Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hsiang-Ting Ho is active.

Publication


Featured researches published by Hsiang-Ting Ho.


PLOS ONE | 2011

MicroRNA-1 and -133 increase arrhythmogenesis in heart failure by dissociating phosphatase activity from RyR2 complex.

Andriy E. Belevych; Sarah E. Sansom; Radmila Terentyeva; Hsiang-Ting Ho; Yoshinori Nishijima; Mickey M. Martin; Hitesh K. Jindal; Jennifer A. Rochira; Yukiko Kunitomo; Maha Abdellatif; Cynthia A. Carnes; Terry S. Elton; Sandor Gyorke; Dmitry Terentyev

In heart failure (HF), arrhythmogenic spontaneous sarcoplasmic reticulum (SR) Ca2+ release and afterdepolarizations in cardiac myocytes have been linked to abnormally high activity of ryanodine receptors (RyR2s) associated with enhanced phosphorylation of the channel. However, the specific molecular mechanisms underlying RyR2 hyperphosphorylation in HF remain poorly understood. The objective of the current study was to test the hypothesis that the enhanced expression of muscle-specific microRNAs (miRNAs) underlies the HF-related alterations in RyR2 phosphorylation in ventricular myocytes by targeting phosphatase activity localized to the RyR2. We studied hearts isolated from canines with chronic HF exhibiting increased left ventricular (LV) dimensions and decreased LV contractility. qRT-PCR revealed that the levels of miR-1 and miR-133, the most abundant muscle-specific miRNAs, were significantly increased in HF myocytes compared with controls (2- and 1.6-fold, respectively). Western blot analyses demonstrated that expression levels of the protein phosphatase 2A (PP2A) catalytic and regulatory subunits, which are putative targets of miR-133 and miR-1, were decreased in HF cells. PP2A catalytic subunit mRNAs were validated as targets of miR-133 by using luciferase reporter assays. Pharmacological inhibition of phosphatase activity increased the frequency of diastolic Ca2+ waves and afterdepolarizations in control myocytes. The decreased PP2A activity observed in HF was accompanied by enhanced Ca2+/calmodulin-dependent protein kinase (CaMKII)-mediated phosphorylation of RyR2 at sites Ser-2814 and Ser-2030 and increased frequency of diastolic Ca2+ waves and afterdepolarizations in HF myocytes compared with controls. In HF myocytes, CaMKII inhibitory peptide normalized the frequency of pro-arrhythmic spontaneous diastolic Ca2+ waves. These findings suggest that altered levels of major muscle-specific miRNAs contribute to abnormal RyR2 function in HF by depressing phosphatase activity localized to the channel, which in turn, leads to the excessive phosphorylation of RyR2s, abnormal Ca2+ cycling, and increased propensity to arrhythmogenesis.


Circulation Research | 2012

Shortened Ca2+ Signaling Refractoriness Underlies Cellular Arrhythmogenesis in a Postinfarction Model of Sudden Cardiac Death

Andriy E. Belevych; Dmitry Terentyev; Radmila Terentyeva; Hsiang-Ting Ho; Inna Györke; Ingrid M. Bonilla; Cynthia A. Carnes; George E. Billman; Sandor Gyorke

Rationale: Diastolic spontaneous Ca2+ waves (DCWs) are recognized as important contributors to triggered arrhythmias. DCWs are thought to arise when [Ca2+] in sarcoplasmic reticulum ([Ca2+]SR) reaches a certain threshold level, which might be reduced in cardiac disease as a consequence of sensitization of ryanodine receptors (RyR2s) to luminal Ca2+. Objective: We investigated the mechanisms of DCW generation in myocytes from normal and diseased hearts, using a canine model of post–myocardial infarction ventricular fibrillation (VF). Methods and Results: The frequency of DCWs, recorded during periodic pacing in the presence of a &bgr;-adrenergic receptor agonist isoproterenol, was significantly higher in VF myocytes than in normal controls. Rather than occurring immediately on reaching a final [Ca2+]SR, DCWs arose with a distinct time delay after attaining steady [Ca2+]SR in both experimental groups. Although the rate of [Ca2+]SR recovery after the SR Ca2+ release was similar between the groups, in VF myocytes the latency to DCWs was shorter, and the [Ca2+]SR at DCW initiation was lower. The restitution of depolarization-induced Ca2+ transients, assessed by a 2-pulse protocol, was significantly faster in VF myocytes than in controls. The VF-related alterations in myocyte Ca2+ cycling were mimicked by the RyR2 agonist, caffeine. The reducing agent, mercaptopropionylglycine, or the CaMKII inhibitor, KN93, decreased DCW frequency and normalized restitution of Ca2+ release in VF myocytes. Conclusions: The attainment of a certain threshold [Ca2+]SR is not sufficient for the generation of DCWs. Postrelease Ca2+ signaling refractoriness critically influences the occurrence of DCWs. Shortened Ca2+ signaling refractoriness due to RyR2 phosphorylation and oxidation is responsible for the increased rate of DCWs observed in VF myocytes and could provide a substrate for synchronization of arrhythmogenic events at the tissue level in hearts prone to VF.


The Journal of Physiology | 2011

Arrhythmogenic adverse effects of cardiac glycosides are mediated by redox modification of ryanodine receptors

Hsiang-Ting Ho; Sarah C.W. Stevens; Radmila Terentyeva; Cynthia A. Carnes; Dmitry Terentyev; Sandor Gyorke

Non‐Technical Summary  Cardiac glycosides (CGs) have been routinely used in the treatment of congestive heart failure (HF). Unfortunately, the therapeutic use of CGs in treating HF is limited by their adverse side effects, including cardiac arrhythmias. The arrhythmic side effects of CGs have been traditionally ascribed to excessive cellular Ca2+ retention (Ca2+ overload) leading to spontaneous discharges of intracellular Ca2+ stores, or Ca2+ waves, in turn causing oscillations of the cardiac membrane potential. In the present study, we demonstrate that the proarrhythmic effects of CGs on Ca2+ cycling in cardiac myocytes involve alterations in the function of ryanodine receptor calcium channels caused by oxidative changes in the channel structure by reactive oxygen species. Our findings reveal a new mechanism for CG‐induced Ca2+ waves and suggest a potential target for antiarrhythmic therapy in HF patients treated with CGs.


Cardiovascular Research | 2015

Neuronal Na+ channel blockade suppresses arrhythmogenic diastolic Ca2+ release

Przemysław B. Radwański; Lucia Brunello; Rengasayee Veeraraghavan; Hsiang-Ting Ho; Qing Lou; Michael A. Makara; Andriy E. Belevych; Mircea Anghelescu; Silvia G. Priori; Pompeo Volpe; Thomas J. Hund; Paul M. L. Janssen; Peter J. Mohler; John H.B. Bridge; Steven Poelzing; Sandor Gyorke

AIMS Sudden death resulting from cardiac arrhythmias is the most common consequence of cardiac disease. Certain arrhythmias caused by abnormal impulse formation including catecholaminergic polymorphic ventricular tachycardia (CPVT) are associated with delayed afterdepolarizations resulting from diastolic Ca2+ release (DCR) from the sarcoplasmic reticulum (SR). Despite high response of CPVT to agents directly affecting Ca2+ cycling, the incidence of refractory cases is still significant. Surprisingly, these patients often respond to treatment with Na+ channel blockers. However, the relationship between Na+ influx and disturbances in Ca2+ handling immediately preceding arrhythmias in CPVT remains poorly understood and is the object of this study. METHODS AND RESULTS We performed optical Ca2+ and membrane potential imaging in ventricular myocytes and intact cardiac muscles as well as surface ECGs on a CPVT mouse model with a mutation in cardiac calsequestrin. We demonstrate that a subpopulation of Na+ channels (neuronal Na+ channels; nNav) colocalize with ryanodine receptor Ca2+ release channels (RyR2). Disruption of the crosstalk between nNav and RyR2 by nNav blockade with riluzole reduced and also desynchronized DCR in isolated cardiomyocytes and in intact cardiac tissue. Such desynchronization of DCR on cellular and tissue level translated into decreased arrhythmias in CPVT mice. CONCLUSIONS Thus, our study offers the first evidence that nNav contribute to arrhythmogenic DCR, thereby providing a conceptual basis for mechanism-based antiarrhythmic therapy.


Nature Communications | 2016

Rationally engineered Troponin C modulates in vivo cardiac function and performance in health and disease

Vikram Shettigar; Bo Zhang; Sean C. Little; Hussam E. Salhi; Brian J. Hansen; Ning Li; Jianchao Zhang; Steve R. Roof; Hsiang-Ting Ho; Lucia Brunello; Jessica K. Lerch; Noah Weisleder; Vadim V. Fedorov; Federica Accornero; Jill A. Rafael-Fortney; Sandor Gyorke; Paul M. L. Janssen; Brandon J. Biesiadecki; Mark T. Ziolo; Jonathan P. Davis

Treatment for heart disease, the leading cause of death in the world, has progressed little for several decades. Here we develop a protein engineering approach to directly tune in vivo cardiac contractility by tailoring the ability of the heart to respond to the Ca2+ signal. Promisingly, our smartly formulated Ca2+-sensitizing TnC (L48Q) enhances heart function without any adverse effects that are commonly observed with positive inotropes. In a myocardial infarction (MI) model of heart failure, expression of TnC L48Q before the MI preserves cardiac function and performance. Moreover, expression of TnC L48Q after the MI therapeutically enhances cardiac function and performance, without compromising survival. We demonstrate engineering TnC can specifically and precisely modulate cardiac contractility that when combined with gene therapy can be employed as a therapeutic strategy for heart disease.


PLOS ONE | 2012

Diesterified Nitrone Rescues Nitroso-Redox Levels and Increases Myocyte Contraction Via Increased SR Ca2+ Handling

Christopher J. Traynham; Steve R. Roof; Honglan Wang; Robert A. Prosak; Lifei Tang; Serge Viatchenko-Karpinski; Hsiang-Ting Ho; Ira Racoma; Dominic J. Catalano; Xin Huang; Yongbin Han; Shang-U Kim; Sandor Gyorke; George E. Billman; Frederick A. Villamena; Mark T. Ziolo

Nitric oxide (NO) and superoxide (O2 −) are important cardiac signaling molecules that regulate myocyte contraction. For appropriate regulation, NO and O2 .− must exist at defined levels. Unfortunately, the NO and O2 .− levels are altered in many cardiomyopathies (heart failure, ischemia, hypertrophy, etc.) leading to contractile dysfunction and adverse remodeling. Hence, rescuing the nitroso-redox levels is a potential therapeutic strategy. Nitrone spin traps have been shown to scavenge O2 .− while releasing NO as a reaction byproduct; and we synthesized a novel, cell permeable nitrone, 2–2–3,4-dihydro-2H-pyrrole 1-oxide (EMEPO). We hypothesized that EMEPO would improve contractile function in myocytes with altered nitroso-redox levels. Ventricular myocytes were isolated from wildtype (C57Bl/6) and NOS1 knockout (NOS1−/−) mice, a known model of NO/O2 .− imbalance, and incubated with EMEPO. EMEPO significantly reduced O2 .− (lucigenin-enhanced chemiluminescence) and elevated NO (DAF-FM diacetate) levels in NOS1−/− myocytes. Furthermore, EMEPO increased NOS1−/− myocyte basal contraction (Ca2+ transients, Fluo-4AM; shortening, video-edge detection), the force-frequency response and the contractile response to β-adrenergic stimulation. EMEPO had no effect in wildtype myocytes. EMEPO also increased ryanodine receptor activity (sarcoplasmic reticulum Ca2+ leak/load relationship) and phospholamban Serine16 phosphorylation (Western blot). We also repeated our functional experiments in a canine post-myocardial infarction model and observed similar results to those seen in NOS1−/− myocytes. In conclusion, EMEPO improved contractile function in myocytes experiencing an imbalance of their nitroso-redox levels. The concurrent restoration of NO and O2 .− levels may have therapeutic potential in the treatment of various cardiomyopathies.


JACC: Basic to Translational Science | 2016

Neuronal Na+ Channels Are Integral Components of Pro-Arrhythmic Na+/Ca2+ Signaling Nanodomain That Promotes Cardiac Arrhythmias During β-Adrenergic Stimulation

Przemysław B. Radwański; Hsiang-Ting Ho; Rengasayee Veeraraghavan; Lucia Brunello; Bin Liu; Andriy E. Belevych; Sathya D. Unudurthi; Michael A. Makara; Silvia G. Priori; Pompeo Volpe; Antonis A. Armoundas; Wolfgang H. Dillmann; Björn C. Knollmann; Peter J. Mohler; Thomas J. Hund; Sandor Gyorke

Summary Although triggered arrhythmias including catecholaminergic polymorphic ventricular tachycardia (CPVT) are often caused by increased levels of circulating catecholamines, the mechanistic link between β-adrenergic receptor (AR) stimulation and the subcellular/molecular arrhythmogenic trigger(s) is unclear. Here, we systematically investigated the subcellular and molecular consequences of β-AR stimulation in the promotion of catecholamine-induced cardiac arrhythmias. Using mouse models of cardiac calsequestrin-associated CPVT, we demonstrate that a subpopulation of Na+ channels, mainly the neuronal Na+ channels (nNav), colocalize with ryanodine receptor 2 (RyR2) and Na+/Ca2+ exchanger (NCX) and are a part of the β-AR-mediated arrhythmogenic process. Specifically, augmented Na+ entry via nNav in the settings of genetic defects within the RyR2 complex and enhanced sarcoplasmic reticulum (SR) Ca2+-ATPase (SERCA)-mediated SR Ca2+ refill is both an essential and a necessary factor for arrhythmogenesis. Furthermore, we show that augmentation of Na+ entry involves β-AR–mediated activation of CAMKII, subsequently leading to nNav augmentation. Importantly, selective pharmacological inhibition as well as silencing of Nav1.6 inhibit myocyte arrhythmic potential and prevent arrhythmias in vivo. Taken together, these data suggest that the arrhythmogenic alteration in Na+/Ca2+ handling evidenced ruing β-AR stimulation results, at least in part, from enhanced Na+ influx through nNav. Therefore, selective inhibition of these channels and of Nav1.6 in particular can serve as a potential antiarrhythmic therapy.


Journal of Applied Physiology | 2012

Endurance exercise training normalizes repolarization and calcium-handling abnormalities, preventing ventricular fibrillation in a model of sudden cardiac death

Ingrid M. Bonilla; Andriy E. Belevych; Arun Sridhar; Yoshinori Nishijima; Hsiang-Ting Ho; Quanhua He; Monica Kukielka; Dmitry Terentyev; Radmila Terentyeva; Bin Liu; Victor P. Long; Sandor Gyorke; Cynthia A. Carnes; George E. Billman

The risk of sudden cardiac death is increased following myocardial infarction. Exercise training reduces arrhythmia susceptibility, but the mechanism is unknown. We used a canine model of sudden cardiac death (healed infarction, with ventricular tachyarrhythmias induced by an exercise plus ischemia test, VF+); we previously reported that endurance exercise training was antiarrhythmic in this model (Billman GE. Am J Physiol Heart Circ Physiol 297: H1171-H1193, 2009). A total of 41 VF+ animals were studied, after random assignment to 10 wk of endurance exercise training (EET; n = 21) or a matched sedentary period (n = 20). Following (>1 wk) the final attempted arrhythmia induction, isolated myocytes were used to test the hypotheses that the endurance exercise-induced antiarrhythmic effects resulted from normalization of cellular electrophysiology and/or normalization of calcium handling. EET prevented VF and shortened in vivo repolarization (P < 0.05). EET normalized action potential duration and variability compared with the sedentary group. EET resulted in a further decrement in transient outward current compared with the sedentary VF+ group (P < 0.05). Sedentary VF+ dogs had a significant reduction in repolarizing K(+) current, which was restored by exercise training (P < 0.05). Compared with controls, myocytes from the sedentary VF+ group displayed calcium alternans, increased calcium spark frequency, and increased phosphorylation of S2814 on ryanodine receptor 2. These abnormalities in intracellular calcium handling were attenuated by exercise training (P < 0.05). Exercise training prevented ischemically induced VF, in association with a combination of beneficial effects on cellular electrophysiology and calcium handling.


Journal of Molecular and Cellular Cardiology | 2015

Obligatory role of neuronal nitric oxide synthase in the heart's antioxidant adaptation with exercise

Steve R. Roof; Hsiang-Ting Ho; Sean C. Little; Joseph E. Ostler; Elizabeth A. Brundage; Muthu Periasamy; Frederick A. Villamena; Sandor Gyorke; Brandon J. Biesiadecki; Christophe Heymes; Steven R. Houser; Jonathan P. Davis; Mark T. Ziolo

Excessive oxidative stress in the heart results in contractile dysfunction. While antioxidant therapies have been a disappointment clinically, exercise has shown beneficial results, in part by reducing oxidative stress. We have previously shown that neuronal nitric oxide synthase (nNOS) is essential for cardioprotective adaptations caused by exercise. We hypothesize that part of the cardioprotective role of nNOS is via the augmentation of the antioxidant defense with exercise by positively shifting the nitroso-redox balance. Our results show that nNOS is indispensable for the augmented anti-oxidant defense with exercise. Furthermore, exercise training of nNOS knockout mice resulted in a negative shift in the nitroso-redox balance resulting in contractile dysfunction. Remarkably, overexpressing nNOS (conditional cardiac-specific nNOS overexpression) was able to mimic exercise by increasing VO2max. This study demonstrates that exercise results in a positive shift in the nitroso-redox balance that is nNOS-dependent. Thus, targeting nNOS signaling may mimic the beneficial effects of exercise by combating oxidative stress and may be a viable treatment strategy for heart disease.


Cardiovascular Research | 2015

Ablation of HRC alleviates cardiac arrhythmia and improves abnormal Ca handling in CASQ2 knockout mice prone to CPVT

Bin Liu; Hsiang-Ting Ho; Lucia Brunello; Sathya D. Unudurthi; Qing Lou; Andriy E. Belevych; Lan Qian; Do Han Kim; Chunghee Cho; Paul M. L. Janssen; Thomas J. Hund; Björn C. Knollmann; Evangelia G. Kranias; Sandor Gyorke

AIMS Cardiac calsequestrin (CASQ2) and histidine-rich Ca-binding protein (HRC) are sarcoplasmic reticulum (SR) Ca-binding proteins that regulate SR Ca release in mammalian heart. Deletion of either CASQ2 or HRC results in relatively mild phenotypes characterized by preserved cardiac structure and function, although CASQ2 knockout (KO), or Cnull, shows increased arrhythmia burden under conditions of catecholaminergic stress. We hypothesized that given the apparent overlap of functions of CASQ2 and HRC, simultaneous ablation of both would deteriorate the cardiac phenotype compared with the single knockouts. METHODS AND RESULTS In contrast to this expectation, double knockout (DKO) mice lacking both CASQ2 and HRC exhibited normal cardiac ejection fraction and ultrastructure. Moreover, the predisposition to catecholamine-dependent arrhythmia that characterizes the Cnull phenotype was alleviated in the DKO mice. At the myocyte level, DKO mice displayed Ca transients of normal amplitude; additionally, the frequency of spontaneous Ca waves and sparks in the presence of isoproterenol were decreased markedly compared with Cnull. Furthermore, restitution of SR Ca release was slowed in DKO myocytes compared with Cnull cells. CONCLUSION Our results suggest that rather than being functionally redundant, CASQ2 and HRC modulate cardiac ryanodine receptor-mediated (RyR2) Ca release in an opposing manner. In particular, while CASQ2 stabilizes RyR2 rendering it refractory in the diastolic phase, HRC enhances RyR2 activity facilitating RyR2 recovery from refractoriness.

Collaboration


Dive into the Hsiang-Ting Ho's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bin Liu

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge