Network


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

Hotspot


Dive into the research topics where Jia-Ning Wang is active.

Publication


Featured researches published by Jia-Ning Wang.


Cardiovascular Research | 2011

VEGF/SDF-1 Promotes Cardiac Stem Cell Mobilization and Myocardial Repair in the Infarcted Heart

Jun-Ming Tang; Jia-Ning Wang; Lei Zhang; Fei Zheng; Jian-Ye Yang; Xia Kong; Linyun Guo; Long Chen; Yongzhang Huang; Yu Wan; Shi-You Chen

AIMS The objective of this study was to investigate whether vascular endothelial growth factor (VEGF) secreted by mesenchymal stem cells (MSC) improves myocardial survival and the engraftment of implanted MSC in infarcted hearts and promotes recruitment of stem cells through paracrine release of myocardial stromal cell-derived factor-1α (SDF-1α). METHODS AND RESULTS VEGF-expressing MSC ((VEGF)MSC)-conditioned medium enhanced SDF-1α expression in heart slices and H9C2 cardiomyoblast cells via VEGF and the vascular endothelial growth factor receptor (VEGFR). The (VEGF)MSC-conditioned medium markedly promoted cardiac stem cell (CSC) migration at least in part via the SDF-1α/CXCR4 pathway and involved binding to VEGFR-1 and VEGFR-3. In vivo, (VEGF)MSC-stimulated SDF-1α expression in infarcted hearts resulted in massive mobilization and homing of bone marrow stem cells and CSC. Moreover, VEGF-induced SDF-1α guided the exogenously introduced CSC in the atrioventricular groove to migrate to the infarcted area, leading to a reduction in infarct size. Functional studies showed that (VEGF)MSC transplantation stimulated extensive angiomyogenesis in infarcted hearts as indicated by the expression of cardiac troponin T, CD31, and von Willebrand factor and improved the left ventricular performance, whereas blockade of SDF-1α or its receptor by RNAi or antagonist significantly diminished the beneficial effects of (VEGF)MSC. CONCLUSION Exogenously expressed VEGF promotes myocardial repair at least in part through SDF-1α/CXCR4-mediated recruitment of CSC.


European Journal of Cardio-Thoracic Surgery | 2009

Mesenchymal stem cells over-expressing SDF-1 promote angiogenesis and improve heart function in experimental myocardial infarction in rats.

Junming Tang; Jia-Ning Wang; Jian-Ye Yang; Xia Kong; Fei Zheng; Linyun Guo; Lei Zhang; Yongzhang Huang

BACKGROUND In addition to its multipotent capability, the mesenchymal stem cell (MSC) can secrete and supply a large amount of vascular endothelial growth factor (VEGF). The stromal-derived factor-1 alpha (SDF-1alpha) plays an important role in the homing of stem cells to the injured tissues of the heart. Therefore, the MSCs over-expressing SDF-1alpha could augment the angiogenesis pathway. METHODS In vitro, the differentiation of the MSCs into endothelial-like cells was induced by cultivation of cells in 10% foetal calf serum and 50 ngml(-1) SDF-1alpha or in specific inhibitors for endothelial nitrous oxide synthase (eNOS). In vivo, the rat model of myocardial infarction was established by occlusion of the left anterior descending coronary artery. Seven days following surgery, 5.0 x 10(9)pfu Ad-SDF-1alpha (adenoviral vector containing human SDF-1alpha gene under the control of the rous sarcoma virus (RSV) promoter), 5.0 x 10(6) Ad-LacZ-MSC or 5.0 x 10(6) Ad-SDF-MSC suspension in a 0.2-ml serum-free medium was injected into four sites in infarcted areas (0.05 ml per site). The rats receiving Ad-SDF-MSC also received the nitrous oxide (NO) synthesis inhibitor N(G)-nitro-l-arginine methyl ester (l-NAME) in drinking water (1 mgkg(-1)). The rats in the control group received the same volume of cell-free medium. Four weeks following transplantation, the heart function was assessed, and histological and molecular analyses were conducted. RESULTS The MSCs could differentiate into endothelial cells in the presence of SDF-1alpha, and the effect could be inhibited by l-NAME in vitro and in vivo. Western Blotting revealed an increased expression of VEGF, Akt and eNOS. Four weeks following transplantation, a reduced infarct size and fibrosis, greater vascular density and thicker left ventricular wall were observed in the Ad-SDF-MSC group. The measurement of haemodynamic parameters showed an improvement in the left ventricular performance in the Ad-SDF-MSC group as compared with other groups. CONCLUSION The MSCs over-expressing the SDF-1alpha can produce effective angiogenesis, resulting in the prevention of progressive heart dysfunction after a myocardial infarction.


Experimental Cell Research | 2009

Vascular endothelial growth factor promotes cardiac stem cell migration via the PI3K/Akt pathway

Junming Tang; Jia-Ning Wang; Xia Kong; Jian-Ye Yang; Linyun Guo; Fei Zheng; Lei Zhang; Yongzhang Huang; Yu Wan

VEGF is a major inducer of angiogenesis. However, the homing role of VEGF for cardiac stem cells (CSCs) is unclear. In in vitro experiments, CSCs were isolated from the rat hearts, and a cellular migration assay was performed using a 24-well transwell system. VEGF induced CSC migration in a concentration-dependent manner, and SU5416 blocked this. Western blot analysis showed that the phosphorylated Akt was markedly increased in the VEGF-treated CSCs and that inhibition of pAkt activity significantly attenuated the VEGF-induced the migration of CSCs. In in vivo experiments, rat heart myocardial infarction (MI) was induced by left coronary artery ligation. One week after MI, the adenoviral vector expressing hVEGF165 and LacZ genes were injected separately into the infarcted myocardium at four sites before endomyocardial transplantation of 2x10(5) PKH26 labeled CSCs (50 muL) at atrioventricular groove. One week after CSC transplantation, RT-PCR, immunohistochemical staining, Western blot, and ELISA analysis were performed to detect the hVEGF mRNA and protein. The expression of hVEGF mRNA and protein was significantly increased in the infarcted and hVEGF165 transfected rat hearts, accompanied by an enhanced PI3K/Ak activity, a greater accumulation of CSCs in the infarcted region, and an improvement in cardiac function. The CSC accumulation was inhibited by either the VEGF receptor blocker SU5416 or the PI3K/Ak inhibitor wortmannin. VEGF signaling may mediate the migration of CSCs via activation of PI3K/Akt.


Molecules and Cells | 2010

Mesenchymal stem cells modified with stromal cell-derived factor 1α improve cardiac remodeling via paracrine activation of hepatocyte growth factor in a rat model of myocardial infarction

Jun-Ming Tang; Jia-Ning Wang; Linyun Guo; Xia Kong; Jian-Ye Yang; Fei Zheng; Lei Zhang; Yongzhang Huang

Mesenchymal stem cells (MSCs) are a promising source for cell-based treatment of myocardial infarction (MI), but existing strategies are restricted by low cell survival and engraftment. We examined whether SDF-1 transfection improve MSC viability and paracrine action in infarcted hearts. We found SDF-1-modified MSCs effectively expressed SDF-1 for at least 21days after exposure to hypoxia. The apoptosis of Ad-SDF-1-MSCs was 42% of that seen in Ad-EGFP-MSCs and 53% of untreated MSCs. In the infarcted hearts, the number of DAPI-labeling cells in the Ad-SDF-1-MSC group was 5-fold that in the Ad-EGFP-MSC group. Importantly, expression of antifibrotic factor, HGF, was detected in cultured MSCs, and HGF expression levels were higher in Ad-SDF-MSC-treated hearts, compared with Ad-EGFP-MSC or control hearts. Compared with the control group, Ad-SDF-MSC transplantation significantly decreased the expression of collagens I and III and matrix metalloproteinase 2 and 9, but heart function was improved in d-SDF-MSC-treated animals. In conclusion, SDF-1-modified MSCs enhanced the tolerance of engrafted MSCs to hypoxic injury in vitro and improved their viability in infarcted hearts, thus helping preserve the contractile function and attenuate left ventricle (LV) remodeling, and this may be at least partly mediated by enhanced paracrine signaling from MSCs via antifibrotic factors such as HGF.


Carcinogenesis | 2013

Apigenin sensitizes doxorubicin-resistant hepatocellular carcinoma BEL-7402/ADM cells to doxorubicin via inhibiting PI3K/Akt/Nrf2 pathway

Ai-Mei Gao; Zun-Ping Ke; Jia-Ning Wang; Jian-Ye Yang; Shi-You Chen; Hui Chen

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a redox- sensitive transcription factor regulating expression of a number of cytoprotective genes. Recently, Nrf2 has emerged as an important contributor to chemoresistance in cancer therapy. In the present study, we found that non-toxic dose of apigenin (APG) significantly sensitizes doxorubicin-resistant BEL-7402 (BEL-7402/ADM) cells to doxorubicin (ADM) and increases intracellular concentration of ADM. Mechanistically, APG dramatically reduced Nrf2 expression at both the messenger RNA and protein levels through downregulation of PI3K/Akt pathway, leading to a reduction of Nrf2-downstream genes. In BEL-7402 xenografts, APG and ADM cotreatment inhibited tumor growth, reduced cell proliferation and induced apoptosis more substantially when compared with ADM treatment alone. These results clearly demonstrate that APG can be used as an effective adjuvant sensitizer to prevent chemoresistance by downregulating Nrf2 signaling pathway.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Response Gene to Complement 32 Promotes Vascular Lesion Formation Through Stimulation of Smooth Muscle Cell Proliferation and Migration

Jia-Ning Wang; Ning Shi; Wei-Bing Xie; Xia Guo; Shi-You Chen

Objective—The objectives of this study were to determine the role of response gene to complement 32 (RGC-32) in vascular lesion formation after experimental angioplasty and to explore the underlying mechanisms. Methods and Results—Using a rat carotid artery balloon-injury model, we documented for the first time that neointima formation was closely associated with a significantly increased expression of RGC-32 protein. Short hairpin RNA knockdown of RGC-32 via adenovirus-mediated gene delivery dramatically inhibited the lesion formation by 62% as compared with control groups 14 days after injury. Conversely, RGC-32 overexpression significantly promoted the neointima formation by 33%. Gain- and loss-of-function studies in primary culture of rat aortic smooth muscle cells (RASMCs) indicated that RGC-32 is essential for both the proliferation and migration of RASMCs. RGC-32 induced RASMC proliferation by enhancing p34CDC2 activity. RGC-32 stimulated the migration of RASMC by inducing focal adhesion contact and stress fiber formation. These effects were caused by the enhanced rho kinase II-&agr; activity due to RGC-32-induced downregulation of Rad GTPase. Conclusion—RGC-32 plays an important role in vascular lesion formation following vascular injury. Increased RGC-32 expression in vascular injury appears to be a novel mechanism underlying the migration and proliferation of vascular smooth muscle cells. Therefore, targeting RGC-32 is a potential therapeutic strategy for the prevention of vascular remodeling in proliferative vascular diseases.


Molecular Biology Reports | 2012

VEGF is essential for the growth and migration of human hepatocellular carcinoma cells.

Lei Zhang; Jia-Ning Wang; Jun-Ming Tang; Xia Kong; Jian-Ye Yang; Fei Zheng; Ling-Yun Guo; Yongzhang Huang; Li Zhang; Lin Tian; Shu-Fen Cao; Chang-Hai Tuo; Hong-Li Guo; Shi-You Chen

Vascular endothelial growth factor (VEGF) plays a crucial role in tumor angiogenesis. VEGF induces new vessel formation and tumor growth by inducing mitogenesis and chemotaxis of normal endothelial cells and increasing vascular permeability. However, little is known about VEGF function in the proliferation, survival or migration of hepatocellular carcinoma cells (HCC). In the present study, we have found that VEGF receptors are expressed in HCC line BEL7402 and human HCC specimens. Importantly, VEGF receptor expression correlates with the development of the carcinoma. By using a comprehensive approaches including TUNEL assay, transwell and wound healing assays, migration and invasion assays, adhesion assay, western blot and quantitative RT-PCR, we have shown that knockdown of VEGF165 expression by shRNA inhibits the proliferation, migration, survival and adhesion ability of BEL7402. Knockdown of VEGF165 decreased the expression of NF-κB p65 and PKCα while increased the expression of p53 signaling molecules, suggesting that VEGF functions in HCC proliferation and migration are mediated by P65, PKCα and/or p53.


Molecules and Cells | 2009

In vivo protein transduction: delivery of PEP-1-SOD1 fusion protein into myocardium efficiently protects against ischemic insult.

You-en Zhang; Jia-Ning Wang; Jun-Ming Tang; Ling-Yun Guo; Jian-Ye Yang; Yongzhang Huang; Yan Tan; Shouzhi Fu; Xia Kong; Fei Zheng

Myocardial ischemia-reperfusion injury is a medical problem occurring as damage to the myocardium following blood flow restoration after a critical period of coronary occlusion. Oxygen free radicals (OFR) are implicated in reperfusion injury after myocardial ischemia. The antioxidant enzyme, Cu, Zn-superoxide dismutase (Cu, Zn-SOD, also called SOD1) is one of the major means by which cells counteract the deleterious effects of OFR after ischemia. Recently, we reported that a PEP-1-SOD1 fusion protein was efficiently delivered into cultured cells and isolated rat hearts with ischemia-reperfusion injury. In the present study, we investigated the protective effects of the PEP-1-SOD1 fusion protein after ischemic insult. Immunofluorescecnce analysis revealed that the expressed and purified PEP-1-SOD1 fusion protein injected into rat tail veins was efficiently transduced into the myocardium with its native protein structure intact. When injected into Sprague-Dawley rat tail veins, the PEP-1- SOD1 fusion protein significantly attenuated myocardial ischemia-reperfusion damage; characterized by improving cardiac function of the left ventricle, decreasing infarct size, reducing the level of malondialdehyde (MDA), decreasing the release of creatine kinase (CK) and lactate dehydrogenase (LDH), and relieving cardiomyocyte apoptosis. These results suggest that the biologically active intact forms of PEP-1-SOD1 fusion protein will provide an efficient strategy for therapeutic delivery in various diseases related to SOD1 or to OFR.


Free Radical Biology and Medicine | 2012

Manganese superoxide dismutase inhibits neointima formation through attenuation of migration and proliferation of vascular smooth muscle cells

Jia-Ning Wang; Ning Shi; Shi-You Chen

Superoxide anion is elevated during neointima development and is essential for neointimal vascular smooth muscle cell (VSMC) proliferation. However, little is known about the role of manganese superoxide dismutase (MnSOD, SOD2) in the neointima formation following vascular injury. SOD2 in the mitochondria plays an important role in cellular defense against oxidative damage. Because of its subcellular localization, SOD2 is considered the first line of defense against oxidative stress and plays a central role in metabolizing superoxide. Because mitochondria are the most important sources of superoxide anion, we speculated that SOD2 may have therapeutic benefits in preventing vascular remodeling. In this study, we used a rat carotid artery balloon-injury model and an adenoviral gene delivery approach to test the hypothesis that SOD2 suppresses vascular lesion formation. SOD2 was activated along with the progression of neointima formation in balloon-injured rat carotid arteries. Depletion of SOD2 by RNA interference markedly promoted the lesion formation, whereas SOD2 overexpression suppressed the injury-induced neointima formation via attenuation of migration and proliferation of VSMCs. SOD2 exerts its inhibitory effect on VSMC migration induced by angiotensin II by scavenging superoxide anion and suppressing the phosphorylation of Akt. Our data indicate that SOD2 is a negative modulator of vascular lesion formation after injury. Therefore, SOD2 augmentation may be a promising therapeutic strategy for the prevention of lesion formation in proliferative vascular diseases such as restenosis.


Journal of Cellular Biochemistry | 2012

Acetylcholine induces mesenchymal stem cell migration via Ca2+/PKC/ERK1/2 signal pathway

Jun-Ming Tang; Jie Yuan; Qing Li; Jia-Ning Wang; Xia Kong; Fei Zheng; Lei Zhang; Long Chen; Linyun Guo; Yong-Hang Huang; Jian-Ye Yang; Shi-You Chen

Acetylcholine (ACh) plays an important role in neural and non‐neural function, but its role in mesenchymal stem cell (MSC) migration remains to be determined. In the present study, we have found that ACh induces MSC migration via muscarinic acetylcholine receptors (mAChRs). Among several mAChRs, MSCs express mAChR subtype 1 (m1AChR). ACh induces MSC migration via interaction with mAChR1. MEK1/2 inhibitor PD98059 blocks ERK1/2 phosphorylation while partially inhibiting the ACh‐induced MSC migration. InsP3Rs inhibitor 2‐APB that inhibits MAPK/ERK phosphorylation completely blocks ACh‐mediated MSC migration. Interestingly, intracellular Ca2+ ATPase‐specific inhibitor thapsigargin also completely blocks ACh‐induced MSC migration through the depletion of intracellular Ca2+ storage. PKCα or PKCβ inhibitor or their siRNAs only partially inhibit ACh‐induced MSC migration, but PKC‐ζ siRNA completely inhibits ACh‐induced MSC migration via blocking ERK1/2 phosphorylation. These results indicate that ACh induces MSC migration via Ca2+, PKC, and ERK1/2 signal pathways. J. Cell. Biochem. 113: 2704–2713, 2012.

Collaboration


Dive into the Jia-Ning Wang's collaboration.

Top Co-Authors

Avatar

Fei Zheng

Hubei University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jian-Ye Yang

Hubei University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Lei Zhang

Hubei University of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jun-Ming Tang

Hubei University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Xia Kong

Hubei University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ling-Yun Guo

Hubei University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yongzhang Huang

Hubei University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Linyun Guo

Hubei University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Lu Wang

Hubei University of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge