51
|
Mittal A, Garg R, Bahl A, Khullar M. Molecular Mechanisms and Epigenetic Regulation in Diabetic Cardiomyopathy. Front Cardiovasc Med 2022; 8:725532. [PMID: 34977165 PMCID: PMC8716459 DOI: 10.3389/fcvm.2021.725532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Diabetes mellitus (DM) is an important lifestyle disease. Type 2 diabetes is one of the prime contributors to cardiovascular diseases (CVD) and diabetic cardiomyopathy (DbCM) and leads to increased morbidity and mortality in patients with DM. DbCM is a typical cardiac disease, characterized by cardiac remodeling in the presence of DM and in the absence of other comorbidities such as hypertension, valvular diseases, and coronary artery disease. DbCM is associated with defective cardiac metabolism, altered mitochondrial structure and function, and other physiological and pathophysiological signaling mechanisms such as oxidative stress, inflammation, myocardial apoptosis, and autophagy. Epigenetic modifiers are crucial players in the pathogenesis of DbCM. Thus, it is important to explore the role of epigenetic modifiers or modifications in regulating molecular pathways associated with DbCM. In this review, we have discussed the role of various epigenetic mechanisms such as histone modifications (acetylation and methylation), DNA methylation and non-coding RNAs in modulating molecular pathways involved in the pathophysiology of the DbCM.
Collapse
Affiliation(s)
- Anupam Mittal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajni Garg
- Council of Scientific and Industrial Research - Institute of Microbial Technology, Chandigarh, India
| | - Ajay Bahl
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
52
|
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: 16] [Impact Index Per Article: 5.3] [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
|
53
|
Chronic Intermittent Hypoxia Regulates CaMKII-Dependent MAPK Signaling to Promote the Initiation of Abdominal Aortic Aneurysm. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:2502324. [PMID: 34970414 PMCID: PMC8714336 DOI: 10.1155/2021/2502324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 02/05/2023]
Abstract
Obstructive sleep apnea (OSA) is highly prevalent in patients with abdominal aortic aneurysm (AAA). However, the effects of OSA on AAA initiation in a murine model of sleep apnea have not been completely studied. In this paper, Apoe−/− C57BL/6 mice infused with angiotensin II (Ang II) were placed in chronic intermittent hypoxia (CIH) condition for inducing OSA-related AAA. CIH significantly promoted the incidence of AAA and inhibited the survival of mice. By performing ultrasonography and elastic Van Gieson staining, CIH was found to be effective in promoting aortic dilation and elastin degradation. Immunohistochemical and zymography results show that CIH upregulated the expression and activity of MMP2 and MMP9 and upregulated MCP1 expression while downregulating α-SMA expression. Also, CIH exposure promoted ROS generation, apoptosis, and mitochondria damage in vascular smooth muscle cells (VSMCs), which were measured by ROS assay, TUNEL staining, and transmission electron microscopy. The result of RNA sequencing of mouse aortas displayed that 232 mRNAs were differently expressed between Ang II and Ang II+CIH groups, and CaMKII-dependent p38/Jnk was confirmed as one downstream signaling of CIH. CaMKII-IN-1, an inhibitor of CaMKII, eliminated the effects of CIH on the loss of primary VSMCs. To conclude, a mouse model of OSA-related AAA, which contains the phenotypes of both AAA and OSA, was established in this study. We suggested CIH as a risk factor of AAA initiation through CaMKII-dependent MAPK signaling.
Collapse
|
54
|
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: 5.5] [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
|
55
|
Kistamás K, Hézső T, Horváth B, Nánási PP. Late sodium current and calcium homeostasis in arrhythmogenesis. Channels (Austin) 2021; 15:1-19. [PMID: 33258400 PMCID: PMC7757849 DOI: 10.1080/19336950.2020.1854986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/26/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
The cardiac late sodium current (INa,late) is the small sustained component of the sodium current active during the plateau phase of the action potential. Several studies demonstrated that augmentation of the current can lead to cardiac arrhythmias; therefore, INa,late is considered as a promising antiarrhythmic target. Fundamentally, enlarged INa,late increases Na+ influx into the cell, which, in turn, is converted to elevated intracellular Ca2+ concentration through the Na+/Ca2+ exchanger. The excessive Ca2+ load is known to be proarrhythmic. This review describes the behavior of the voltage-gated Na+ channels generating INa,late in health and disease and aims to discuss the physiology and pathophysiology of Na+ and Ca2+ homeostasis in context with the enhanced INa,late demonstrating also the currently accessible antiarrhythmic choices.
Collapse
Affiliation(s)
- Kornél Kistamás
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Hézső
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter P Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Dental Physiology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
56
|
Simon M, Ko CY, Rebbeck RT, Baidar S, Cornea RL, Bers DM. CaMKIIδ post-translational modifications increase affinity for calmodulin inside cardiac ventricular myocytes. J Mol Cell Cardiol 2021; 161:53-61. [PMID: 34371035 PMCID: PMC8716136 DOI: 10.1016/j.yjmcc.2021.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 11/18/2022]
Abstract
Persistent over-activation of CaMKII (Calcium/Calmodulin-dependent protein Kinase II) in the heart is implicated in arrhythmias, heart failure, pathological remodeling, and other cardiovascular diseases. Several post-translational modifications (PTMs)-including autophosphorylation, oxidation, S-nitrosylation, and O-GlcNAcylation-have been shown to trap CaMKII in an autonomously active state. The molecular mechanisms by which these PTMs regulate calmodulin (CaM) binding to CaMKIIδ-the primary cardiac isoform-has not been well-studied particularly in its native myocyte environment. Typically, CaMKII activates upon Ca-CaM binding during locally elevated [Ca]free and deactivates upon Ca-CaM dissociation when [Ca]free returns to basal levels. To assess the effects of CaMKIIδ PTMs on CaM binding, we developed a novel FRET (Förster resonance energy transfer) approach to directly measure CaM binding to and dissociation from CaMKIIδ in live cardiac myocytes. We demonstrate that autophosphorylation of CaMKIIδ increases affinity for CaM in its native environment and that this increase is dependent on [Ca]free. This leads to a 3-fold slowing of CaM dissociation from CaMKIIδ (time constant slows from ~0.5 to 1.5 s) when [Ca]free is reduced with physiological kinetics. Moreover, oxidation further slows CaM dissociation from CaMKIIδ T287D (phosphomimetic) upon rapid [Ca]free chelation and increases FRET between CaM and CaMKIIδ T287A (phosphoresistant). The CaM dissociation kinetics-measured here in myocytes-are similar to the interval between heartbeats, and integrative memory would be expected as a function of heart rate. Furthermore, the PTM-induced slowing of dissociation between beats would greatly promote persistent CaMKIIδ activity in the heart. Together, these findings suggest a significant role of PTM-induced changes in CaMKIIδ affinity for CaM and memory under physiological and pathophysiological processes in the heart.
Collapse
Affiliation(s)
- Mitchell Simon
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Christopher Y Ko
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Robyn T Rebbeck
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Sonya Baidar
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, CA, USA.
| |
Collapse
|
57
|
Gaitán-González P, Sánchez-Hernández R, Arias-Montaño JA, Rueda A. Tale of two kinases: Protein kinase A and Ca 2+/calmodulin-dependent protein kinase II in pre-diabetic cardiomyopathy. World J Diabetes 2021; 12:1704-1718. [PMID: 34754372 PMCID: PMC8554373 DOI: 10.4239/wjd.v12.i10.1704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/28/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome is a pre-diabetic state characterized by several biochemical and physiological alterations, including insulin resistance, visceral fat accumulation, and dyslipidemias, which increase the risk for developing cardiovascular disease. Metabolic syndrome is associated with augmented sympathetic tone, which could account for the etiology of pre-diabetic cardiomyopathy. This review summarizes the current knowledge of the pathophysiological consequences of enhanced and sustained β-adrenergic response in pre-diabetes, focusing on cardiac dysfunction reported in diet-induced experimental models of pre-diabetic cardiomyopathy. The research reviewed indicates that both protein kinase A and Ca2+/calmodulin-dependent protein kinase II play important roles in functional responses mediated by β1-adrenoceptors; therefore, alterations in the expression or function of these kinases can be deleterious. This review also outlines recent information on the role of protein kinase A and Ca2+/calmodulin-dependent protein kinase II in abnormal Ca2+ handling by cardiomyocytes from diet-induced models of pre-diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Pamela Gaitán-González
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Rommel Sánchez-Hernández
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - José-Antonio Arias-Montaño
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Angélica Rueda
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| |
Collapse
|
58
|
Federico M, Zavala M, Vico T, López S, Portiansky E, Alvarez S, Abrille MCV, Palomeque J. CaMKII activation in early diabetic hearts induces altered sarcoplasmic reticulum-mitochondria signaling. Sci Rep 2021; 11:20025. [PMID: 34625584 PMCID: PMC8501049 DOI: 10.1038/s41598-021-99118-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/20/2021] [Indexed: 01/01/2023] Open
Abstract
Prediabetic myocardium, induced by fructose-rich diet (FRD), is prone to increased sarcoplasmic reticulum (SR)-Ca2+ leak and arrhythmias due to increased activity of the Ca2+/calmodulin protein kinase II (CaMKII). However, little is known about the role of SR-mitochondria microdomains, mitochondrial structure, and mitochondrial metabolisms. To address this knowledge gap we measured SR-mitochondrial proximity, intracellular Ca2+, and mitochondrial metabolism in wild type (WT) and AC3-I transgenic mice, with myocardial-targeted CaMKII inhibition, fed with control diet (CD) or with FRD. Confocal images showed significantly increased spontaneous Ca2+ release events in FRD vs. CD WT cardiomyocytes. [3H]-Ryanodine binding assay revealed higher [3H]Ry binding in FRD than CD WT hearts. O2 consumption at State 4 and hydrogen peroxide (H2O2) production rate were increased, while respiratory control rate (RCR) and Ca2+ retention capacity (CRC) were decreased in FRD vs. CD WT isolated mitochondria. Transmission Electron Microscopy (TEM) images showed increased proximity at the SR-mitochondria microdomains, associated with increased tethering proteins, Mfn2, Grp75, and VDAC in FRD vs. CD WT. Mitochondria diameter was decrease and roundness and density were increased in FRD vs. CD WT specimens. The fission protein, Drp1 was significantly increased while the fusion protein, Opa1 was unchanged in FRD vs. CD WT hearts. These differences were prevented in AC3-I mice. We conclude that SR-mitochondria microdomains are subject to CaMKII-dependent remodeling, involving SR-Ca2+ leak and mitochondria fission, in prediabetic mice induced by FRD. We speculate that CaMKII hyperactivity induces SR-Ca2+ leak by RyR2 activation which in turn increases mitochondria Ca2+ content due to the enhanced SR-mitochondria tethering, decreasing CRC.
Collapse
Affiliation(s)
- Marilen Federico
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina
| | - Maite Zavala
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina
| | - Tamara Vico
- Instituto de Bioquímica y Medicina Molecular, UBA-CONICET, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Sofía López
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina
| | - Enrique Portiansky
- Laboratorio de Análisis de Imágenes, UNLP, Facultad de Ciencias Veterinarias, La Plata, Argentina
| | - Silvia Alvarez
- Instituto de Bioquímica y Medicina Molecular, UBA-CONICET, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Maria Celeste Villa Abrille
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina
| | - Julieta Palomeque
- Centro de Investigaciones Cardiovasculares, UNLP-CONICET-CCT La Plata, Facultad de Ciencias Médicas, UNLP, 60 y 120 s/n, La Plata, CP 1900, Argentina.
| |
Collapse
|
59
|
MicroRNAs and Calcium Signaling in Heart Disease. Int J Mol Sci 2021; 22:ijms221910582. [PMID: 34638924 PMCID: PMC8508866 DOI: 10.3390/ijms221910582] [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: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 01/02/2023] Open
Abstract
In hearts, calcium (Ca2+) signaling is a crucial regulatory mechanism of muscle contraction and electrical signals that determine heart rhythm and control cell growth. Ca2+ signals must be tightly controlled for a healthy heart, and the impairment of Ca2+ handling proteins is a key hallmark of heart disease. The discovery of microRNA (miRNAs) as a new class of gene regulators has greatly expanded our understanding of the controlling module of cardiac Ca2+ cycling. Furthermore, many studies have explored the involvement of miRNAs in heart diseases. In this review, we aim to summarize cardiac Ca2+ signaling and Ca2+-related miRNAs in pathological conditions, including cardiac hypertrophy, heart failure, myocardial infarction, and atrial fibrillation. We also discuss the therapeutic potential of Ca2+-related miRNAs as a new target for the treatment of heart diseases.
Collapse
|
60
|
Hegyi B, Shimkunas R, Jian Z, Izu LT, Bers DM, Chen-Izu Y. Mechanoelectric coupling and arrhythmogenesis in cardiomyocytes contracting under mechanical afterload in a 3D viscoelastic hydrogel. Proc Natl Acad Sci U S A 2021; 118:e2108484118. [PMID: 34326268 PMCID: PMC8346795 DOI: 10.1073/pnas.2108484118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The heart pumps blood against the mechanical afterload from arterial resistance, and increased afterload may alter cardiac electrophysiology and contribute to life-threatening arrhythmias. However, the cellular and molecular mechanisms underlying mechanoelectric coupling in cardiomyocytes remain unclear. We developed an innovative patch-clamp-in-gel technology to embed cardiomyocytes in a three-dimensional (3D) viscoelastic hydrogel that imposes an afterload during regular myocyte contraction. Here, we investigated how afterload affects action potentials, ionic currents, intracellular Ca2+ transients, and cell contraction of adult rabbit ventricular cardiomyocytes. We found that afterload prolonged action potential duration (APD), increased transient outward K+ current, decreased inward rectifier K+ current, and increased L-type Ca2+ current. Increased Ca2+ entry caused enhanced Ca2+ transients and contractility. Moreover, elevated afterload led to discordant alternans in APD and Ca2+ transient. Ca2+ alternans persisted under action potential clamp, indicating that the alternans was Ca2+ dependent. Furthermore, all these afterload effects were significantly attenuated by inhibiting nitric oxide synthase 1 (NOS1). Taken together, our data reveal a mechano-chemo-electrotransduction (MCET) mechanism that acutely transduces afterload through NOS1-nitric oxide signaling to modulate the action potential, Ca2+ transient, and contractility. The MCET pathway provides a feedback loop in excitation-Ca2+ signaling-contraction coupling, enabling autoregulation of contractility in cardiomyocytes in response to afterload. This MCET mechanism is integral to the individual cardiomyocyte (and thus the heart) to intrinsically enhance its contractility in response to the load against which it has to do work. While this MCET is largely compensatory for physiological load changes, it may also increase susceptibility to arrhythmias under excessive pathological loading.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Rafael Shimkunas
- Department of Pharmacology, University of California, Davis, CA 95616
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Zhong Jian
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Leighton T Izu
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, CA 95616;
- Department of Biomedical Engineering, University of California, Davis, CA 95616
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| |
Collapse
|
61
|
Siri-Angkul N, Dadfar B, Jaleel R, Naushad J, Parambathazhath J, Doye AA, Xie LH, Gwathmey JK. Calcium and Heart Failure: How Did We Get Here and Where Are We Going? Int J Mol Sci 2021; 22:ijms22147392. [PMID: 34299010 PMCID: PMC8306046 DOI: 10.3390/ijms22147392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/23/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
Abstract
The occurrence and prevalence of heart failure remain high in the United States as well as globally. One person dies every 30 s from heart disease. Recognizing the importance of heart failure, clinicians and scientists have sought better therapeutic strategies and even cures for end-stage heart failure. This exploration has resulted in many failed clinical trials testing novel classes of pharmaceutical drugs and even gene therapy. As a result, along the way, there have been paradigm shifts toward and away from differing therapeutic approaches. The continued prevalence of death from heart failure, however, clearly demonstrates that the heart is not simply a pump and instead forces us to consider the complexity of simplicity in the pathophysiology of heart failure and reinforces the need to discover new therapeutic approaches.
Collapse
Affiliation(s)
- Natthaphat Siri-Angkul
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Behzad Dadfar
- Department of General Medicine, School of Medicine, Mazandaran University of Medical Sciences, Sari 1471655836, Iran
| | - Riya Jaleel
- School of International Education, Zhengzhou University, Zhengzhou 450001, China
| | - Jazna Naushad
- Weill Cornell Medicine Qatar, Doha P. O. Box 24144, Qatar
| | | | | | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA
| | - Judith K. Gwathmey
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Correspondence: ; Tel.: +973-972-2411; Fax: +973-972-7489
| |
Collapse
|
62
|
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: 3.3] [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
|
63
|
Melatonin, Its Metabolites and Their Interference with Reactive Nitrogen Compounds. Molecules 2021; 26:molecules26134105. [PMID: 34279445 PMCID: PMC8271479 DOI: 10.3390/molecules26134105] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Melatonin and several of its metabolites are interfering with reactive nitrogen. With the notion of prevailing melatonin formation in tissues that exceeds by far the quantities in blood, metabolites come into focus that are poorly found in the circulation. Apart from their antioxidant actions, both melatonin and N1-acetyl-5-methoxykynuramine (AMK) downregulate inducible and inhibit neuronal NO synthases, and additionally scavenge NO. However, the NO adduct of melatonin redonates NO, whereas AMK forms with NO a stable product. Many other melatonin metabolites formed in oxidative processes also contain nitrosylatable sites. Moreover, AMK readily scavenges products of the CO2-adduct of peroxynitrite such as carbonate radicals and NO2. Protein AMKylation seems to be involved in protective actions.
Collapse
|
64
|
Salvatore T, Pafundi PC, Galiero R, Albanese G, Di Martino A, Caturano A, Vetrano E, Rinaldi L, Sasso FC. The Diabetic Cardiomyopathy: The Contributing Pathophysiological Mechanisms. Front Med (Lausanne) 2021; 8:695792. [PMID: 34277669 PMCID: PMC8279779 DOI: 10.3389/fmed.2021.695792] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Individuals with diabetes mellitus (DM) disclose a higher incidence and a poorer prognosis of heart failure (HF) than non-diabetic people, even in the absence of other HF risk factors. The adverse impact of diabetes on HF likely reflects an underlying “diabetic cardiomyopathy” (DM–CMP), which may by exacerbated by left ventricular hypertrophy and coronary artery disease (CAD). The pathogenesis of DM-CMP has been a hot topic of research since its first description and is still under active investigation, as a complex interplay among multiple mechanisms may play a role at systemic, myocardial, and cellular/molecular levels. Among these, metabolic abnormalities such as lipotoxicity and glucotoxicity, mitochondrial damage and dysfunction, oxidative stress, abnormal calcium signaling, inflammation, epigenetic factors, and others. These disturbances predispose the diabetic heart to extracellular remodeling and hypertrophy, thus leading to left ventricular diastolic and systolic dysfunction. This Review aims to outline the major pathophysiological changes and the underlying mechanisms leading to myocardial remodeling and cardiac functional derangement in DM-CMP.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
65
|
Hegyi B, Fasoli A, Ko CY, Van BW, Alim CC, Shen EY, Ciccozzi MM, Tapa S, Ripplinger CM, Erickson JR, Bossuyt J, Bers DM. CaMKII Serine 280 O-GlcNAcylation Links Diabetic Hyperglycemia to Proarrhythmia. Circ Res 2021; 129:98-113. [PMID: 33926209 PMCID: PMC8221539 DOI: 10.1161/circresaha.120.318402] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 04/22/2021] [Accepted: 04/28/2021] [Indexed: 12/16/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Action Potentials
- Adult
- Aged
- Animals
- Arrhythmias, Cardiac/enzymology
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Biomarkers/blood
- Blood Glucose/metabolism
- Calcium Signaling
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Case-Control Studies
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Excitation Contraction Coupling
- Female
- Glycosylation
- Heart Rate
- Humans
- Male
- Mice, Inbred C57BL
- Mice, Inbred ICR
- Mice, Transgenic
- Middle Aged
- Mutation
- Myocardial Contraction
- Myocytes, Cardiac/enzymology
- NADPH Oxidase 2/genetics
- NADPH Oxidase 2/metabolism
- Phosphorylation
- Protein Processing, Post-Translational
- Mice
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Anna Fasoli
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Christopher Y. Ko
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Benjamin W. Van
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Chidera C. Alim
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Erin Y. Shen
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Marisa M. Ciccozzi
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Srinivas Tapa
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Crystal M. Ripplinger
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Jeffrey R. Erickson
- Department of Physiology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand (J.R.E.)
| | - Julie Bossuyt
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| | - Donald M. Bers
- Department of Pharmacology, University of California, Davis (B.H., A.F., C.Y.K., B.W.V., C.C.A., E.Y.S., M.M.C., S.T., C.M.R., J.B., D.M.B.)
| |
Collapse
|
66
|
Ca 2+/calmodulin kinase II-dependent regulation of β IV-spectrin modulates cardiac fibroblast gene expression, proliferation, and contractility. J Biol Chem 2021; 297:100893. [PMID: 34153319 PMCID: PMC8294584 DOI: 10.1016/j.jbc.2021.100893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/10/2021] [Accepted: 06/17/2021] [Indexed: 01/26/2023] Open
Abstract
Fibrosis is a pronounced feature of heart disease and the result of dysregulated activation of resident cardiac fibroblasts (CFs). Recent work identified stress-induced degradation of the cytoskeletal protein βIV-spectrin as an important step in CF activation and cardiac fibrosis. Furthermore, loss of βIV-spectrin was found to depend on Ca2+/calmodulin-dependent kinase II (CaMKII). Therefore, we sought to determine the mechanism for CaMKII-dependent regulation of βIV-spectrin and CF activity. Computational screening and MS revealed a critical serine residue (S2250 in mouse and S2254 in human) in βIV-spectrin phosphorylated by CaMKII. Disruption of βIV-spectrin/CaMKII interaction or alanine substitution of βIV-spectrin Ser2250 (βIV-S2254A) prevented CaMKII-induced degradation, whereas a phosphomimetic construct (βIV-spectrin with glutamic acid substitution at serine 2254 [βIV-S2254E]) showed accelerated degradation in the absence of CaMKII. To assess the physiological significance of this phosphorylation event, we expressed exogenous βIV-S2254A and βIV-S2254E constructs in βIV-spectrin-deficient CFs, which have increased proliferation and fibrotic gene expression compared with WT CFs. βIV-S2254A but not βIV-S2254E normalized CF proliferation, gene expression, and contractility. Pathophysiological targeting of βIV-spectrin phosphorylation and subsequent degradation was identified in CFs activated with the profibrotic ligand angiotensin II, resulting in increased proliferation and signal transducer and activation of transcription 3 nuclear accumulation. While therapeutic delivery of exogenous WT βIV-spectrin partially reversed these trends, βIV-S2254A completely negated increased CF proliferation and signal transducer and activation of transcription 3 translocation. Moreover, we observed βIV-spectrin phosphorylation and associated loss in total protein within human heart tissue following heart failure. Together, these data illustrate a considerable role for the βIV-spectrin/CaMKII interaction in activating profibrotic signaling.
Collapse
|
67
|
RIPK3-Mediated Necroptosis in Diabetic Cardiomyopathy Requires CaMKII Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6617816. [PMID: 34194608 PMCID: PMC8203407 DOI: 10.1155/2021/6617816] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/01/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023]
Abstract
Activation of Ca2+/calmodulin-dependent protein kinase (CaMKII) has been proved to play a vital role in cardiovascular diseases. Receptor-interaction protein kinase 3- (RIPK3-) mediated necroptosis has crucially participated in cardiac dysfunction. The study is aimed at investigating the effect as well as the mechanism of CaMKII activation and necroptosis on diabetic cardiomyopathy (DCM). Wild-type (WT) and the RIPK3 gene knockout (RIPK3−/−) mice were intraperitoneally injected with 60 mg/kg/d streptozotocin (STZ) for 5 consecutive days. After 12 w of feeding, 100 μL recombinant adenovirus solution carrying inhibitor 1 of protein phosphatase 1 (I1PP1) gene was injected into the caudal vein of mice. Echocardiography, myocardial injury, CaMKII activity, necroptosis, RIPK1 expression, mixed lineage kinase domain-like protein (MLKL) phosphorylation, and mitochondrial ultrastructure were measured. The results showed that cardiac dysfunction, CaMKII activation, and necroptosis were aggravated in streptozotocin- (STZ-) stimulated mice, as well as in (Lepr) KO/KO (db/db) mice. RIPK3 deficiency alleviated cardiac dysfunction, CaMKII activation, and necroptosis in DCM. Furthermore, I1PP1 overexpression reversed cardiac dysfunction, myocardial injury and necroptosis augment, and CaMKII activity enhancement in WT mice with DCM but not in RIPK3−/− mice with DCM. The present study demonstrated that CaMKII activation and necroptosis augment in DCM via a RIPK3-dependent manner, which may provide therapeutic strategies for DCM.
Collapse
|
68
|
Liu Y, Shao Q, Cheng HJ, Li T, Zhang X, Callahan MF, Herrington D, Kitzman D, Zhao D, Cheng CP. Chronic Ca 2+/Calmodulin-Dependent Protein Kinase II Inhibition Rescues Advanced Heart Failure. J Pharmacol Exp Ther 2021; 377:316-325. [PMID: 33722881 PMCID: PMC8140392 DOI: 10.1124/jpet.120.000361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/11/2021] [Indexed: 11/22/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is upregulated in congestive heart failure (CHF), contributing to electrical, structural, and functional remodeling. CaMKII inhibition is known to improve CHF, but its direct cardiac effects in CHF remain unclear. We hypothesized that CaMKII inhibition improves cardiomyocyte function, [Ca2+]i regulation, and β-adrenergic reserve, thus improving advanced CHF. In a 16-week study, we compared plasma neurohormonal levels and left ventricular (LV)- and myocyte-functional and calcium transient ([Ca2+]iT) responses in male Sprague-Dawley rats (10/group) with CHF induced by isoproterenol (170 mg/kg sq for 2 days). In rats with CHF, we studied the effects of the CaMKII inhibitor KN-93 or its inactive analog KN-92 (n = 4) (70 µg/kg per day, mini-pump) for 4 weeks. Compared with controls, isoproterenol-treated rats had severe CHF with 5-fold-increased plasma norepinephrine and about 50% decreases in ejection fraction (EF) and LV contractility [slope of LV end-systolic pressure-LV end-systolic volume relation (EES)] but increased time constant of LV relaxation (τ). They also showed significantly reduced myocyte contraction [maximum rate of myocyte shortening (dL/dtmax)], relaxation (dL/dtmax), and [Ca2+]iT Isoproterenol superfusion caused significantly fewer increases in dL/dtmax and [Ca2+]iT KN-93 treatment prevented plasma norepinephrine elevation, with increased basal and acute isoproterenol-stimulated increases in EF and EES and decreased τ in CHF. KN-93 treatment preserved normal myocyte contraction, relaxation, [Ca2+]iT, and β-adrenergic reserve, whereas KN-92 treatment failed to improve LV and myocyte function, and plasma norepinephrine remained high in CHF. Thus, chronic CaMKII inhibition prevented CHF-induced activation of the sympathetic nervous system, restoring normal LV and cardiomyocyte basal and β-adrenergic-stimulated contraction, relaxation, and [Ca2+]iT, thereby playing a rescue role in advanced CHF. SIGNIFICANCE STATEMENT: We investigated the therapeutic efficacy of late initiation of chronic Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibition on progression of advanced congestive heart failure (CHF). Chronic CaMKII inhibition prevented CHF-induced activation of the sympathetic nervous system and restored normal intrinsic cardiomyocyte basal and β-adrenergic receptor-stimulated relaxation, contraction, and [Ca2+]i regulation, leading to reversal of CHF progression. These data provide new evidence that CaMKII inhibition is able and sufficient to rescue a failing heart, and thus cardiac CaMKII inhibition is a promising target for improving CHF treatment.
Collapse
Affiliation(s)
- Yixi Liu
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Qun Shao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Heng-Jie Cheng
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Tiankai Li
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Xiaowei Zhang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Michael F Callahan
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - David Herrington
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Dalane Kitzman
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - David Zhao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Che-Ping Cheng
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| |
Collapse
|
69
|
Mishra S, Kass DA. Cellular and molecular pathobiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 2021; 18:400-423. [PMID: 33432192 PMCID: PMC8574228 DOI: 10.1038/s41569-020-00480-6] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 01/30/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) affects half of all patients with heart failure worldwide, is increasing in prevalence, confers substantial morbidity and mortality, and has very few effective treatments. HFpEF is arguably the greatest unmet medical need in cardiovascular disease. Although HFpEF was initially considered to be a haemodynamic disorder characterized by hypertension, cardiac hypertrophy and diastolic dysfunction, the pandemics of obesity and diabetes mellitus have modified the HFpEF syndrome, which is now recognized to be a multisystem disorder involving the heart, lungs, kidneys, skeletal muscle, adipose tissue, vascular system, and immune and inflammatory signalling. This multiorgan involvement makes HFpEF difficult to model in experimental animals because the condition is not simply cardiac hypertrophy and hypertension with abnormal myocardial relaxation. However, new animal models involving both haemodynamic and metabolic disease, and increasing efforts to examine human pathophysiology, are revealing new signalling pathways and potential therapeutic targets. In this Review, we discuss the cellular and molecular pathobiology of HFpEF, with the major focus being on mechanisms relevant to the heart, because most research has focused on this organ. We also highlight the involvement of other important organ systems, including the lungs, kidneys and skeletal muscle, efforts to characterize patients with the use of systemic biomarkers, and ongoing therapeutic efforts. Our objective is to provide a roadmap of the signalling pathways and mechanisms of HFpEF that are being characterized and which might lead to more patient-specific therapies and improved clinical outcomes.
Collapse
Affiliation(s)
- Sumita Mishra
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David A. Kass
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,
| |
Collapse
|
70
|
Implications of SGLT Inhibition on Redox Signalling in Atrial Fibrillation. Int J Mol Sci 2021; 22:ijms22115937. [PMID: 34073033 PMCID: PMC8198069 DOI: 10.3390/ijms22115937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained (atrial) arrhythmia, a considerable global health burden and often associated with heart failure. Perturbations of redox signalling in cardiomyocytes provide a cellular substrate for the manifestation and maintenance of atrial arrhythmias. Several clinical trials have shown that treatment with sodium-glucose linked transporter inhibitors (SGLTi) improves mortality and hospitalisation in heart failure patients independent of the presence of diabetes. Post hoc analysis of the DECLARE-TIMI 58 trial showed a 19% reduction in AF in patients with diabetes mellitus (hazard ratio, 0.81 (95% confidence interval: 0.68-0.95), n = 17.160) upon treatment with SGLTi, regardless of pre-existing AF or heart failure and independent from blood pressure or renal function. Accordingly, ongoing experimental work suggests that SGLTi not only positively impact heart failure but also counteract cellular ROS production in cardiomyocytes, thereby potentially altering atrial remodelling and reducing AF burden. In this article, we review recent studies investigating the effect of SGLTi on cellular processes closely interlinked with redox balance and their potential effects on the onset and progression of AF. Despite promising insight into SGLTi effect on Ca2+ cycling, Na+ balance, inflammatory and fibrotic signalling, mitochondrial function and energy balance and their potential effect on AF, the data are not yet conclusive and the importance of individual pathways for human AF remains to be established. Lastly, an overview of clinical studies investigating SGLTi in the context of AF is provided.
Collapse
|
71
|
Ren J, Wu NN, Wang S, Sowers JR, Zhang Y. Obesity cardiomyopathy: evidence, mechanisms, and therapeutic implications. Physiol Rev 2021; 101:1745-1807. [PMID: 33949876 PMCID: PMC8422427 DOI: 10.1152/physrev.00030.2020] [Citation(s) in RCA: 206] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The prevalence of heart failure is on the rise and imposes a major health threat, in part, due to the rapidly increased prevalence of overweight and obesity. To this point, epidemiological, clinical, and experimental evidence supports the existence of a unique disease entity termed “obesity cardiomyopathy,” which develops independent of hypertension, coronary heart disease, and other heart diseases. Our contemporary review evaluates the evidence for this pathological condition, examines putative responsible mechanisms, and discusses therapeutic options for this disorder. Clinical findings have consolidated the presence of left ventricular dysfunction in obesity. Experimental investigations have uncovered pathophysiological changes in myocardial structure and function in genetically predisposed and diet-induced obesity. Indeed, contemporary evidence consolidates a wide array of cellular and molecular mechanisms underlying the etiology of obesity cardiomyopathy including adipose tissue dysfunction, systemic inflammation, metabolic disturbances (insulin resistance, abnormal glucose transport, spillover of free fatty acids, lipotoxicity, and amino acid derangement), altered intracellular especially mitochondrial Ca2+ homeostasis, oxidative stress, autophagy/mitophagy defect, myocardial fibrosis, dampened coronary flow reserve, coronary microvascular disease (microangiopathy), and endothelial impairment. Given the important role of obesity in the increased risk of heart failure, especially that with preserved systolic function and the recent rises in COVID-19-associated cardiovascular mortality, this review should provide compelling evidence for the presence of obesity cardiomyopathy, independent of various comorbid conditions, underlying mechanisms, and offer new insights into potential therapeutic approaches (pharmacological and lifestyle modification) for the clinical management of obesity cardiomyopathy.
Collapse
Affiliation(s)
- Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China.,Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Ne N Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shuyi Wang
- School of Medicine, Shanghai University, Shanghai, China.,University of Wyoming College of Health Sciences, Laramie, Wyoming
| | - James R Sowers
- Dalton Cardiovascular Research Center, Diabetes and Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
72
|
Ma K, Ma G, Guo Z, Liu G, Liang W. Regulatory mechanism of calcium/calmodulin-dependent protein kinase II in the occurrence and development of ventricular arrhythmia (Review). Exp Ther Med 2021; 21:656. [PMID: 33968186 PMCID: PMC8097202 DOI: 10.3892/etm.2021.10088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/05/2021] [Indexed: 11/24/2022] Open
Abstract
Ventricular arrhythmia (VA) is a highly fatal arrhythmia that involves multiple ion channels. Of all sudden cardiac death events, ~85% result from VAs, including ventricular tachycardia and ventricular fibrillation. Calcium/calmodulin-dependent pro-tein kinase II (CaMKII) is an important ion channel regulator that participates in the excitation-contraction coupling of the heart, and as such is important for regulating its electrophysiological function. CaMKII can be activated in a Ca2+/calmodulin (CaM)-dependent or Ca2+/CaM-independent manner, serving a key role in the occurrence and development of VA. The present review aimed to determine whether activated CaMKII induces early afterdepolarizations and delayed afterdepolarizations that result in VA by regulating sodium, potassium and calcium ions. Assessing VA mechanisms based on the CaMKII pathway is of great significance to the clinical treatment of VA and the de-velopment of effective drugs for use in clinical practice.
Collapse
Affiliation(s)
- Kexin Ma
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Guoping Ma
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Zijing Guo
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Gang Liu
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Wenjie Liang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
73
|
Khajehlandi M, Bolboli L, Siahkuhian M, Rami M, Tabandeh M, Khoramipour K, Suzuki K. Endurance Training Regulates Expression of Some Angiogenesis-Related Genes in Cardiac Tissue of Experimentally Induced Diabetic Rats. Biomolecules 2021; 11:biom11040498. [PMID: 33806202 PMCID: PMC8066303 DOI: 10.3390/biom11040498] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Exercise can ameliorate cardiovascular dysfunctions in the diabetes condition, but its precise molecular mechanisms have not been entirely understood. The aim of the present study was to determine the impact of endurance training on expression of angiogenesis-related genes in cardiac tissue of diabetic rats. Thirty adults male Wistar rats were randomly divided into three groups (N = 10) including diabetic training (DT), sedentary diabetes (SD), and sedentary healthy (SH), in which diabetes was induced by a single dose of streptozotocin (50 mg/kg). Endurance training (ET) with moderate-intensity was performed on a motorized treadmill for six weeks. Training duration and treadmill speed were increased during five weeks, but they were kept constant at the final week, and slope was zero at all stages. Real-time polymerase chain reaction (RT-PCR) analysis was used to measure the expression of myocyte enhancer factor-2C (MEF2C), histone deacetylase-4 (HDAC4) and Calmodulin-dependent protein kinase II (CaMKII) in cardiac tissues of the rats. Our results demonstrated that six weeks of ET increased gene expression of MEF2C significantly (p < 0.05), and caused a significant reduction in HDAC4 and CaMKII gene expression in the DT rats compared to the SD rats (p < 0.05). We concluded that moderate-intensity ET could play a critical role in ameliorating cardiovascular dysfunction in a diabetes condition by regulating the expression of some angiogenesis-related genes in cardiac tissues.
Collapse
Affiliation(s)
- Mojdeh Khajehlandi
- Department of Exercise Physiology, Faculty of Educational Sciences and Psychology, University of Mohaghegh Ardabili, Ardabil 5619913131, Iran; (M.K.); (M.S.)
| | - Lotfali Bolboli
- Department of Exercise Physiology, Faculty of Educational Sciences and Psychology, University of Mohaghegh Ardabili, Ardabil 5619913131, Iran; (M.K.); (M.S.)
- Correspondence: (L.B.); (K.S.); Tel.: +98-91-4351-2590 (L.B.); +81-4-2947-6898 (K.S.)
| | - Marefat Siahkuhian
- Department of Exercise Physiology, Faculty of Educational Sciences and Psychology, University of Mohaghegh Ardabili, Ardabil 5619913131, Iran; (M.K.); (M.S.)
| | - Mohammad Rami
- Department of Sport Physiology, Faculty of Sport Sciences, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran;
| | - Mohammadreza Tabandeh
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran;
| | - Kayvan Khoramipour
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Physiology Research Center and Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616913555, Iran;
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Saitama, Japan
- Correspondence: (L.B.); (K.S.); Tel.: +98-91-4351-2590 (L.B.); +81-4-2947-6898 (K.S.)
| |
Collapse
|
74
|
Duran J, Nickel L, Estrada M, Backs J, van den Hoogenhof MMG. CaMKIIδ Splice Variants in the Healthy and Diseased Heart. Front Cell Dev Biol 2021; 9:644630. [PMID: 33777949 PMCID: PMC7991079 DOI: 10.3389/fcell.2021.644630] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/22/2021] [Indexed: 01/16/2023] Open
Abstract
RNA splicing has been recognized in recent years as a pivotal player in heart development and disease. The Ca2+/calmodulin dependent protein kinase II delta (CaMKIIδ) is a multifunctional Ser/Thr kinase family and generates at least 11 different splice variants through alternative splicing. This enzyme, which belongs to the CaMKII family, is the predominant family member in the heart and functions as a messenger toward adaptive or detrimental signaling in cardiomyocytes. Classically, the nuclear CaMKIIδB and cytoplasmic CaMKIIδC splice variants are described as mediators of arrhythmias, contractile function, Ca2+ handling, and gene transcription. Recent findings also put CaMKIIδA and CaMKIIδ9 as cardinal players in the global CaMKII response in the heart. In this review, we discuss and summarize the new insights into CaMKIIδ splice variants and their (proposed) functions, as well as CaMKII-engineered mouse phenotypes and cardiac dysfunction related to CaMKIIδ missplicing. We also discuss RNA splicing factors affecting CaMKII splicing. Finally, we discuss the translational perspective derived from these insights and future directions on CaMKIIδ splicing research in the healthy and diseased heart.
Collapse
Affiliation(s)
- Javier Duran
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Lennart Nickel
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Manuel Estrada
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Johannes Backs
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Maarten M G van den Hoogenhof
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| |
Collapse
|
75
|
King DR, Entz M, Blair GA, Crandell I, Hanlon AL, Lin J, Hoeker GS, Poelzing S. The conduction velocity-potassium relationship in the heart is modulated by sodium and calcium. Pflugers Arch 2021; 473:557-571. [PMID: 33660028 PMCID: PMC7940307 DOI: 10.1007/s00424-021-02537-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/28/2021] [Accepted: 02/04/2021] [Indexed: 01/27/2023]
Abstract
The relationship between cardiac conduction velocity (CV) and extracellular potassium (K+) is biphasic, with modest hyperkalemia increasing CV and severe hyperkalemia slowing CV. Recent studies from our group suggest that elevating extracellular sodium (Na+) and calcium (Ca2+) can enhance CV by an extracellular pathway parallel to gap junctional coupling (GJC) called ephaptic coupling that can occur in the gap junction adjacent perinexus. However, it remains unknown whether these same interventions modulate CV as a function of K+. We hypothesize that Na+, Ca2+, and GJC can attenuate conduction slowing consequent to severe hyperkalemia. Elevating Ca2+ from 1.25 to 2.00 mM significantly narrowed perinexal width measured by transmission electron microscopy. Optically mapped, Langendorff-perfused guinea pig hearts perfused with increasing K+ revealed the expected biphasic CV-K+ relationship during perfusion with different Na+ and Ca2+ concentrations. Neither elevating Na+ nor Ca2+ alone consistently modulated the positive slope of CV-K+ or conduction slowing at 10-mM K+; however, combined Na+ and Ca2+ elevation significantly mitigated conduction slowing at 10-mM K+. Pharmacologic GJC inhibition with 30-μM carbenoxolone slowed CV without changing the shape of CV-K+ curves. A computational model of CV predicted that elevating Na+ and narrowing clefts between myocytes, as occur with perinexal narrowing, reduces the positive and negative slopes of the CV-K+ relationship but do not support a primary role of GJC or sodium channel conductance. These data demonstrate that combinatorial effects of Na+ and Ca2+ differentially modulate conduction during hyperkalemia, and enhancing determinants of ephaptic coupling may attenuate conduction changes in a variety of physiologic conditions.
Collapse
Affiliation(s)
- D Ryan King
- Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Michael Entz
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Grace A Blair
- Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Ian Crandell
- Center for Biostatistics and Health Data Science, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | - Alexandra L Hanlon
- Center for Biostatistics and Health Data Science, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | - Joyce Lin
- Department of Mathematics, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Gregory S Hoeker
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Steven Poelzing
- Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
- School of Medicine, Virginia Tech Carilion, Roanoke, VA, USA.
| |
Collapse
|
76
|
Gbr AA, Abdel Baky NA, Mohamed EA, Zaky HS. Cardioprotective effect of pioglitazone and curcumin against diabetic cardiomyopathy in type 1 diabetes mellitus: impact on CaMKII/NF-κB/TGF-β1 and PPAR-γ signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:349-360. [PMID: 32984914 DOI: 10.1007/s00210-020-01979-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022]
Abstract
Diabetic cardiomyopathy (DCM) is a leading cause of death in diabetic patients, which is currently without available specific treatment. This study aimed to investigate the potential protective effects of pioglitazone (Pio) and curcumin (Cur) against DCM in type 1 diabetes mellitus (T1DM), with pointing to their role on Ca+2/calmodulin-dependent protein kinase II (CaMKII) and peroxisome proliferator-activated receptor gamma (PPAR-γ) expression. Diabetes was induced in adult male Sprague Dawley rats by administration of single intraperitoneal injection of streptozotocin (STZ) (52.5 mg/kg). Diabetic rats were administered either Pio (20 mg/kg/day) or Cur (100 mg/kg/day) orally for 6 weeks. Treatment with Pio and/or Cur markedly reduced serum cardiac injury markers and lipid profile markers in diabetic animals. Additionally, Pio and/or Cur treatment mitigated oxidative stress and fibrosis in diabetic rats as evident from the significant suppression in myocardial lipid peroxidation and tumor growth factor beta 1 (TGF-β1) level, with concomitant significant elevation in total antioxidant capacity (TAC) and improvement in histopathological architecture of heart tissue. Pio/Cur treatment protocol accomplished its cardioprotective effect by depressing cardiac CaMKII/NF-κB signaling accompanied by enhancement in PPAR-γ expression. Conclusively, these findings demonstrated the therapeutic potential of Pio/Cur regimen in alleviating DCM in T1DM through modulation of CaMKII and PPAR-γ expression. Graphical Abstract.
Collapse
Affiliation(s)
- Aya A Gbr
- Egypt Ministry of Health and Population, Cairo, Egypt
| | - Nayira A Abdel Baky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Naser City, Cairo, P.N.11754, Egypt.
| | - Eman A Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Naser City, Cairo, P.N.11754, Egypt
| | - Heba S Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Naser City, Cairo, P.N.11754, Egypt
| |
Collapse
|
77
|
Hegyi B, Pölönen RP, Hellgren KT, Ko CY, Ginsburg KS, Bossuyt J, Mercola M, Bers DM. Cardiomyocyte Na + and Ca 2+ mishandling drives vicious cycle involving CaMKII, ROS, and ryanodine receptors. Basic Res Cardiol 2021; 116:58. [PMID: 34648073 PMCID: PMC8516771 DOI: 10.1007/s00395-021-00900-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 12/19/2022]
Abstract
Cardiomyocyte Na+ and Ca2+ mishandling, upregulated Ca2+/calmodulin-dependent kinase II (CaMKII), and increased reactive oxygen species (ROS) are characteristics of various heart diseases, including heart failure (HF), long QT (LQT) syndrome, and catecholaminergic polymorphic ventricular tachycardia (CPVT). These changes may form a vicious cycle of positive feedback to promote cardiac dysfunction and arrhythmias. In HF rabbit cardiomyocytes investigated in this study, the inhibition of CaMKII, late Na+ current (INaL), and leaky ryanodine receptors (RyRs) all attenuated the prolongation and increased short-term variability (STV) of action potential duration (APD), but in age-matched controls these inhibitors had no or minimal effects. In control cardiomyocytes, we enhanced RyR leak (by low [caffeine] plus isoproterenol mimicking CPVT) which markedly increased STV and delayed afterdepolarizations (DADs). These proarrhythmic changes were significantly attenuated by both CaMKII inhibition and mitochondrial ROS scavenging, with a slight synergy with INaL inhibition. Inducing LQT by elevating INaL (by Anemone toxin II, ATX-II) caused markedly prolonged APD, increased STV, and early afterdepolarizations (EADs). Those proarrhythmic ATX-II effects were largely attenuated by mitochondrial ROS scavenging, and partially reduced by inhibition of CaMKII and pathological leaky RyRs using dantrolene. In human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) bearing LQT3 mutation SCN5A N406K, dantrolene significantly attenuated cell arrhythmias and APD prolongation. Targeting critical components of the Na+-Ca2+-CaMKII-ROS-INaL arrhythmogenic vicious cycle may exhibit important on-target and also trans-target effects (e.g., INaL and RyR inhibition can alter INaL-mediated LQT3 effects). Incorporating this vicious cycle into therapeutic strategies provides novel integrated insight for treating cardiac arrhythmias and diseases.
Collapse
Affiliation(s)
- Bence Hegyi
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Risto-Pekka Pölönen
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA ,grid.168010.e0000000419368956Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Kim T. Hellgren
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Christopher Y. Ko
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Kenneth S. Ginsburg
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Julie Bossuyt
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Mark Mercola
- grid.168010.e0000000419368956Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Donald M. Bers
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| |
Collapse
|
78
|
Wang J, Cheng X, Zhao H, Yang Q, Xu Z. Downregulation of the zinc transporter SLC39A13 (ZIP13) is responsible for the activation of CaMKII at reperfusion and leads to myocardial ischemia/reperfusion injury in mouse hearts. J Mol Cell Cardiol 2020; 152:69-79. [PMID: 33307093 DOI: 10.1016/j.yjmcc.2020.12.002] [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: 07/08/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
While Zn2+ dyshomeostasis is known to contribute to ischemia/reperfusion (I/R) injury, the roles of zinc transporters that are responsible for Zn2+ homeostasis in the pathogenesis of I/R injury remain to be addressed. This study reports that ZIP13 (SLC39A13), a zinc transporter, plays a role in myocardial I/R injury by modulating the Ca2+ signaling pathway rather than by regulating Zn2+ transport. ZIP13 is downregulated upon reperfusion in mouse hearts or in H9c2 cells at reoxygenation. Ca2+ but not Zn2+ was responsible for ZIP13 downregulation, implying that ZIP13 may play a role in I/R injury through the Ca2+ signaling pathway. In line with our assumption, knockout of ZIP13 resulted in phosphorylation (Thr287) of Ca2+-calmodulin-dependent protein kinase (CaMKII), indicating that downregulation of ZIP13 leads to CaMKII activation. Further studies showed that the heart-specific knockout of ZIP13 enhanced I/R-induced CaMKII phosphorylation in mouse hearts. In contrast, overexpression of ZIP13 suppressed I/R-induced CaMKII phosphorylation. Moreover, the heart-specific knockout of ZIP13 exacerbated myocardial infarction in mouse hearts subjected to I/R, whereas overexpression of ZIP13 reduced infarct size. In addition, knockout of ZIP13 induced increases of mitochondrial Ca2+, ROS, mitochondrial swelling, decrease in the mitochondrial respiration control rate (RCR), and dissipation of mitochondrial membrane potential (ΔΨm) in a CaMKII-dependent manner. These data suggest that downregulation of ZIP13 at reperfusion contributes to myocardial I/R injury through activation of CaMKII and the mitochondrial death pathway.
Collapse
Affiliation(s)
- Jie Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Xinxin Cheng
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Huanhuan Zhao
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Qing Yang
- Department of Cardiology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Zhelong Xu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China; Department of Cardiology, General Hospital, Tianjin Medical University, Tianjin 300052, China.
| |
Collapse
|
79
|
MD1 Depletion Predisposes to Ventricular Arrhythmias in the Setting of Myocardial Infarction. Heart Lung Circ 2020; 30:869-881. [PMID: 33257269 DOI: 10.1016/j.hlc.2020.09.938] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND Myeloid differentiation protein 1 (MD1) is expressed in the human heart and is a negative regulator of Toll-like receptor 4 (TLR4) signalling. MD1 exerts anti-arrhythmic effects. AIM The aim of this study was to determine the role of MD1 in myocardial infarction (MI)-related ventricular arrhythmias (VAs). METHOD Myocardial infarction was induced by surgical ligation of the left anterior coronary artery in MD1 knockout (KO) mice and their wild-type littermates. Myocardial infarction-induced vulnerability to VAs and its underlying mechanisms were evaluated. RESULTS Myeloid differentiation protein 1 was downregulated in the MI mice. Myeloid differentiation protein 1 deficiency decreased post-MI left ventricular (LV) function and increased the infarct size. The MI mice exhibited prolonged action potential duration (APD), enhanced APD alternans thresholds, and a higher incidence of VAs. Myocardial infarction-induced LV fibrosis and inflammation decreased the expression levels of Kv4.2, Kv4.3, Kv1.5, and Kv2.1, increased Cav1.2 expression, and disturbed Ca2+ handling protein expression. These MI-induced adverse effects were further exacerbated in KO mice. Mechanistically, MD1 deletion markedly enhanced the activation of the TLR4/calmodulin-dependent protein kinase II (CaMKII) signalling pathway in post-MI mice. CONCLUSIONS Myeloid differentiation protein 1 deletion increases the vulnerability to VAs in post-MI mice. This is mainly caused by the aggravated maladaptive LV fibrosis and inflammation and interference with the expressions of ion channels and Ca2+ handling proteins, which is related to enhanced activation of the TLR4/CaMKII signalling pathway.
Collapse
|
80
|
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: 54] [Impact Index Per Article: 10.8] [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
|
81
|
Yang Y, He J, Yuan M, Tse G, Zhang K, Ma Z, Li J, Zhang Y, Gao Y, Zhang Y, Wang R, Li G, Liu T. Xanthine oxidase inhibitor allopurinol improves atrial electrical remodeling in diabetic rats by inhibiting CaMKII/NCX signaling. Life Sci 2020; 259:118290. [PMID: 32822713 DOI: 10.1016/j.lfs.2020.118290] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/09/2020] [Accepted: 08/13/2020] [Indexed: 02/08/2023]
Abstract
AIMS Atrial fibrillation (AF) is a common arrhythmia which is associated with higher risk of stroke, heart failure and all-cause mortality. Abnormal Ca2+ handling in diabetes mellitus (DM) can cause delayed depolarization involved with increased NCX activity. Complicated mechanisms are involved in atrial remodeling, of which CaMKII may be a key node signal. Therefore, we intend to explore whether CaMKII activation induces atrial electrical remodeling by regulating NCX expression in this study. MAIN METHODS Adult male SD rats were used to establish a diabetic rat model, divided into three groups: the control group, DM group and allopurinol group. Hemodynamic and ECG indicators were recorded, after which electrophysiological studies were conducted. The protein expression of CaMKII, p-CaMKII, XO, MnSOD and NCX was measured by Western blot and immunohistochemistry. H&E and Masson staining were applied for observing myocardial fibrosis. HL-1 cells were cultured for the measurement of ROS generation. KEY FINDINGS The arrangement of atrial myocytes was disordered and the collagen volume fraction of the atrium tissue was elevated in the DM group compared with the control group, and improved by allopurinol. Higher incidence of inducible AF, reduced conduction velocity and higher conduction inhomogeneity were observed in diabetic rats. These electrophysiological abnormalities were accompanied by higher oxidative stress and protein expression of p-CaMKII and NCX. Allopurinol prevented the development of these abnormal changes. SIGNIFICANCE Allopurinol can improve atrial electrical remodeling by inhibiting CaMKII activity and protein expression of NCX. These data indicate xanthine oxidase inhibition can reduce oxidative stress and ameliorate atrial electrical remodeling.
Collapse
Affiliation(s)
- Yajuan Yang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China; Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jinli He
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Ming Yuan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Kai Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zuowang Ma
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Jian Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yue Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yunlai Gao
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Yu Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Ruxing Wang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
82
|
Romero-García T, Landa-Galvan HV, Pavón N, Mercado-Morales M, Valdivia HH, Rueda A. Autonomous activation of CaMKII exacerbates diastolic calcium leak during beta-adrenergic stimulation in cardiomyocytes of metabolic syndrome rats. Cell Calcium 2020; 91:102267. [PMID: 32920522 DOI: 10.1016/j.ceca.2020.102267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023]
Abstract
Autonomous Ca2+/calmodulin-dependent protein kinase II (CaMKII) activation induces abnormal diastolic Ca2+ leak, which leads to triggered arrhythmias in a wide range of cardiovascular diseases, including diabetic cardiomyopathy. In hyperglycemia, Ca2+ handling alterations can be aggravated under stress conditions via the β-adrenergic signaling pathway, which also involves CaMKII activation. However, little is known about intracellular Ca2+ handling disturbances under β-adrenergic stimulation in cardiomyocytes of the prediabetic metabolic syndrome (MetS) model with obesity, and the participation of CaMKII in these alterations. MetS was induced in male Wistar rats by administering 30 % sucrose in drinking water for 16 weeks. Fluo 3-loaded MetS cardiomyocytes exhibited augmented diastolic Ca2+ leak (in the form of spontaneous Ca2+ waves) under basal conditions and that Ca2+ leakage was exacerbated by isoproterenol (ISO, 100 nM). At the molecular level, [3H]-ryanodine binding and basal phosphorylation of cardiac ryanodine receptor (RyR2) at Ser2814, a CaMKII site, were increased in heart homogenates of MetS rats with no changes in RyR2 expression. These alterations were not further augmented by Isoproterenol. SERCA pump activity was augmented 48 % in MetS hearts before β-adrenergic stimuli, which is associated to augmented PLN phosphorylation at T17, a target of CaMKII. In MetS hearts. CaMKII auto-phosphorylation (T287) was increased by 80 %. The augmented diastolic Ca2+ leak was prevented by CaMKII inhibition with AIP. In conclusion, CaMKII autonomous activation in cardiomyocytes of MetS rats with central obesity significantly contributes to abnormal diastolic Ca2+ leak, increasing the propensity for β-adrenergic receptor-driven lethal arrhythmias.
Collapse
Affiliation(s)
- Tatiana Romero-García
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del IPN, Cinvestav-IPN, Mexico City, 07300 Mexico
| | - Huguet V Landa-Galvan
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del IPN, Cinvestav-IPN, Mexico City, 07300 Mexico
| | - Natalia Pavón
- Department of Pharmacology, Instituto Nacional de Cardiología "Ignacio Chavez", Mexico City, Mexico
| | - Martha Mercado-Morales
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del IPN, Cinvestav-IPN, Mexico City, 07300 Mexico
| | - Héctor H Valdivia
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Angélica Rueda
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del IPN, Cinvestav-IPN, Mexico City, 07300 Mexico.
| |
Collapse
|
83
|
Bhattacharyya M, Karandur D, Kuriyan J. Structural Insights into the Regulation of Ca 2+/Calmodulin-Dependent Protein Kinase II (CaMKII). Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035147. [PMID: 31653643 DOI: 10.1101/cshperspect.a035147] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a highly conserved serine/threonine kinase that is ubiquitously expressed throughout the human body. Specialized isoforms of CaMKII play key roles in neuronal and cardiac signaling. The distinctive holoenzyme architecture of CaMKII, with 12-14 kinase domains attached by flexible linkers to a central hub, poses formidable challenges for structural characterization. Nevertheless, progress in determining the structural mechanisms underlying CaMKII functions has come from studying the kinase domain and the hub separately, as well as from a recent electron microscopic investigation of the intact holoenzyme. In this review, we discuss our current understanding of the structure of CaMKII. We also discuss the intriguing finding that the CaMKII holoenzyme can undergo activation-triggered subunit exchange, a process that has implications for the potentiation and perpetuation of CaMKII activity.
Collapse
Affiliation(s)
- Moitrayee Bhattacharyya
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California 94720.,Howard Hughes Medical Institute, University of California, Berkeley, California 94720
| | - Deepti Karandur
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California 94720.,Howard Hughes Medical Institute, University of California, Berkeley, California 94720
| | - John Kuriyan
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720.,California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California 94720.,Howard Hughes Medical Institute, University of California, Berkeley, California 94720.,Department of Chemistry, University of California, Berkeley, California 94720.,Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| |
Collapse
|
84
|
Skrzycki M, Kaźmierczak B. The hidden role of the Sigma1 receptor in muscle cells. J Recept Signal Transduct Res 2020; 40:201-208. [PMID: 32054378 DOI: 10.1080/10799893.2020.1727924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 10/25/2022]
Abstract
This review describes the very specific role of Sigma1 receptor in different types of muscle cells. Sigma1 receptor is a transmembrane protein residing in such structures like MAM. It has chaperoning activity supporting function of many proteins, particularly ion channels, including Ca2+ channels. This latter function is of particular meaning for muscle cells, due to their calcium-based/regulated metabolism. Here we discuss new reports pointing to participation of Sigma1 receptor in muscle specific processes like contraction, EC-coupling, calcium currents and in diseases like left ventricular hypertrophy, transverse aortic stenosis and hypertension-induced heart dysfunction.
Collapse
Affiliation(s)
- Michał Skrzycki
- Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | - Beata Kaźmierczak
- Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
85
|
Amelioration of diastolic dysfunction by dapagliflozin in a non-diabetic model involves coronary endothelium. Pharmacol Res 2020; 157:104781. [PMID: 32360273 DOI: 10.1016/j.phrs.2020.104781] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 01/13/2023]
Abstract
The results of trials with sodium-glucose cotransporter 2 (SGLT2) inhibitors raised the possibility that this class of drugs provides cardiovascular benefits independently from their anti-diabetic effects, although the mechanisms are unknown. Therefore, we tested the effects of SGLT2 inhibitor dapagliflozin on the progression of experimental heart disease in a non-diabetic model of heart failure with preserved ejection fraction. Dahl salt-sensitive rats were fed a high-salt diet to induce hypertension and diastolic dysfunction and were then treated with dapagliflozin for six weeks. Dapagliflozin ameliorated diastolic function as documented by echo-Doppler and heart catheterization, while blood pressure remained markedly elevated. Chronic in vivo treatment with dapagliflozin reduced diastolic Ca2+ and Na+ overload and increased Ca2+ transient amplitude in ventricular cardiomyocytes, although no direct action of dapagliflozin on isolated cardiomyocytes was observed. Dapagliflozin reversed endothelial activation and endothelial nitric oxide synthase deficit, with reduced cardiac inflammation and consequent attenuation of pro-fibrotic signaling. The potential involvement of coronary endothelium was supported by the endothelial upregulation of Na+/H+ exchanger 1in vivo and direct effects on dapagliflozin on the activity of this exchanger in endothelial cells in vitro. In conclusions, several mechanisms may cumulatively play a significant role in the dapagliflozin-associated cardioprotection. Dapagliflozin ameliorates diastolic function and exerts a positive effect on the myocardium, possibly targeting coronary endothelium. The lower degree of endothelial dysfunction, inflammation and fibrosis translate into improved myocardial performance.
Collapse
|
86
|
Hegyi B, Chen-Izu Y, Izu LT, Rajamani S, Belardinelli L, Bers DM, Bányász T. Balance Between Rapid Delayed Rectifier K + Current and Late Na + Current on Ventricular Repolarization: An Effective Antiarrhythmic Target? Circ Arrhythm Electrophysiol 2020; 13:e008130. [PMID: 32202931 PMCID: PMC7331791 DOI: 10.1161/circep.119.008130] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/16/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Rapid delayed rectifier K+ current (IKr) and late Na+ current (INaL) significantly shape the cardiac action potential (AP). Changes in their magnitudes can cause either long or short QT syndromes associated with malignant ventricular arrhythmias and sudden cardiac death. METHODS Physiological self AP-clamp was used to measure INaL and IKr during the AP in rabbit and porcine ventricular cardiomyocytes to test our hypothesis that the balance between IKr and INaL affects repolarization stability in health and disease conditions. RESULTS We found comparable amount of net charge carried by IKr and INaL during the physiological AP, suggesting that outward K+ current via IKr and inward Na+ current via INaL are in balance during physiological repolarization. Remarkably, IKr and INaL integrals in each control myocyte were highly correlated in both healthy rabbit and pig myocytes, despite high overall cell-to-cell variability. This close correlation was lost in heart failure myocytes from both species. Pretreatment with E-4031 to block IKr (mimicking long QT syndrome 2) or with sea anemone toxin II to impair Na+ channel inactivation (mimicking long QT syndrome 3) prolonged AP duration (APD); however, using GS-967 to inhibit INaL sufficiently restored APD to control in both cases. Importantly, INaL inhibition significantly reduced the beat-to-beat and short-term variabilities of APD. Moreover, INaL inhibition also restored APD and repolarization stability in heart failure. Conversely, pretreatment with GS-967 shortened APD (mimicking short QT syndrome), and E-4031 reverted APD shortening. Furthermore, the amplitude of AP alternans occurring at high pacing frequency was decreased by INaL inhibition, increased by IKr inhibition, and restored by combined INaL and IKr inhibitions. CONCLUSIONS Our data demonstrate that IKr and INaL are counterbalancing currents during the physiological ventricular AP and their integrals covary in individual myocytes. Targeting these ionic currents to normalize their balance may have significant therapeutic potential in heart diseases with repolarization abnormalities. Visual Overview: A visual overview is available for this article.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis
- Department of Biomedical Engineering, University of California, Davis
- Department of Internal Medicine/Cardiology, University of California, Davis
| | | | - Sridharan Rajamani
- Amgen, Inc., South San Francisco, University of Debrecen, Debrecen, Hungary
| | - Luiz Belardinelli
- InCarda Therapeutics, Inc., Newark, CA, University of Debrecen, Debrecen, Hungary
| | - Donald M. Bers
- Department of Pharmacology, University of California, Davis
| | - Tamás Bányász
- Department of Pharmacology, University of California, Davis
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
87
|
Cardiomyocyte calcium handling in health and disease: Insights from in vitro and in silico studies. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 157:54-75. [PMID: 32188566 DOI: 10.1016/j.pbiomolbio.2020.02.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/31/2019] [Accepted: 02/29/2020] [Indexed: 02/07/2023]
Abstract
Calcium (Ca2+) plays a central role in cardiomyocyte excitation-contraction coupling. To ensure an optimal electrical impulse propagation and cardiac contraction, Ca2+ levels are regulated by a variety of Ca2+-handling proteins. In turn, Ca2+ modulates numerous electrophysiological processes. Accordingly, Ca2+-handling abnormalities can promote cardiac arrhythmias via various mechanisms, including the promotion of afterdepolarizations, ion-channel modulation and structural remodeling. In the last 30 years, significant improvements have been made in the computational modeling of cardiomyocyte Ca2+ handling under physiological and pathological conditions. However, numerous questions involving the Ca2+-dependent regulation of different macromolecular complexes, cross-talk between Ca2+-dependent regulatory pathways operating over a wide range of time scales, and bidirectional interactions between electrophysiology and mechanics remain to be addressed by in vitro and in silico studies. A better understanding of disease-specific Ca2+-dependent proarrhythmic mechanisms may facilitate the development of improved therapeutic strategies. In this review, we describe the fundamental mechanisms of cardiomyocyte Ca2+ handling in health and disease, and provide an overview of currently available computational models for cardiomyocyte Ca2+ handling. Finally, we discuss important uncertainties and open questions about cardiomyocyte Ca2+ handling and highlight how synergy between in vitro and in silico studies may help to answer several of these issues.
Collapse
|
88
|
Kistamás K, Veress R, Horváth B, Bányász T, Nánási PP, Eisner DA. Calcium Handling Defects and Cardiac Arrhythmia Syndromes. Front Pharmacol 2020; 11:72. [PMID: 32161540 PMCID: PMC7052815 DOI: 10.3389/fphar.2020.00072] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
Calcium ions (Ca2+) play a major role in the cardiac excitation-contraction coupling. Intracellular Ca2+ concentration increases during systole and falls in diastole thereby determining cardiac contraction and relaxation. Normal cardiac function also requires perfect organization of the ion currents at the cellular level to drive action potentials and to maintain action potential propagation and electrical homogeneity at the tissue level. Any imbalance in Ca2+ homeostasis of a cardiac myocyte can lead to electrical disturbances. This review aims to discuss cardiac physiology and pathophysiology from the elementary membrane processes that can cause the electrical instability of the ventricular myocytes through intracellular Ca2+ handling maladies to inherited and acquired arrhythmias. Finally, the paper will discuss the current therapeutic approaches targeting cardiac arrhythmias.
Collapse
Affiliation(s)
- Kornél Kistamás
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Roland Veress
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter P Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Dental Physiology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - David A Eisner
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
89
|
Qin L, Wang J, Zhao R, Zhang X, Mei Y. Ginsenoside-Rb1 Improved Diabetic Cardiomyopathy through Regulating Calcium Signaling by Alleviating Protein O-GlcNAcylation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:14074-14085. [PMID: 31793297 DOI: 10.1021/acs.jafc.9b05706] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Ginsenoside-Rb1 (Rb1), a major active component of ginseng, has many benefits for cardiovascular disease and diabetes mellitus (DM), but the effect and mechanism on diabetic cardiomyopathy are not clear. In the present study, we found that Rb1-feeding significantly improved cardiac dysfunction and abnormal cardiomyocytes calcium signaling caused by diabetes. This improved calcium signaling was because Rb1 reduced Ca2+ leakage caused by overactivated ryanodine receptor 2 (RyR2) and increased Ca2+ uptake by sarcoplasmic reticulum Ca2+-ATPase 2a (SERCA 2a). Furthermore, we found that Rb1 not only enhanced energy metabolism like metformin and eliminated O-GlcNAcylation of calcium handling proteins to regulate calcium signaling but also directly inhibited RyR2 activity to regulate calcium signaling. The present study indicated that as a health supplement or drug, Rb1 was a relatively effective auxiliary therapeutic substance for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Linhui Qin
- School of Basic Medical Sciences , Zhengzhou University , Zhengzhou 450001 , Henan , China
| | - Jianping Wang
- Chengdu Slan Biotechnology Co., Ltd. , Chengdu 610041 , Sichuan , China
| | - RongRong Zhao
- School of Basic Medical Sciences , Zhengzhou University , Zhengzhou 450001 , Henan , China
| | - Xiao Zhang
- School of Basic Medical Sciences , Zhengzhou University , Zhengzhou 450001 , Henan , China
| | - Yingwu Mei
- School of Basic Medical Sciences , Zhengzhou University , Zhengzhou 450001 , Henan , China
| |
Collapse
|
90
|
Li K, Lin Y, Li C. MiR-338-5p ameliorates pathological cardiac hypertrophy by targeting CAMKIIδ. Arch Pharm Res 2019; 42:1071-1080. [PMID: 31820396 DOI: 10.1007/s12272-019-01199-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/29/2019] [Indexed: 12/11/2022]
Abstract
Pathological cardiac hypertrophy (PCH) is characterized by an increase in cardiomyocyte size and thickening of the ventricular walls during the adaptive response to maintain cardiac function, which often progresses to a maladaptive response and, ultimately, to heart failure. Previous studies have demonstrated that miRNAs play roles in the pathogenesis of PCH. In this study, we first found that the regulation of miR-338-5p was aberrant in cardiac tissues of heart failure patients and transverse aortic constriction (TAC)-induced PCH mice. Overexpression of miR-338-5p in the heart using recombinant adeno-associated virus serotype 9 (rAAV9) ameliorated TAC-induced PCH, as indicated by a decreased heart weight/body weight (HW/BW) ratio. Furthermore, miR-338-5p mitigated the TAC-induced damage in heart contraction and relaxation function, as measured by echocardiography and a cardio hemodynamic measurement, respectively. We also identified CAMKIIδ as a direct target of miR-338-5p using bioinformatics tools and the luciferase reporter assay. Finally, we observed that the miR-338-5p-mediated downregulation of CAMKIIδ reversed the cell surface area enlargement induced by the Ang-II treatment in H9c2 cells. Therefore, we highlight a novel molecular mechanism of the miR-338-5p/CAMKIIδ axis that contributes to the pathogenesis of PCH.
Collapse
Affiliation(s)
- Kailong Li
- Department of Ultrasound, Affiliated Hospital of Jining Medical University, Jining, 272000, Shandong, PR China
| | - Yuedong Lin
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, PR China
| | - Cong Li
- Department of Ultrasound, Affiliated Hospital of Jining Medical University, Jining, 272000, Shandong, PR China.
| |
Collapse
|
91
|
Abstract
Advances in the treatment of heart failure with reduced ejection fraction due to systolic dysfunction are engaging an ever-expanding compendium of molecular signaling targets. Well established approaches modifying hemodynamics and cell biology by neurohumoral receptor blockade are evolving, exploring the role and impact of modulating intracellular signaling pathways with more direct myocardial effects. Even well-tread avenues are being reconsidered with new insights into the signaling engaged and thus opportunity to treat underlying myocardial disease. This review explores therapies that have proven successful, those that have not, those that are moving into the clinic but whose utility remains to be confirmed, and those that remain in the experimental realm. The emphasis is on signaling pathways that are tractable for therapeutic manipulation. Of the approaches yet to be tested in humans, we chose those with a well-established experimental history, where clinical translation may be around the corner. The breadth of opportunities bodes well for the next generation of heart failure therapeutics.
Collapse
Affiliation(s)
| | | | - David A. Kass
- Division of Cardiology, Department of Medicine
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore Maryland, 21205
| |
Collapse
|
92
|
Hegyi B, Chen-Izu Y, Izu LT, Bányász T. Altered K + current profiles underlie cardiac action potential shortening in hyperkalemia and β-adrenergic stimulation. Can J Physiol Pharmacol 2019; 97:773-780. [PMID: 31091413 DOI: 10.1139/cjpp-2019-0056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hyperkalemia is known to develop in various conditions including vigorous physical exercise. In the heart, hyperkalemia is associated with action potential (AP) shortening that was attributed to altered gating of K+ channels. However, it remains unknown how hyperkalemia changes the profiles of each K+ current under a cardiac AP. Therefore, we recorded the major K+ currents (inward rectifier K+ current, IK1; rapid and slow delayed rectifier K+ currents, IKr and IKs, respectively) using AP-clamp in rabbit ventricular myocytes. As K+ may accumulate at rapid heart rates during sympathetic stimulation, we also examined the effect of isoproterenol on these K+ currents. We found that IK1 was significantly increased in hyperkalemia, whereas the reduction of driving force for K+ efflux dominated over the altered channel gating in case of IKr and IKs. Overall, the markedly increased IK1 in hyperkalemia overcame the relative decreases of IKr and IKs during AP, resulting in an increased net repolarizing current during AP phase 3. β-Adrenergic stimulation of IKs also provided further repolarizing power during sympathetic activation, although hyperkalemia limited IKs upregulation. These results indicate that facilitation of IK1 in hyperkalemia and β-adrenergic stimulation of IKs represent important compensatory mechanisms against AP prolongation and arrhythmia susceptibility.
Collapse
Affiliation(s)
- Bence Hegyi
- a Department of Pharmacology, University of California, Davis, CA 95616, USA
| | - Ye Chen-Izu
- a Department of Pharmacology, University of California, Davis, CA 95616, USA.,b Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.,c Department of Internal Medicine/Cardiology, University of California, Davis, CA 95616, USA
| | - Leighton T Izu
- a Department of Pharmacology, University of California, Davis, CA 95616, USA
| | - Tamás Bányász
- a Department of Pharmacology, University of California, Davis, CA 95616, USA.,d Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
93
|
Hamilton S, Terentyev D. Altered Intracellular Calcium Homeostasis and Arrhythmogenesis in the Aged Heart. Int J Mol Sci 2019; 20:ijms20102386. [PMID: 31091723 PMCID: PMC6566636 DOI: 10.3390/ijms20102386] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/29/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022] Open
Abstract
Aging of the heart is associated with a blunted response to sympathetic stimulation, reduced contractility, and increased propensity for arrhythmias, with the risk of sudden cardiac death significantly increased in the elderly population. The altered cardiac structural and functional phenotype, as well as age-associated prevalent comorbidities including hypertension and atherosclerosis, predispose the heart to atrial fibrillation, heart failure, and ventricular tachyarrhythmias. At the cellular level, perturbations in mitochondrial function, excitation-contraction coupling, and calcium homeostasis contribute to this electrical and contractile dysfunction. Major determinants of cardiac contractility are the intracellular release of Ca2+ from the sarcoplasmic reticulum by the ryanodine receptors (RyR2), and the following sequestration of Ca2+ by the sarco/endoplasmic Ca2+-ATPase (SERCa2a). Activity of RyR2 and SERCa2a in myocytes is not only dependent on expression levels and interacting accessory proteins, but on fine-tuned regulation via post-translational modifications. In this paper, we review how aberrant changes in intracellular Ca2+ cycling via these proteins contributes to arrhythmogenesis in the aged heart.
Collapse
Affiliation(s)
- Shanna Hamilton
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Dmitry Terentyev
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
94
|
Hegyi B, Morotti S, Liu C, Ginsburg KS, Bossuyt J, Belardinelli L, Izu LT, Chen-Izu Y, Bányász T, Grandi E, Bers DM. Enhanced Depolarization Drive in Failing Rabbit Ventricular Myocytes: Calcium-Dependent and β-Adrenergic Effects on Late Sodium, L-Type Calcium, and Sodium-Calcium Exchange Currents. Circ Arrhythm Electrophysiol 2019; 12:e007061. [PMID: 30879336 PMCID: PMC6720130 DOI: 10.1161/circep.118.007061] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Heart failure (HF) is characterized by electrophysiological remodeling resulting in increased risk of cardiac arrhythmias. Previous reports suggest that elevated inward ionic currents in HF promote action potential (AP) prolongation, increased short-term variability of AP repolarization, and delayed afterdepolarizations. However, the underlying changes in late Na+ current (INaL), L-type Ca2+ current, and NCX (Na+/Ca2+ exchanger) current are often measured in nonphysiological conditions (square-pulse voltage clamp, slow pacing rates, exogenous Ca2+ buffers). METHODS We measured the major inward currents and their Ca2+- and β-adrenergic dependence under physiological AP clamp in rabbit ventricular myocytes in chronic pressure/volume overload-induced HF (versus age-matched control). RESULTS AP duration and short-term variability of AP repolarization were increased in HF, and importantly, inhibition of INaL decreased both parameters to the control level. INaL was slightly increased in HF versus control even when intracellular Ca2+ was strongly buffered. But under physiological AP clamp with normal Ca2+ cycling, INaL was markedly upregulated in HF versus control (dependent largely on CaMKII [Ca2+/calmodulin-dependent protein kinase II] activity). β-Adrenergic stimulation (often elevated in HF) further enhanced INaL. L-type Ca2+ current was decreased in HF when Ca2+ was buffered, but CaMKII-mediated Ca2+-dependent facilitation upregulated physiological L-type Ca2+ current to the control level. Furthermore, L-type Ca2+ current response to β-adrenergic stimulation was significantly attenuated in HF. Inward NCX current was upregulated at phase 3 of AP in HF when assessed by combining experimental data and computational modeling. CONCLUSIONS Our results suggest that CaMKII-dependent upregulation of INaL in HF significantly contributes to AP prolongation and increased short-term variability of AP repolarization, which may lead to increased arrhythmia propensity, and is further exacerbated by adrenergic stress.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California Davis, Davis
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis, Davis
| | - Caroline Liu
- Department of Pharmacology, University of California Davis, Davis
| | | | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, Davis
| | | | - Leighton T. Izu
- Department of Pharmacology, University of California Davis, Davis
| | - Ye Chen-Izu
- Department of Pharmacology, University of California Davis, Davis
- Department of Biomedical Engineering, University of California Davis, Davis
- Department of Internal Medicine/Cardiology, University of California Davis, Davis
| | - Tamás Bányász
- Department of Pharmacology, University of California Davis, Davis
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis
| | - Donald M. Bers
- Department of Pharmacology, University of California Davis, Davis
| |
Collapse
|