Network


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

Hotspot


Dive into the research topics where Xiaoxiang Tian is active.

Publication


Featured researches published by Xiaoxiang Tian.


Journal of Molecular and Cellular Cardiology | 2010

Cellular repressor of E1A-stimulated genes inhibits human vascular smooth muscle cell apoptosis via blocking P38/JNK MAP kinase activation

Yaling Han; Guangzhe Wu; Jie Deng; Jie Tao; Liang Guo; Xiaoxiang Tian; Jian Kang; Xiaolin Zhang; Chenghui Yan

Vascular smooth muscle cell (VSMC) apoptosis accelerates atherosclerosis and promotes restenosis following vascular injury. The current study examined the effects of cellular repressor of E1A-stimulated genes (CREG), a novel glycoprotein inhibiting transcription activation, on the regulation of VSMC apoptosis. Serum starvation or treatment of human VSMCs with apoptosis inducers (STS or VP-16) significantly reduced CREG expression and caused caspase-3 activation. CREG downregulation and caspase-3 activation were inversely related, suggesting that reduced CREG expression may contribute to VSMC apoptosis. Both loss-of-function (CREG-DW produced by retroviruses expressing CREG shRNAs) and gain-of-function (CREG-UP produced by retroviral infection with vector pLNCX-CREG) studies were performed to confirm this hypothesis. CREG-DW significantly increased VSMC apoptosis, whereas CREG-UP significantly reduced apoptosis. Moreover, p38 and JNK mitogen-activated protein kinases were significantly upregulated in CREG-DW and significantly reduced in CREG-UP VSMCs. More importantly, CREG-DW-induced VSMC apoptosis was blocked by the p38-specific inhibitor SB203580 or by overexpression of a dominant-negative P38 alpha (p38 alpha AGF). Balloon injury-induced vascular caspase-3 activation was significantly inhibited by treatment with recombinant CREG protein. These results demonstrated for the first time that CREG plays a key role in modulating VSMC apoptosis through the p38 and JNK signal transduction pathways, both in vitro and in situ.


Apoptosis | 2010

Overexpressing cellular repressor of E1A-stimulated genes protects mesenchymal stem cells against hypoxia- and serum deprivation-induced apoptosis by activation of PI3K/Akt

Jie Deng; Yaling Han; Chenhui Yan; Xiaoxiang Tian; Jie Tao; Jian Kang; Shaohua Li

Bone marrow-derived mesenchymal stem cells (MSCs) have great potential for repair after myocardial infarction. However, poor viability of transplanted MSCs in the ischemic heart has limited their therapeutic potential. Cellular repressor of E1A-stimulated genes (CREG) has been identified as a potent inhibitor of apoptosis. The aim of this study was to investigate the anti-apoptotic effects of CREG on MSCs under hypoxic and serum deprivation (SD) conditions. We also investigated the potential mechanism(s) that may mediate the actions of CREG. All experiments were performed on rat bone marrow MSCs. Apoptosis was induced by exposure of cells to hypoxia/SD in a sealed GENbox hypoxic chamber. Effects of CREG were investigated in the absence or presence of inhibitors that target phosphoinositide 3-kinase (PI3K). We found that the overexpression of CREG markedly protected MSCs from hypoxia/SD-induced apoptosis through inhibition of the mitochondrial apoptotic pathway, leading to attenuation of caspase-3. Moreover, CREG enhanced Akt phosphorylation and decreased the expression of p53 in MSCs under hypoxic/SD conditions. The PI3K/Akt inhibitor LY294002 significantly increased the amount of p53 protein and attenuated the anti-apoptotic effects of CREG on MSCs. This study indicates that CREG is a novel and potent survival factor for MSCs, therefore, it may be a useful therapeutic adjunct for transplanting MSCs into damaged heart after myocardial infarction.


Journal of Physiological Sciences | 2010

Endogenous transforming growth factor (TGF) beta1 promotes differentiation of smooth muscle cells from embryonic stem cells: stable plasmid-based siRNA silencing of TGF beta1 gene expression

Yaling Han; Na Li; Xiaoxiang Tian; Jian Kang; Chenghui Yan; Yanmei Qi

Transforming growth factor (TGF) beta1 has been shown to promote differentiation of smooth muscle cells (SMC) from some precursor cells. Whether endogenous TGF beta1 also contributes to SMC differentiation during embryogenesis, however, remains unclear. In this study, a plasmid-based TGF beta1 RNA interference embryonic stem (ES) cell line was constructed. Morphological observation showed that TGF beta1 knockdown significantly prevented differentiated cells from outgrowing from ES cells-derived embryoid bodies (EBs). Immunofluorescence staining indicated that SM alpha-actin-positive cells were confluent and dense in the control group but dispersed in the TGF beta1 knockdown group. RT-PCR and western blot suggested that TGF beta1 knockdown resulted in a decrease in the expression of early SMC markers SM alpha-actin and myocardin in EBs. Both the retarded extension of cell outgrowth and the decrease in SM alpha-actin and myocardin expression could not be rescued by addition of exogenous TGF beta1. These data suggest that endogenous TGF beta1 promotes differentiation of SMC from ES cells.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Contribution of Homeostatic Chemokines CCL19 and CCL21 and Their Receptor CCR7 to Coronary Artery Disease

Wenzhi Cai; Jie Tao; Xiaolin Zhang; Xiaoxiang Tian; Tengfei Liu; Xueyao Feng; Jing Bai; Chenghui Yan; Yaling Han

Objective— Our aim was to identify the role of the homeostatic chemokines CCL19 and CCL21 and their common receptor CCR7 in atherogenesis and to study the relationships between CCL19, CCL21, and CCR7 gene variants and coronary artery disease in a Chinese Han population. Approach and Results— Immunohistochemical analysis of samples with atherosclerosis of various stages showed increased CCL19, CCL21, and CCR7 expression in atherosclerotic coronary plaques compared with nonatherosclerotic controls. Expression levels increased in positive correlation with coronary lesion stage. Cell adhesion assays confirmed that CCL19 promoted monocyte adhesion, which was induced by CCR7, to human umbilical vein endothelial cells, an effect partially antagonized by atorvastatin. After the human umbilical vein endothelial cells were treated with CCR7-neutralizing antibody, both CCL19- and CCL21-induced monocyte to human umbilical vein endothelial cell migration and CCL19-induced monocyte to human umbilical vein endothelial cell adhesion were abolished. The associations between genetic variants of CCL19, CCL21, CCR7, and coronary artery disease in a Chinese Han population were determined by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. The following single nucleotide polymorphisms were associated with coronary artery disease: CCL19 rs2227302, CCL21 rs2812377, and CCR7 rs588019. Individuals with the CCL19 rs2227302 T allele or CCL21 rs2812377 G allele had higher plasma CCL19 levels than those with C/C genotype and higher CCL21 levels than those with T/T genotype in both case and control subjects. Conclusion— CCL19/CCL21–CCR7 is a novel homeostatic chemokine system that modulates human monocyte adhesion and migration, promoting atherogenesis. It is associated with coronary artery disease risk in Chinese Han individuals. These data suggest that the CCL19/CCL21–CCR7 axis plays an important role in atherosclerosis progression.


Atherosclerosis | 2012

Cellular repressor E1A-stimulated genes controls phenotypic switching of adventitial fibroblasts by blocking p38MAPK activation

Yang Li; Jie Tao; Jian Zhang; Xiaoxiang Tian; Shaowei Liu; Mingyu Sun; Xiaolin Zhang; Chenghui Yan; Yaling Han

AIMS Phenotypic modulation of adventitial fibroblasts (AFs) plays an important role in the pathogenesis of proliferative vascular diseases. The current study aimed to identify the role of cellular repressor E1A-stimulated genes (CREG), a critical mediator in the maintenance of vascular homeostasis, in AF phenotypic modulation and adventitial remodeling. METHOD AND RESULTS Using in situ double-immunofluorescence staining, we ascertained that CREG expression was significantly down-regulated in the adventitia after vascular injury, and its expression pattern was conversely correlated with the expression of smooth muscle α-actin (α-SMA), a marker for differentiation of AFs into myofibroblasts. In vitro data confirmed the association of CREG in angiotensin II (Ang II)-induced AF differentiation. Additionally, overexpression of CREG attenuated Ang II-induced α-SMA expression in AFs. CREGoverexpressing AFs showed decreased levels of proliferation on days 2-5 following stimulation by Ang II compared with controls, with changes in the cell cycle profile as shown by BrdU incorporation assay and fluorescence activated cell sorting analysis. Moreover, wound healing assay and transwell migration model demonstrated that upregulation of CREG expression inhibited Ang II-induced AF migration. We found that CREG-mediated its counterbalancing effects in Ang II-induced phenotypic modulation, proliferation and migration by inhibition of the p38MAPK signaling pathway, validated by pharmacological blockade of p38MAPK with SB 203580 and by overexpression of p38MAPK with transfectants expressing constitutively active p38αMAPK. CONCLUSION Our findings suggest that CREG is a novel AF phenotypic modulator in a p38MAPK-dependent manner. Modulating CREG on the local vascular wall may become a new therapeutic target against proliferative vascular diseases.


Biochimica et Biophysica Acta | 2017

CREG protects from myocardial ischemia/reperfusion injury by regulating myocardial autophagy and apoptosis

Haixu Song; Chenghui Yan; Xiaoxiang Tian; Nan Zhu; Yang Li; Dan Liu; Yanxia Liu; Meili Liu; Chengfei Peng; Quanyu Zhang; Erhe Gao; Yaling Han

AIMS Human cellular repressor of E1A-stimulated genes (CREG) is a secreted glycoprotein that regulates tissue and cell homeostasis and has been shown to antagonize heart fibrosis, which indicates a potential protective effect of CREG against cardiomyocyte chronic damage. However, little is known about the role of CREG in myocardial tissue acute injury, in this study, we aimed to investigate the role of CREG in myocardial ischemia/reperfusion (MI/R) injury and clarify the mechanism of action. METHODS AND RESULTS Wild-type Creg (Creg+/+), heterozygous Creg (Creg+/-) mice and mice pretreated with infusion of recombinant 0.3mg/kg·d CREG protein (reCreg+/+) were subjected to 30min of left ascending coronary ischemia and 24h of reperfusion. Evans Blue-triphenyl- tetrazolium chloride (TTC) solution and echocardiography analysis were used to evaluate the effects of CREG on MI/R mice. The underlying mechanisms were further determined by cultured myocardial cells in vitro. Our findings revealed that the level of CREG protein in mouse hearts was significantly decreased after mice were subjected to MI/R. Moreover, Creg+/- mice had larger infarction size 2h after reperfusion and worse cardiac function 28days after MI/R injury compared to that in Creg+/+ mice. However, reCreg+/+ mice could maintain CREG at a high level even after MI/R injury, and mitigated infarction size and improved cardiac function significantly. In Creg+/- mice, myocardial autophagy was dysfunctional characterized by accumulation of LC3A and p62, while apoptotic cell number increase was detected by cleaved caspase-3 blotting and TUNEL staining. Conversely, decreased apoptosis and activated autophagy were detected in reCreg+/+ mice. Furthermore, chloroquine, a kind of autophagy blocker, was used to demonstrate recombinant CREG protected cardiomyocytes against apoptosis mediated by activating autophagy both in vivo and in vitro. Finally, we found CREG was involved into lysosomal protein transfer and improve cellular autophagy. CONCLUSION CREG protects heart against MI/R injury-induced cardiomyocytes apoptosis by activating lysosomal autophagy. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren and Megan Yingmei Zhang.


International Journal of Cardiology | 2015

Cellular repressor of E1A-stimulated gene overexpression in bone mesenchymal stem cells protects against rat myocardial infarction

Chengfei Peng; Haifeng Pei; Feipeng Wei; Xiaoxiang Tian; Jie Deng; Chenghui Yan; Yang Li; Mingyu Sun; Jian Zhang; Dan Liu; Jingjing Rong; Jie Wang; Erhe Gao; Shaohua Li; Yaling Han

BACKGROUND Bone mesenchymal stem cell (BMSC) therapy has modest success in ischemic heart disease but has been limited by poor survival in diseased microenvironments. Cellular repressor of E1A-stimulated genes (CREG) can prevent BMSCs from apoptosis in vitro; however, the effects of CREG-modified BMSCs on ischemic heart disease and the related mechanism remain undefined. Therefore, we designed to study the cardioprotective effects of CREG overexpression in BMSCs ((CREG)BMSCs) after transplantation into infarcted heart of rats. METHODS In vivo studies, 50 μl PBS or 1.5×10(6)(Norm)BMSCs, (GFP)BMSCs or (CREG)BMSCs were implanted intramyocardially in myocardial infarction rat models. 3 or 14 days later, cardiac function, fibrosis, apoptosis and angiogenesis were analyzed by echocardiography, masson, western blot and immunofluorescence staining, respectively. ELISA, western blot and matrigel assay were used in vitro to detect vascular endothelial growth factor (VEGF) secretion, signaling molecule expression, and angiogenic tube formation. RESULTS In vivo, prolonged cardiac function (14d LVEF: 50.87 ± 0.94%; LVFS: 23.41 ± 1.12%), decreased fibrosis (14d Fibrotic area: 27.37 ± 1.03%) and apoptosis and increased angiogenesis were observed in (CREG)BMSCs, compared with other groups. In vivo and in vitro, VEGF secretion from (CREG)BMSCs was markedly enhanced. In vitro, angiogenic tube formation in (CREG)BMSC supernatants significantly increased. Moreover, CREG activated hypoxia-inducible factor-1α (HIF-1α), but not HIF-1β. Knockdown of HIF-1α with siRNA decreased VEGF secretion and angiogenic tube formation. Notably, CREG did not influence HIF-1α mRNA synthesis but inhibited the expression of Von Hippel-Lindau (VHL), a key protein that regulates HIF-1α degradation. CONCLUSIONS The (CREG)BMSC transplantation, directly or indirectly, may promote VEGFs anti-apoptosis and angiogenesis via the inhibition of VHL-mediated HIF-1α degradation, consequently protecting against myocardial infarction.


Journal of Molecular and Cellular Cardiology | 2015

Cellular repressor of E1A-stimulated genes inhibits inflammation to decrease atherosclerosis in ApoE−/− mice

Mingyu Sun; Xiaoxiang Tian; Yanxia Liu; Nan Zhu; Yang Li; Guitang Yang; Chengfei Peng; Chenghui Yan; Yaling Han

AIMS Macrophage inflammation response is important in the pathogenesis of atherosclerosis. We investigated the role and mechanism of cellular repressor of E1A-stimulated genes (CREG) in regulating TNF-α induced inflammation response in macrophages and explore whether CREG might be a therapeutic target for atherosclerosis. METHOD AND RESULTS Immunostaining and western blotting showed that expression of CREG was reduced in human atherosclerotic coronary artery. In vivo experiments demonstrated that supplementation of recombinant CREG protein to ApoE(-/-) mice fed with high fat diet alleviated aortic atherosclerosis development and inflammation. In vitro, macrophage from ApoE(-/-) mice fed with high fat diet had lower level of CREG compared to control mice fed with normal diet. Immunohistochemical staining and western blotting further confirmed that CREG inhibited inflammatory response of macrophages induced by TNF-α. Supplementation of exogenous recombinant CREG protein or CREG gene silencing showed that CREG promoted autophagy in TNF-α treated macrophages. The use of autophagy inhibitors, 3-methyladenine and bafilomycin A, identified that CREG attenuated TNF-α induced inflammation by activate autophagy. In addition, supplementation of exogenous CREG protein stimulated expression and maturity of cathepsin B and cathepsin L and induced lysosome formation, whereas CREG deficiency reduced lysosomal formation. CONCLUSION CREG inhibits inflammation and promotes autophagy mediated by lysosome formation; it might be a potential therapeutic target in atherosclerosis.


International Journal of Molecular Sciences | 2013

CREG promotes the proliferation of human umbilical vein endothelial cells through the ERK/cyclin E signaling pathway.

Jie Tao; Chenghui Yan; Xiaoxiang Tian; Shaowei Liu; Yang Li; Jian Zhang; Mingyu Sun; Xinliang Ma; Yaling Han

Cellular repressor of E1A-stimulated genes (CREG) is a recently discovered secreted glycoprotein involved in homeostatic modulation. We previously reported that CREG is abundantly expressed in the adult vascular endothelium and dramatically downregulated in atherosclerotic lesions. In addition, CREG participates in the regulation of apoptosis, inflammation and wound healing of vascular endothelial cells. In the present study, we attempted to investigate the effect of CREG on the proliferation of vascular endothelial cells and to decipher the underlying molecular mechanisms. Overexpression of CREG in human umbilical vein endothelial cells (HUVEC) was obtained by infection with adenovirus carrying CREG. HUVEC proliferation was investigated by flow cytometry and 5-bromo-2′-deoxy-uridine (BrdU) incorporation assays. The expressions of cyclins, cyclin-dependent kinases and signaling molecules were also examined. In CREG-overexpressing cells, we observed a marked increase in the proportion of the S and G2 population and a decrease in the G0/G1 phase population. The number of BrdU positively-stained cells also increased, obviously. Furthermore, silencing of CREG expression by specific short hairpin RNA effectively inhibited the proliferation of human umbilical vein endothelial cells (HUVEC). CREG overexpression induced the expression of cyclin E in both protein and mRNA levels to regulate cell cycle progression. Further investigation using inhibitor blocking analysis identified that ERK activation mediated the CREG modulation of the proliferation and cyclin E expression in HUVEC. In addition, blocking vascular endothelial growth factor (VEGF) in CREG-overexpressed HUVEC and supplementation of VEGF in CREG knocked-down HUVEC identified that the pro-proliferative effect of CREG was partially mediated by VEGF-induced ERK/cyclin E activation. These results suggest a novel role of CREG to promote HUVEC proliferation through the ERK/cyclin E signaling pathway.


Journal of Geriatric Cardiology | 2013

Purification and functional assessment of smooth muscle cells derived from mouse embryonic stem cells

Xiaoxiang Tian; Jian Kang; Chenghui Yan; Kai Xu; Jie Tao; Guitang Yang; Yaling Han

Objective To obtain a pure population of smooth muscle cells (SMC) derived from mouse embryonic stem cells (ESC) and further assess their functions. Methods A vector, expressing both puromycin resistance gene (puror) and enhanced green fluorescent protein (EGFP) gene driven by smooth muscle 22α (SM22α) promoter, named pSM22α-puror-IRES2-EGFP was constructed and used to transfect ESC. Transgenic ESC (Tg-ESC) clones were selected by G418 and identified by PCR amplification of puror gene. The characteristics of Tg-ESC were detected by alkaline phosphatase (ALP) staining, SSEA-1 immunofluorescence and teratoma formation test in vivo. After induction of SMC differentiation by all-trans retinoic acid, differentiated Tg-ESC were treated with 10 µg/mL puromycin for three days to obtain purified SMC (P-SMC). Percentage of EGFP+ cells in P-SMC was assessed by flow cytometer. Expressions of smooth muscle specific markers were detected by immunostaining and Western blotting. Proliferation, migration and contractility of P-SMC were analyzed by growth curve, trans-well migration assay, and carbachol treatment, respectively. Finally, both P-SMC and unpurified SMC (unP-SMC) were injected into syngeneic mouse to see teratoma development. Results Tg-ESC clone was successfully established and confirmed by PCR detection of puror gene in its genomic DNA. The Tg-ESC was positive for ALP staining, SSEA-1 staining and formed teratoma containing tissues derived from three germ layers. After retinoic acid induction, large amount of EGFP positive cells outgrew from differentiated Tg-ESC. Three days of puromycin treatment produced a population of P-SMC with an EGFP+ percentage as high as 98.2% in contrast to 29.47% of unP-SMC. Compared with primary mouse vascular smooth muscle cells (VSMC), P-SMC displayed positive, but lowered expression of SMC-specific markers including SM α-actin and myosin heavy chain (SM-MHC) detected either, by immunostaining, or immunoblotting, accelerated proliferation, improved migration (99.33 ± 2.04 vs. 44.00 ± 2.08 migrated cells/field, P < 0.05), and decreased contractility in response to carbachol (7.75 ± 1.19 % vs. 16.50 ± 3.76 % in cell area reduction, P < 0.05). In vivo injection of unP-SMC developed apparent teratoma while P-SMC did not. Conclusions We obtained a pure population of ESC derived SMC with less mature (differentiated) phenotypes, which will be of great use in research of vascular diseases and in bio-engineered vascular grafts for regenerative medicine.

Collaboration


Dive into the Xiaoxiang Tian's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dan Liu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge