Network


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

Hotspot


Dive into the research topics where Han-Gang Yu is active.

Publication


Featured researches published by Han-Gang Yu.


Circulation Research | 2000

Effects of the Renin-Angiotensin System on the Current Ito in Epicardial and Endocardial Ventricular Myocytes From the Canine Heart

Han-Gang Yu; Junyuan Gao; Hong-Sheng Wang; Randy S. Wymore; Susan F. Steinberg; David McKinnon; Michael R. Rosen; Ira S. Cohen

The Ca(2+)-independent portion of transient outward K(+) current (I(to)) exhibits a transmural gradient in ventricle. To investigate control mechanisms for this gradient, we studied canine epicardial and endocardial ventricular myocytes with use of the whole-cell patch-clamp technique. I(to) was larger in amplitude, had a more negative voltage threshold for activation, and had a more negative midpoint of inactivation in epicardium. Recovery from inactivation was >10-fold slower in endocardium. Incubation of epicardial myocytes with angiotensin II for 2 to 52 hours altered I(to) to resemble unincubated endocardium and reduced the amplitude of the phase 1 notch of the action potential. In contrast, incubation of endocardial myocytes with losartan for 2 to 52 hours altered I(to) to resemble unincubated epicardium and induced a phase 1 notch in the action potential. With RNase protection assays, we determined that incubations with angiotensin II or losartan did not alter mRNA levels for either Kv4.3 or Kv1.4; thus, a change in the alpha subunit for I(to) is unlikely to be responsible. To test whether posttranslational modification produced the effects of angiotensin II, we coexpressed Kv4.3 and the angiotensin II type 1a receptor in Xenopus oocytes. Incubation with angiotensin II increased the time constant for recovery from inactivation of the expressed current by 2-fold with an incubation time constant of 3.7 hours. No effect on activation or inactivation voltage dependence was observed. These results demonstrate that the properties of I(to) in endocardium and epicardium are plastic and likely under the tonic-differing influence of the renin-angiotensin system.


Circulation | 2003

Role of L-Type Calcium Channels in Pacing-Induced Short-Term and Long-Term Cardiac Memory in Canine Heart

Alexei N. Plotnikov; Han-Gang Yu; J.Christoph Geller; Ravil Z. Gainullin; Parag Chandra; Kornelis W. Patberg; Steven Friezema; Peter Danilo; Ira S. Cohen; Steven J. Feinmark; Michael R. Rosen

Background—We tested the hypothesis that ICa,L is important to the development of cardiac memory. Methods and Results—The effects of L-type Ca2+ channel blockade and &bgr;-blockade were tested on acutely anesthetized and on chronically instrumented, conscious dogs. Short-term memory (STM) was induced by 2 hours of ventricular pacing and long-term memory (LTM) by ventricular pacing for 21 days. STM dogs received placebo, nifedipine, or propranolol, and LTM dogs received placebo, atenolol, or amlodipine. AT1 receptor blockade (candesartan) and ACE inhibition (trandolapril) were also tested in LTM. Microelectrodes were used to record transmembrane potentials from isolated epicardial and endocardial slabs using a protocol simulating STM in intact animals. Left ventricular epicardial myocytes from LTM or sham control dogs were dissociated, and ICa,L was recorded (whole-cell patch-clamp technique). Evolution of STM and LTM was attenuated by ICa,L blockers but not &bgr;-blockers. Neither AT1 receptor blockade nor ACE inhibition suppressed LTM. In microelectrode experiments, pacing induced an epicardial-endocardial gradient change mimicking STM that was suppressed by nifedipine. In patch-clamp experiments, peak ICa,L density in LTM and control were equivalent, but activation was more positive and time constants of inactivation longer in LTM (P <0.05). Conclusions—ICa,L blockade but not &bgr;-adrenergic blockade suppresses cardiac memory. LTM evolution is unaffected by angiotensin II blockade and is associated with altered ICa,L kinetics.


Journal of Cardiovascular Pharmacology | 2006

Constitutively Active Src Tyrosine Kinase Changes Gating of HCN4 Channels Through Direct Binding to the Channel Proteins

Suzanne S. Arinsburg; Ira S. Cohen; Han-Gang Yu

Cardiac pacemaker current, if, is generated by hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Our previous studies demonstrated that altered tyrosine phosphorylation can modulate the properties of both if and HCN channels. To assess a hypothesis that the intracellular tyrosine kinase Src may play a role in modulation by tyrosine phosphorylation of if, we cotransfected HEK293 cells with HCN4 and Src proteins. When HCN4 was cotransfected with a constitutively activated Src protein (Src529), the resultant voltage-dependent HCN4 activation was positively shifted (HCN4: V1/2=−93 mV; Src529: V1/2=−80 mV). The activation kinetics were accelerated at some potentials but not over the entire voltage range tested (eg, at −95 mV, τ_act(HCN4)=3243 ms; τ_act(Src529)=1113 ms). When HCN4 was cotransfected with a dominant negative Src protein (Src296), the HCN4 activation was shifted more negative to a smaller degree (HCN4: V1/2=−93 mV; Src296: V1/2=−98 mV; statistically insignificant) and the activation kinetics were slowed at most test potentials (eg, at −95 mV, τ_act(Src296)=7396 ms). Neither Src529 nor Src296 significantly altered HCN4 current density. Coimmunoprecipitation experiments revealed that Src forms a complex with HCN4 in HEK293 cells and in rat ventricular myocytes. Our data provide a novel mechanism of if regulation by Src tyrosine phosphorylation.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2009

Integration of skeletal muscle resistance arteriolar reactivity for perfusion responses in the metabolic syndrome

Jefferson C. Frisbee; John M. Hollander; Robert W. Brock; Han-Gang Yu; Matthew A. Boegehold

Previous study suggests that with evolution of the metabolic syndrome, patterns of arteriolar reactivity are profoundly altered and may constrain functional hyperemia. This study investigated interactions between parameters of vascular reactivity at two levels of resistance arterioles in obese Zucker rats (OZR), translating these observations into perfusion regulation for in situ skeletal muscle. Dilation of isolated and in situ resistance arterioles from OZR to acetylcholine, arachidonic acid (AA), and hypoxia (isolated arterioles only) were blunted vs. lean Zucker rats (LZR), although dilation to adenosine was intact. Increased adrenergic tone (phenylephrine) or intralumenal pressure (ILP) impaired dilation in both strains (OZR>LZR). Treatment of OZR arterioles with Tempol (superoxide dismutase mimetic) or SQ-29548 (prostaglandin H2/thromboxane A2 receptor antagonist) improved dilator reactivity under control conditions and with increased ILP, but had minimal effect with increased adrenergic tone. Arteriolar dilation to adenosine was well maintained in both strains under all conditions. For in situ cremasteric arterioles, muscle contraction-induced elevations in metabolic demand elicited arteriolar dilations and hyperemic responses that were blunted in OZR vs. LZR, although distal parallel arterioles were characterized by heterogeneous dilator and perfusion responses. alpha-Adrenoreceptor blockade improved outcomes at rest but had minimal effect with elevated metabolic demand. Treatment with Tempol or SQ-29548 had minimal impact at rest, but lessened distal arteriolar perfusion heterogeneity with increased metabolic demand. In blood-perfused gastrocnemius of OZR, perfusion was constrained primarily by adrenergic tone, while myogenic activation and endothelium-dependent dilation did not appear to contribute significantly to ischemia. These results of this novel, integrated approach suggest that adrenergic tone and metabolic dilation are robust determinants of bulk perfusion to skeletal muscle of OZR, while endothelial dysfunction may more strongly regulate perfusion distribution homogeneity via the impact of oxidant stress and AA metabolism.


American Journal of Physiology-heart and Circulatory Physiology | 2012

Defective calcium inactivation causes long QT in obese insulin-resistant rat

Yen-Chang Lin; Jianying Huang; Hong Kan; Vincent Castranova; Jefferson C. Frisbee; Han-Gang Yu

The majority of diabetic patients who are overweight or obese die of heart disease. We suspect that the obesity-induced insulin resistance may lead to abnormal cardiac electrophysiology. We tested this hypothesis by studying an obese insulin-resistant rat model, the obese Zucker rat (OZR). Compared with the age-matched control, lean Zucker rat (LZR), OZR of 16-17 wk old exhibited an increase in QTc interval, action potential duration, and cell capacitance. Furthermore, the L-type calcium current (I(CaL)) in OZR exhibited defective inactivation and lost the complete inactivation back to the closed state, leading to increased Ca(2+) influx. The current density of I(CaL) was reduced in OZR, whereas the threshold activation and the current-voltage relationship of I(CaL) were not significantly altered. L-type Ba(2+) current (I(BaL)) in OZR also exhibited defective inactivation, and steady-state inactivation was not significantly altered. However, the current-voltage relationship and activation threshold of I(BaL) in OZR exhibited a depolarized shift compared with LZR. The total and membrane protein expression levels of Cav1.2 [pore-forming subunit of L-type calcium channels (LTCC)], but not the insulin receptors, were decreased in OZR. The insulin receptor was found to be associated with the Cav1.2, which was weakened in OZR. The total protein expression of calmodulin was reduced, but that of Cavβ2 subunit was not altered in OZR. Together, these results suggested that the 16- to 17-wk-old OZR has 1) developed cardiac hypertrophy, 2) exhibited altered electrophysiology manifested by the prolonged QTc interval, 3) increased duration of action potential in isolated ventricular myocytes, 4) defective inactivation of I(CaL) and I(BaL), 5) weakened the association of LTCC with the insulin receptor, and 6) decreased protein expression of Cav1.2 and calmodulin. These results also provided mechanistic insights into a remodeled cardiac electrophysiology under the condition of insulin resistance, enhancing our understanding of long QT associated with obese type 2 diabetic patients.


Journal of Biological Chemistry | 2008

Novel Mechanism for Suppression of Hyperpolarization-activated Cyclic Nucleotide-gated Pacemaker Channels by Receptor-like Tyrosine Phosphatase-α

Jianying Huang; Aijie Huang; Qi Zhang; Yen-Chang Lin; Han-Gang Yu

We have previously reported an important role of increased tyrosine phosphorylation activity by Src in the modulation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Here we provide evidence showing a novel mechanism of decreased tyrosine phosphorylation on HCN channel properties. We found that the receptor-like protein-tyrosine phosphatase-α (RPTPα) significantly inhibited or eliminated HCN2 channel expression in HEK293 cells. Biochemical evidence showed that the surface expression of HCN2 was remarkably reduced by RPTPα, which was in parallel to the decreased tyrosine phosphorylation of the channel protein. Confocal imaging confirmed that the membrane surface distribution of the HCN2 channel was inhibited by RPTPα. Moreover, we detected the presence of RPTPα proteins in cardiac ventricles with expression levels changed during development. Inhibition of tyrosine phosphatase activity by phenylarsine oxide or sodium orthovanadate shifted ventricular hyperpolarization-activated current (If, generated by HCN channels) activation from nonphysiological voltages into physiological voltages associated with accelerated activation kinetics. In conclusion, we showed a critical role RPTPα plays in HCN channel function via tyrosine dephosphorylation. These findings are also important to neurons where HCN and RPTPα are richly expressed.


Journal of Cardiovascular Pharmacology | 1998

Cesium Effects on if and iK in Rabbit Sinoatrial Node Myocytes: Implications for SA Node Automaticity

Liu Ym; Han-Gang Yu; Li Cz; Cohen Is; Mario Vassalle

Cesium blocks the hyperpolarization-activated current i(f) but blocks neither the delayed-rectifier current i(K) nor the sinoatrial (SA) node discharge. It has been proposed that the failure of Cs+ to block SA discharge is either an incomplete block or a negative shift of i(f). However, an alternative possibility is that i(K) (rather than i(f)) has a predominant role in the SA-pacemaker potential. To investigate this point, the effects of Cs+ on both i(f) and i(K) in the pacemaker range of potentials were studied in the same single SA node cell at the same time by means of the perforated patch-clamp technique. Hyperpolarizing steps from a holding potential (Vh) of -35 mV into and past the pacemaker-potential range resulted in a progressively larger i(f) associated with an increasing slope conductance. Cs+ (2 mM) reversibly blocked both i(f) and the slope conductance increase, suggesting that the current activated was indeed predominantly i(f). Subsequently, hyperpolarizing steps to -50, -60, and -70 mV were applied in the absence (to activate only i(f)) and in the presence of a prior depolarizing step to +10 mV (to activate i(K) as well, as the action potential normally does). Cs+ almost abolished i(f) but only slightly decreased i(K). It is concluded that the failure of Cs+ to block the SA- node spontaneous discharge is not due to a shift of i(f) out of the pacemaker range (due to run-down) or an incomplete block of i(f). Instead, the resistance of i(K) to block by Cs+ is consistent with a predominant role of i(K) for the discharge of the SA node, although i(f) can contribute under normal or special circumstances. The reduction of i(K) by Cs+ raises the question whether the Cs+ slows the SA-node discharge not only by suppressing I(f), but also by reducing i(K).


Biochimica et Biophysica Acta | 2009

Associated Changes in HCN2 and HCN4 Transcripts and If Pacemaker Current in Myocytes

Qi Zhang; Aijie Huang; Yen-Chang Lin; Han-Gang Yu

The time- and voltage-dependent inward current generated by the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels contributes to the tissue-specific rhythmic activities in the brain and heart. Four isoforms (HCN1-HCN4) have been identified. Previous studies showed that different HCN isoforms may form functional heteromeric channels. We report here that when HCN2 and HCN4 mRNA were injected into Xenopus oocytes with various ratios of HCN2 over HCN4 at 1:1, 10:1, and 1:10, respectively, the resultant channels showed a depolarized current activation and significantly faster activation kinetics near the midpoint of activation compared with HCN4 homomeric channels. In adult rat myocytes overexpressing HCN4, there was an associated increase in HCN2 mRNA. In neonatal rat myocytes in which HCN2 was knocked down, there was also a simultaneous decrease in HCN4 mRNA. Coimmunoprecipitation experiments showed that HCN2 and HCN4 channel proteins can associate with each other in adult rat ventricles. Finally, in adult myocytes overexpressing HCN4, the hyperpolarization-activated inward current activation, I(f), was shifted to physiological voltages from non-physiological voltages, associated with faster activation kinetics. These data suggested that different ratios of HCN2 and HCN4 transcripts overlapping in different tissues also contribute to the tissue-specific properties of I(f).


Journal of Biological Chemistry | 2009

Rescue of a Trafficking Defective Human Pacemaker Channel via a Novel Mechanism ROLES OF Src, Fyn, AND Yes TYROSINE KINASES

Yen-Chang Lin; Jianying Huang; Hong Kan; Jefferson C. Frisbee; Han-Gang Yu

Therapeutic strategies such as using channel blockers and reducing culture temperature have been used to rescue some long QT-associated voltage-gated potassium Kv trafficking defective mutant channels. A hyperpolarization-activated cyclic nucleotide-gated HCN4 pacemaker channel mutant (D553N) has been recently found in a patient associated with cardiac arrhythmias including long QT. D553N showed the defective trafficking to the cell surface, leading to little ionic current expression (loss-of-function). We show in this report that enhanced tyrosine phosphorylation mediated by Src, Fyn, and Yes kinases was able to restore the surface expression of D553N for normal current expression. Src or Yes, but not Fyn, significantly increased the current density and surface expression of D553N. Fyn accelerated the activation kinetics of the rescued D553N. Co-expression of D553N with Yes exhibited the slowest activation kinetics of D553N. Src, Fyn, and Yes significantly enhanced the tyrosine phosphorylation of D553N. A combination of Src, Fyn, and Yes rescued the current expression and the gating of D553N comparable with those of wild-type HCN4. In conclusion, we demonstrate a novel mechanism using three endogenous Src kinases to rescue a trafficking defective HCN4 mutant channel (D553N) by enhancing the tyrosine phosphorylation of the mutant channel protein.


American Journal of Physiology-heart and Circulatory Physiology | 2015

Leptin decreases heart rate associated with increased ventricular repolarization via its receptor.

Yen-Chang Lin; Jianying Huang; Stan Hileman; Karen H. Martin; Robert Hull; Mary E Davis; Han-Gang Yu

Leptin has been proposed to modulate cardiac electrical properties via β-adrenergic receptor activation. The presence of leptin receptors and adipocytes in myocardium raised a question as to whether leptin can directly modulate cardiac electrical properties such as heart rate and QT interval via its receptor. In this work, the role of local direct actions of leptin on heart rate and ventricular repolarization was investigated. We identified the protein expression of leptin receptors at cell surface of sinus node, atrial, and ventricular myocytes isolated from rat heart. Leptin at low doses (0.1-30 μg/kg) decreased resting heart rate; at high doses (150-300 μg/kg), leptin induced a biphasic effect (decrease and then increase) on heart rate. In the presence of high-dose propranolol (30 mg/kg), high-dose leptin only reduced heart rate and sometimes caused sinus pauses and ventricular tachycardia. The leptin-induced inhibition of resting heart rate was fully reversed by leptin antagonist. Leptin also increased heart rate-corrected QT interval (QTc), and leptin antagonist did not. In isolated ventricular myocytes, leptin (0.03-0.3 μg/ml) reversibly increased the action potential duration. These results supported our hypothesis that in addition to indirect pathway via sympathetic tone, leptin can directly decrease heart rate and increase QT interval via its receptor independent of β-adrenergic receptor stimulation. During inhibition of β-adrenergic receptor activity, high concentration of leptin in myocardium can cause deep bradycardia, prolonged QT interval, and ventricular arrhythmias.

Collaboration


Dive into the Han-Gang Yu's collaboration.

Top Co-Authors

Avatar

Jianying Huang

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Yen-Chang Lin

West Virginia University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qi Zhang

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Yen-Chang Lin

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Robert Hull

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Jefferson C. Frisbee

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Nam Nhut Phan

Ton Duc Thang University

View shared research outputs
Top Co-Authors

Avatar

Aijie Huang

West Virginia University

View shared research outputs
Researchain Logo
Decentralizing Knowledge