Network


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

Hotspot


Dive into the research topics where Xuewei Xia is active.

Publication


Featured researches published by Xuewei Xia.


PLOS ONE | 2013

The relative contribution of paracine effect versus direct differentiation on adipose-derived stem cell transplantation mediated cardiac repair.

Dezhong Yang; Wei Wang; Liangpeng Li; Yulan Peng; Peng Chen; Haiyun Huang; Yanli Guo; Xuewei Xia; Yuanyuan Wang; Hongyong Wang; Wei Eric Wang; Chunyu Zeng

Background Recent studies have demonstrated that transplantation of adipose-derived stem cell (ADSC) can improve cardiac function in animal models of myocardial infarction (MI). However, the mechanisms underlying the beneficial effect are not fully understood. In this study, we characterized the paracrine effect of transplanted ADSC and investigated its relative importance versus direct differentiation in ADSC transplantation mediated cardiac repair. Methodology/Principal Findings MI was experimentally induced in mice by ligation of the left anterior descending coronary artery. Either human ADSC, conditioned medium (CM) collected from the same amount of ADSC or control medium was injected into the peri-infarct region immediately after MI. Compared with the control group, both ADSC and ADSC-CM significantly reduced myocardial infarct size and improved cardiac function. The therapeutic efficacy of ADSC was moderately superior to ADSC-CM. ADSC-CM significantly reduced cardiomyocyte apoptosis in the infarct border zone, to a similar degree with ADSC treatment. ADSC enhanced angiogenesis in the infarct border zone, but to a stronger degree than that seen in the ADSC-CM treatment. ADSC was able to differentiate to endothelial cell and smooth muscle cell in post-MI heart; these ADSC-derived vascular cells amount to about 9% of the enhanced angiogenesis. No cardiomyocyte differentiated from ADSC was found. Conclusions ADSC-CM is sufficient to improve cardiac function of infarcted hearts. The therapeutic function of ADSC transplantation is mainly induced by paracrine-mediated cardioprotection and angiogenesis, while ADSC differentiation contributes a minor benefit by being involved in angiogenesis. Highlights 1 ADSC-CM is sufficient to exert a therapeutic potential. 2. ADSC was able to differentiate to vascular cells but not cardiomyocyte. 3. ADSC derived vascular cells amount to about 9% of the enhanced angiogenesis. 4. Paracrine effect is the major mechanism of ADSC therapeutic function for MI.


Circulation Research | 2013

Prolyl Hydroxylase Domain Protein 2 Silencing Enhances the Survival and Paracrine Function of Transplanted Adipose-Derived Stem Cells in Infarcted Myocardium

Wei Eric Wang; Dezhong Yang; Liangpeng Li; Wei Wang; Yulan Peng; Caiyu Chen; Peng Chen; Xuewei Xia; Hongyong Wang; Jiahui Jiang; Qiao Liao; Yuan Li; Ganfeng Xie; Haiyun Huang; Yanli Guo; Linda Ye; Dayue Darrel Duan; Xiongwen Chen; Steven R. Houser; Chunyu Zeng

Rationale: Transplantation of stem cells into damaged hearts has had modest success as a treatment for ischemic heart disease. One of the limitations is the poor stem cell survival in the diseased microenvironment. Prolyl hydroxylase domain protein 2 (PHD2) is a cellular oxygen sensor that regulates 2 key transcription factors involved in cell survival and inflammation: hypoxia-inducible factor and nuclear factor-&kgr;B. Objective: We studied whether and how PHD2 silencing in human adipose-derived stem cells (ADSCs) enhances their cardioprotective effects after transplantation into infarcted hearts. Methods and Results: ADSCs were transduced with lentiviral short hairpin RNA against prolyl hydroxylase domain protein 2 (shPHD2) to silence PHD2. ADSCs, with or without shPHD2, were transplanted after myocardial infarction in mice. ADSCs reduced cardiomyocyte apoptosis, fibrosis, and infarct size and improved cardiac function. shPHD2-ADSCs exerted significantly more protection. PHD2 silencing induced greater ADSC survival, which was abolished by short hairpin RNA against hypoxia-inducible factor-1&agr;. Conditioned medium from shPHD2-ADSCs decreased cardiomyocyte apoptosis. Insulin-like growth factor-1 (IGF-1) levels were significantly higher in the conditioned medium of shPHD2-ADSCs versus ADSCs, and depletion of IGF-1 attenuated the cardioprotective effects of shPHD2-ADSC–conditioned medium. Nuclear factor-&kgr;B activation was induced by shPHD2 to induce IGF-1 secretion via binding to IGF-1 gene promoter. Conclusions: PHD2 silencing promotes ADSCs survival in infarcted hearts and enhances their paracrine function to protect cardiomyocytes. The prosurvival effect of shPHD2 on ADSCs is hypoxia-inducible factor-1&agr; dependent, and the enhanced paracrine function of shPHD2-ADSCs is associated with nuclear factor-&kgr;B–mediated IGF-1 upregulation. PHD2 silencing in stem cells may be a novel strategy for enhancing the effectiveness of stem cell therapy after myocardial infarction.


Journal of Cellular Physiology | 2015

Mitochondrial DNA oxidative damage contributes to cardiomyocyte ischemia/reperfusion-injury in rats: cardioprotective role of lycopene.

Rongchuan Yue; Xuewei Xia; Jiahui Jiang; Dezhong Yang; Yu Han; Xiongwen Chen; Yue Cai; Liangpeng Li; Wei Eric Wang; Chunyu Zeng

Mitochondrial (mt) dysfunction and oxidative stress are involved in the pathogenesis of ischemia/reperfusion (I/R)‐injury. Lycopene, a lipophilic antioxidant found mainly in tomatoes and in other vegetables and fruits, can protect mtDNA against oxidative damage. However, the role of mtDNA in myocardial I/R‐injury is unclear. In the present study, we aimed to determine if and how lycopene protects cardiomyocytes from I/R‐injury. In both in vitro and in vivo studies, I/R‐injury increased mt 8‐hydroxyguanine (8‐OHdG) content, decreased mtDNA content and mtDNA transcription levels, and caused mitochondrial dysfunction in cardiomyocytes. These effects of I/R injury on cardiomycoytes were blocked by pre‐treatment with lycopene. MtDNA depletion alone was sufficient to induce cardiomyocyte death. I/R‐injury decreased the protein level of a key activator of mt transcription, mitochondrial transcription factor A (Tfam), which was blocked by lycopene. The protective effect of lycopene on mtDNA was associated with a reduction in mitochondrial ROS production and stabilization of Tfam. In conclusion, lycopene protects cardiomyocytes from the oxidative damage of mtDNA induced by I/R‐injury. J. Cell. Physiol. 230: 2128–2141, 2015.


Circulation | 2017

Dedifferentiation, Proliferation, and Redifferentiation of Adult Mammalian Cardiomyocytes After Ischemic Injury

Wei Eric Wang; Liangpeng Li; Xuewei Xia; Wenbin Fu; Qiao Liao; Cong Lan; Dezhong Yang; Hongmei Chen; Rongchuan Yue; C. Zeng; Lin Zhou; Bin Zhou; Dayue Darrel Duan; Xiongwen Chen; Steven R. Houser; Chunyu Zeng

Background: Adult mammalian hearts have a limited ability to generate new cardiomyocytes. Proliferation of existing adult cardiomyocytes (ACMs) is a potential source of new cardiomyocytes. Understanding the fundamental biology of ACM proliferation could be of great clinical significance for treating myocardial infarction (MI). We aim to understand the process and regulation of ACM proliferation and its role in new cardiomyocyte formation of post-MI mouse hearts. Methods: &bgr;-Actin-green fluorescent protein transgenic mice and fate-mapping Myh6-MerCreMer-tdTomato/lacZ mice were used to trace the fate of ACMs. In a coculture system with neonatal rat ventricular myocytes, ACM proliferation was documented with clear evidence of cytokinesis observed with time-lapse imaging. Cardiomyocyte proliferation in the adult mouse post-MI heart was detected by cell cycle markers and 5-ethynyl-2-deoxyuridine incorporation analysis. Echocardiography was used to measure cardiac function, and histology was performed to determine infarction size. Results: In vitro, mononucleated and bi/multinucleated ACMs were able to proliferate at a similar rate (7.0%) in the coculture. Dedifferentiation proceeded ACM proliferation, which was followed by redifferentiation. Redifferentiation was essential to endow the daughter cells with cardiomyocyte contractile function. Intercellular propagation of Ca2+ from contracting neonatal rat ventricular myocytes into ACM daughter cells was required to activate the Ca2+-dependent calcineurin-nuclear factor of activated T-cell signaling pathway to induce ACM redifferentiation. The properties of neonatal rat ventricular myocyte Ca2+ transients influenced the rate of ACM redifferentiation. Hypoxia impaired the function of gap junctions by dephosphorylating its component protein connexin 43, the major mediator of intercellular Ca2+ propagation between cardiomyocytes, thereby impairing ACM redifferentiation. In vivo, ACM proliferation was found primarily in the MI border zone. An ischemia-resistant connexin 43 mutant enhanced the redifferentiation of ACM-derived new cardiomyocytes after MI and improved cardiac function. Conclusions: Mature ACMs can reenter the cell cycle and form new cardiomyocytes through a 3-step process: dedifferentiation, proliferation, and redifferentiation. Intercellular Ca2+ signal from neighboring functioning cardiomyocytes through gap junctions induces the redifferentiation process. This novel mechanism contributes to new cardiomyocyte formation in post-MI hearts in mammals.


Stem Cell Research & Therapy | 2017

Metformin promotes the survival of transplanted cardiosphere-derived cells thereby enhancing their therapeutic effect against myocardial infarction

Rongchuan Yue; Wenbin Fu; Xiang Liao; Cong Lan; Qiao Liao; Liangpeng Li; Dezhong Yang; Xuewei Xia; Xiongwen Chen; Chunyu Zeng; Wei Eric Wang

BackgroundTransplantation of cardiosphere-derived cells (CDCs) has been shown to exert a therapeutic effect in patients with myocardial infarction (MI). However, poor survival of transplanted CDCs limits their beneficial effect. Metformin (MET) activates AMP-activated protein kinase (AMPK) which is associated with cell survival. The aim of this study is to determine whether MET improves CDC survival in the transplantation microenvironment and enhances the therapeutic effect of CDC transplantation against MI.MethodsCDCs were isolated and expanded from transgenic β-actin-GFP mice. CDCs were pretreated with MET and intramyocardially injected into wild-type C57 mouse heart with MI injury. The survival of CDCs was quantified, and the infarct size and cardiac function of treated hearts were evaluated.ResultsCDC transplantation modestly reduced infarct size and improved cardiac function in the post-MI heart, which was further improved by MET treatment. MET pretreatment significantly increased the survival of CDCs transplanted into the myocardium. MET also reduced CDC apoptosis induced by oxidative stress in vitro. The anti-apoptotic effect of MET was blocked by the AMPK inhibitor compound C. MET increased AMPK phosphorylation and upregulated endothelial nitric oxide synthase (eNOS) in CDCs under oxidative stress, which might be associated with the anti-apoptotic effect of MET.ConclusionsMET improves the survival of transplanted CDCs in the myocardium, thereby enhancing their therapeutic effect against MI injury. The pro-survival function of MET on CDCs might be associated with an AMPK-eNOS-dependent mechanism.


Clinical Science | 2017

Therapeutic Effect of a Novel Wnt Pathway Inhibitor on Cardiac Regeneration after Myocardial Infarction

Dezhong Yang; Wenbin Fu; Liangpeng Li; Xuewei Xia; Qiao Liao; Rongchuan Yue; Hongmei Chen; Xiongwen Chen; Songzhu An; Chunyu Zeng; Wei Eric Wang

After myocardial infarction (MI), the heart is difficult to repair because of great loss of cardiomyoctyes and lack of cardiac regeneration. Novel drug candidates that aim at reducing pathological remodeling and stimulating cardiac regeneration are highly desirable. In the present study, we identified if and how a novel porcupine inhibitor CGX1321 influenced MI and cardiac regeneration. Permanent ligation of left anterior descending (LAD) coronary artery was performed in mice to induce MI injury. Cardiac function was measured by echocardiography, infarct size was examined by TTC staining. Fibrosis was evaluated with Massons trichrome staining and vimentin staining. As a result, CGX1321 administration blocked the secretion of Wnt proteins, and inhibited both canonical and non-canonical Wnt signaling pathways. CGX1321 improved cardiac function, reduced myocardial infarct size, and fibrosis of post-MI hearts. CGX1321 significantly increased newly formed cardiomyocytes in infarct border zone of post-MI hearts, evidenced by the increased EdU+ cardiomyocytes. Meanwhile, CGX1321 increased Ki67+ and phosphohistone H3 (PH3+) cardiomyocytes in culture, indicating enhanced cardiomyocyte proliferation. The mRNA microarray showed that CGX1321 up-regulated cell cycle regulating genes such as Ccnb1 and Ccne1 CGX1321 did not alter YAP protein phosphorylation and nuclear translocation in cardiomyocytes. In conclusion, porcupine inhibitor CGX1321 reduces MI injury by limiting fibrosis and promoting regeneration. It promotes cardiomyocyte proliferation by stimulating cell cycle regulating genes with a Hippo/YAP-independent pathway.


Circulation Research | 2013

PHD2 Silencing Enhances the Survival and Paracrine Function of Transplanted Adipose-Derived Stem Cells in Infarcted Myocardium

Wei Eric Wang; Dezhong Yang; Liangpeng Li; Wei Wang; Yulan Peng; Caiyu Chen; Chen Peng; Xuewei Xia; Hongyong Wang; Jiahui Jiang; Qiao Liao; Yuan Li; Ganfeng Xie; Haiyun Huang; Yanli Guo; Linda Ye; Dayue Darrel Duan; Xiongwen Chen; Steven R. Houser; Chunyu Zeng

Rationale: Transplantation of stem cells into damaged hearts has had modest success as a treatment for ischemic heart disease. One of the limitations is the poor stem cell survival in the diseased microenvironment. Prolyl hydroxylase domain protein 2 (PHD2) is a cellular oxygen sensor that regulates 2 key transcription factors involved in cell survival and inflammation: hypoxia-inducible factor and nuclear factor-&kgr;B. Objective: We studied whether and how PHD2 silencing in human adipose-derived stem cells (ADSCs) enhances their cardioprotective effects after transplantation into infarcted hearts. Methods and Results: ADSCs were transduced with lentiviral short hairpin RNA against prolyl hydroxylase domain protein 2 (shPHD2) to silence PHD2. ADSCs, with or without shPHD2, were transplanted after myocardial infarction in mice. ADSCs reduced cardiomyocyte apoptosis, fibrosis, and infarct size and improved cardiac function. shPHD2-ADSCs exerted significantly more protection. PHD2 silencing induced greater ADSC survival, which was abolished by short hairpin RNA against hypoxia-inducible factor-1&agr;. Conditioned medium from shPHD2-ADSCs decreased cardiomyocyte apoptosis. Insulin-like growth factor-1 (IGF-1) levels were significantly higher in the conditioned medium of shPHD2-ADSCs versus ADSCs, and depletion of IGF-1 attenuated the cardioprotective effects of shPHD2-ADSC–conditioned medium. Nuclear factor-&kgr;B activation was induced by shPHD2 to induce IGF-1 secretion via binding to IGF-1 gene promoter. Conclusions: PHD2 silencing promotes ADSCs survival in infarcted hearts and enhances their paracrine function to protect cardiomyocytes. The prosurvival effect of shPHD2 on ADSCs is hypoxia-inducible factor-1&agr; dependent, and the enhanced paracrine function of shPHD2-ADSCs is associated with nuclear factor-&kgr;B–mediated IGF-1 upregulation. PHD2 silencing in stem cells may be a novel strategy for enhancing the effectiveness of stem cell therapy after myocardial infarction.


Biochimica et Biophysica Acta | 2018

A novel porcupine inhibitor blocks WNT pathways and attenuates cardiac hypertrophy

Jiahui Jiang; Cong Lan; Liangpeng Li; Dezhong Yang; Xuewei Xia; Qiao Liao; Wenbin Fu; Xiongwen Chen; Songzhu An; Wei Eric Wang; Chunyu Zeng

WNT pathways are critically involved in the cardiac hypertrophy growth. Porcupine, an acyltransferase that specifically enables secretion of all WNT ligands, became a highly druggable target for inhibiting WNT pathways. Here we test if a novel small-molecule porcupine inhibitor CGX1321, which has entered human clinical trials as an anti-cancer agent, exerts an anti-hypertrophic effect. Transverse aortic constriction (TAC) was performed to induce cardiac hypertrophy on four-month-old male C57 mice. Cardiac function was measured with echocardiography. Histological analysis was performed to detect cardiomyocyte size and molecular expressions. CGX1321 was administrated daily for 4 weeks post TAC injury. As a result, CGX1321 improved cardiac function and animal survival of post-TAC mice. CGX1321 significantly reduced cardiomyocyte hypertrophy, cardiomyocyte apoptosis and fibrosis induced by TAC injury. CGX1321 significantly inhibited TAC induced nuclear translocation of β-catenin and the elevation of Frizzled-2, cyclin-D1 and c-myc expression, indicating its inhibitory effect on canonical WNT pathway. Furthermore, CGX1321 inhibited TAC induced nuclear translocation of nuclear factor of activated T-cells and the elevation of phosphorylated c-Jun expression, suggesting its inhibitory function on non-canonical WNT pathway. We conclude that CGX1321 inhibits both canonical and non-canonical WNT pathways, and attenuates cardiac hypertrophy. Our findings support the porcupine inhibitors as a class of new drugs to be potentially used for treating patients with cardiac hypertrophy.


Heart | 2015

ASSA14-03-14 Mitochondrial DNA damage contribute to ischemia/reperfusion-injury in rat cardiac myocytes: the protective effects of lycopene

Rongchuan Yue; Wei Eric Wang; Xuewei Xia; Jiahui Jiang; Dezhong Yang; Yu Han; C. Zeng

Objectives Recent studies suggest that oxidative stress and mitochondrial dysfunction are involved in the pathogenesis of ischemia/reperfusion (I/R)-injury. Mitochondrial DNA (mtDNA) is highly vulnerable to oxidative stress and lycopene is found to protect mtDNA against oxidative damage. Our recent study indicates lycopene reduced I/R-injury in vitro by alleviating oxidative stress and preventing mitochondrial dysfunction. This study was aimed to determine whether mtDNA damage is involved in the I/R-injury and whether lycopene can protect cardiac myocytes from I/R-injury by inhibiting mtDNA damage. Methods We established I/R-injury model with rat in vivo and we also established hypoxia/ reoxygenation-injury model with H9c2 cells to simulate I/R-injury in vitro. Reactive oxygen species (ROS) and mitochondrial superoxide levels were determined. Mitochondrial 8-hydroxyguanine (8-OHdG), mtDNA content and mtDNA transcript levels were detected to find out if mtDNA were damaged; The protein expression of mitochondrial transcription factor A (Tfam) in mitochondrial, a key protein for mtDNA transcription, replication and component for nucleoid organisation were also determined by western blot. Results I/R significantly increased reactive oxygen species (ROS) production and mitochondrial superoxide levels. In addition, I/R increased mitochondrial 8-hydroxyguanine (8-OHdG) content, while reduced mtDNA content and mtDNA transcript levels. Consistent with these findings, I/R was found to decrease the protein expression of Tfam in mitochondrial. Lycopene pretreatment efficiently attenuated the oxidative damage to mtDNA induced by I/R both in vivo and in vitro. Conclusion Our results suggest that mtDNA damage may account for I/R-injury. Lycopene has a great pharmacological potential in protecting mtDNA against the I/R-injury in the heart.


Circulation Research | 2013

Prolyl Hydroxylase Domain Protein 2 Silencing Enhances the Survival and Paracrine Function of Transplanted Adipose-Derived Stem Cells in Infarcted MyocardiumNovelty and Significance

Wei Eric Wang; Dezhong Yang; Liangpeng Li; Wei Wang; Yulan Peng; Caiyu Chen; Peng Chen; Xuewei Xia; Hongyong Wang; Jiahui Jiang; Qiao Liao; Yuan Li; Ganfeng Xie; Haiyun Huang; Yanli Guo; Linda Ye; Dayue Darrel Duan; Xiongwen Chen; Steven R. Houser; Chunyu Zeng

Rationale: Transplantation of stem cells into damaged hearts has had modest success as a treatment for ischemic heart disease. One of the limitations is the poor stem cell survival in the diseased microenvironment. Prolyl hydroxylase domain protein 2 (PHD2) is a cellular oxygen sensor that regulates 2 key transcription factors involved in cell survival and inflammation: hypoxia-inducible factor and nuclear factor-&kgr;B. Objective: We studied whether and how PHD2 silencing in human adipose-derived stem cells (ADSCs) enhances their cardioprotective effects after transplantation into infarcted hearts. Methods and Results: ADSCs were transduced with lentiviral short hairpin RNA against prolyl hydroxylase domain protein 2 (shPHD2) to silence PHD2. ADSCs, with or without shPHD2, were transplanted after myocardial infarction in mice. ADSCs reduced cardiomyocyte apoptosis, fibrosis, and infarct size and improved cardiac function. shPHD2-ADSCs exerted significantly more protection. PHD2 silencing induced greater ADSC survival, which was abolished by short hairpin RNA against hypoxia-inducible factor-1&agr;. Conditioned medium from shPHD2-ADSCs decreased cardiomyocyte apoptosis. Insulin-like growth factor-1 (IGF-1) levels were significantly higher in the conditioned medium of shPHD2-ADSCs versus ADSCs, and depletion of IGF-1 attenuated the cardioprotective effects of shPHD2-ADSC–conditioned medium. Nuclear factor-&kgr;B activation was induced by shPHD2 to induce IGF-1 secretion via binding to IGF-1 gene promoter. Conclusions: PHD2 silencing promotes ADSCs survival in infarcted hearts and enhances their paracrine function to protect cardiomyocytes. The prosurvival effect of shPHD2 on ADSCs is hypoxia-inducible factor-1&agr; dependent, and the enhanced paracrine function of shPHD2-ADSCs is associated with nuclear factor-&kgr;B–mediated IGF-1 upregulation. PHD2 silencing in stem cells may be a novel strategy for enhancing the effectiveness of stem cell therapy after myocardial infarction.

Collaboration


Dive into the Xuewei Xia's collaboration.

Top Co-Authors

Avatar

Chunyu Zeng

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Dezhong Yang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Liangpeng Li

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Eric Wang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Qiao Liao

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiahui Jiang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Rongchuan Yue

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wenbin Fu

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge