Network


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

Hotspot


Dive into the research topics where Zhanqin Huang is active.

Publication


Featured researches published by Zhanqin Huang.


Cellular Physiology and Biochemistry | 2008

The Protective Effect of Egr-1 Antisense Oligodeoxyribonucleotide on Myocardial Injury Induced by Ischemia-reperfusion and Hypoxia- reoxygenation

Yanmei Zhang; Ganggang Shi; Jinhong Zheng; Yanqiu Lv; Ping Gao; Zhanqin Huang; Fenfei Gao; Yanqiong Zhou

Aims: Our previous studies have shown that myocardial ischemia-reperfusion (I/R) injury is related closely with early growth response (Egr)-1 overexpression. The present study is to confirm thoroughly the effects of Egr-1 on the occurrance and development of myocardial I/R injury. Methods: The Sprague-Dawley rat myocardial I/R model and cultured cardiomyocyte hypoxia-reoxygenation (H/R) model were established. The synthesized Egr-1 antisense oligodeoxyribonucleotide (AS-ODN) was transfected into myocardial tissues and cells. Hemodynamic parameters, myeloperoxidase (MPO), cardiac troponin I (cTnI), tumor necrosis factor-α (TNF-α), morphology, spontaneous beat and cell viability were measured to assess the degree of injury and inflammation of myocardial tissues and cells. Results: In vivo, Egr-1 AS-ODN significantly attenuated injury and inflammation of myocardial tissues caused by I/R evidenced by the amelioration of hemodynamics and the reduction in MPO activity. In vitro, Egr-1 AS-ODN significantly relieved injury and inflammation of cultured cardiomyocyte caused by H/R evidenced by the improvement in morphology, structure and beat as well as the decrease in leakage of cTnI and release of TNF-α from cultured cardiomyocyte. Conclusions: These data suggest that Egr-1 plays a vital role in the pathogenesis of myocardial I/R injury and Egr-1 AS-ODN could protect the myocardium from I/R injury.


Scientific Reports | 2015

Effect of N -n-butyl haloperidol iodide on ROS/JNK/Egr-1 signaling in H9c2 cells after hypoxia/reoxygenation

Yanmei Zhang; Han Liao; Shuping Zhong; Fenfei Gao; Yicun Chen; Zhanqin Huang; Shishi Lu; Ting Sun; Bin Wang; Weiqiu Li; Han Xu; Fuchun Zheng; Ganggang Shi

Reactive oxygen species (ROS)-induced oxidative stress in cells is an important pathophysiological process during myocardial ischemia/reperfusion (I/R) injury, and the transcription factor Egr-1 is a master switch for various damage pathways during reperfusion injury. An in vitro model of myocardial I/R injury and H9c2 cardiomyoblast cells hypoxia/reoxygenation (H/R) was used to assess whether there is abnormal intracellular ROS/JNK/Egr-1 signaling. We also assessed whether N-n-butyl haloperidol (F2), which exerts protective effects during myocardial I/R injury, can modulate this pathway. H/R induced ROS generation, JNK activation, and increased the expression of Egr-1 protein in H9c2 cells. The ROS scavengers edaravone (EDA) and N-acetyl-L-cysteine (NAC) reduced ROS level, downregulated JNK activation, and Egr-1 expression in H9c2 cells after H/R. The JNK inhibitor SP600125 inhibited Egr-1 overexpression in H9c2 cells caused by H/R. F2 could downregulate H/R-induced ROS level, JNK activation, and Egr-1 expression in H9c2 cells in a dose-dependent manner. The ROS donor hypoxanthine-xanthine oxidase (XO/HX) and the JNK activator ANISO antagonized the effects of F2. Therefore, H/R activates ROS/Egr-1 signaling pathway in H9c2 cells, and JNK activation plays an important role in this pathway. F2 regulates H/R-induced ROS/JNK/Egr-1 signaling, which might be an important mechanism by which it antagonizes myocardial I/R injury.


Cellular Physiology and Biochemistry | 2009

Egr-1, the Potential Target of Calcium Channel Blockers in Cardioprotection with Ischemia/Reperfusion Injury in Rats

Zhanqin Huang; Haiqing Li; Fuxiao Guo; Qiangyong Jia; Yanmei Zhang; Xing-Ping Liu; Ganggang Shi

Aims: In this study, we tested whether Egr-1 is a potential target of calcium channel blockers in cardioprotection with I/R injury. Methods: We treated rats in vivo I/R and rat cultured cardiomyocytes in vitro hypoxia/reoxygenation (H/R) models with three types of classical calcium channel blockers (verapamil, diltiazem and nifedipine). Activity of creatine kinase (CK), lactate dehydrogenase (LDH), myeloperoxidase (MPO) superoxide dismutase (SOD) and level of malondialdehyde (MDA) in plasma and culture medium were measured to assess the degree of injury and inflammation of myocardial tissues and cells. Egr-1 mRNA and protein expressions were examined by RT-PCR and Western-blot analyses. Results: Calcium channel blockers (verapamil, diltiazem and nifedipine) significantly attenuated myocardial injury, as shown by reduced release of CK and LDH, preserved SOD activity and decreased MDA production and MPO activity. Concomitant with cardioprotection by calcium channel blockers, the mRNA and protein expression of Egr-1 increased with I/R and H/R injury was significantly reduced in myocardial tissue and cultured cardiomyocytes. Conclusions: These results suggested that the cardioprotective effects of calcium channel blockers with I/R or H/R injury might be mediated by downregulating Egr-1 expression. Egr-1 might be the potential target of calcium channel blockers in cardioprotection with ischemia/reperfusion injury.


Cellular Physiology and Biochemistry | 2009

N-4-tert-butyl benzyl haloperidol chloride suppresses Ca2+-dependent Egr-1 expression and subsequently inhibits vascular smooth muscle cell proliferation induced by angiotensin II.

Yicun Chen; Jinhong Zheng; Yanmei Zhang; Jinzhi Wang; Qing Liu; Zhanqin Huang; Fenfei Gao; Yanqiong Zhou; Ganggang Shi

Background: N-4-Tert-Butyl benzyl haloperidol chloride (C3) was a novel calcium antagonist synthesized in our laboratory. The present study is to explore the effect of C3 on vascular smooth muscle cell proliferation and the mechanism involved. Methods: The effects of C3 on Ang II-induced cytosolic free Ca2+ concentration change, VSMC proliferation, the key early growth response factor 1 (Egr-1) were evaluated by laser scanning confocal microscopy, microtiter tetrazolium (MTT) proliferation assay, flow cytometry analysis, Western blot and RT-PCR analysis, respectively. An extracellular Ca2+ chelator EGTA and antisense Egr-1 oligodeoxyribonucleotides (ODNs) were used to establish the relation between Ca2+-dependent Egr-1 expression induced by Ang II and VSMC proliferation. Results: C3 attenuated the Ang II-induced extracellular Ca2+ influx, inhibited VSMCs proliferation and arrested VSMCs in G1-phase. C3 also triggered a significant reduction in PDGF-A and cyclin D1, Cdk2 along with an overexpression of p21Cip1. Antisense Egr-1 ODNs inhibited VSMCs proliferation, which was related to G1-phase arrest, due to inhibiting the expression of Egr-1 and C3 inhibited the overexpression of Egr-1. Conclusion: Egr-1 may play a key role in Ang II-induced proliferation of VSMCs. C3 inhibits vascular smooth muscle cell proliferation and the mechanism is involved with the inhibition of over-expression of Egr-1.


Cellular Physiology and Biochemistry | 2010

Cardiac Electrophysiological and Antiarrhythmic Effects of N-n-butyl Haloperidol Iodide

Fenfei Gao; Si-Yuan Hao; Zhanqin Huang; Yanmei Zhang; Yan Qiong Zhou; Yicun Chen; Xing-Ping Liu; Ganggang Shi

Aims: N-n-butyl haloperidol (F<sub>2</sub>), a novel compound of quaternary ammonium salt derivatives of haloperidol, was reported to antagonize myocardial ischemia/reperfusion injuries. The antiarrhythmic potential and electrophysiological effects of F<sub>2</sub> on rat cardiac tissues were investigated. Methods and Results: In Langendorff-perfused rat hearts, the ventricular arrhythmias were induced by left anterior descending coronary artery of rat heart ligated for 20 min before the release of the ligature. F<sub>2</sub> provided some inhibitive effects against ischemia- and reperfusion-induced ventricular arrhythmias. In His bundle electrogram and epicardial ECG recordings, the drug produced bradycardia, delayed the conduction through the atrioventricular node and prolonged the Wenckebach cycle length and atrioventricular nodal effective refractory period. In whole-cell patch-clamp study, F<sub>2</sub> primarily inhibited the L-type Ca<sup>2+</sup> current (I<sub>Ca,L</sub>) (IC<sub>50</sub> = 0.17 µM) with tonic blocking properties and little use-dependence. And the drug also decreased the Na<sup>+</sup> current (IC<sub>50</sub> = 77.5 µM), the transient outward K<sup>+</sup> current (IC<sub>50</sub> = 20.4 µM), the steady-state outward K<sup>+</sup> current (IC<sub>50</sub> = 56.2 µM) and the inward rectifier K<sup>+</sup> current (IC<sub>50</sub> = 127.3 µM). Conclusion: F<sub>2</sub> may be a promising drug for the treatment of ischemic heart disease with cardiac arrhythmia.


Cellular Physiology and Biochemistry | 2011

N-n-butyl haloperidol iodide preserves cardiomyocyte calcium homeostasis during hypoxia/ischemia.

Jian-Feng Xiao; Chun-Yan Wang; Yong-Pan Huang; Jian-Xin Shen; Fenfei Gao; Zhanqin Huang; Yan-Shan Zheng; Ganggang Shi

Aims: N-n-Butyl haloperidol iodide (F2) is a novel compound derived from haloperidol. In our previous work, F2 was found to be an L-type calcium channel blocker which played a protective role in rat heart ischemic–reperfusion injury in a dose-dependent manner. In the current study, we aimed to investigate the effects and some possible mechanisms of F2 on calcium transients in hypoxic/ischemic rat cardiac myocytes. Methods and Results: Calcium transients’ images of rat cardiac myocytes were recorded during simulated hypoxia, using a confocal calcium imaging system. The amplitude, rising time from 25% to 75% (RT25-75), decay time from 75% to 25% (DT75-25) of calcium transients, and resting [Ca2+]i were extracted from the images by self-coding programs. In this study, hypoxia produced a substantial increase in diastolic [Ca2+]i and reduced the amplitude of calcium transients. Both RT25-75 and DT75-25 of Ca2+ transients were significantly prolonged. And F2 could reduce the increase in resting [Ca2+]iand the prolongation of RT25-75 and DT75-25 of Ca2+ transients during hypoxia. F2 also inhibited the reduction in amplitude of calcium transients which was caused by 30-min hypoxia. The activity of SERCA2a (sarcoplasmic reticulum Ca2+-ATPase, determined by test kits) decreased after 30-min ischemia, and intravenous F2 in rats could ameliorate the decreased activity of SERCA2a. The inward and outward currents of NCX (recorded by whole-cell patch-clamp analysis) were reduced during 10-min hypoxia, and F2 further inhibited the outward currents of NCX during 10-min hypoxia. All these data of SERCA2a and NCX might be responsible for the changes in calcium transients during hypoxia. Conclusion: Our data suggest that F2 reduced changes in calcium transients that caused by hypoxia/ischemia, which was regarded to be a protective role in calcium homeostasis of ventricular myocytes, probably via changing the function of SERCA2a.


Cellular Physiology and Biochemistry | 2009

Egr-1, a Central and Unifying Role in Cardioprotection from Ischemia-Reperfusion Injury?

Fenfei Gao; Qiangyong Jia; Fuxiao Guo; Yanmei Zhang; Zhanqin Huang; Yanqiong Zhou; Yicun Chen; Ganggang Shi

Aims: Our previous studies have shown that N-n-butyl haloperidol iodide (F2) can antagonize myocardial ischemia/reperfusion (I/R) injury by blocking intracellular Ca2+ overload and suppressing Egr-1 overexpression. The present study is to investigate the relation between the reduction of Ca2+ overload and the inhibition of Egr-1 overexpression. Methods: The Sprague-Dawley rat myocardial I/R model and cultured cardiomyocyte hypoxia-reoxygenation (H/R) model were established. Administration of Egr-1 antisense oligodeoxyribonucleotide (AS-ODN) only or combining with F2, Egr-1 protein expression was examined by Western-blot analyses. Hemodynamic parameters, creatine kinase (CK) and lactate dehydrogenase (LDH), superoxide dismutase (SOD) and malondialdehyde (MDA), myeloperoxidase (MPO), cardiac troponin I (cTnI), and tumor necrosis factor-α (TNF-α) were measured to assess the degree of injury and inflammation of myocardial tissues and cells. Results: Treatment with Egr-1 AS-ODN significantly reduced Egr-1 protein expression and attenuated injury and inflammation of myocardium caused by I/R or H/R evidenced by the amelioration of hemodynamics, the decrease in leakage of CK, LDH, cTnI, the increase in MDA generation, the decrease in SOD activity, the reduction of MPO activity in myocardial tissues and release of TNF-α from cultured cardiomyocytes. Treatment with F2 combined with Egr-1 AS-ODN, the inhibition of Egr-1 protein expression and inflammation (MPO activity and TNF-α level) were not enhanced, but the protection from myocardial I/R (or H/R) injury was significantly increased in hemodynamics and cytomembrane permeability relative to the using of Egr-1 AS-ODN only. Conclusion: These data suggest that the inhibition of Egr-1 overexpression cannot involve all mechanisms of cardioprotection from I/R injury.


Oxidative Medicine and Cellular Longevity | 2013

N-n-Butyl Haloperidol Iodide Ameliorates Cardiomyocytes Hypoxia/Reoxygenation Injury by Extracellular Calcium-Dependent and -Independent Mechanisms

Yanmei Zhang; Gaoyong Chen; Shuping Zhong; Fuchun Zheng; Fenfei Gao; Yicun Chen; Zhanqin Huang; Wenfeng Cai; Weiqiu Li; Xing-Ping Liu; Yan-Shan Zheng; Han Xu; Ganggang Shi

N-n-butyl haloperidol iodide (F2) has been shown to antagonize myocardial ischemia/reperfusion injury by blocking calcium channels. This study explores the biological functions of ERK pathway in cardiomyocytes hypoxia/reoxygenation injury and clarifies the mechanisms by which F2 ameliorates cardiomyocytes hypoxia/reoxygenation injury through the extracellular-calcium-dependent and -independent ERK1/2-related pathways. In extracellularcalcium-containing hypoxia/reoxygenation cardiomyocytes, PKCα and ERK1/2 were activated, Egr-1 protein level and cTnI leakage increased, and cell viability decreased. The ERK1/2 inhibitors suppressed extracellular-calcium-containing-hypoxia/reoxygenation-induced Egr-1 overexpression and cardiomyocytes injury. PKCα inhibitor downregulated extracellularcalcium-containing-hypoxia/reoxygenation-induced increase in p-ERK1/2 and Egr-1 expression. F2 downregulated hypoxia/reoxygenation-induced elevation of p-PKCα, p-ERK1/2, and Egr-1 expression and inhibited cardiomyocytes damage. The ERK1/2 and PKCα activators antagonized F2s effects. In extracellular-calcium-free-hypoxia/reoxygenation cardiomyocytes, ERK1/2 was activated, LDH and cTnI leakage increased, and cell viability decreased. F2 and ERK1/2 inhibitors antagonized extracellular-calcium-free-hypoxia/reoxygenation-induced ERK1/2 activation and suppressed cardiomyocytes damage. The ERK1/2 activator antagonized F2s above effects. F2 had no effect on cardiomyocyte cAMP content or PKA and Egr-1 expression. Altogether, ERK activation in extracellular-calcium-containing and extracellular-calcium-free hypoxia/reoxygenation leads to cardiomyocytes damage. F2 may ameliorate cardiomyocytes hypoxia/reoxygenation injury by regulating the extracellular-calcium-dependent PKCα/ERK1/2/Egr-1 pathway and through the extracellular-calcium-independent ERK1/2 activation independently of the cAMP/PKA pathway or Egr-1 overexpression.


Drug Design Development and Therapy | 2014

N-n-butyl haloperidol iodide inhibits H2O2-induced Na+/Ca2+-exchanger activation via the Na+/H+ exchanger in rat ventricular myocytes.

Yong-Pan Huang; Fenfei Gao; Bin Wang; Fuchun Zheng; Yanmei Zhang; Yicun Chen; Zhanqin Huang; Yan-Shan Zheng; Shuping Zhong; Ganggang Shi

N-n-butyl haloperidol iodide (F2), a novel compound, has shown palliative effects in myocardial ischemia/reperfusion (I/R) injury. In this study, we investigated the effects of F2 on the extracellular signal-regulated kinase kinase (MEK)/extracellular signal-regulated kinase (ERK)/Na+/H+ exchanger (NHE)/Na+/Ca2+ exchanger (NCX) signal-transduction pathway involved in H2O2-induced Ca2+ overload, in order to probe the underlying molecular mechanism by which F2 antagonizes myocardial I/R injury. Acute exposure of rat cardiac myocytes to 100 μM H2O2 increased both NHE and NCX activities, as well as levels of phosphorylated MEK and ERK. The H2O2-induced increase in NCX current (INCX) was nearly completely inhibited by the MEK inhibitor U0126 (1,4-diamino-2,3-dicyano-1,4-bis[o-aminophenylmercapto] butadiene), but only partly by the NHE inhibitor 5-(N,N-dimethyl)-amiloride (DMA), indicating the INCX increase was primarily mediated by the MEK/mitogen-activated protein kinase (MAPK) pathway, and partially through activation of NHE. F2 attenuated the H2O2-induced INCX increase in a concentration-dependent manner. To determine whether pathway inhibition was H2O2-specific, we examined the ability of F2 to inhibit MEK/ERK activation by epidermal growth factor (EGF), and NHE activation by angiotensin II. F2 not only inhibited H2O2-induced and EGF-induced MEK/ERK activation, but also completely blocked both H2O2-induced and angiotensin II-induced increases in NHE activity, suggesting that F2 directly inhibits MEK/ERK and NHE activation. These results show that F2 exerts multiple inhibitions on the signal-transduction pathway involved in H2O2-induced INCX increase, providing an additional mechanism for F2 alleviating intracellular Ca2+ overload to protect against myocardial I/R injury.


Biochemical and Biophysical Research Communications | 2012

Effects of N-n-butyl haloperidol iodide on the rat myocardial sarcoplasmic reticulum Ca 2+ -ATPase during ischemia/reperfusion

Yanmei Zhang; Chunyan Wang; Fuchun Zheng; Fenfei Gao; Yicun Chen; Zhanqin Huang; Zhengyuan Xia; Michael G. Irwin; Weiqiu Li; Xing-Ping Liu; Yan-Shan Zheng; Han Xu; Ganggang Shi

We have previously shown that N-n-butyl haloperidol iodide (F(2)), a newly synthesized compound, reduces ischemia/reperfusion (I/R) injury by preventing intracellular Ca(2+) overload through inhibiting L-type calcium channels and outward current of Na(+)/Ca(2+) exchanger. This study was to investigate the effects of F(2) on activity and protein expression of the rat myocardial sarcoplasmic reticulum Ca(2+)-ATPase (SERCA) during I/R to discover other molecular mechanisms by which F(2) maintains intracellular Ca(2+) homeostasis. In an in vivo rat model of myocardial I/R achieved by occluding coronary artery for 30-60 min followed by 0-120 min reperfusion, treatment with F(2) (0.25, 0.5, 1, 2 and 4 mg/kg, respectively) dose-dependently inhibited the I/R-induced decrease in SERCA activity. However, neither different durations of I/R nor different doses of F(2) altered the expression levels of myocardial SERCA2a protein. These results indicate that F(2) exerts cardioprotective effects against I/R injury by inhibiting I/R-mediated decrease in SERCA activity by a mechanism independent of SERCA2a protein levels modulation.

Collaboration


Dive into the Zhanqin Huang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge