Network


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

Hotspot


Dive into the research topics where Zhitu Zhu is active.

Publication


Featured researches published by Zhitu Zhu.


International Journal of Molecular Sciences | 2012

Bufalin Induces Lung Cancer Cell Apoptosis via the Inhibition of PI3K/Akt Pathway

Zhitu Zhu; Hongzhi Sun; Guangyou Ma; Zhenghua Wang; Enze Li; Yangyang Liu; Yunpeng Liu

Bufalin is a class of toxic steroids which could induce the differentiation and apoptosis of leukemia cells, and induce the apoptosis of gastric, colon and breast cancer cells. However, the anti-tumor effects of bufalin have not been demonstrated in lung cancer. In this study we used A549 human lung adenocarcinoma epithelial cell line as the experimental model to evaluate the potential of bufalin in lung cancer chemotherapy. A549 cells were treated with bufalin, then the proliferation was detected by MTT assay and apoptosis was detected by flow cytometry analysis and Giemsa staining. In addition, A549 cells were treated by Akt inhibitor LY294002 in combination with bufalin and the activation of Akt and Caspase-3 as well as the expression levels of Bax, Bcl-2 and livin were examined by Western blot analysis. The results showed that Bufalin inhibited the proliferation of A549 cells and induced the apoptosis of A549 cells in a dose and time dependent manner. Mechanistically, we found that bufalin inhibited the activation of Akt. Moreover, bufalin synergized with Akt inhibitor to induce the apoptosis of A549 cells and this was associated with the upregulation of Bax expression, the downregulation of Bcl-2 and livin expression, and the activation of Caspase-3. In conclusion, our findings demonstrate that bufalin induces lung cancer cell apoptosis via the inhibition of PI3K/Akt pathway and suggest that bufalin is a potential regimen for combined chemotherapy to overcome the resistance of lung cancer cells to chemotherapeutics induced apoptosis.


Journal of Cancer Research and Clinical Oncology | 2012

Down-regulation of Cbl-b by bufalin results in up-regulation of DR4/DR5 and sensitization of TRAIL-induced apoptosis in breast cancer cells.

Shunchao Yan; Xiujuan Qu; Chong’an Xu; Zhitu Zhu; Lingyun Zhang; Ling Xu; Na Song; Yuee Teng; Yunpeng Liu

PurposeTNF-related apoptosis-inducing ligand (TRAIL) is a potential cancer therapeutic agent that preferentially induces apoptosis in cancer cells. However, breast cancer cells are generally resistant to TRAIL. Bufalin is a major active ingredient of the traditional Chinese medicine ChanSu. The present study aimed to assess the synergistic effect of bufalin and TRAIL and elucidate the underlying mechanisms in breast cancer cells.MethodsCell proliferation and apoptosis were measured by MTT assay and flow cytometry, respectively. The expression of proteins was assayed by flow cytometry and/or Western blotting. Transfection studies were used to determine the involvement of DR4, DR5 and Cbl-b in the synergistic effect of bufalin and TRAIL.ResultsMCF-7 and MDA-MB-231 cells were resistant to TRAIL. Both cell lines were dramatically sensitized to TRAIL-induced apoptosis by bufalin. Further experiments indicated that bufalin up-regulated DR4 and DR5, activated ERK, JNK and p38 MAPK and down-regulated Cbl-b. Blocking the up-regulation of DR4 and DR5 by siRNA rendered cells less sensitive to apoptosis induced by the combination of bufalin and TRAIL. Inhibition of the activation of ERK, JNK and p38 MAPK by specific inhibitors attenuated DR4 and DR5 up-regulation. Moreover, down-regulation of Cbl-b by shRNA led to stronger activation of ERK, JNK and p38 MAPK, more up-regulation of DR4 and DR5, and a stronger synergistic effect of bufalin and TRAIL.ConclusionsBufalin enhanced TRAIL-induced apoptosis by up-regulating the expression of DR4 and DR5. Bufalin-induced down-regulation of Cbl-b contributed to the up-regulation of DR4 and DR5, which might be partially mediated by the activation of ERK, JNK and p38 MAPK.


BMC Cancer | 2014

The role of RhoC in epithelial-to-mesenchymal transition of ovarian carcinoma cells

Wen-feng Gou; Yang Zhao; Hang Lu; Xue-feng Yang; Yin-Ling Xiu; Shuang Zhao; Jian-min Liu; Zhitu Zhu; Hongzhi Sun; Yunpeng Liu; Feng Xu; Yasuo Takano; Hua-chuan Zheng

BackgroundRhoC is a small G protein/GTPase and involved in tumor mobility, invasion and metastasis. Previously, up-regulated RhoC expression is found to play an important role in ovarian carcinogenesis and subsequent progression by modulating proliferation, apoptosis, migration and invasion.MethodsWe transfected RhoC-expressing plasmid and RhoC siRNA into CAOV3 and OVCAR3 cells respectively. These cells and transfectants were exposed to vascular epithelial growth factor (VEGF), transforming growth factor (TGF)-β1 or their receptor inhibitors with the phenotypes and their related-molecules examined.ResultsTGF-β1R or VEGFR inhibitor suppressed the proliferation, migration, invasion and lamellipodia formation, the expression of N-cadherin, α-SMA, snail and Notch1 mRNA or protein, and enhanced E-cadherin mRNA and protein expression in CAOV3 and its RhoC-overexpressing transfectants, whereas both growth factors had the opposite effects in OVCAR3 cells and their RhoC-hypoexpressing transfectants. Ectopic RhoC expression enhanced migration, invasion, lamellipodia formation and the alteration in epithelial to mesenchymal transition (EMT) markers of CAOV3 cells regardless of the treatment of VEGFR or TGF-β1R inhibitor, whereas RhoC knockdown resulted in the converse in OVCAR3 cells even with the exposure to VEGF or TGF-β1.ConclusionRhoC expression might be involved in EMT of ovarian epithelial carcinoma cells, stimulated by TGF-β1 and VEGF.


World Journal of Surgical Oncology | 2012

Inhibition of Jak-STAT3 pathway enhances bufalin-induced apoptosis in colon cancer SW620 cells

Zhitu Zhu; Enze Li; Yangyang Liu; Yu Gao; Hongzhi Sun; Guangyou Ma; Zhenghua Wang; Xiaomei Liu; Qingjun Wang; Xiujuan Qu; Yunpeng Liu; Yunlong Yu

BackgroundThe purpose of the research is to investigate the roles of Jak-STAT3 signaling pathway in bufalin-induced apoptosis in colon cancer SW620 cells.MethodsThe inhibitory effects of bufalin on cell proliferation were determined by MTT (Methyl thiazolyltetrazolium) assay. The morphological changes of cells were measured by Wright-Giemsa staining. The cell cycle arrest and apoptosis were tested by flow cytometry analysis. Western Blot was used to determine the protein expression of the apoptosis inhibitors livin and caspase-3, the apoptosis-related proteins Bax and Bcl-2, as well as the key protein kinases in the Jak-stat3 signaling pathway, stat3 and p-stat3.Results(1) Bufalin inhibited the proliferation of SW620 cells. IC50 at 24 h, 48 h and 72 h were 76.72 ± 6.21 nmol/L, 34.05 ± 4.21 nmol/L and 16.7 ± 6.37 nmol/L. (2) Bufalin induced SW620 cell cycle arrest and apoptosis, indicated by the appearance of apoptotic bodies; (3) The results from flow cytometry demonstrated that there was cell cycle G2/M phase arrest in 20 nmol/L bufalin treatment group (36.29 ± 2.11% vs 18.39 ± 1.74%, P<0.01); there was a sub-diploid apoptosis peak in 80 nmol/L bufalin treatment group (19.69 ± 1.63% vs 0.99 ± 0.23%, P <0.01). The apoptosis rate was 34.63 ± 2.57% (vs 19.69 ± 1.63%, P = 0.002) in JAK kinase inhibitor AG490 plus bufalin treatment group. (4) During the process of bufalin-induced apoptosis in SW620 cells, transient activation of p-stat3 inhibited the activation of stat3, up-regulated Bax expression, down-regulated livin and Bcl-2 expression (P<0.01), and activated caspase-3. Inhibition of Jak-stat3 signaling pathway by pre-treatment with AG490 significantly enhanced the bufalin-induced apoptosis (P<0.01), further up-regulated Bax protein expression, down-regulated livin and Bcl-2 protein expression and enhanced caspase-3 activation.ConclusionsBufalin not only inhibited the growth of colon cancer SW620 cells, but also induced apoptosis of SW620 cells. Activation of caspase-3, up-regulation of Bax, down-regulation of livin and Bcl-2, as well as inhibition of Jak-stat3 signaling pathway might be the important mechanisms for the bufalin-induced apoptosis.


Oncology Letters | 2015

Insulin-like growth factor-I induces epithelial to mesenchymal transition via GSK-3β and ZEB2 in the BGC-823 gastric cancer cell line

Heming Li; Ling Xu; Lei Zhao; Yanju Ma; Zhitu Zhu; Yunpeng Liu; Xiujuan Qu

Metastasis is the most common cause of mortality in patients with gastric cancer. Epithelial-to-mesenchymal transition (EMT), which may be stimulated by insulin-like growth factor-I (IGF-I) is involved in the metastasis of numerous tumors; however, the molecular mechanism by which IGF-I may induce tumor cell EMT remains to be elucidated in gastric cancer. The present study aimed to investigate the induction of EMT in BGC-823 gastric cancer cells. It was identified that IGF-I induced EMT by upregulating the levels of ZEB2 transcription factor, and this was dependent on the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway in these cells. In addition, glycogen synthase kinase 3β (GSK-3β), an intracellular downstream effector of PI3K/Akt, sustained the epithelial phenotype by repressing ZEB2 expression and the subsequent inhibition of EMT induced by IGF-I, suggesting the involvement of a potential PI3K/Akt-GSK-3β-ZEB2 signaling pathway in IGF-I-induced EMT in gastric cancer BGC-823 cells. Overall, the results of the present study suggest that IGF-I induced EMT by the activation of a PI3K/Akt-GSK-3β-ZEB2 signaling pathway in gastric cancer BGC-823 cells. Therefore, this study may provide more useful information regarding the mechanism of gastric cancer metastasis.


FEBS Letters | 2013

Lipid raft-regulated IGF-1R activation antagonizes TRAIL-induced apoptosis in gastric cancer cells

Ling Xu; Xiujuan Qu; Xuejun Hu; Zhitu Zhu; Ce Li; Enze Li; Yanju Ma; Na Song; Yunpeng Liu

Gastric cancer cells are resistant to tumor necrosis factor‐related apoptosis‐inducing ligand (TRAIL) and the resistance mechanism is not fully understood. In human gastric cancer MGC803 and BGC823 cells, TRAIL induces insulin‐like growth factor‐1 receptor (IGF‐1R) pathway activation. Treatment with IGF‐1R inhibitor OSI‐906 or small interfering RNAs against IGF‐1R, prevents IGF‐1R pathway activation and increases TRAIL‐induced apoptosis. The TRAIL‐induced IGF‐1R pathway activation is promoted by IGF‐1R translocation into lipid rafts. Moreover, the translocation of IGF‐1R into lipid rafts is regulated by Casitas B‐lineage lymphoma b (Cbl‐b). Taken together, TRAIL‐induced IGF‐1R activation antagonizes TRAIL‐induced apoptosis by Cbl‐b‐regulated distribution of IGF‐1R in lipid rafts.


Tumor Biology | 2014

Nrdp1 inhibits growth of colorectal cancer cells by nuclear retention of p27

Hang Lu; Hua Li; Dong Mao; Zhitu Zhu; Hongzhi Sun

The molecular mechanism underlying the proliferation of colorectal cancer (CRC) cells is not completely understood. Here, we found that the level of neuregulin receptor degradation protein-1 (Nrdp1) E3 ubiquitin ligase was significantly decreased in CRC tissues, compared with the adjacent normal tissues from human patients. Knockdown of Nrdp1 enhanced the proliferation of CRC cells, while overexpression of Nrdp1 inhibited the proliferation of CRC cells. Further analysis showed that Nrdp1 may induce degradation of its target ErbB3 to inhibit activation of both ERK/MAPK and PI3K/Akt pathways in CRC cells, which seemed to affect cell proliferation via nuclear retention of a major cell-cycle inhibitor, p27. Taken together, these findings suggest that Nrdp1-mediated ErbB3 degradation suppresses cellular growth of CRC and that Nrdp1 loss in CRC may promote tumor progression, thus highlighting Nrdp1 as a novel target for CRC therapy.


Oncology Letters | 2017

Transforming growth factor‑β1 contributes to oxaliplatin resistance in colorectal cancer via epithelial to mesenchymal transition

Liang Mao; Yan Li; Jinping Zhao; Qia Li; Bin Yang; Yuanyuan Wang; Zhitu Zhu; Hongzhi Sun; Zhenhua Zhai

Transforming growth factor-β1 (TGF-β1), secreted by main components of tumor microenvironment, is considered to be closely associated with cancer development and chemoresistance. The present study aimed to analyze the effects and mechanisms underlying TGF-β1-induced chemoresistance to oxaliplatin (LOH) in human colorectal cancer (CRC) cell lines. The cytotoxic effects of LOH subsequent to TGF-β1 treatment were assessed in three CRC cell lines by MTT assay. In addition, epithelial to mesenchymal transition (EMT), DNA damage and apoptosis assays were performed to evaluate the mechanisms of drug resistance in vitro. It was revealed that an exposure of CRC cells to TGF-β1 induced EMT. This was followed by a decrease in the levels of DNA damage and LOH-induced apoptotic cell death at certain TGF-β1 concentrations compared with untreated cells, which was responsible for LOH resistance. TGF-β1 leads to resistance to LOH in CRC cells, primarily through EMT. These data not only provide insight into the understanding of the chemoresistant mechanisms, but also may guide the clinical applications of reducing EMT to enhance the sensitivity to chemotherapy, by targeting TGF-β1.


Iubmb Life | 2016

Identification of arginine and its “Downstream” molecules as potential markers of breast cancer

Lu Hu; Yu Gao; Yunfeng Cao; Yinxu Zhang; Minghao Xu; Yuanyuan Wang; Yu Jing; Shengnan Guo; Fangyu Jing; Xiaodan Hu; Zhitu Zhu

Breast cancer (BC) is the most commonly diagnosed cancer in women worldwide. Arginine is a semiessential amino acid in humans and is essential for several biological pathways in malignant and normal cells, such as ornithine and N1, N12‐diacetylspermine (DiAcSpm). This study aimed to determine the role of arginine and these downstream molecules in BC. Plasma arginine, ornithine, and arginine–to–ornithine ratio (AOR) were analysed by liquid chromatography‐tandem mass spectrometry (LC‐MS/MS). Urine samples were measured by the colloid gold aggregation to test determination of urinary diAcSpm. A principal component analysis was performed to evaluate the results observed between breast tumor and control characteristics. Differences in individual metabolite concentrations between BC patients and controls were tested by receiver operating characteristics (ROC) analyses. Students t tests were used to detect the differences between two groups of normally distributed variables, and Wilcoxon sign rank tests were performed for asymmetrically distributed variables. As we analyzed, BC patients had lower plasma arginine and arginine/ornithine level, and higher plasma ornithine and urinary DiAcSpm concentrations as compared with control patients (P = 0.028, 0.020, 0.002, and 0.011, respectively). And the ROC curve was drawn and the area under the curve of the metabolites was calculated to be 0.659 (P = 0.028), 0.645 (P = 0.045), 0.7233 (P = 0.002), 0.683 (P = 0.011), respectively. In addition, our analysis showed that arginine concentrations and AOR had a positive correlation with ER status, while ornithine had a negative correlation with T stage (P = 0.042, 0.083, 0.023, respectively).In conclusion, arginine and these downstream molecules were biomarkers for BC. More studies are needed to highlight the theoretical strengths.


PLOS ONE | 2013

Optimal Duration of Fluorouracil-Based Adjuvant Chemotherapy for Patients with Resectable Gastric Cancer

Jinglei Qu; Xin Li; Xiujuan Qu; Zhitu Zhu; Lizhong Zhou; Yuee Teng; Jingdong Zhang; Bo Jin; Mingfang Zhao; Ping Yu; Yunpeng Liu

Background Although several clinical trials have suggested that postoperative adjuvant chemotherapy can improve survival of patients with gastric cancer, the optimal treatment duration has not been studied. This retrospective analysis evaluated the outcomes of patients with gastric cancer treated with six cycles of fluorouracil-based treatment compared with a cohort treated with four or eight cycles. Methods We retrospectively identified 237 patients with stage IB–IIIC gastric cancer who received four, six, or eight cycles of fluorouracil-based adjuvant chemotherapy administered every 3 weeks after radical gastrectomy. The endpoint was overall survival (OS). Factors associated with prognosis were also analyzed. Results The estimated 3-year OS rates for the four-, six-, and eight-cycle cohorts were 54.4%, 76.1%, and 68.9%, respectively; and the estimated 5-year OS rates were 41.2%, 74.0%, and 65.8%, respectively. Patients who received six cycles were more likely to have a better OS than those who received four cycles (P = 0.002). Eight cycles failed to show an additional survival benefit (P = 0.454). In the multivariate analysis, the number of chemotherapy cycles was associated with OS independent of clinical covariates (P<0.05). Subgroup analysis suggested that among patients in all age groups examined, male patients, and subgroups of fluorouracil plus oxaliplatin combined chemotherapy, stage III, poor differentiation, and gastrectomy with D2 lymphadenectomy, six cycles of adjuvant chemotherapy were associated with a statistically significant benefit of OS compared with four cycles (P<0.05). Conclusions Six cycles of adjuvant chemotherapy might lead to a favorable outcome for patients with gastric cancer, and two further cycles could not provide an additional clinical benefit.

Collaboration


Dive into the Zhitu Zhu's collaboration.

Top Co-Authors

Avatar

Hongzhi Sun

Liaoning Medical University

View shared research outputs
Top Co-Authors

Avatar

Enze Li

Liaoning Medical University

View shared research outputs
Top Co-Authors

Avatar

Qingjun Wang

Liaoning Medical University

View shared research outputs
Top Co-Authors

Avatar

Yangyang Liu

Liaoning Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhenghua Wang

Liaoning Medical University

View shared research outputs
Top Co-Authors

Avatar

Guangyou Ma

Liaoning Medical University

View shared research outputs
Top Co-Authors

Avatar

Hang Lu

Liaoning Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaomei Liu

Liaoning Medical University

View shared research outputs
Top Co-Authors

Avatar

Yun-Feng Cao

Dalian Institute of Chemical Physics

View shared research outputs
Researchain Logo
Decentralizing Knowledge