Network


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

Hotspot


Dive into the research topics where Xinyu Zhao is active.

Publication


Featured researches published by Xinyu Zhao.


PLOS ONE | 2011

BIM-Mediated AKT Phosphorylation Is a Key Modulator of Arsenic Trioxide-Induced Apoptosis in Cisplatin-Sensitive and -Resistant Ovarian Cancer Cells

Zhu Yuan; Fang Wang; Zhiwei Zhao; Xinyu Zhao; Ji Qiu; Chunlai Nie; Yuquan Wei

Background Chemo-resistance to cisplatin-centered cancer therapy is a major obstacle to the effective treatment of human ovarian cancer. Previous reports indicated that arsenic trioxide (ATO) induces cell apoptosis in both drug-sensitive and -resistant ovarian cancer cells. Principal Findings In this study, we determined the molecular mechanism of ATO-induced apoptosis in ovarian cancer cells. Our data demonstrated that ATO induced cell apoptosis by decreasing levels of phosphorylated AKT (p-AKT) and activating caspase-3 and caspase-9. Importantly, BIM played a critical role in ATO-induced apoptosis. The inhibition of BIM expression prevented AKT dephosphorylation and inhibited caspase-3 activation during cell apoptosis. However, surprisingly, gene silencing of AKT or FOXO3A had little effect on BIM expression and phosphorylation. Moreover, the activation of caspase-3 by ATO treatment improved AKT dephosphorylation, not only by cleaving the regulatory A subunit of protein phosphatase 2A (PP2A), but also by increasing its activation. Furthermore, our data indicated that the c-Jun N-terminal kinases (JNK) pathway is involved in the regulation of BIM expression. Conclusions We demonstrated the roles of BIM in ATO-induced apoptosis and the molecular mechanisms of BIM expression regulated by ATO during ovarian cancer cell apoptosis. Our findings suggest that BIM plays an important role in regulating p-AKT by activating caspase-3 and that BIM mediates the level of AKT phosphorylation to determine the threshold for overcoming cisplatin resistance in ovarian cancer cells.


Electrophoresis | 2009

Quantitative protein expression profiling of 14-3-3 isoforms in human renal carcinoma shows 14-3-3 epsilon is involved in limitedly increasing renal cell proliferation.

Shufang Liang; Yuhuan Xu; Guobo Shen; Qingping Liu; Xinyu Zhao; Zhizhong Xu; Xi Xie; Fengming Gong; Ronghui Li; Yuquan Wei

14‐3‐3 proteins regulate many cellular processes that are implicated in cancer development, and the seven 14‐3‐3 isoforms have different expression level and isoform‐specific roles in different tumors. However, the biological functions of 14‐3‐3 proteins and their correlations with renal carcinoma have not been investigated so far. In our study, the expression profiles and functional characterization of 14‐3‐3 proteins were discovered by a sensitive stable isotope labeling with amino acids in cell culture based quantitative proteomics analysis in human renal carcinoma tissues. We found that 14‐3‐3ε was up‐regulated with 1.44‐fold changes in renal cancerous tissues compared with that in counterpart kidney tissues, and 14‐3‐3σ was almost not detected in both tissues due to its DNA highly methylated in our previous reports. The other five isoforms almost have similar expression level in two states of renal tissues. The following RT‐PCR, Western blot and immunohistochemistry analysis for specific 14‐3‐3 isoform expression were all consistent with the quantitative proteomic data. Furthermore, the overexpression of 14‐3‐3ε in vitro can limitedly prompt the abnormal growth of renal tumor cells.


Journal of Translational Medicine | 2011

Comparisons of three polyethyleneimine-derived nanoparticles as a gene therapy delivery system for renal cell carcinoma

Zhizhong Xu; Guobo Shen; Xiangying Xia; Xinyu Zhao; Peng Zhang; Huanhuan Wu; QingFa Guo; Zhiyong Qian; Yuquan Wei; Shufang Liang

BackgroundPolyethyleneimine (PEI), which can interact with negatively charged DNA through electrostatic interaction to form nanocomplexes, has been widely attempted to use as a gene delivery system. However, PEI has some defects that are not fit for keeping on gene expression. Therefore, some modifications against PEI properties have been done to improve their application value in gene delivery. In this study, three modified PEI derivatives, including poly(ε-caprolactone)-pluronic-poly(ε-caprolactone) grafted PEI (PCFC-g-PEI), folic acid-PCFC-isophorone diidocyanate-PEI (FA-PEAs) and heparin-PEI (HPEI), were evaluated in terms of their cytotoxicity and transfection efficiency in vitro and in vivo in order to ascertain their potential application in gene therapy.MethodsMTT assay and a marker GFP gene, encoding green fluorescent protein, were used to evaluate cell toxicity and transfection activity of the three modified PEI in vitro. Renal cell carcinoma (RCC) models were established in BALB/c nude mice inoculated with OS-RC-2 cells to detect the gene therapy effects using the three PEI-derived nanoparticles as gene delivery vehicles. The expression status of a target gene Von Hippel-Lindau (VHL) in treated tumor tissues was analyzed by semiquantitative RT-PCR and immunohistochemistry.ResultsEach of three modified PEI-derived biomaterials had an increased transfection efficiency and a lower cytotoxicity compared with its precursor PEI with 25-kD or 2-kD molecule weight in vitro. And the mean tumor volume was obviously decreased 30% by using FA-PEAs to transfer VHL plasmids to treat mice RCC models. The VHL gene expression was greatly improved in the VHL-treated group. While there was no obvious tumor inhibition treated by PCFC-g-PEI:VHL and HPEI:VHL complexes.ConclusionsThe three modified PEI-derived biomaterials, including PCFC-g-PEI, FA-PEAs and HPEI, had an increased transfection efficiency in vitro and obviously lower toxicities compared with their precursor PEI molecules. The FA-PEAs probably provide a potential gene delivery system to treat RCC even other cancers in future.


Iubmb Life | 2008

Gene expression and methylation status of 14‐3‐3σ in human renal carcinoma tissues

Shufang Liang; Yuhuan Xu; Guobo Shen; Xinyu Zhao; Jin Zhou; Xingbin Li; Fengming Gong; Bo Ling; Li Fang; Canhua Huang; Yuquan Wei

Loss of 14‐3‐3σ expression mainly by methylation‐mediated silencing has been reported in several human cancers, but the methylation status of 14‐3‐3σ in human renal carcinoma is rarely studied so far. In this report, 14‐3‐3σ expression was first examined by RT‐PCR and immunohistochemistry, and further we investigated the methylation status by methylation‐specific PCR and the correlation between 14‐3‐3σ expression and its methylation. We found 14‐3‐3σ expression was lost in 27 of 31 renal tissues including 16 renal carcinoma tissues, eight para‐cancerous kidney tissues and seven normal kidney tissues. Among 16 renal carcinoma tissues, 14 cases had complete hypermethylation of 14‐3‐3σ. Eight para‐cancerous kidney tissues were almost completely methylated except one case had both methylation and unmethylation. Among seven normal kidney tissues, five cases had partial methylation, and the other two cases were completely methylated. In addition, 14‐3‐3σ mRNA had weak expression in OS‐RC‐2 cells, but it increased with gradual demethylation after treatment by a demethylation agent, 5‐aza‐2′‐deoxycytidine. In general, 14‐3‐3σ mRNA was mostly unexpressed, and its DNA frequently hypermethylated within 14‐3‐3σ coding region was closely associated with the gene silencing in cancerous and para‐cancerous kidney tissues. 14‐3‐3σ was also frequently methylated and almost silencing in normal kidney tissues. However, the methylation frequency was gradually reinforced with the extent of malignancy from normal to para‐cancerous and cancerous kidney tissues.


Annals of Oncology | 2008

Efficient inhibition of cisplatin-resistant human ovarian cancer growth and prolonged survival by gene transferred vesicular stomatitis virus matrix protein in nude mice

Qian Zhong; Y.-J. Wen; H.-S. Yang; Hong Luo; A.-F. Fu; Fan Yang; L.-J. Chen; X. Chen; Xiaorong Qi; Hong-gang Lin; Y. Wan; X.-C. Chen; Y.-Q. Wei; Xinyu Zhao

BACKGROUNDnThe vesicular stomatitis virus matrix protein (VSVMP) has been receiving attention as an anticancer agent because of its ability of inducing apoptosis.nnnMATERIALS AND METHODSnNude mice bearing A2780s and A2780cp ovarian tumors were treated twice weekly with i.v. administration of 50 microg VSVMP/250 mug liposome complex, 50 microg empty plasmid/250 microg liposome complex, 0.9% NaCl solution or weekly with i.p. administration of cisplatin (5 mg/kg) for 3 weeks. Tumor volume and survival time were observed. TUNEL assay and CD34 vessel staining were conducted in tumor tissue. Antiangiogenesis in vivo were determined by sponge assay. Antiproliferative and apoptosis-inducing activities of VSVMP in vitro were tested on MS1 murine endothelial cells and four human ovarian cancer cell lines: A2780s, A2780cp, HO8910 and COC1.nnnRESULTSnAdministration of VSVMP resulted in significant inhibition (87%-98% maximum inhibition relative to controls) in the growth of A2780s and A2780cp tumor xenografts, and prolonged the survival of the treated mice. Complete tumor regression happened in VSVMP-treated mice in both tumor models. These antitumor responses were associated with marked increases in tumor apoptosis and reductions in intratumoral microvessel density.nnnCONCLUSIONSnOur data indicate that VSVMP may provide an effective approach to inhibit both cisplatin-sensitive and -resistant human ovarian cancer growth with minimal side-effects.


Journal of Biological Chemistry | 2015

PNAS-4, an Early DNA Damage Response Gene, Induces S Phase Arrest and Apoptosis by Activating Checkpoint Kinases in Lung Cancer Cells.

Zhu Yuan; Wenhao Guo; Jun Yang; Lei Li; Meiliang Wang; Yi Lei; Yang Wan; Xinyu Zhao; Na Luo; Ping Cheng; Xinyu Liu; Chunlai Nie; Yong Peng; Aiping Tong; Yuquan Wei

Background: Elevated PNAS-4 induces S phase arrest and apoptosis in vitro and inhibits tumor growth in vivo. Results: PNAS-4 activates Chk1/2 to cause inhibition of the Cdc25A-CDK2-cyclin E/A pathway, causing S phase arrest and apoptosis. Conclusion: Activation of Chk1/2 is a determinant of S phase arrest and apoptosis. Significance: We provide a novel action mechanism for PNAS-4 as a potential therapeutic target for lung cancer. PNAS-4, a novel pro-apoptotic gene, was activated during the early response to DNA damage. Our previous study has shown that PNAS-4 induces S phase arrest and apoptosis when overexpressed in A549 lung cancer cells. However, the underlying action mechanism remains far from clear. In this work, we found that PNAS-4 expression in lung tumor tissues is significantly lower than that in adjacent lung tissues; its expression is significantly increased in A549 cells after exposure to cisplatin, methyl methane sulfonate, and mitomycin; and its overexpression induces S phase arrest and apoptosis in A549 (p53 WT), NCI-H460 (p53 WT), H526 (p53 mutation), and Calu-1 (p53−/−) lung cancer cells, leading to proliferation inhibition irrespective of their p53 status. The S phase arrest is associated with up-regulation of p21Waf1/Cip1 and inhibition of the Cdc25A-CDK2-cyclin E/A pathway. Up-regulation of p21Waf1/Cip1 is p53-independent and correlates with activation of ERK. We further showed that the intra-S phase checkpoint, which occurs via DNA-dependent protein kinase-mediated activation of Chk1 and Chk2, is involved in the S phase arrest and apoptosis. Gene silencing of Chk1/2 rescues, whereas that of ATM or ATR does not affect, S phase arrest and apoptosis. Furthermore, human PNAS-4 induces DNA breaks in comet assays and γ-H2AX staining. Intriguingly, caspase-dependent cleavage of Chk1 has an additional role in enhancing apoptosis. Taken together, our findings suggest a novel mechanism by which elevated PNAS-4 first causes DNA-dependent protein kinase-mediated Chk1/2 activation and then results in inhibition of the Cdc25A-CDK2-cyclin E/A pathway, ultimately causing S phase arrest and apoptosis in lung cancer cells.


Cancer Science | 2009

Improved therapeutic efficacy against murine carcinoma by combining honokiol with gene therapy of PNAS-4, a novel pro-apoptotic gene.

Zhu Yuan; Huan-yi Liu; Fei Yan; Yongsheng Wang; Lantu Gou; Chunlai Nie; Zhenyu Ding; Songtao Lai; Yuwei Zhao; Xinyu Zhao; Jiong Li; Hongxin Deng; Yong-Qiu Mao; Lijuan Chen; Yuquan Wei; Xia Zhao

PNAS‐4, a novel pro‐apoptotic gene activated during the early response to DNA damage, can inhibit proliferation via apoptosis when overexpressed in some tumor cells. Recent studies have indicated that honokiol can induce apoptosis, inhibit angiogenesis, and suppress tumor growth. In the present study, we investigated whether mouse PNAS‐4 (mPNAS‐4) could augment the apoptosis of tumor cells induced by honokiol in vitro, and whether the antiangiogenic activity of honokiol and induction of apoptosis by mPNAS‐4 could work cooperatively to improve the antitumor efficacy in vivo. In vitro, mPNAS‐4 inhibited proliferation of murine colorectal carcinoma CT26 and Lewis lung carcinoma LL2 cells through induction of apoptosis, and significantly augmented the apoptosis of CT26 and LL2 cells induced by honokiol. Compared with treatment with mPNAS‐4 or honokiol alone, in vivo systemic administration of an expression plasmid encoding mPNAS‐4 and low‐dose honokiol significantly suppressed tumor growth through the enhanced induction of apoptosis and the augmented inhibition of angiogenesis. Our data suggest that the combined treatment with mPNAS‐4 plus honokiol augments antitumor effects in vitro and in vivo, and that the improved antitumor activity in vivo may be associated with enhanced induction of apoptosis and augmented inhibition of angiogenesis. The present study may provide a novel and effective method for the treatment of cancer. (Cancer Sci 2009; 100: 1757–1766)


Cancer Letters | 2009

Prokaryotic expression, purification of a new tumor-relative protein FAM92A1-289 and its characterization in renal cell carcinoma

Shufang Liang; Fengming Gong; Xinyu Zhao; Xianhuo Wang; Guobo Shen; Yuhuan Xu; Hanshuo Yang; Xu-Zhi Ruan; Yuquan Wei

In order to study the characterization of a new tumor-relative FAM92A1-289 protein, we first constructed plasmid FAM92A1-pQE30 for fusion expression in Escherichia coli. The recombinant protein FAM92A1-289 was affinity-purified by Ni2+-charged resin and separated by HPLC chromatography with high purity, and it was further identified by electrospray ionization-mass spectrometry. Furthermore, the expression and cell localization of FAM92A1-289 by immunohistochemistry using our self-prepared polyclonal antibody showed it was expressed in cytoplasm of renal carcinoma. FAM92A1-289 mRNA was expressed in 2 of 10 kidney tissues and in 6 of 12 primary renal tumors. FAM92A1-289 can promote cell growth in vitro and in vivo by colony formation and mouse xenograft assay. Our present data indicated FAM92A1-289 is a new tumor-related gene with oncogenic potentials to probably play roles in renal carcinogenesis.


Cancer Chemotherapy and Pharmacology | 2009

PNAS-4, a novel pro-apoptotic gene, can potentiate antineoplastic effects of cisplatin

Zhu Yuan; Fei Yan; Yongsheng Wang; Huan-yi Liu; Lantu Gou; Xinyu Zhao; Songtao Lai; Hongxin Deng; Jiong Li; Zhenyu Ding; Shao-qun Xiong; Bing Kan; Yong-Qiu Mao; Lijuan Chen; Yuquan Wei; Xia Zhao

PurposePNAS-4, a novel pro-apoptotic gene activated during the early response to DNA damage, can inhibit proliferation via apoptosis when overexpressed in some tumor cells. The objectives of this study were to determine whether PNAS-4 could enhance apoptosis induced by cisplatin besides its induction of apoptosis, and to evaluate the usefulness of combined treatment with mouse PNAS-4 (mPNAS-4) gene therapy and low-dose cisplatin chemotherapy in the inhibition of tumor growth in colon carcinoma (CT26) and Lewis lung carcinoma (LL/2) murine models.MethodsIn this study, the inxa0vitro growth-inhibitory and pro-apoptotic effects of PNAS-4 and/or cisplatin on CT26, LL/2, and SKOV3 cancer cells were assessed by MTT assay, flow cytometric analysis, DNA fragmentation, and morphological analysis, respectively. The inxa0vivo antitumor activity of combined treatment with mPNAS-4 gene therapy and low-dose cisplatin were evaluated in the inhibition of tumor growth in colon carcinoma (CT26) and Lewis lung carcinoma (LL/2) murine models. Tumor volume and survival time were observed. Induction of apoptosis was also assessed in tumor tissues.ResultsInxa0vitro, PNAS-4 inhibited proliferation of colon carcinoma (CT26), Lewis lung carcinoma (LL/2) and human ovarian cancer (SKOV3) cell lines via apoptosis, and significantly enhanced the apoptosis of CT26, LL/2, and SKOV3 cells induced by cisplatin. Inxa0vivo systemic administration of expression plasmid encoding mPNAS-4 (pcDNA3.1-mPS) and cisplatin, significantly decreased tumor growth through increased tumor cell apoptosis compared to treatment with mPNAS-4 or cisplatin alone.ConclusionsOur data suggests that the combined treatment with mPNAS-4 plus cisplatin may augment the induction of apoptosis in tumor cells inxa0vitro and inxa0vivo, and that the augmented antitumor activity inxa0vivo may result from the increased induction of apoptosis. The present study may provide a novel way to augment the antitumor efficacy of cytotoxic chemotherapy.


PLOS ONE | 2012

NOXA-Induced Alterations in the Bax/Smac Axis Enhance Sensitivity of Ovarian Cancer Cells to Cisplatin

Chao Lin; Xinyu Zhao; Lei Li; Huan-yi Liu; Kang Cao; Yang Wan; Xinyu Liu; Chunlai Nie; Lei Liu; Aiping Tong; Hongxin Deng; Jiong Li; Zhu Yuan; Yuquan Wei

Ovarian cancer is the most common cause of death from gynecologic malignancy. Deregulation of p53 and/or p73-associated apoptotic pathways contribute to the platinum-based resistance in ovarian cancer. NOXA, a pro-apoptotic BH3-only protein, is identified as a transcription target of p53 and/or p73. In this study, we found that genetic variants of Bcl-2 proteins exist among cisplatin-sensitive and -resistant ovarian cancer cells, and the responses of NOXA and Bax to cisplatin are regulated mainly by p53. We further evaluated the effect of NOXA on cisplatin. NOXA induced apoptosis and sensitized A2780s and SKOV3 cells to cisplatin in vitro and in vivo. The effects were mediated by elevated Bax expression, enhanced caspase activation, release of Cyt C and Smac into the cytosol. Furthermore, gene silencing of Bax or Smac significantly attenuated NOXA and/or cisplatin-induced apoptosis in chemosensitive A2780s cells, whereas overexpression of Bax or addition of Smac-N7 peptide significantly increased NOXA and/or cisplatin-induced apoptosis in chemoresistant SKOV3 cells. To our knowledge, these data suggest a new mechanism by which NOXA chemosensitized ovarian cancer cells to cisplatin by inducing alterations in the Bax/Smac axis. Taken together, our findings show that NOXA is potentially useful as a chemosensitizer in ovarian cancer therapy.

Collaboration


Dive into the Xinyu Zhao's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fei Yan

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge