Network


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

Hotspot


Dive into the research topics where Jianying Huang is active.

Publication


Featured researches published by Jianying Huang.


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 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.


American Journal of Physiology-cell Physiology | 2010

Inactivation of L-type calcium channel modulated by HCN2 channel

Yen-Chang Lin; Jianying Huang; Qi Zhang; John M. Hollander; Jefferson C. Frisbee; Karen H. Martin; Casey C Nestor; Robert L. Goodman; Han-Gang Yu

Ca(2+) entry is delicately controlled by inactivation of L-type calcium channel (LTCC) composed of the pore-forming subunit alpha1C and the auxiliary subunits beta1 and alpha2delta. Calmodulin is the key protein that interacts with the COOH-terminal motifs of alpha1C, leading to the fine control of LTCC inactivation. In this study we show evidence that a hyperpolarization-activated cyclic nucleotide-gated channel, HCN2, can act as a nonchannel regulatory protein to narrow the L-type Ca(2+) channel current-voltage curve. In the absence of LTCC auxiliary subunits, HCN2 can induce alpha1C inactivation. Without alpha2delta, HCN2-induced fast inactivation of alpha1C requires calmodulin. With alpha2delta, the alpha1C/HCN2/alpha2delta channel inactivation does not require calmodulin. In contrast, beta1-subunit plays a relatively minor role in the interaction of alpha1C with HCN2. The NH(2) terminus of HCN2 and the IQ motif of alpha1C subunit are required for alpha1C/HCN2 channel interaction. Ca(2+) channel inactivation is significantly slowed in hippocampus neurons (HNs) overexpressing HCN2 mutant lacking NH(2) terminus and accelerated in HNs overexpressing the wild-type HCN2 compared with HN controls. Collectively, these results revealed a potentially novel protection mechanism for achieving the LTCC inactivation via interaction with HCN2.


Journal of Cardiovascular Pharmacology | 2015

PP2 prevents isoproterenol stimulation of cardiac pacemaker activity.

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

Abstract: Increasing evidence has demonstrated the potential risks of cardiac arrhythmias (such as prolonged QT interval) using tyrosine kinase inhibitors for cancer therapy. We report here that a widely used selective inhibitor of Src tyrosine kinases, PP2, can inhibit and prevent isoproterenol stimulation of cardiac pacemaker activity. In dissected rat sinus node, PP2 inhibited and prevented isoproterenol stimulation of spontaneous beating rate. In isolated sinus node myocytes, PP2 suppressed the hyperpolarization-activated “funny” current (If) by negatively shifting the activation curve and decelerating activation kinetics, associated with decreased cell surface expression and reduced tyrosine phosphorylation of hyperpolarization-activated cyclic nucleotide-modulated channel 4 (HCN4) channel proteins. In human embryonic kidney 293 cells overexpressing recombinant human HCN4 channels, PP2 reversed isoproterenol stimulation of HCN4 and inhibited HCN4-573x, a cAMP-insensitive human HCN4 mutant. Isoprotenrenol had little effects on HCN4-573x. These results demonstrated that inhibition of presumably tyrosine Src kinase activity in heart by PP2 decreased and prevented the potential &bgr;-adrenergic stimulation of cardiac pacemaker activity. These effects are mediated, at least partially, by a cAMP-independent attenuation of channel activity and cell surface expression of HCN4, the key channel protein that controls the heart rate.


Journal of Biological Chemistry | 2009

Non-proteolytic HCN2 in the heart.

Han-Gang Yu; Jianying Huang; Yen-Chang Lin

We read with interest this article, “Proteolytic Processing of HCN2 and Co-assembly with HCN4 in the Generation of Cardiac Pacemaker Channels” (1). We would like to point out that the cleaved HCN2, which appeared near 60 kDa in the mouse heart, is not a general experimental observation in the heart from other species. Non-proteolytic HCN2 appearing near 100 kDa has been reported in rat (2–4) and human heart ventricles (5). The possibility for the co-assembly of non-proteolytic HCN2 with HCN4 to form a cardiac pacemaker channel has also been reported (3). In Fig. 5, the normalized onset current was used to obtain the midpoint activation (V½), which is supposed to be derived from the activation curve constructed from tail currents that are missing (Fig. 5A). The V½ value is more positive for HCN4 (triangle) than for HCN2 (square) in Fig. 5B, but more positive for HCN2 (white square) than for HCN4 (dark square) in Fig. 5C; one of them must be mislabeled. Additionally, the authors cited a V½ value of −92 mV for If (Fig. 5C), which did not appear in the cited reference. The reference they cited was investigating the co-assembly of HCN1/HCN4 as a potential molecular basis of sinus node If that has a V½ of about −63 mV. Cardiac pacemaker currents have dramatically different gating properties in the sinus node and in the ventricles. The biochemical and patch clamp data of HCN2 and HCN4 obtained in HEK293 cells are not close to that obtained from the sinus node If.


American Journal of Physiology-cell Physiology | 2008

Src Tyrosine Kinase Alters Gating of Hyperpolarization-Activated HCN4 Pacemaker Channel through Tyr531

Chenhong Li; Qi Zhang; Bunyen Teng; S. Jamal Mustafa; Jianying Huang; Han-Gang Yu


Journal of Cardiovascular Pharmacology | 2014

PP2 Prevents beta-Adrenergic Stimulation of Cardiac Pacemaker Activity

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


Circulation | 2013

Abstract 13439: Leptin Inhibits Heart Rate Via its Receptor

Jianying Huang; Yen-Chang Lin; Kimberly Harding; Han-Gang Yu

Collaboration


Dive into the Jianying Huang's collaboration.

Top Co-Authors

Avatar

Han-Gang Yu

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

Yen-Chang Lin

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Qi Zhang

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

Aijie Huang

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Hong Kan

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Bunyen Teng

West Virginia University

View shared research outputs
Researchain Logo
Decentralizing Knowledge