Network


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

Hotspot


Dive into the research topics where Jialiang Liang is active.

Publication


Featured researches published by Jialiang Liang.


Stem Cells International | 2015

Exosomes Secreted from CXCR4 Overexpressing Mesenchymal Stem Cells Promote Cardioprotection via Akt Signaling Pathway following Myocardial Infarction

Kai Kang; Ruilian Ma; Wenfeng Cai; Wei Huang; Christian Paul; Jialiang Liang; Yuhua Wang; Tiejun Zhao; Ha Won Kim; Meifeng Xu; Ronald W. Millard; Zhili Wen; Yigang Wang

Background and Objective. Exosomes secreted from mesenchymal stem cells (MSC) have demonstrated cardioprotective effects. This study examined the role of exosomes derived from MSC overexpressing CXCR4 for recovery of cardiac functions after myocardial infarction (MI). Methods. In vitro, exosomes from MSC transduced with lentiviral CXCR4 (ExoCR4) encoding a silencing sequence or null vector were isolated and characterized by transmission electron microscopy and dynamic light scattering. Gene expression was then analyzed by qPCR and Western blotting. Cytoprotective effects on cardiomyocytes were evaluated and effects of exosomes on angiogenesis analyzed. In vivo, an exosome-pretreated MSC-sheet was implanted into a region of scarred myocardium in a rat MI model. Angiogenesis, infarct size, and cardiac functions were then analyzed. Results. In vitro, ExoCR4 significantly upregulated IGF-1α and pAkt levels and downregulated active caspase 3 level in cardiomyocytes. ExoCR4 also enhanced VEGF expression and vessel formation. However, effects of ExoCR4 were abolished by an Akt inhibitor or CXCR4 knockdown. In vivo, ExoCR4 treated MSC-sheet implantation promoted cardiac functional restoration by increasing angiogenesis, reducing infarct size, and improving cardiac remodeling. Conclusions. This study reveals a novel role of exosomes derived from MSCCR4 and highlights a new mechanism of intercellular mediation of stem cells for MI treatment.


PLOS ONE | 2014

MicroRNA-377 Regulates Mesenchymal Stem Cell-Induced Angiogenesis in Ischemic Hearts by Targeting VEGF

Zhili Wen; Wei Huang; Yuliang Feng; Wenfeng Cai; Yuhua Wang; Xiaohong Wang; Jialiang Liang; Mashhood Wani; Jing Chen; Pin Zhu; Jimei Chen; Ronald W. Millard; Guo-Chang Fan; Yigang Wang

MicroRNAs have been appreciated in various cellular functions, including the regulation of angiogenesis. Mesenchymal-stem-cells (MSCs) transplanted to the MI heart improve cardiac function through paracrine-mediated angiogenesis. However, whether microRNAs regulate MSC induced angiogenesis remains to be clarified. Using microRNA microarray analysis, we identified a microRNA expression profile in hypoxia-treated MSCs and observed that among all dysregulated microRNAs, microRNA-377 was decreased the most significantly. We also validated that vascular endothelial growth factor (VEGF) is a target of microRNA-377 using dual-luciferase reporter assay and Western-blotting. Knockdown of endogenous microRNA-377 promoted tube formation in human umbilical vein endothelial cells. We then engineered rat MSCs with lentiviral vectors to either overexpress microRNA-377 (MSCmiR-377) or knockdown microRNA-377 (MSCAnti-377) to investigate whether microRNA-377 regulated MSC-induced myocardial angiogenesis, using MSCs infected with lentiviral empty vector to serve as controls (MSCNull). Four weeks after implantation of the microRNA-engineered MSCs into the infarcted rat hearts, the vessel density was significantly increased in MSCAnti-377-hearts, and this was accompanied by reduced fibrosis and improved myocardial function as compared to controls. Adverse effects were observed in MSCmiR-377-treated hearts, including reduced vessel density, impaired myocardial function, and increased fibrosis in comparison with MSCNull-group. These findings indicate that hypoxia-responsive microRNA-377 directly targets VEGF in MSCs, and knockdown of endogenous microRNA-377 promotes MSC-induced angiogenesis in the infarcted myocardium. Thus, microRNA-377 may serve as a novel therapeutic target for stem cell-based treatment of ischemic heart disease.


PLOS ONE | 2012

Suicide Gene Reveals the Myocardial Neovascularization Role of Mesenchymal Stem Cells Overexpressing CXCR4 (MSCCXCR4)

Jialiang Liang; Wei Huang; Xi-Yong Yu; Atif Ashraf; Kishore K. Wary; Meifeng Xu; Ronald W. Millard; Muhammad Ashraf; Yigang Wang

Background Our previous studies indicated that MSCCXCR4 improved cardiac function after myocardial infarction (MI). This study was aimed to investigate the specific role of MSCCXCR4 in neovascularization of infarcted myocardium using a suicide gene approach. Methods MSCs were transduced with either lentivirus-null vector/GFP (MSCNull as control) or vector encoding for overexpressing CXCR4/GFP. The MSC derived-endothelial cell (EC) differentiation was assessed by a tube formation assay, Dil-ac-LDL uptake, EC marker expression, and VE-cadherin promoter activity assay. Gene expression was analyzed by quantitative RT-PCR or Western blot. The suicide gene approach was under the control of VE-cadherin promoter. In vivo studies: Cell patches containing MSCNull or MSCCXCR4 were transduced with suicide gene and implanted into the myocardium of MI rat. Rats received either ganciclovir (GCV) or vehicle after cell implantation. After one month, the cardiac functional changes and neovascularization were assessed by echocardiography, histological analysis, and micro-CT imaging. Results The expression of VEGF-A and HIF-1α was significantly higher in MSCCXCR4 as compared to MSCNull under hypoxia. Additionally, MSCCXCR4 enhanced new vessel formation and EC differentiation, as well as STAT3 phosphorylation under hypoxia. STAT3 participated in the transcription of VE-cadherin in MSCCXCR4 under hypoxia, which was inhibited by WP1066 (a STAT3 inhibitor). In addition, GCV specifically induced death of ECs with suicide gene activation. In vivo studies: MSCCXCR4 implantation promoted cardiac functional restoration, reduced infarct size, improved cardiac remodeling, and enhanced neovascularization in ischemic heart tissue. New vessels derived from MSCCXCR4 were observed at the injured heart margins and communicated with native coronary arteries. However, the derived vessel networks were reduced by GCV, reversing improvement of cardiac function. Conclusion The transplanted MSCCXCR4 enhanced neovascularization after MI by boosting release of angiogenic factors and increasing the potential of endothelial differentiation.


Stem Cells | 2017

Inhibition of microRNA‐495 Enhances Therapeutic Angiogenesis of Human Induced Pluripotent Stem Cells

Jialiang Liang; Wei Huang; Wenfeng Cai; Lei Wang; Linlin Guo; Christian Paul; Xi-Yong Yu; Yigang Wang

Therapeutic angiogenesis has emerged as a promising strategy to regenerate the damaged blood vessels resulting from ischemic diseases such as myocardial infarction (MI). However, the functional integration of implanted endothelial cells (ECs) in infarcted heart remains challenging. We herein develop an EC generation approach by inhibiting microRNA‐495 (miR‐495) in human induced pluripotent stem cells (hiPSCs) and assess the angiogenic potential for MI treatment. The anti‐angiogenic miR‐495 belonging to Dlk1‐Dio3 miR cluster was identified through expression profiling and computational analysis. Loss‐of‐function experiments for miR‐495 were performed using a lentiviral transfer of antisense sequence in hiPSCs. The pluripotency of hiPSCs was not impacted by the genetic modification. Induced with differentiation medium, miR‐495 inhibition enhanced the expression of EC genes of hiPSCs, as well as the yield of ECs. Newly derived ECs displayed prominent angiogenic characteristics including tube formation, cell migration, and proliferation. Mechanistically, miR‐495 mediated the expression of endothelial or angiogenic genes by directly targeting vascular endothelial zinc finger 1. After transplantation in immunodeficient MI mice, the derived ECs significantly increased neovascularization in the infarcted heart, prevented functional worsening, and attenuated expansion of infarct size. The functional integration of the implanted ECs into coronary networks was also enhanced by inhibiting miR‐495. miR‐495 represents a new target not only for promoting EC generation from hiPSCs but also for enhancing angiogenesis and engraftment of hiPSC‐derived ECs in ischemic heart. Stem Cells 2017;35:337–350


Cell Biochemistry and Function | 2017

Paracrine effect of CXCR4‐overexpressing mesenchymal stem cells on ischemic heart injury

Shizheng Wu; Ying-Lan Li; Wei Huang; Wenfeng Cai; Jialiang Liang; Christian Paul; Lin Jiang; Zhi-Chao Wu; Meifeng Xu; Ping Zhu; Yigang Wang

It has been reported that CXCR4‐overexpressing mesenchymal stem cells (MSCCX4) can repair heart tissue post myocardial infarction. This study aims to investigate the MSCCX4‐derived paracrine cardio‐protective signaling in the presence of myocardial infarction. Mesenchymal stem cells (MSCs) were divided into 3 groups: MSC only, MSCCX4, and CXCR4 gene‐specific siRNA‐transduced MSC. Mesenchymal stem cells were exposed to hypoxia, and then MSCs‐conditioned culture medium was incubated with neonatal and adult cardiomyocytes, respectively. Cell proliferation–regulating genes were assessed by real‐time polymerase chain reaction (RT‐PCR). In vitro: The number of cardiomyocytes undergoing DNA synthesis, cytokinesis, and mitosis was increased to a greater extent in MSCCX4 medium‐treated group than control group, while this proproliferative effect was reduced in CXCR4 gene‐specific siRNA‐transduced MSC–treated cells. Accordingly, the maximal enhancement of vascular endothelial growth factor, cyclin 2, and transforming growth factor‐β2 was observed in hypoxia‐exposed MSCCX4. In vivo: MSCs were labeled with enhanced green fluorescent protein (EGFP) and engrafted into injured myocardium in rats. The number of EGFP and CD31 positive cells in the MSCCX4 group was significantly increased than other 2 groups, associated with the reduced left ventricular (LV) fibrosis, the increased LV free wall thickness, the enhanced angiogenesis, and the improved contractile function. CXCR4 overexpression can mobilize MSCs into ischemic area, whereby these cells can promoted angiogenesis and alleviate LV remodeling via paracrine signaling mechanism.


Journal of Cellular and Molecular Medicine | 2015

CXCR4 attenuates cardiomyocytes mitochondrial dysfunction to resist ischaemia-reperfusion injury

Wenfeng Cai; Kai Kang; Wei Huang; Jialiang Liang; Yuliang Feng; Guan-Sheng Liu; Dehua Chang; Zhili Wen; Christian Paul; Meifeng Xu; Ronald W. Millard; Yigang Wang

The chemokine (C‐X‐C motif) receptor 4 (CXCR4) is expressed on native cardiomyocytes and can modulate isolated cardiomyocyte contractility. This study examines the role of CXCR4 in cardiomyocyte response to ischaemia‐reperfusion (I/R) injury. Isolated adult rat ventricular cardiomyocytes were subjected to hypoxia/reoxygenation (H/R) to simulate I/R injury. In response to H/R injury, the decrease in CXCR4 expression was associated with dysfunctional energy metabolism indicated by an increased adenosine diphosphate/adenosine triphosphate (ADP/ATP) ratio. CXCR4‐overexpressing cardiomyocytes were used to determine whether such overexpression (OE) can prevent bio‐energetic disruption‐associated cell death. CXCR4 OE was performed with adenoviral infection with CXCR4 encoding‐gene or non‐translated nucleotide sequence (Control). The increased CXCR4 expression was observed in cardiomyocytes post CXCR4‐adenovirus transduction and this OE significantly reduced the cardiomyocyte contractility under basal conditions. Although the same extent of H/R‐provoked cytosolic calcium overload was measured, the hydrogen peroxide‐induced decay of mitochondrial membrane potential was suppressed in CXCR4 OE group compared with control group, and the mitochondrial swelling was significantly attenuated in CXCR4 group, implicating that CXCR4 OE prevents permeability transition pore opening exposure to overload calcium. Interestingly, this CXCR4‐induced mitochondrial protective effect is associated with the enhanced signal transducer and activator of transcription 3 (expression in mitochondria. Consequently, in the presence of H/R, mitochondrial dysfunction was mitigated and cardiomyocyte death was decreased to 65% in the CXCR4 OE group as compared with the control group. I/R injury leads to the reduction in CXCR4 in cardiomyocytes associated with the dysfunctional energy metabolism, and CXCR4 OE can alleviate mitochondrial dysfunction to improve cardiomyocyte survival.


Circulation | 2016

Abstract 15792: Reprogramming of Fibroblasts Into Induced Cardiac Progenitor Cells With the CRISPR-dCas9 System

Lin Jiang; Jialiang Liang; Wei Huang; Wenfeng Cai; Christian Paul; Yigang Wang


Circulation | 2015

Abstract 13646: Electrical Stimulation Upregulates the miR-99 Family to Promote Cardiac Differentiation and Maturation of Human iPS Cells

Linlin Guo; Jialiang Liang; Wei Huang; Wenfeng Cai; Lei Wang; Jiapeng Wang; Christian Paul; Ronald W. Millard; Meifeng Xu; Yigang Wang


Circulation | 2014

Abstract 13222: Hax1 Overexpression Promotes Sca1 + Cardiac Stem Cell Proliferation and Protects Against Hypoxia-induced Apoptosis

Wenfeng Cai; Wei Huang; Sun Wook Kim; Jialiang Liang; Ruilian Ma; Hideyuki Kondo; Ha Won Kim; Meifeng Xu; Ronald W. Millard; Yigang Wang


Circulation | 2013

Abstract 10728: MicroRNA-377 Regulates Angiogenesis by Targeting VEGF: Implications for Mesenchymal Stem Cells Based Therapy in Ischemic Heart Disease

Zhili Wen; Wei Huang; Yuliang Feng; Yuhua Wang; Jialiang Liang; Wenfeng Cai; Kai Kang; Dehua Chang; Ping Zhu; Ronald W. Millard; Muhammad Ashraf; Yigang Wang

Collaboration


Dive into the Jialiang Liang's collaboration.

Top Co-Authors

Avatar

Wei Huang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Yigang Wang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meifeng Xu

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Wenfeng Cai

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Yuhua Wang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Kai Kang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Zhili Wen

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Muhammad Ashraf

China University of Geosciences

View shared research outputs
Top Co-Authors

Avatar

Atif Ashraf

University of Cincinnati

View shared research outputs
Researchain Logo
Decentralizing Knowledge