Zhangping Liao
Nanchang University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Zhangping Liao.
European Journal of Pharmacology | 2015
Zhen Guo; Zhangping Liao; Liqing Huang; Dan Liu; Dong Yin; Ming He
Mitochondria-mediated apoptosis is a critical mechanism of anoxia/ reoxygenation (A/R)-induced injury in cardiomyocytes. Kaempferol (Kae) is a natural polyphenol and a type of flavonoid, which has been demonstrated to protect myocardium against ischemia/reperfusion (I/R) injury. However, the mechanism is still not fully elucidated. We hypothesize that Kae may improve the mitochondrial function during I/R injury via a potential signal pathway. In this study, an in vitro I/R model was replicated on neonatal rat primary cardiomyocytes by A/R treatment. Cell viability was monitored by the 3-(4,5-dimethylthiazol- 2-yl)-5-(3- carboxymethoxyphenyl)-2-(4-sulfophenyl)-2 H-tetrazolium (MTS) assay. The levels of intracellular reactive oxygen species, mitochondrial membrane potential (Δψm) and apoptosis were determined by flow cytometry. Protein expression was detected by Western Blotting. mPTP opening and the activity of caspase-3 were measured by colorimetric method. The results showed that Kae effectively enhanced the cell viability and decreased the LDH release in cardiomyocytes subjected to A/R injury. Kae reduced the A/R-induced reactive oxygen species generation, the loss of Δψm, and the release of cytochrome c from mitochondria into cytosol. Kae inhibited the A/R-stimulated mPTP opening and activation of caspase-3, and ultimate decrease in cardiomyocytes apoptosis. Furthermore, we found Kae up-regulated Human Silent Information Regulator Type 1 (SIRT1) expression, indicating SIRT1 signal pathway likely involved the cardioprotection of Kae. Sirtinol, a SIRT1 inhibitor, abolished the protective effect of Kae in cardiomyocytes subjected to A/R. Additionally, Kae significantly increased the expression of Bcl-2. Thus, we firstly demonstrate that Kae protects cardiomyocytes against A/R injury through mitochondrial pathway mediated by SIRT1.
European Journal of Pharmacology | 2009
He-Ping Chen; Zhangping Liao; Qi-Ren Huang; Ming He
Development of intracellular calcium overload is an important pathophysiological factor in myocardial ischemia/reperfusion or anoxia/reoxygenation injury. Recent studies have shown that Sodium Ferulate (SF) stimulates nitric oxide (NO) production and exerts a cardioprotective effect in the ischemia-reperfused heart. However, it has not been determined whether the cardioprotection of SF is associated with suppression of Ca(2+) overload via NO/cyclic GMP (cGMP)/cGMP-dependent protein kinase (PKG) pathway. In this work, after cardiomyocytes were incubated with 100, 200, 400, or 800 microM SF for 3 h, anoxia/reoxygenation injury was induced and intracellular Ca(2+) concentration, NO synthase (NOS) activity, guanylate cyclase activity, NO, and cGMP formation were measured appropriately. The results showed that treatment with SF concentration-dependently inhibited calcium overload induced by anoxia/reoxygenation. We also demonstrated that SF (100-800 microM) concentration dependently enhanced NO and cGMP formation through increasing NOS activity and guanylate cyclase activity in the cardiomyocytes. On the contrary, inhibition of calcium overload by SF was markedly attenuated by addition of an NOS inhibitor, an NO scavenger, an soluble guanylate cyclase inhibitor, and a PKG inhibitor: N(G)-nitro-l-arginine methyl ester (L-NAME, 100 microM), 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazole-1-oxyl-3-oxide (c-PTIO, 1.0 microM), 1H-[1, 2, 4] oxadiazolo [4, 3-alpha] quinoxalin-1-one (ODQ, 20 microM) and KT5823 (0.2 microM), respectively. Our findings indicate that SF significantly attenuates anoxia/reoxygenation-induced Ca(2+) overload and improves cell survival in cultured cardiomyocytes through NO/cGMP/PKG signal pathway.
Molecular Medicine Reports | 2014
Zeyu Zhang; Dan Liu; Bo Yi; Zhangping Liao; Lei Tang; Dong Yin; Ming He
We previously demonstrated that iron overload induces liver damage by causing the formation of reactive oxygen species (ROS). Taurine is a potent free radical scavenger that attenuates the damage caused by excessive oxygen free radicals. Therefore, the aim of the present study was to investigate whether taurine could reduce the hepatotoxicity of iron overload with regard to ROS production. Mice were intraperitoneally injected with iron 5 days/week for 13 weeks to achieve iron overload. It was found that iron overload resulted in liver dysfunction, increased apoptosis and elevated oxidative stress. Taurine supplementation increased liver taurine levels by 40% and led to improved liver function, as well as a reduction in apoptosis, ROS formation and mitochondrial swelling and an attenuation in the loss of the mitochondrial membrane potential. Treatment with taurine mediated a reduction in oxidative stress in iron-overloaded mice, attenuated liver lipid peroxidation, elevated antioxidant enzyme activities and maintained reduced glutathione levels. These results indicate that taurine reduces iron-induced hepatic oxidative stress, preserves liver function and inhibits hepatocyte apoptosis. Therefore, taurine may be a potential therapeutic drug to reduce liver damage caused by iron overload.
Acta Pharmacologica Sinica | 2010
Li-ping Liu; Zhangping Liao; Dong Yin; Wei-dong Li; Dan Liu; Qing Li; Qi-Ren Huang; Yao-fang Yang; Ming He
AbstractAim:To investigate the protective effects of preconditioning human umbilical vein endothelial cells (HUVECs) with Polygonum multiflorum stilbeneglycoside (PMS) under anoxia/reoxygenation (A/R), and the mechanism of protection.Methods:Prior to A/R, HUVECs were incubated with PMS (0.6×10−11, 1.2×10−11, or 2.4×10−11 mol/L) for 3 h. Cell injury was subsequently evaluated by measuring cell viability with an MTT assay and lactate dehydrogenase (LDH) release, whereas lipid peroxidation was assayed by measuring malondialdehyde (MDA) content. Antioxidant capacity was quantified by superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activity. Nitric oxide (NO) production was determined by nitrite accumulation. Endothelial NO synthase (eNOS) and inducible NOS (iNOS) protein expression was detected by Western blotting. Guanylate cyclase activity and cyclic GMP (cGMP) activity were assessed by an enzyme immunoassay kit.Results:PMS incubation attenuated A/R-induced injury in a concentration-dependent manner, as evidenced by a decrease in LDH activity and an increase in cell viability. PMS exerted its protective effect by inhibiting the A/R-mediated elevation of MDA content, as well as by promoting the recovery of SOD and GSH-Px activities. Additionally, PMS incubation enhanced NO and cGMP formation by increasing iNOS expression and guanylate cyclase activity. The protective effects of PMS were markedly attenuated by NOS inhibitor L-NAME, soluble guanylate cyclase inhibitor ODQ or PKG inhibitor KT5823.Conclusion:PMS preincubation resulted in the enhancement of antioxidant activity and anti-lipid peroxidation. The NO/cGMP/cGMP-dependent protein kinase (PKG) signaling pathway was involved in the effect of PMS on HUVECs.
International Immunopharmacology | 2014
Dan Liu; Bo Yi; Zhangping Liao; Lei Tang; Dong Yin; Shu Zeng; Jianguo Yao; Ming He
Previous studies have indicated that 14-3-3γ is upregulated by stress in LPS-induced cardiovascular injury. In this study, we investigated the interaction of 14-3-3γ and Bcl-2 family members in the control of the mitochondrial permeability transition (MPT) to test the hypothesis that abundant levels of 14-3-3γ can protect against LPS-induced injury via a Bcl-2 family/mitochondria pathway. The cardiomyocytes were treated with LPS (1mg l(-1)) for 6h; the interaction between 14-3-3γ and phospho-Bad(S112) was detected by co-immunoprecipitation (co-IP); the levels of Bcl-2 family members in the cytosolic and mitochondrial fractions were examined by Western blot; the apoptosis and mitochondrial membrane potential (ΔΨm) were detected by flow cytometry; and the mitochondrial permeability transition pore (mPTP) opening was tested by mitochondrial swelling. Our results revealed that LPS treatment results in cardiomyocyte injury, and these effects were significantly attenuated by pFLAG-14-3-3γ. Moreover, LPS treatment induced Bax translocation to the mitochondria, ΔΨm loss, mitochondrial swelling, and cytochrome c release, and pFLAG-14-3-3γ reversed these effects induced by LPS. Moreover, overexpressed 14-3-3γ protein could assist Bad(S112) phosphorylation and interact with it to form a complex, which might result in the disassociation of Bcl-2 from the Bad/Bcl-2 complex and its translocation from the cytosol to the mitochondria. Our data firstly confirmed that a high level of 14-3-3γ protects against LPS-induced cardiomyocyte injury likely through a pathway associated with the regulation of the subcellular localizations of Bcl-2 and Bad that results in the prevention of mPTP opening, the maintenance of ΔΨm, and ultimately the inhibition of apoptosis.
Phytotherapy Research | 2009
Zhangping Liao; Dong Yin; Wufeng Wang; Guohua Zeng; Dan Liu; He-Ping Chen; Qi-Ren Huang; Ming He
Sasanquasaponin (SQS) is an effective component of Camellia oleifera Abel. This study was designed to investigate the cardioprotective effect of SQS against ischemia‐reperfusion (I/R) injury and the possible mechanism in isolated rat hearts. These hearts were pretreated by SQS only or SQS and HOE140 in different groups, and then subjected to I/R injury. Hemodynamic parameters, oxidative injury, and NO level were measured. The results showed that SQS preconditioning could decrease the incidences of arrhythmias and improve the heart functions. In addition, SQS preconditioning could protect isolated I/R injured heart against lipid peroxidation, as evidenced by increases in SOD and GSH‐Px activity, and by decreases in contents of MDA, ROS generation. However, HOE140 treatment reversed all these indexes. NO production was significantly decreased after a treatment with HOE140. So we can propose that SQS preconditioning could induce the cardioprotective effects and the possible mechanism was that the activation of bradykinin‐NO system by SQS preconditioning had an inhibition effect on ROS generation in isolated heart. Copyright
Molecular Nutrition & Food Research | 2015
Zhangping Liao; Dan Liu; Lei Tang; Dong Yin; Shuhua Yin; Songqing Lai; Jianguo Yao; Ming He
SCOPE This study elucidates the effects of long-term nutritional preconditioning by resveratrol on ischemia/reperfusion (I/R) injury and its underlying mechanisms. METHODS AND RESULTS Mice were treated with resveratrol at 2.0 mg/kg/day by gastric gavages for 6 wk. Then hearts were isolated and subjected to I/R injury in a Langendorff apparatus. Resveratrol significantly improved left ventricular pressure, ±dp/dtmax, and coronary flow; decreased the lactate dehydrogenase and creatine phosphokinase activities; and reduced the infarction size. Additionally, long-term oral resveratrol intake prevented mitochondrial permeability transition pore opening and subsequently inhibited mitochondria-mediated apoptosis, as demonstrated by decrease of cytochrome c release, inactivation of caspase-3, and reduction of terminal deoxynucleotidyl transferase mediated nick end labeling positive cells. Furthermore, resveratrol inhibited the upregulation of voltage-dependent anion channel 1 (VDAC1) expression induced by I/R injury. Local left-ventricle overexpression of VDAC1 by adenovirus diminished the protective effect of resveratrol against I/R injury, indicating that VDAC1 plays an important role in resveratrol-mediated cardioprotection. CONCLUSION Our data revealed that long-term oral intake of resveratrol sets nutritional preconditioning to cope with myocardial I/R injury. Strikingly, we found that resveratrol downregulates VDAC1, leading to prevention of mitochondrial permeability transition pore opening and cardiomyocyte apoptosis.
Molecular Medicine Reports | 2015
Yu-Feng Yan; Wen-Jie Yang; Qiang Xu; He-Ping Chen; Xiao-Shan Huang; Ling-Yu Qiu; Zhangping Liao; Qi-Ren Huang
DJ-1 protein, as a multifunctional intracellular protein, has an important role in transcriptional regulation and anti-oxidant stress. A recent study by our group showed that DJ-1 can regulate the expression of certain anti‑oxidant enzymes and attenuate hypoxia/re‑oxygenation (H/R)‑induced oxidative stress in the cardiomyocyte cell line H9c2; however, the detailed molecular mechanisms have remained to be elucidated. Nuclear factor erythroid 2‑like 2 (Nrf2) is an essential transcription factor that regulates the expression of several anti‑oxidant genes via binding to the anti‑oxidant response element (ARE). The present study investigated whether activation of the Nrf2 pathway is responsible for the induction of anti‑oxidative enzymes by DJ‑1 and contributes to the protective functions of DJ‑1 against H/R‑induced oxidative stress in H9c2 cells. The results demonstrated that DJ‑1‑overexpressing H9c2 cells exhibited anti‑oxidant enzymes, including manganese superoxide dismutase, catalase and glutathione peroxidase, to a greater extent and were more resistant to H/R‑induced oxidative stress compared with native cells, whereas DJ‑1 knockdown suppressed the induction of these enzymes and further augmented the oxidative stress injury. Determination of the importance of Nrf2 in DJ‑1‑mediated anti‑oxidant enzymes induction and cytoprotection against oxidative stress induced by H/R showed that overexpression of DJ‑1 promoted the dissociation of Nrf2 from its cytoplasmic inhibitor Keap1, resulting in enhanced levels of nuclear translocation, ARE‑binding and transcriptional activity of Nrf2. Of note, Nrf2 knockdown abolished the DJ‑1‑mediated induction of anti‑oxidant enzymes and cytoprotection against oxidative stress induced by H/R. In conclusion, these findings indicated that activation of the Nrf2 pathway is a critical mechanism by which DJ-1 upregulates anti-oxidative enzymes and attenuates H/R-induced oxidative stress in H9c2 cells.
Molecular Medicine Reports | 2013
Dan Liu; Huan He; Dong Yin; Ailing Que; Lei Tang; Zhangping Liao; Qi-Ren Huang; Ming He
Chronic iron overload may result in hepatic fibrosis and even neoplastic transformation due to a burst of reactive oxygen species (ROS). Mitochondria have been proposed to be important in the production of ROS. The purpose of this study was to investigate the role of the mitochondrial permeability transition pore (mPTP) in the burst of ROS, and to clarify the mechanism whereby ROS induced by iron overload results in hepatic damage. It has been demonstrated that when ferrocene-induced iron-overloaded mice were fed the cyclosporin A (CsA), a specific inhibitor of the mPTP, diet (10 mg/kg/day) for 50 days, liver-to-body weight ratio, serum levels of alanine transaminase (ALT) and aspartate transaminase (AST), ROS production, mitochondrial swelling, loss of mitochondrial membrane potential (Δψ) and hepatocyte apoptosis decreased. However, the total antioxidant status, including superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase activities, increased. The protective effect of CsA on the liver of iron-overloaded mice may be due to inhibition of the ROS burst and a successive antioxidant effect. To the best of our knowledge, these data provide the first support for the theory that ROS-induced ROS release (RIRR) may be involved in the burst of ROS in the liver and greatly contribute to the hepatic damage initiated by iron overload.
Journal of Cardiovascular Pharmacology | 2015
Yu-Feng Yan; He-Ping Chen; Xiao-Shan Huang; Ling-Yu Qiu; Zhangping Liao; Qi-Ren Huang
Abstract: We have recently shown that DJ-1 is implicated in the delayed cardioprotective effect of hypoxic preconditioning (HPC) against hypoxia/reoxygenation (H/R) injury as an endogenous protective protein. This study aims to further investigate the underlying mechanism by which DJ-1 mediates the delayed cardioprotection of HPC against H/R-induced oxidative stress. Using a well-characterized cellular model of HPC from rat heart–derived H9c2 cells, we found that HPC promoted nuclear factor erythroid 2–related factor 2 (Nrf2) and its cytoplasmic inhibitor Kelch-like ECH-associated protein-1 (Keap1) dissociation and resulted in increased nuclear translocation, antioxidant response element–binding, and transcriptional activity of Nrf2 24 hours after HPC, with subsequent upregulation of manganese superoxide dismutase (MnSOD) and heme oxygenase-1 (HO-1), which provided delayed protection against H/R-induced oxidative stress in normal H9c2 cells. However, the aforementioned effects of HPC were abolished in DJ-1–knockdown H9c2 cells, which were restored by restoration of DJ-1 expression. Importantly, we showed that inhibition of the Nrf2 pathway in H9c2 cells mimicked the effects of DJ-1 knockdown and abolished HPC-derived induction of antioxidative enzymes (MnSOD and HO-1) and the delayed cardioprotection. In addition, inhibition of Nrf2 also reversed the effects of restored DJ-1 expression on induction of antioxidative enzymes and delayed cardioprotection by HPC in DJ-1–knockdown H9c2 cells. Taken together, this work revealed that activation of Nrf2 pathway and subsequent upregulation of antioxidative enzymes could be a critical mechanism by which DJ-1 mediates the delayed cardioprotection of HPC against H/R-induced oxidative stress in H9c2 cells.