Network


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

Hotspot


Dive into the research topics where Yongshun Wang is active.

Publication


Featured researches published by Yongshun Wang.


Biochemical and Biophysical Research Communications | 2012

MiR-499 induces cardiac differentiation of rat mesenchymal stem cells through wnt/β-catenin signaling pathway

Lulu Zhang; Jingjin Liu; Fang Liu; Wenhua Liu; Yongshun Wang; Bin Zhu

OBJECTIVE To test the hypothesis that over-expressing miR-499 in rat bone marrow-derived mesenchymal stem cells (BM-MSCs) induces them to differentiate into cardiomyocyte-like cells through the wnt/β-catenin signaling pathway. METHODS Rat BM-MSCs were infected with lentiviral vectors bearing miR-499. The expression of cardiac-specific markers, NKx2.5, GATA4, MEF2C, and cTnI in these cells were examined by rtPCR or Western blot analysis and the activity of the wnt/β-catenin signaling pathway was evaluated by measuring the phosphorylation status of β-catenin. RESULTS Over-expression of miR-499 in rat BM-MSCs increased the expression of cardiac-specific genes, such as NKx2.5, GATA4, MEF2C, and cTnI and decreased the ratio of phosphorylated/dephosphorylated β-catenin in the wnt/β-catenin signaling pathway, thus activating the pathway. Knocking down the expression of Dvl, an adaptor molecule in the wnt/β-catenin signaling, partially blocked the role of the miR-499 and decreased those cardiac-specific genes. CONCLUSION Over-expression of miR-499 in rat BM-MSCs induces them toward cardiac differentiation through the activating the wnt/β-catenin signal pathway.


PLOS ONE | 2014

Angiopoietin-like 4 confers resistance to hypoxia/serum deprivation-induced apoptosis through PI3K/Akt and ERK1/2 signaling pathways in mesenchymal stem cells.

Meng Hou; Jinjin Cui; Jingjin Liu; Fang Liu; Rui Jiang; Kai Liu; Yongshun Wang; Li Yin; Wenhua Liu

Angiopoietin-like 4 (ANGPTL4) is a potential anti-apoptotic agent for various cells. We examined the protective effect of ANGPTL4 on hypoxia/serum deprivation (SD)-induced apoptosis of MSCs, as well as the possible mechanisms. MSCs were obtained from rat bone marrow and cultured in vitro. Apoptosis was induced by hypoxia/SD for up to 24 hr, and assessed by flow cytometry and TUNEL assay. Expression levels of Akt, ERK1/2, focal adhesion kinase (FAK), Src, Bcl-2, Bax, cytochrome C and cleaved caspase-3 were detected by Western blotting. Integrin β1 mRNA was detected by qRT-PCR. Mitochondrial membrane potential was assayed using a membrane-permeable dye. Hypoxia/SD-induced apoptosis was significantly attenuated by recombinant rat ANGPTL4 in a concentration dependent manner. Moreover, ANGPTL4 decreased the hypoxia/SD-induced caspase-3 cleavage and the cytochrome C release, but increased the Bcl-2/Bax ratio and the mitochondrial membrane potential. Decreased expression of integrin β1, the ANGPTL4 receptor was observed during hypoxia/SD conditions, however, such decrease was reversed by ANGPTL4. In addition, ANGPTL4 induced integrin β1-associated FAK and Src phosphorylation, which was blocked by anti-integrin β1 antibody. ANGPTL4 also reversed the hypoxia/SD-induced decrease of Akt and ERK 1/2 phosphorylation, and the effect of ANGPTL4 was abolished by inhibitors of either integrins, ERK1/2, or phosphatidylinositol 3-kinase (PI3K). Blocking integrinβ1, Akt or ERK largely attenuated anti-apoptotic effect of ANGPTL4. ANGPTL4 protects MSCs from hypoxia/SD-induced apoptosis by interacting with integrins to stimulate FAK complex, leading to downstream ERK1/2 and PI3K/Akt signaling pathways and mimicking the pathway in which MSCs contact with the extracellular matrix.


PLOS ONE | 2013

Wnt1 Inhibits Hydrogen Peroxide-Induced Apoptosis in Mouse Cardiac Stem Cells

Jingjin Liu; Yongshun Wang; Wenjuan Du; Wenhua Liu; Fang Liu; Lulu Zhang; Maomao Zhang; Meng Hou; Kai Liu; Shuo Zhang

Background Because of their regenerative and paracrine abilities, cardiac stem cells (CSCs) are the most appropriate, optimal and promising candidates for the development of cardiac regenerative medicine strategies. However, native and exogenous CSCs in ischemic hearts are exposed to various pro-apoptotic or cytotoxic factors preventing their regenerative and paracrine abilities. Methods and Results We examined the effects of H2O2 on mouse CSCs (mCSCs), and observed that hydrogen peroxide (H2O2) treatment induces mCSCs apoptosis via the caspase 3 pathway, in a dose-dependent manner. We then examined the effects of Wnt1 over-expression on H2O2-induced apoptosis in mCSCs and observed that Wnt1 significantly decreased H2O2-induced apoptosis in mCSCs. On the other hand, inhibition of the canonical Wnt pathway by the secreted frizzled related protein 2 (SFRP2) or knockdown of β-catenin in mCSCs reduced cells resistance to H2O2-induced apoptosis, suggesting that Wnt1 predominantly prevents H2O2-induced apoptosis through the canonical Wnt pathway. Conclusions Our results provide the first evidences that Wnt1 plays an important role in CSCs’ defenses against H2O2-induced apoptosis through the canonical Wnt1/GSK3β/β-catenin signaling pathway.


Scientific Reports | 2016

MiR218 Modulates Wnt Signaling in Mouse Cardiac Stem Cells by Promoting Proliferation and Inhibiting Differentiation through a Positive Feedback Loop.

Yongshun Wang; Jingjin Liu; Jinjin Cui; Meng Sun; Wenjuan Du; Tao Chen; Xing Ming; Lulu Zhang; Jiangtian Tian; Ji Li; Li Yin; Fang Liu; Zhongyue Pu; Bo Lv; Jingbo Hou

MiRNA expression was determined in both proliferating and differentiated cardiac stem cells (CSCs) through a comprehensive miRNA microarray analysis. We selected miR218 for functional follow-up studies to examine its significance in CSCs. First, we observed that the expression of miR218 was altered in CSCs during differentiation into cardiomyocytes, and transfection of an miR218 mimic or miR218 inhibitor affected the myocardial differentiation of CSCs. Furthermore, we observed that a negative regulator of Wnt signaling, sFRP2, was a direct target of miR218, and the protein levels of sFRP2 were increased in cells transfected with the synthetic miR218 inhibitor. In contrast, transfection with the miR218 mimic decreased the expression of sFRP2 and potentiated Wnt signaling. The subsequent down-regulation of sFRP2 by shRNA potentiated Wnt signaling, contributing to a gene expression program that is important for CSC proliferation and cardiac differentiation. Specifically, canonical Wnt signaling induced miR218 transcription. Thus, miR218 and Wnt signaling were coupled through a feed-forward positive feedback loop, forming a biological regulatory circuit. Together, these results provide the first evidence that miR218 plays an important role in CSC proliferation and differentiation through the canonical Wnt signaling pathway.


International Journal of Molecular Medicine | 2012

Trimetazidine improves right ventricular function by increasing miR-21 expression

Fang Liu; Li Yin; Lulu Zhang; Wenhua Liu; Jingjin Liu; Yongshun Wang

Trimetazidine (TMZ) improves left ventricular (LV) function and alleviates angina. TMZ is a metabolism-related drug, but its molecular actions and non-metabolic effects have not yet been elucidated. In this study, we investigated whether TMZ improves right ventricular (RV) function and decreases apoptosis in RV myocardial cells (RVMCs) by regulating miRNA-21 (miR-21) expression in vitro and in vivo. We used cultivated RVMCs and LV myocardial cells (LVMCs) and provided hypoxic stimulation for 24 h to induce MC apoptosis. RVMCs showed more severe apoptosis as indicated by terminal deoxynucleotidyl-transferase-mediated dUTP nick end-labeling (TUNEL) staining and caspase-3 protein and activity assays. The decrease in miR-21 expression was more dramatic in RVMCs. Subsequently, TMZ (10 µM) was added to the RVMCs prior to hypoxic stimulation. The TMZ-treated RVMCs showed less apoptosis and an increased expression of miR-21. The transfection of RVMCs with a miR-21-specific inhibitor weakened the protective effects of TMZ. To evaluate TMZ effectiveness in right heart failure, we used a combination treatment of hypoxia and the vascular endothelial growth factor receptor blocker, Su5416, to construct a stable model, and administered TMZ. TMZ improved RV function (as indicated by an increase in tricuspid annular plane systolic excursion), and inhibited fibrosis. TMZ also protects RVMCs againts apoptosis and increases miR-21 expression.


International Journal of Molecular Medicine | 2016

Macrophage migration inhibitory factor promotes cardiac stem cell proliferation and endothelial differentiation through the activation of the PI3K/Akt/mTOR and AMPK pathways

Jinjin Cui; Fengyun Zhang; Yongshun Wang; Jingjin Liu; Xing Ming; Jingbo Hou; Bo Lv; Shaohong Fang

Macrophage migration inhibitory factor (MIF) has pleiotropic immune functions in a number of inflammatory diseases. Recent evidence from expression and functional studies has indicated that MIF is involved in various aspects of cardiovascular disease. In this study, we aimed to determine whether MIF supports in vitro c-kit+CD45− cardiac stem cell (CSC) survival, proliferation and differentiation into endothelial cells, as well as the possible mechanisms involved. We observed MIF receptor (CD74) expression in mouse CSCs (mCSCs) using PCR and immunofluorescence staining, and MIF secretion by mCSCs using PCR and ELISA in vitro. Increasing amounts of exogenous MIF did not affect CD74 expression, but promoted mCSC survival, proliferation and endothelial differentiation. By contrast, treatment with an MIF inhibitor (ISO-1) or siRNA targeting CD74 (CD74-siRNA) suppressed the biological changes induced by MIF in the mCSCs. Increasing amounts of MIF increased the phosphorylation of Akt and mammalian target of rapamycin (mTOR), which are known to support cell survival, proliferation and differentiation. These effects of MIF on the mCSCs were abolished by LY294002 [a phosphoinositide 3-kinase (PI3K) inhibitor] and MK-2206 (an Akt inhibitor). Moreover, adenosine monophosphate-activated protein kinase (AMPK) phosphorylation increased following treatment with MIF. The AMPK inhibitor, compound C, partly blocked the pro-proliferative effects of MIF on the mCSCs. In conclusion, our results suggest that MIF promotes mCSC survival, proliferation and endothelial differentiation through the activation of the PI3K/Akt/mTOR and AMPK signaling pathways. Thus, MIF may prove to be a potential therapeutic factor in the treatment of heart failure and myocardial infarction by activating CSCs.


Journal of Vascular Research | 2017

Cables1 Inhibits Proliferation and Induces Senescence by Angiotensin II via a p21-Dependent Pathway in Human Umbilical Vein Endothelial Cells

Zhongyue Pu; Yongshun Wang; Xinxin Liu; Jingjin Liu; Jinjin Cui; Yan Wang; Bo Lv

Cables1 (Cdk5 and Abl enzyme substrate 1) is a vital cell cycle regulator and a candidate tumor suppressor that negatively regulates cell growth by inhibiting cyclin-dependent kinases. Here, we report on the critical role of the Cables1/p21 pathway, which inhibits cell proliferation and induces cell senescence in human umbilical vein endothelial cells. Moreover, we confirmed that silencing of Cables1 promoted cell proliferation as well as increased resistance to angiotensin II-induced senescence, at least in part, by altering Cables1 activation. We further demonstrated that knockdown of p21 reverses Cables1-mediated cell growth inhibition and cell senescence. Taken together, these results suggest that the Cables1/p21 pathway has a strong effect on the induction of cell senescence and inhibition of cell growth, and acts as a novel regulatory mechanism in which p21 is probably one of several downstream effector molecules to mediate Cables1.


Biochemical and Biophysical Research Communications | 2018

Fibroblast growth factor 19 improves cardiac function and mitochondrial energy homoeostasis in the diabetic heart

Wei Xu; Yongshun Wang; Yibo Guo; Jingjin Liu; Lijia Ma; Wei Cao; Yuhong Zhou

In diabetic cardiomyopathy, mitochondrial fatty acid oxidation dominates over mitochondrial glucose oxidation, leading to metabolic disturbances. Fibroblast growth factor 19 (FGF19) acts as a metabolic regulator and may have a cardioprotective role on diabetic cardiomyopathy. In this study, we investigated the effects of FGF19 on energy metabolism. FGF19 treatment of diabetic hearts exhibited higher glucose uptake and lower lipid profiles, suggesting changes in energy metabolism. The protective effects of FGF19 prevented ventricular dysfunction in diabetic hearts and improved mitochondrial function by the upregulation of PGC-1α expression. On the other side, knockdown of PGC-1α by siRNA attenuated the effects of FGF19 on the enhancement of mitochondrial function and energy efficiency. Taken together, these results show that FGF19 exhibited improved mitochondrial efficiency, which might be associated with higher cardiac contractility in diabetic hearts. It is also of note that modulation of PGC-1α, which is responsible for the activation by FGF19, may be a therapeutic target for diabetic cardiomyopathy.


Experimental and Therapeutic Medicine | 2017

ERK1/2 pathway regulates coxsackie and adenovirus receptor expression in mouse cardiac stem cells

Jingjin Liu; Qiang Sun; Yongshun Wang; Jinjin Cui; Maomao Zhang; Lili Li; Haibo Jia; Lulu Zhang; Bin Zhu; Miaomiao Jiang; Shuo Zhang

Cardiac stem cells (CSCs) are the most promising and effective candidates for the therapy of cardiac regenerative diseases; however, they have marked limitations. For instance, the implantation of CSCs is hampered by factors such as their sustainability and long-term durability. Gene modification appears to be the most effective method of optimizing CSCs and gene therapy trials have demonstrated that efficient gene transfer is key to achieving therapeutic efficacy. However, the transduction ability of adenovirus (Ad) is limited. Previous studies have reported that low expression of coxsackie and adenovirus receptor (CAR) in target cells decreases the transduction efficiency. A promising method for improving Ad-mediated gene transfer is to increase CAR expression in target cells. The present study investigated the effect of the Raf-mitogen-associated protein kinase (MAPK) kinase (MEK)-extracellular signal-associated protein kinase (ERK) signaling pathway on the expression of CAR on CSCs, as this pathway decreases cell-cell adhesion via cell surface molecules. The results demonstrated that interference with the Raf-MEK-ERK signaling pathway by knockdown of ERK1/2 upregulated the expression of CAR. The entry of the Ad into the cells was increased following inhibition of ERK1/2. Moreover, following knockdown of CAR, the entry of Ad into cells was decreased. However, knockdown of c-Jun N-terminal kinase and p38 as other components of the MAPK pathway did not affect CAR expression. Therefore, CAR expression in CSCs may be mediated via the Raf-MEK-ERK signaling pathway. Upregulation of CAR by knockdown of ERK1/2 may significantly improve Ad-mediated genetic modification of CSCs in the treatment of cardiovascular diseases.


Inflammation | 2013

Sca-1-positive cardiac stem cell migration in a cardiac infarction model.

Jingjin Liu; Yongshun Wang; Wenjuan Du

Collaboration


Dive into the Yongshun Wang's collaboration.

Top Co-Authors

Avatar

Jingjin Liu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Fang Liu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Jinjin Cui

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Lulu Zhang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Li Yin

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Wenhua Liu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Wenjuan Du

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Jingbo Hou

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Shuo Zhang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Xinxin Liu

Harbin Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge