms) and QTc (431 ± 25 vs 424 ± 19 ms) intervals or QT dispersion (44 ± 13 vs 42 ± 17 ms). However, STVQT was significantly higher in IGT patients (5.0 ± 0.7 vs 3.7 ± 0.7, P < 0.0001). The elevated temporal STVQT in patients with IGT may be an early indicator of increased instability of cardiac repolarization during prediabetic conditions.
Collapse
Affiliation(s)
- Andrea Orosz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Szabolcs Nyiraty
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Anna E. Körei
- First Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Putz
- First Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Róbert Takács
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Attila Nemes
- Second Department of Medicine and Cardiology Centre, University of Szeged, Szeged, Hungary
| | | | - László Balogh
- Juhász Gyula Faculty of Education, Institute of Physical Education and Sport Science, University of Szeged, Szeged, Hungary
| | - György Ábrahám
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Kempler
- First Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Julius Gy. Papp
- Department of Pharmacology and Pharmacotherapy, 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, University of Szeged, Szeged, Hungary
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Csaba Lengyel
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
- First Department of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
30
|
Huang X, Zhong N, Zhang H, Ma A, Yuan Z, Guo N. Reduced expression of HCN channels in the sinoatrial node of streptozotocin-induced diabetic rats. Can J Physiol Pharmacol 2016; 95:586-594. [PMID: 28177679 DOI: 10.1139/cjpp-2016-0418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diabetes mellitus (DM) is associated with an electrical remodeling of the heart, increasing the risk of arrhythmias. However, knowledge of electrical remodeling in the sinoatrial node (SAN) by DM is limited. We investigated the expression of HCN channel isoforms, HCN1-HCN4, in SAN from streptozotocin (STZ)-induced diabetic rats and the age-matched controls. We found that the STZ-induced diabetic rats have a lower intrinsic heart rate, a lengthened sinoatrial conduction time, and rate-corrected maximal sinoatrial node recovery time in vivo as well as a longer cycle length (CL) in vitro, as compared with the control. Optical mapping of the SAN demonstrated an inferior leading pacemaker site, reduced SAN conduction velocity and diastolic depolarization slope, and a longer action potential duration in the STZ-induced diabetic rats than in the control. The transcripts and proteins of HCN2 and HCN4 in diabetic SAN were reduced. Specific blockade of HCN channels by 3 μmol/L ivabradine significantly prolonged the CL of a Langendorff heart by 18% in the diabetic rats and 26% in the control. The reduced expression of HCN channel isoforms in the SAN of the STZ-induced diabetic rat may be an important contributor to the reduced SAN function in DM.
Collapse
Affiliation(s)
- Xin Huang
- a Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China, 710061
| | - Nier Zhong
- b Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, P.R. China, 710068
| | - Hong Zhang
- c School of Electrical Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China, 710049
| | - Aiqun Ma
- a Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China, 710061
| | - Zuyi Yuan
- a Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China, 710061
| | - Ning Guo
- a Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China, 710061
| |
Collapse
|
31
|
Pini A, Obara I, Battell E, Chazot PL, Rosa AC. Histamine in diabetes: Is it time to reconsider? Pharmacol Res 2016; 111:316-324. [DOI: 10.1016/j.phrs.2016.06.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 10/21/2022]
|
32
|
Wang K, Li L, Wu Y, Yang Y, Chen J, Zhang D, Liu Z, Xu J, Cao M, Mao X, Liu C. Increased serum gamma-glutamyltransferase levels are associated with ventricular instability in type 2 diabetes. Endocrine 2016; 52:63-72. [PMID: 26433737 DOI: 10.1007/s12020-015-0760-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 09/23/2015] [Indexed: 12/26/2022]
Abstract
The purpose of our study is to examine the association between serum GGT levels and ventricular instability in Chinese patients with T2DM. We conducted a cross-sectional, community-based study in Nanjing, China from June to November 2011. Among 10,050 patients aged 40-79 years, we enrolled 2444 with pre-diabetes, 2496 with T2DM, and 4521 without diabetes (non-diabetes). Electrocardiograms were performed to measure the QT interval corrected for heart rate (QTc) and QT interval dispersion (QTd). Serum GGT levels, metabolic parameters, body mass index, and blood pressure were also measured. We found that there were no significant associations of increased QTc/QTd with serum GGT levels in participants with pre-existing T2DM and non-diabetes, after adjusting for age, duration of diabetes, and metabolic parameters. Even after adjustment, higher risks of QTc ≥ 440 ms/√s and QTd ≥ 58 ms were found in participants with serum GGT levels ≥49 U/L compared with those with <15 U/L in the pre-diabetes (QTc: OR 1.96, 95 % CI 1.23-2.47; QTd: OR 1.34, 95 % CI 1.07-1.94) and newly diagnosed T2DM (QTc: OR 2.01, 95 % CI 1.39-2.51; QTd: OR 1.53, 95 % CI 1.03-1.99) groups. We conclude that Increased serum GGT levels are associated with some markers of ventricular repolarization abnormalities in the early stage of T2DM.
Collapse
Affiliation(s)
- Kun Wang
- Department of Endocrinology, Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, 100, Hongshan Road, Nanjing, 210028, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, 87, Dingjiaqiao Road, Nanjing, 210009, China
| | - Yang Wu
- Department of Endocrinology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Suzhou University, 185, Juqian Road, Changzhou, 213003, China
| | - Yu Yang
- Department of Endocrinology, Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, 100, Hongshan Road, Nanjing, 210028, China
| | - Jie Chen
- Department of Endocrinology, Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, 100, Hongshan Road, Nanjing, 210028, China
| | - Danyu Zhang
- Department of Endocrinology, Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, 100, Hongshan Road, Nanjing, 210028, China
| | - Zhoujun Liu
- Department of Endocrinology, Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, 100, Hongshan Road, Nanjing, 210028, China
| | - Juan Xu
- Department of Endocrinology, Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, 100, Hongshan Road, Nanjing, 210028, China
| | - Meng Cao
- Department of Endocrinology, Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, 100, Hongshan Road, Nanjing, 210028, China
| | - Xiaodong Mao
- Department of Endocrinology, Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, 100, Hongshan Road, Nanjing, 210028, China
| | - Chao Liu
- Department of Endocrinology, Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, 100, Hongshan Road, Nanjing, 210028, China.
| |
Collapse
|
33
|
Dubó S, Gallegos D, Cabrera L, Sobrevia L, Zúñiga L, González M. Cardiovascular Action of Insulin in Health and Disease: Endothelial L-Arginine Transport and Cardiac Voltage-Dependent Potassium Channels. Front Physiol 2016; 7:74. [PMID: 27014078 PMCID: PMC4791397 DOI: 10.3389/fphys.2016.00074] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/15/2016] [Indexed: 12/19/2022] Open
Abstract
Impairment of insulin signaling on diabetes mellitus has been related to cardiovascular dysfunction, heart failure, and sudden death. In human endothelium, cationic amino acid transporter 1 (hCAT-1) is related to the synthesis of nitric oxide (NO) and insulin has a vascular effect in endothelial cells through a signaling pathway that involves increases in hCAT-1 expression and L-arginine transport. This mechanism is disrupted in diabetes, a phenomenon potentiated by excessive accumulation of reactive oxygen species (ROS), which contribute to lower availability of NO and endothelial dysfunction. On the other hand, electrical remodeling in cardiomyocytes is considered a key factor in heart failure progression associated to diabetes mellitus. This generates a challenge to understand the specific role of insulin and the pathways involved in cardiac function. Studies on isolated mammalian cardiomyocytes have shown prolongated action potential in ventricular repolarization phase that produces a long QT interval, which is well explained by attenuation in the repolarizing potassium currents in cardiac ventricles. Impaired insulin signaling causes specific changes in these currents, such a decrease amplitude of the transient outward K(+) (Ito) and the ultra-rapid delayed rectifier (IKur) currents where, together, a reduction of mRNA and protein expression levels of α-subunits (Ito, fast; Kv 4.2 and IKs; Kv 1.5) or β-subunits (KChIP2 and MiRP) of K(+) channels involved in these currents in a MAPK mediated pathway process have been described. These results support the hypothesis that lack of insulin signaling can produce an abnormal repolarization in cardiomyocytes. Furthermore, the arrhythmogenic potential due to reduced Ito current can contribute to an increase in the incidence of sudden death in heart failure. This review aims to show, based on pathophysiological models, the regulatory function that would have insulin in vascular system and in cardiac electrophysiology.
Collapse
Affiliation(s)
- Sebastián Dubó
- Department of Kinesiology, Faculty of Medicine, Universidad de Concepción Concepción, Chile
| | - David Gallegos
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción Concepción, Chile
| | - Lissette Cabrera
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de ConcepciónConcepción, Chile; Department of Morphophysiology, Faculty of Medicine, Universidad Diego PortalesSantiago, Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynecology, Faculty of Medicine, School of Medicine, Pontificia Universidad Católica de ChileSantiago, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de SevillaSeville, Spain; Faculty of Medicine and Biomedical Sciences, University of Queensland Centre for Clinical Research (UQCCR), University of QueenslandHerston, QLD, Queensland, Australia
| | - Leandro Zúñiga
- Centro de Investigaciones Médicas, Escuela de Medicina, Universidad de Talca Talca, Chile
| | - Marcelo González
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de ConcepciónConcepción, Chile; Group of Research and Innovation in Vascular Health (GRIVAS-Health)Chillán, Chile
| |
Collapse
|
34
|
Bilodeau C, Bardou O, Maillé É, Berthiaume Y, Brochiero E. Deleterious impact of hyperglycemia on cystic fibrosis airway ion transport and epithelial repair. J Cyst Fibros 2016; 15:43-51. [DOI: 10.1016/j.jcf.2015.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 04/07/2015] [Accepted: 04/10/2015] [Indexed: 02/08/2023]
|
35
|
Murfitt L, Whiteley G, Iqbal MM, Kitmitto A. Targeting caveolin-3 for the treatment of diabetic cardiomyopathy. Pharmacol Ther 2015; 151:50-71. [PMID: 25779609 DOI: 10.1016/j.pharmthera.2015.03.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 03/09/2015] [Indexed: 12/21/2022]
Abstract
Diabetes is a global health problem with more than 550 million people predicted to be diabetic by 2030. A major complication of diabetes is cardiovascular disease, which accounts for over two-thirds of mortality and morbidity in diabetic patients. This increased risk has led to the definition of a diabetic cardiomyopathy phenotype characterised by early left ventricular dysfunction with normal ejection fraction. Here we review the aetiology of diabetic cardiomyopathy and explore the involvement of the protein caveolin-3 (Cav3). Cav3 forms part of a complex mechanism regulating insulin signalling and glucose uptake, processes that are impaired in diabetes. Further, Cav3 is key for stabilisation and trafficking of cardiac ion channels to the plasma membrane and so contributes to the cardiac action potential shape and duration. In addition, Cav3 has direct and indirect interactions with proteins involved in excitation-contraction coupling and so has the potential to influence cardiac contractility. Significantly, both impaired contractility and rhythm disturbances are hallmarks of diabetic cardiomyopathy. We review here how changes to Cav3 expression levels and altered relationships with interacting partners may be contributory factors to several of the pathological features identified in diabetic cardiomyopathy. Finally, the review concludes by considering ways in which levels of Cav3 may be manipulated in order to develop novel therapeutic approaches for treating diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Lucy Murfitt
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK
| | - Gareth Whiteley
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK
| | - Mohammad M Iqbal
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK
| | - Ashraf Kitmitto
- Institute of Cardiovascular Sciences, Faculty of Medical and Human Sciences, University of Manchester, M13 9NT, UK.
| |
Collapse
|
36
|
Ballou LM, Lin RZ, Cohen IS. Control of cardiac repolarization by phosphoinositide 3-kinase signaling to ion channels. Circ Res 2015; 116:127-37. [PMID: 25552692 DOI: 10.1161/circresaha.116.303975] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Upregulation of phosphoinositide 3-kinase (PI3K) signaling is a common alteration in human cancer, and numerous drugs that target this pathway have been developed for cancer treatment. However, recent studies have implicated inhibition of the PI3K signaling pathway as the cause of a drug-induced long-QT syndrome in which alterations in several ion currents contribute to arrhythmogenic drug activity. Surprisingly, some drugs that were thought to induce long-QT syndrome by direct block of the rapid delayed rectifier (IKr) also seem to inhibit PI3K signaling, an effect that may contribute to their arrhythmogenicity. The importance of PI3K in regulating cardiac repolarization is underscored by evidence that QT interval prolongation in diabetes mellitus also may result from changes in multiple currents because of decreased insulin activation of PI3K in the heart. How PI3K signaling regulates ion channels to control the cardiac action potential is poorly understood. Hence, this review summarizes what is known about the effect of PI3K and its downstream effectors, including Akt, on sodium, potassium, and calcium currents in cardiac myocytes. We also refer to some studies in noncardiac cells that provide insight into potential mechanisms of ion channel regulation by this signaling pathway in the heart. Drug development and safety could be improved with a better understanding of the mechanisms by which PI3K regulates cardiac ion channels and the extent to which PI3K inhibition contributes to arrhythmogenic susceptibility.
Collapse
Affiliation(s)
- Lisa M Ballou
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.)
| | - Richard Z Lin
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.).
| | - Ira S Cohen
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.).
| |
Collapse
|
37
|
Uysal F, Ozboyaci E, Bostan O, Saglam H, Semizel E, Cil E. Evaluation of electrocardiographic parameters for early diagnosis of autonomic dysfunction in children and adolescents with type-1 diabetes mellitus. Pediatr Int 2014; 56:675-80. [PMID: 24617770 DOI: 10.1111/ped.12329] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 01/14/2014] [Accepted: 02/04/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND The aim of this study was to identify the sensitivity of electrocardiogram (ECG) in early diagnosis of cardiac autonomic function disorder in children with type 1 diabetes mellitus. METHODS A total of 150 children and adolescents with type 1 diabetes mellitus were enrolled between June 2009 and June 2010, as well as 100 age- and sex-matched healthy control children. Twelve-lead ECG was done in all cases and heart rate, QT and QTc interval, dispersion of P wave (Pd), and of QT (QTd) and QTc interval (QTcd) were measured. The clinical and demographic features such as age, gender, duration of follow up and level of HbA1c and fasting glucose were obtained and the effects of these parameters on ECG measurements were investigated. RESULTS The mean age of the patients and controls was 11.61 ± 3.72 years and 10.92 ± 3.2 years, respectively. QT and QTc interval and QTcd interval were significantly higher in diabetic children compared to healthy controls but these ECG findings were not associated with the duration of diabetes or glycemic state. Pd was significantly higher in the diabetic patients with HbA1c >7.5% compared to control, and this was also found in patients that were followed up >1 year. CONCLUSIONS Cardiac autonomic function disorder, which is one of the most important causes of morbidity and mortality, may emerge in the course of type 1 diabetes mellitus. It can be diagnosed on ECG even when the patients are asymptomatic.
Collapse
Affiliation(s)
- Fahrettin Uysal
- Department of Pediatric Cardiology, Medical Faculty, University of Uludag, Gorukle, Bursa, Turkey
| | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- J. P. Morrow
- Division of Cardiology; Department of Medicine; College of Physicians and Surgeons of Columbia University; New York NY USA
| |
Collapse
|
39
|
Biet M, Morin N, Benrezzak O, Naimi F, Bellanger S, Baillargeon JP, Chouinard L, Gallo-Payet N, Carpentier AC, Dumaine R. Lasting alterations of the sodium current by short-term hyperlipidemia as a mechanism for initiation of cardiac remodeling. Am J Physiol Heart Circ Physiol 2014; 306:H291-7. [DOI: 10.1152/ajpheart.00715.2013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Clinical and animal studies indicate that increased fatty acid delivery to lean tissues induces cardiac electrical remodeling and alterations of cellular calcium homeostasis. Since this may represent a mechanism initiating cardiac dysfunction during establishment of insulin resistance and diabetes or anaerobic cardiac metabolism (ischemia), we sought to determine if short-term exposure to high plasma concentration of fatty acid in vivo was sufficient to alter the cardiac sodium current ( INa) in dog ventricular myocytes. Our results show that delivery of triglycerides and nonesterified fatty acids by infusion of Intralipid + heparin (IH) for 8 h increased the amplitude of INa by 43% and shifted its activation threshold by −5 mV, closer to the resting membrane potential. Steady-state inactivation (availability) of the channels was reduced by IH with no changes in recovery from inactivation. As a consequence, INa “window” current, a strong determinant of intracellular Na+ and Ca2+ concentrations, was significantly increased. The results indicate that increased circulating fatty acids alter INa gating in manners consistent with an increased cardiac excitability and augmentation of intracellular calcium. Moreover, these changes could still be measured after the dogs were left to recover for 12 h after IH perfusion, suggesting lasting changes in INa. Our results indicate that fatty acids rapidly induce cardiac remodeling and suggest that this process may be involved in the development of cardiac dysfunctions associated to insulin resistance and diabetes.
Collapse
Affiliation(s)
- M. Biet
- Department of Physiology and Biophysics, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada; and
| | - N. Morin
- Department of Physiology and Biophysics, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada; and
| | - O. Benrezzak
- Department of Medecine (Endocrinology), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - F. Naimi
- Department of Physiology and Biophysics, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada; and
| | - S. Bellanger
- Department of Physiology and Biophysics, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada; and
| | - J. P. Baillargeon
- Department of Medecine (Endocrinology), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - L. Chouinard
- Department of Medecine (Endocrinology), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - N. Gallo-Payet
- Department of Medecine (Endocrinology), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - A. C. Carpentier
- Department of Medecine (Endocrinology), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - R. Dumaine
- Department of Physiology and Biophysics, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada; and
| |
Collapse
|
40
|
Stables CL, Musa H, Mitra A, Bhushal S, Deo M, Guerrero-Serna G, Mironov S, Zarzoso M, Vikstrom KL, Cawthorn W, Pandit SV. Reduced Na⁺ current density underlies impaired propagation in the diabetic rabbit ventricle. J Mol Cell Cardiol 2014; 69:24-31. [PMID: 24412579 DOI: 10.1016/j.yjmcc.2013.12.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 12/23/2013] [Accepted: 12/31/2013] [Indexed: 12/12/2022]
Abstract
Diabetes is associated with an increased risk of sudden cardiac death, but the underlying mechanisms remain unclear. Our goal was to investigate changes occurring in the action potential duration (APD) and conduction velocity (CV) in the diabetic rabbit ventricle, and delineate the principal ionic determinants. A rabbit model of alloxan-induced diabetes was utilized. Optical imaging was used to record electrical activity in isolated Langendorff-perfused hearts in normo-, hypo- and hyper-kalemia ([K(+)]o=4, 2, 12 mM respectively). Patch clamp experiments were conducted to record Na(+) current (I(Na)) in isolated ventricular myocytes. The mRNA/protein expression levels for Nav1.5 (the α-subunit of I(Na)) and connexin-43 (Cx43), as well as fibrosis levels were examined. Computer simulations were performed to interpret experimental data. We found that the APD was not different, but the CV was significantly reduced in diabetic hearts in normo-, hypo-, and, hyper-kalemic conditions (13%, 17% and 33% reduction in diabetic vs. control, respectively). The cell capacitance (Cm) was increased (by ~14%), and the density of INa was reduced by ~32% in diabetic compared to control hearts, but the other biophysical properties of I(Na) were unaltered. The mRNA/protein expression levels for Cx43 were unaltered. For Nav1.5, the mRNA expression was not changed, and though the protein level tended to be less in diabetic hearts, this reduction was not statistically significant. Staining showed no difference in fibrosis levels between the control and diabetic ventricles. Computer simulations showed that the reduced magnitude of I(Na) was a key determinant of impaired propagation in the diabetic ventricle, which may have important implications for arrhythmogenesis.
Collapse
Affiliation(s)
- Catherine L Stables
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Hassan Musa
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Aditi Mitra
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Sandesh Bhushal
- Department of Engineering, Norfolk State University, Norfolk, VA, USA
| | - Makarand Deo
- Department of Engineering, Norfolk State University, Norfolk, VA, USA
| | - Guadalupe Guerrero-Serna
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Sergey Mironov
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Manuel Zarzoso
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Karen L Vikstrom
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - William Cawthorn
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Sandeep V Pandit
- Center for Arrhythmia Research, Department of Internal Medicine-Cardiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
41
|
Lopez-Izquierdo A, Pereira RO, Wende AR, Punske BB, Abel ED, Tristani-Firouzi M. The absence of insulin signaling in the heart induces changes in potassium channel expression and ventricular repolarization. Am J Physiol Heart Circ Physiol 2013; 306:H747-54. [PMID: 24375641 DOI: 10.1152/ajpheart.00849.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Diabetes mellitus increases the risk for cardiac dysfunction, heart failure, and sudden death. The wide array of neurohumoral changes associated with diabetes pose a challenge to understanding the roles of specific pathways that alter cardiac function. Here, we use a mouse model with cardiomyocyte-restricted deletion of insulin receptors (CIRKO, cardiac-specific insulin receptor knockout) to study the specific effects of impaired cardiac insulin signaling on ventricular repolarization, independent of the generalized metabolic derangements associated with diabetes. Impaired insulin action caused a reduction in mRNA and protein expression of several key K(+) channels that dominate ventricular repolarization. Specifically, components of transient outward K(+) current fast component (Ito,fast; Kv4.2 and KChiP2) were reduced, consistent with a reduction in the amplitude of Ito,fast in isolated left ventricular CIRKO myocytes, compared with littermate controls. The reduction in Ito,fast resulted in ventricular action potential prolongation and prolongation of the QT interval on the surface ECG. These results support the notion that the lack of insulin signaling in the heart is sufficient to cause the repolarization abnormalities described in other animal models of diabetes.
Collapse
Affiliation(s)
- Angelica Lopez-Izquierdo
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | | | | | | | | | | |
Collapse
|
42
|
Lu Z, Jiang YP, Wu CYC, Ballou LM, Liu S, Carpenter ES, Rosen MR, Cohen IS, Lin RZ. Increased persistent sodium current due to decreased PI3K signaling contributes to QT prolongation in the diabetic heart. Diabetes 2013; 62:4257-65. [PMID: 23974924 PMCID: PMC3837031 DOI: 10.2337/db13-0420] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diabetes is an independent risk factor for sudden cardiac death and ventricular arrhythmia complications of acute coronary syndrome. Prolongation of the QT interval on the electrocardiogram is also a risk factor for arrhythmias and sudden death, and the increased prevalence of QT prolongation is an independent risk factor for cardiovascular death in diabetic patients. The pathophysiological mechanisms responsible for this lethal complication are poorly understood. Diabetes is associated with a reduction in phosphoinositide 3-kinase (PI3K) signaling, which regulates the action potential duration (APD) of individual myocytes and thus the QT interval by altering multiple ion currents, including the persistent sodium current INaP. Here, we report a mechanism for diabetes-induced QT prolongation that involves an increase in INaP caused by defective PI3K signaling. Cardiac myocytes of mice with type 1 or type 2 diabetes exhibited an increase in APD that was reversed by expression of constitutively active PI3K or intracellular infusion of phosphatidylinositol 3,4,5-trisphosphate (PIP3), the second messenger produced by PI3K. The diabetic myocytes also showed an increase in INaP that was reversed by activated PI3K or PIP3. The increases in APD and INaP in myocytes translated into QT interval prolongation for both types of diabetic mice. The long QT interval of type 1 diabetic hearts was shortened by insulin treatment ex vivo, and this effect was blocked by a PI3K inhibitor. Treatment of both types of diabetic mouse hearts with an INaP blocker also shortened the QT interval. These results indicate that downregulation of cardiac PI3K signaling in diabetes prolongs the QT interval at least in part by causing an increase in INaP. This mechanism may explain why the diabetic population has an increased risk of life-threatening arrhythmias.
Collapse
Affiliation(s)
- Zhongju Lu
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Ya-Ping Jiang
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Chia-Yen C. Wu
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Lisa M. Ballou
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Shengnan Liu
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Eileen S. Carpenter
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | - Michael R. Rosen
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
- Department of Pharmacology, Columbia University, New York, New York
| | - Ira S. Cohen
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
- Corresponding author: Ira S. Cohen, , or Richard Z. Lin,
| | - Richard Z. Lin
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
- Medical Service, Northport VA Medical Center, Northport, New York
- Corresponding author: Ira S. Cohen, , or Richard Z. Lin,
| |
Collapse
|
43
|
Wu M, Obara Y, Norota I, Nagasawa Y, Ishii K. Insulin suppresses IKs (KCNQ1/KCNE1) currents, which require β-subunit KCNE1. Pflugers Arch 2013; 466:937-46. [PMID: 24068254 DOI: 10.1007/s00424-013-1352-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 09/06/2013] [Accepted: 09/07/2013] [Indexed: 11/29/2022]
Abstract
Abnormal QT prolongation in diabetic patients has become a clinical problem because it increases the risk of lethal ventricular arrhythmia. In an animal model of type 1 diabetes mellitus, several ion currents, including the slowly activating delayed rectifier potassium current (IKs), are altered. The IKs channel is composed of KCNQ1 and KCNE1 subunits, whose genetic mutations are well known to cause long QT syndrome. Although insulin is known to affect many physiological and pathophysiological events in the heart, acute effects of insulin on cardiac ion channels are poorly understood at present. This study was designed to investigate direct electrophysiological effects of insulin on IKs (KCNQ1/KCNE1) currents. KCNQ1 and KCNE1 were co-expressed in Xenopus oocytes, and whole cell currents were measured by a two-microelectrode voltage-clamp method. Acute application of insulin suppressed the KCNQ1/KCNE1 currents and phosphorylated Akt and extracellular signal-regulated kinase (ERK), the two major downstream effectors, in a concentration-dependent manner. Wortmannin (10(-6) M), a phosphoinositide 3-kinase (PI3K) inhibitor, attenuated the suppression of the currents and phosphorylation of Akt by insulin, whereas U0126 (10(-5) M), a mitogen-activated protein kinase kinase (MEK) inhibitor, had no effect on insulin-induced suppression of the currents. In addition, insulin had little effect on KCNQ1 currents without KCNE1, which indicated an essential role of KCNE1 in the acute suppressive effects of insulin. Mutagenesis studies revealed amino acid residues 111-118 within the distal third C-terminus of KCNE1 as an important region. Insulin has direct electrophysiological effects on IKs currents, which may affect cardiac excitability.
Collapse
Affiliation(s)
- Minghua Wu
- Department of Pharmacology, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata, 990-9585, Japan
| | | | | | | | | |
Collapse
|
44
|
Kistamás K, Szentandrássy N, Hegyi B, Ruzsnavszky F, Váczi K, Bárándi L, Horváth B, Szebeni A, Magyar J, Bányász T, Kecskeméti V, Nánási PP. Effects of pioglitazone on cardiac ion currents and action potential morphology in canine ventricular myocytes. Eur J Pharmacol 2013; 710:10-9. [PMID: 23588116 DOI: 10.1016/j.ejphar.2013.03.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 03/21/2013] [Accepted: 03/28/2013] [Indexed: 12/17/2022]
Abstract
Despite its widespread therapeutical use there is little information on the cellular cardiac effects of the antidiabetic drug pioglitazone in larger mammals. In the present study, therefore, the concentration-dependent effects of pioglitazone on ion currents and action potential configuration were studied in isolated canine ventricular myocytes using standard microelectrode, conventional whole cell patch clamp, and action potential voltage clamp techniques. Pioglitazone decreased the maximum velocity of depolarization and the amplitude of phase-1 repolarization at concentrations ≥3 μM. Action potentials were shortened by pioglitazone at concentrations ≥10 μM, which effect was accompanied with significant reduction of beat-to-beat variability of action potential duration. Several transmembrane ion currents, including the transient outward K(+) current (Ito), the L-type Ca(2+) current (ICa), the rapid and slow components of the delayed rectifier K(+) current (IKr and IKs, respectively), and the inward rectifier K(+) current (IK1) were inhibited by pioglitazone under conventional voltage clamp conditions. Ito was blocked significantly at concentrations ≥3 μM, ICa, IKr, IKs at concentrations ≥10 μM, while IK1 at concentrations ≥30 μM. Suppression of Ito, ICa, IKr, and IK1 has been confirmed also under action potential voltage clamp conditions. ATP-sensitive K(+) current, when activated by lemakalim, was effectively blocked by pioglitazone. Accordingly, action potentials were prolonged by 10 μM pioglitazone when the drug was applied in the presence of lemakalim. All these effects developed rapidly and were readily reversible upon washout. In conclusion, pioglitazone seems to be a harmless agent at usual therapeutic concentrations.
Collapse
Affiliation(s)
- Kornél Kistamás
- Department of Physiology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Torres-Jacome J, Gallego M, Rodríguez-Robledo JM, Sanchez-Chapula JA, Casis O. Improvement of the metabolic status recovers cardiac potassium channel synthesis in experimental diabetes. Acta Physiol (Oxf) 2013. [PMID: 23181465 DOI: 10.1111/apha.12043] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIMS The fast transient outward current, I(to,fast) , is the most extensively studied cardiac K(+) current in diabetic animals. Two hypotheses have been proposed to explain how type-1 diabetes reduces this current in cardiac muscle. The first one is a deficiency in channel expression due to a defect in the trophic effect of insulin. The second one proposes flawed glucose metabolism as the cause of the reduced I(to,fast) . Moreover, little information exists about the effects and possible mechanisms of diabetes on the other repolarizing currents of the human heart: I(to,slow) , I(Kr) , I(Ks) , I(Kur) , I(Kslow) and I(K1) . METHODS We recorded cardiac action potentials and K(+) currents in ventricular cells isolated from control and streptozotocin- or alloxan-induced diabetic mice and rabbits. Channel protein expression was determined by immunofluorescence. RESULTS Diabetes reduces the amplitude of I(to,fast) , I(to,slow) and I(Kslow) , in ventricular myocytes from mouse and rabbit, with no effect on I(ss) , I(Kr) or I(K1) . The absence of changes in the biophysical properties of the currents and the immunofluorescence experiments confirmed the reduction in channel protein synthesis. Six-hour incubation of myocytes with insulin or pyruvate recovered current amplitudes and fluorescent staining. The activation of AMP-K reduced the same K(+) currents in healthy myocytes and prevented the pyruvate-induced current recovery. CONCLUSION Diabetes reduces K(+) current densities in ventricular myocytes due to a defect in channel protein synthesis. Activation of AMP-K secondary to deterioration in the metabolic status of the cells is responsible for K(+) channel reductions.
Collapse
Affiliation(s)
- J. Torres-Jacome
- Unidad de Investigación ‘Carlos Médez’ del Centro Universitario de Investigaciones Biomédicas; Universidad de Colima; Colima; México
| | | | - J. M. Rodríguez-Robledo
- Department of Physiology; School of Pharmacy; University of the Basque Country UPV/EHU; Vitoria; Spain
| | - J. A. Sanchez-Chapula
- Unidad de Investigación ‘Carlos Médez’ del Centro Universitario de Investigaciones Biomédicas; Universidad de Colima; Colima; México
| | | |
Collapse
|
46
|
Xie C, Biary N, Tocchetti CG, Aon MA, Paolocci N, Kauffman J, Akar FG. Glutathione oxidation unmasks proarrhythmic vulnerability of chronically hyperglycemic guinea pigs. Am J Physiol Heart Circ Physiol 2013; 304:H916-26. [PMID: 23376824 DOI: 10.1152/ajpheart.00026.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic hyperglycemia in type-1 diabetes mellitus is associated with oxidative stress (OS) and sudden death. Mechanistic links remain unclear. We investigated changes in electrophysiological (EP) properties in a model of chronic hyperglycemia before and after challenge with OS by GSH oxidation and tested reversibility of EP remodeling by insulin. Guinea pigs survived for 1 mo following streptozotocin (STZ) or saline (sham) injection. A treatment group received daily insulin for 2 wk to reverse STZ-induced hyperglycemia (STZ + Ins). EP properties were measured using high-resolution optical action potential mapping before and after challenge of hearts with diamide. Despite elevation of glucose levels in STZ compared with sham-operated (P = 0.004) and STZ + Ins (P = 0.002) animals, average action potential duration (APD) and arrhythmia propensity were not altered at baseline. Diamide promoted early (<10 min) formation of arrhythmic triggers reflected by a higher arrhythmia scoring index in STZ (P = 0.045) and STZ + Ins (P = 0.033) hearts compared with sham-operated hearts. APD heterogeneity underwent a more pronounced increase in response to diamide in STZ and STZ + Ins hearts compared with sham-operated hearts. Within 30 min, diamide resulted in spontaneous incidence of ventricular tachycardia and ventricular fibrillation (VT/VF) in 3/6, 2/5, 1/5, and 0/4 STZ, STZ + Ins, sham-operated, and normal hearts, respectively. Hearts prone to VT/VF exhibited greater APD heterogeneity (P = 0.010) compared with their VT/VF-free counterparts. Finally, altered EP properties in STZ were not rescued by insulin. In conclusion, GSH oxidation enhances APD heterogeneity and increases arrhythmia scoring index in a guinea pig model of chronic hyperglycemia. Despite normalization of glycemic levels by insulin, these proarrhythmic properties are not reversed, suggesting the importance of targeting antioxidant defenses for arrhythmia suppression.
Collapse
Affiliation(s)
- Chaoqin Xie
- Cardiovascular Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Xiang YL, He L, Xiao J, Xia S, Deng SB, Xiu Y, She Q. Effect of trimetazidine treatment on the transient outward potassium current of the left ventricular myocytes of rats with streptozotocin-induced type 1 diabetes mellitus. Braz J Med Biol Res 2012; 45:205-11. [PMID: 22331134 PMCID: PMC3854190 DOI: 10.1590/s0100-879x2012007500019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2011] [Accepted: 02/06/2012] [Indexed: 02/22/2023] Open
Abstract
Cardiovascular complications are a leading cause of mortality in patients with diabetes mellitus (DM). The present study was designed to investigate the effects of trimetazidine (TMZ), an anti-angina drug, on transient outward potassium current (Ito) remodeling in ventricular myocytes and the plasma contents of free fatty acid (FFA) and glucose in DM. Sprague-Dawley rats, 8 weeks old and weighing 200-250 g, were randomly divided into three groups of 20 animals each. The control group was injected with vehicle (1 mM citrate buffer), the DM group was injected with 65 mg/kg streptozotocin (STZ) for induction of type 1 DM, and the DM+TMZ group was injected with the same dose of STZ followed by a 4-week treatment with TMZ (60 mg·kg−1·day−1). All animals were then euthanized and their hearts excised and subjected to electrophysiological measurements or gene expression analyses. TMZ exposure significantly reversed the increased plasma FFA level in diabetic rats, but failed to change the plasma glucose level. The amplitude of Ito was significantly decreased in left ventricular myocytes from diabetic rats relative to control animals (6.25 ± 1.45 vs 20.72 ± 2.93 pA/pF at +40 mV). The DM-associated Ito reduction was attenuated by TMZ. Moreover, TMZ treatment reversed the increased expression of the channel-forming alpha subunit Kv1.4 and the decreased expression of Kv4.2 and Kv4.3 in diabetic rat hearts. These data demonstrate that TMZ can normalize, or partially normalize, the increased plasma FFA content, the reduced Ito of ventricular myocytes, and the altered expression Kv1.4, Kv4.2, and Kv4.3 in type 1 DM.
Collapse
Affiliation(s)
- Yu-luan Xiang
- Department of Cardiology, the Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
48
|
Virág L, Jost N, Papp R, Koncz I, Kristóf A, Kohajda Z, Harmati G, Carbonell-Pascual B, Ferrero JM, Papp JG, Nánási PP, Varró A. Analysis of the contribution of I(to) to repolarization in canine ventricular myocardium. Br J Pharmacol 2012; 164:93-105. [PMID: 21410683 DOI: 10.1111/j.1476-5381.2011.01331.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The contribution of the transient outward potassium current (I(to)) to ventricular repolarization is controversial as it depends on the experimental conditions, the region of myocardium and the species studied. The aim of the present study was therefore to characterize I(to) and estimate its contribution to repolarization reserve in canine ventricular myocardium. EXPERIMENTAL APPROACH Ion currents were recorded using conventional whole-cell voltage clamp and action potential voltage clamp techniques in canine isolated ventricular cells. Action potentials were recorded from canine ventricular preparations using microelectrodes. The contribution of I(to) to repolarization was studied using 100 µM chromanol 293B in the presence of 0.5 µM HMR 1556, which fully blocks I(Ks). KEY RESULTS The high concentration of chromanol 293B used effectively suppressed I(to) without affecting other repolarizing K(+) currents (I(K1), I(Kr), I(p)). Action potential clamp experiments revealed a slowly inactivating and a 'late' chromanol-sensitive current component occurring during the action potential plateau. Action potentials were significantly lengthened by chromanol 293B in the presence of HMR 1556. This lengthening effect induced by I(to) inhibition was found to be reverse rate-dependent. It was significantly augmented after additional attenuation of repolarization reserve by 0.1 µM dofetilide and this caused the occurrence of early afterdepolarizations. The results were confirmed by computer simulation. CONCLUSIONS AND IMPLICATIONS The results indicate that I(to) is involved in regulating repolarization in canine ventricular myocardium and that it contributes significantly to the repolarization reserve. Therefore, blockade of I(to) may enhance pro-arrhythmic risk.
Collapse
Affiliation(s)
- L Virág
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Szebeni A, Szentandrássy N, Pacher P, Simkó J, Nánási PP, Kecskeméti V. Can the electrophysiological action of rosiglitazone explain its cardiac side effects? Curr Med Chem 2011; 18:3720-8. [PMID: 21774756 DOI: 10.2174/092986711796642364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Accepted: 07/09/2011] [Indexed: 01/30/2023]
Abstract
Recent large clinical trials found an association between the antidiabetic drug rosiglitazone therapy and increased risk of cardiovascular adverse events. The aim of this report is to elucidate the cardiac electrophysiological properties of rosiglitazone (R) on isolated rat and murine ventricular papillary muscle cells and canine ventricular myocytes using conventional microelectrode, whole cell voltage clamp, and action potential (AP) voltage clamp techniques. In histidine-decarboxylase knockout mice as well as in their wild types R (1-30 µM) shortened AP duration at 90% level of repolarization (APD(90)) and increased the AP amplitude (APA) in a concentration-dependent manner. In rat ventricular papillary muscle cells R (1-30 µM) caused a significant reduction of APA and maximum velocity of depolarization (V(max)) which was accompanied by lengthening of APD(90). In single canine ventricular myocytes at concentrations ≥10 µM R decreased the amplitude of phase-1 repolarization, the plateau potential and reduced V(max). R suppressed several ion currents in a concentration-dependent manner under voltage clamp conditions. The EC(50) value for this inhibition was 25.2±2.7 µM for the transient outward K(+ ) current (I(to)), 72.3±9.3 µM for the rapid delayed rectifier K(+ ) current (I(Kr)), and 82.5±9.4 µM for the L-type Ca(2+ ) current (I(Ca)) with Hill coefficients close to unity. The inward rectifier K(+ ) current (I(K1)) was not affected by R up to concentrations of 100 µM. Suppression of I(to), I(Kr), and I(Ca) has been confirmed under action potential voltage clamp conditions as well. The observed alterations in the AP morphology and densities of ion currents may predict serious proarrhythmic risk in case of intoxication with R as a consequence of overdose or decreased elimination of the drug, particularly in patients having multiple cardiovascular risk factors, such as elderly diabetic patients.
Collapse
Affiliation(s)
- A Szebeni
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Nagyvárad tér 4, P.O.B. 370, 1445, Hungary
| | | | | | | | | | | |
Collapse
|
50
|
Varró A, Baczkó I. Cardiac ventricular repolarization reserve: a principle for understanding drug-related proarrhythmic risk. Br J Pharmacol 2011; 164:14-36. [PMID: 21545574 PMCID: PMC3171857 DOI: 10.1111/j.1476-5381.2011.01367.x] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 02/22/2011] [Accepted: 03/02/2011] [Indexed: 12/11/2022] Open
Abstract
Cardiac repolarization abnormalities can be caused by a wide range of cardiac and non-cardiac compounds and may lead to the development of life-threatening Torsades de Pointes (TdP) ventricular arrhythmias. Drug-induced torsades de pointes is associated with unexpected and unexplained sudden cardiac deaths resulting in the withdrawal of several compounds in the past. To better understand the mechanism of such unexpected sudden cardiac deaths, the concept of repolarization reserve has recently emerged. According to this concept, pharmacological, congenital or acquired impairment of one type of transmembrane ion channel does not necessarily result in excessive repolarization changes because other repolarizing currents can take over and compensate. In this review, the major factors contributing to repolarization reserve are discussed in the context of their clinical significance in physiological and pathophysiological conditions including drug administration, genetic defects, heart failure, diabetes mellitus, gender, renal failure, hypokalaemia, hypothyroidism and athletes' sudden deaths. In addition, pharmacological support of repolarization reserve as a possible therapeutic option is discussed. Some methods for the quantitative estimation of repolarization reserve are also recommended. It is concluded that repolarization reserve should be considered by safety pharmacologists to better understand, predict and prevent previously unexplained drug-induced sudden cardiac deaths.
Collapse
Affiliation(s)
- András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary.
| | | |
Collapse
|