Network


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

Hotspot


Dive into the research topics where Xiaotang Yu is active.

Publication


Featured researches published by Xiaotang Yu.


Cell Death and Disease | 2014

Roles of Wnt/β-catenin signaling in the gastric cancer stem cells proliferation and salinomycin treatment

Jun Mao; Shujun Fan; Wei Ma; P Fan; Bo Wang; Jian Zhang; Huan Wang; B Tang; Q Zhang; Xiaotang Yu; Lianhui Wang; Bo Song; Lianhong Li

The Wnt1 protein, a secreted ligand that activates Wnt signaling pathways, contributes to the self-renewal of cancer stem cells (CSCs) and thus may be a major determinant of tumor progression and chemoresistance. In a series of gastric cancer specimens, we found strong correlations among Wnt1 expression, CD44 expression, and the grade of gastric cancer. Stable overexpression of Wnt1 increased AGS gastric cancer cells’ proliferation rate and spheroids formation, which expressed CSC surface markers Oct4 and CD44. Subcutaneous injection of nude mice with Wnt1-overexpressing AGS cells resulted in larger tumors than injection of control AGS cells. Salinomycin, an antitumor agent, significantly reduced the volume of tumor caused by Wnt1-overexpressing AGS cells in vivo. This is achieved by inhibiting the proliferation of CD44+Oct4+ CSC subpopulation, at least partly through the suppression of Wnt1 and β-catenin expression. Taken together, activation of Wnt1 signaling accelerates the proliferation of gastric CSCs, whereas salinomycin acts to inhibit gastric tumor growth by suppressing Wnt signaling in CSCs. These results suggest that Wnt signaling might have a critical role in the self-renewal of gastric CSCs, and salinomycin targeting Wnt signaling may have important clinical applications in gastric cancer therapy.


Stem Cell Research & Therapy | 2013

Genistein decreases the breast cancer stem-like cell population through Hedgehog pathway

Panhong Fan; Shujun Fan; Huan Wang; Jun Mao; Yu Shi; Mohammed Mohammed Ibrahim; Wei Ma; Xiaotang Yu; Zhenhuan Hou; Bo Wang; Lianhong Li

IntroductionThe existence of breast cancer stem-like cells (BCSCs) has profound implications for cancer prevention. Genistein, a predominant isoflavone found in soy products, has multiple robust anti-tumor effects in various cancers, especially in the breast and prostate cancer. In this study, we aimed to evaluate genistein inhibition of BCSCs and its potential mechanism by culturing MCF-7 breast cancer cells and implanting these cells into nude mice.MethodsCell counting, colony formation and cell apoptosis analysis were used to evaluate the effect of genistein on breast cancer cells’ growth, proliferation and apoptosis. We then used mammosphere formation assay and CD44CD24 staining to evaluate the effect of genistein on BCSCs in vitro. A nude mice xenograft model was employed to determine whether genistein could target BCSCs in vivo, as assessed by real-time polymerase chain reaction (PCR) and immunohistochemical staining. The potential mechanism was investigated utilizing real-time PCR, western blotting analysis and immunohistochemical staining.ResultsGenistein inhibited the MCF-7 breast cancer cells’ growth and proliferation and promoted apoptosis. Both in vitro and in vivo genistein decreased breast cancer stem cells, and inhibited breast cancer stem-like cells through down-regulation of the Hedgehog-Gli1 Signaling Pathway.ConclusionsWe demonstrated for the first time that genistein inhibits BCSCs by down-regulating Hedgehog-Gli1 signaling pathway. These findings provide support and rationale for investigating the clinical application of genistein in treating breast cancer, and specifically by targeting breast cancer stem cells.


The International Journal of Biochemistry & Cell Biology | 2013

ShRNA targeting Notch1 sensitizes breast cancer stem cell to paclitaxel

Jun Mao; Bo Song; Yu Shi; Bo Wang; Shujun Fan; Xiaotang Yu; Jianwu Tang; Lianhong Li

Breast cancer is currently the most lethal gynecologic malignancy in many countries, and paclitaxel is a cornerstone in the treatment of this malignancy. Unfortunately, the efficacy of paclitaxel is limited due to the development of drug resistance. Evidence has suggested that cancer stem cells (CSCs) are involved in resistance to various forms of therapies, including chemotherapy. However, the interaction between paclitaxel resistance and CSCs and its underlying mechanisms have not been previously explored. In this study, we confirmed that paclitaxel enriched breast CSCs (CD44+/CD24-) in a dose-dependent manner in MCF-7 human breast cancer cell line. We then demonstrated that Notch1 was overexpressed in breast CSCs isolated from paclitaxel-treated MCF-7 cells compared to non-CSCs. The short hairpin RNA (shRNA) mediated knock-down of Notch1 inhibited MCF-7 cell proliferation and induced cell apoptosis. The anti-apoptosis protein NF-κB was decreased significantly when treated with shRNA-Notch1, and this effect was sharply improved by combination with paclitaxel. Paclitaxel decreased CD44+/CD24- cell population in MCF-7 cells and reduced the size and number of primary mammospheres after down-regulating the Notch1. Furthermore, shRNA-Notch1 inhibited the growth of tumor xenografts in nude mice noticeably. RT-PCR and Western blotting analysis showed that the expressions of ALDH1, NICD, Hes-1 and the drug transporter ABCG2 were decreased both in vitro and in vivo. These results suggest that Notch1 might play a critical role in the resistance to paclitaxel, and targeting Notch1 may have important clinical applications in cancer therapy.


Chemico-Biological Interactions | 2015

Salinomycin exerts anticancer effects on human breast carcinoma MCF-7 cancer stem cells via modulation of Hedgehog signaling

Ying Lu; Wei Ma; Jun Mao; Xiaotang Yu; Zhenhuan Hou; Shujun Fan; Bo Song; Huan Wang; Jiazhi Li; Le Kang; Pixu Liu; Quentin Liu; Lianhong Li

Breast cancer tissue contains a small population of cells that have the ability to self-renew, these cells are known as breast cancer stem cells (BCSCs). The Hedgehog signal transduction pathway plays a central role in stem cell development, its aberrant activation has been shown to contribute to the development of breast cancer, making this pathway an attractive therapeutic target. Salinomycin (Sal) is a novel identified cancer stem cells (CSCs) killer, however, the molecular basis for its anticancer effects is not yet clear. In the current study, Sals ability to modulate the activity of key elements in the Hedgehog pathway was examined in the human breast cancer cell line MCF-7, as well as in a subpopulation of cancer stem cells identified within this cancer cell line. We show here that Sal inhibits proliferation, invasion, and migration while also inducing apoptosis in MCF-7 cells. Interestingly, in a subpopulation of MCF-7 cells with the CD44(+)/CD24(-) markers and high ALDH1 levels indicative of BCSCs, modulators of Hedgehog signaling Smo and Gli1 were significantly down-regulated upon treatment with Sal. These results demonstrate that Sal also inhibits proliferation and induces apoptosis of BCSCs, further establishing it as therapeutically relevant in the context of breast cancers and also indicating that modulation of Hedgehog signaling is one potential mechanism by which it exerts these anticancer effects.


Biomedicine & Pharmacotherapy | 2016

miR-221/222 enhance the tumorigenicity of human breast cancer stem cells via modulation of PTEN/Akt pathway

Bailong Li; Ying Lu; Honghai Wang; Xiaocui Han; Jun Mao; Jiazhi Li; Lihui Yu; Bo Wang; Shujun Fan; Xiaotang Yu; Bo Song

BACKGROUND The miR-221/222 cluster has been discovered to function as oncogene in human malignancies including breast cancer. However, the role of miR-221/222 in the self-renewal of breast cancer stem cells (BCSCs) is not fully understood. In this study, we examined the impact and mechanism of miR-221/222 on the breast cancer cell viability, migration and invasion, and propagation of BCSCs. METHODS Human breast cancer cell line MCF-7 was transfected with miR-221/222 mimics or inhibitors to overexpress or knock down miR-221/222 respectively using Lipofactamine 2000. The biological effects of miR-221 and miR-222 were then assessed by cell proliferation assay, colony formation assay and transwell chamber assays. CD44/CD24 staining and mammosphere formation assay were performed to evaluate the ability of BCSCs self-renewal. Potential target gene phosphatase and tensin homolog (PTEN) and its downstream effector, phosphorylated Akt (p-Akt) were identified by Western blot and qRT-PCR methods. RESULTS PTEN, a tumor suppressor gene, was confirmed as a target of miR-221/222 in breast cancer cell line MCF-7. Downregulation of PTEN by miR-221/222 increased the phosphorylation of Akt. Enforced expression of miR-221/222 promoted breast cancer cell proliferation, migration and invasion via targeting PTEN/Akt pathway. Importantly, ectopic expression of miR-221/222 enriched the proportion of CD44(+)/CD24(-) BCSCs and improved the mammosphere formation capacity through targeting PTEN/Akt pathway. Blocking the endogenous miR-221/222 restored PTEN expression and subsequently decreased Akt phosphorylation, and thereby reversed this phenotype. CONCLUSIONS Our results suggested that miR-221/222 enhance breast cancer growth, migration and invasion, meanwhile propagate the self-renewal of BCSCs. This is achieved possibly through targeting PTEN/Akt pathway. miR-221/222 might be a novel therapeutic candidate for human breast cancer.


Chemico-Biological Interactions | 2016

Salinomycin suppresses TGF-β1-induced epithelial-to-mesenchymal transition in MCF-7 human breast cancer cells.

Chunying Zhang; Ying Lu; Qing Li; Jun Mao; Zhenhuan Hou; Xiaotang Yu; Shujun Fan; Jiazhi Li; Tong Gao; Bing Yan; Bo Wang; Bo Song; Lianhong Li

Epithelial-to-mesenchymal transition (EMT) is the major cause of breast cancer to initiate invasion and metastasis. Salinomycin (Sal) has been found as an effective chemical compound to kill breast cancer stem cells. However, the effect of Sal on invasion and metastasis of breast cancer is unclear. In the present study, we showed that Sal reversed transforming growth factor-β1 (TGF-β1) induced invasion and metastasis accompanied with down-regulation of MMP-2 by experiments on human breast cancer cell line MCF-7. Sal was able to inhibit TGF-β1-induced EMT phenotypic transition and the activation of key signaling molecules involved in Smad (p-Smad2/3,Snail1) and non-Smad (β-catenin, p-p38 MAPK) signals which cooperatively regulate the induction of EMT. Importantly, in a series of breast cancer specimens, we found strong correlation among E-cadherin expression, β-catenin expression, and the lymph node metastatic potential of breast cancer. Our research suggests that Sal is promised to be a chemotherapeutic drug by suppressing the metastasis of breast cancer.


Biomedicine & Pharmacotherapy | 2014

Smoothened activates breast cancer stem-like cell and promotes tumorigenesis and metastasis of breast cancer.

Lixia Wang; Wei Duan; Le Kang; Jun Mao; Xiaotang Yu; Shujun Fan; Lianhong Li; Yajun Tao

Smoothened (Smo) is a G protein-coupled receptor protein encoded by the Smo gene of the hedgehog signalling pathway, which is thought to play an important role in maintaining organ patterning, cell differentiation and self-renewal. The possible role of Smo in the process of tumorigenesis and metastasis of breast cancer still remains unclear. The present experiments were to investigate the effect of Smo on activating breast cancer stem-like CD44(+)CD24(-) cells and the tumorigenesis and metastasis of breast cancer. By injected CD44(+)CD24(-) cells (1×10(4)) into the cleared fat pad of NOD/SCID mice, it was observed that CD44(+)CD24(-) cells possess higher tumor-initiating capacity and metastasis properties than equal numbers of non-CD44(+)CD24(-) cells. The mRNA and protein expressions of Smo in CD44(+)CD24(-) cells were higher than those in non-CD44(+)CD24(-) cells, indicating that Smo may play a role in maintaining breast cancer stem cell features. qRT-PCR results revealed that expressions of STAT3, Bcl-2 and cyclinD1 mRNA in MCF-7 cells were decreased after transfected by Smo siRNA. In addition, the expressions of MMP-2 and MMP-9 were downregulated in MCF-7 cells after Smo expression was inhibited. Smo inhibition may be a possible therapeutic target that potentially suppresses breast tumor formation and development.


Journal of Toxicologic Pathology | 2015

Expression and activation of EGFR and STAT3 during the multistage carcinogenesis of intrahepatic cholangiocarcinoma induced by 3′-methyl-4 dimethylaminoazobenzene in rats

Fan Zhang; Lianhong Li; Xingwu Yang; Bo Wang; Jinyao Zhao; Shilun Lu; Xiaotang Yu

The purpose of this study was to investigate whether the epidermal growth factor receptor (EGFR) and signal transducer and activator of transcription-3 (STAT3) signal pathway contributes to the carcinogenesis of intrahepatic cholangiocarcinoma (ICC) induced by 3’-methyl-4 dimethylaminoazobenzene (3’Me-DAB) in rats. EGFR, TGFα, STAT3 and p-STAT3 in different stages of carcinogenesis were detected by immunohistochemistry (IHC). In situ hybridization (ISH) was applied to investigate the expression of STAT3 mRNA. Oval cells were verified by the immunohistochemical staining of alpha-fetoprotein (AFP), CD133 and epithelial cell adhesion molecules (EpCAM). Sequential development of necrosis, oval cell proliferation, cholangiofibrosis (CF) and ICC was observed in the liver of rats administered 3’Me-DAB. Oval cells showed positive expression of AFP, CD133 and EpCAM. The expression of EGFR was significantly higher in the ICC than in oval cells, CF or normal bile ducts (p<0.05), but there was no difference in EGFR expression between the other groups. The highest expression of p-STAT3 and TGFα was observed in CF. The expression of these two molecules in the ICC and oval cells was significantly higher than in normal bile ducts (p<0.05). Elevation of STAT3 mRNA was detected during carcinogenesis as shown by ISH, strong intensity was observed in the ICC and moderate intensity was observed in oval cells and CF. These observations suggest that the EGFR and STAT3 signal pathway contributes to the carcinogenesis of ICC. High activity of STAT3 during the carcinogenesis of ICC may be the result of high activity of EGFR triggered by TGFα.


Oncotarget | 2017

Protein tyrosine phosphatase receptor-type δ acts as a negative regulator suppressing breast cancer

Xiaotang Yu; Fan Zhang; Jun Mao; Ying Lu; Jiazhi Li; Wei Ma; Shujun Fan; Chunying Zhang; Qing Li; Bo Wang; Bo Song; Lianhong Li

Protein tyrosine phosphatase receptor-type δ (PTPRD) is frequently inactivated in human cancers. This study investigated the role of PTPRD in the regulation of stemness, epithelial-mesenchymal transition (EMT), and migration and invasion in breast cancer cells. In vitro, PTPRD silencing using siRNA enhanced the stem cell-like properties of breast cancer cells, including their mammosphere- and holoclone-forming abilities, and it promoted tumorigenicity in vivo. PTPRD knockdown also increased the CD44+/CD24− breast cancer stem cell (BCSC) population and the expression of the stem cell markers ALDH1 and OCT4. It also promoted migration and invasion by breast cancer cell, EMT, and activation of signal transducer and activator of transcription 3 (STAT3). BCSCs expressed low levels of PTPRD, displayed mesenchymal phenotypes, and were more sensitive to IL-6-mediated STAT3 activation than non-BCSCs. PTPRD expression was upregulated by IL-6 in breast cancer cells, thereby establishing a negative feedback circuit by which IL-6 induced canonical STAT3 phosphorylation and transiently upregulated PTPRD, which in turn dephosphorylated STAT3 and prevented downstream signaling via the IL-6/STAT3 cascade. These data suggest that therapies aimed at restoring or enhancing PTPRD expression may be effective in controlling breast cancer progression and metastasis.


Oncology Letters | 2016

Epidermal growth factor receptor expression and gene copy number analysis in gastric carcinoma samples from Chinese patients.

Fan Zhang; Xingwu Yang; Lianhong Li; Lei Sun; Bo Wang; Xiaotang Yu

Epidermal growth factor receptor (EGFR) expression and gene copy number have been observed to be associated with a positive clinical response to EGFR inhibitors. The present study aimed to evaluate EGFR expression and gene copy number in samples of gastric carcinoma (GC) from Chinese patients. EGFR expression and gene copy number were detected using immunohistochemistry and fluorescence in situ hybridization, in tissue array slides containing 150 individual samples of GC tissue. The association between EGFR status, clinicopathological features and overall patient survival was analyzed. Out of the 150 cases of GC evaluated, 63 (42.00%) demonstrated weak EGFR expression and 20 (13.33%) demonstrated EGFR overexpression. EGFR expression was observed to be associated with tumor location (P<0.05). Out of 104 cases of GC, which produced a clear FISH signal, 6 (5.77%) exhibited EGFR gene amplification and 5 (4.80%) exhibited balanced polysomy. Patients exhibiting GC, who demonstrated weak EGFR expression, EGFR overexpression or increased EGFR gene copy number, possessed an unfavorable prognosis. Multivariate analysis revealed that EGFR expression, tumor/node/metastasis stage and tumor location were potential independent unfavorable prognostic factors for GC patients. In conclusion, EGFR overexpression, gene amplification and polysomy were observed in GC patients and were associated with an unfavorable prognosis. Evaluation of EGFR status may therefore facilitate the identification of a subset of GC patients sensitive to treatment with EGFR-targeted therapies.

Collaboration


Dive into the Xiaotang Yu's collaboration.

Top Co-Authors

Avatar

Lianhong Li

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Jun Mao

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Shujun Fan

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Bo Wang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Bo Song

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Jiazhi Li

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Ma

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Ying Lu

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Huan Wang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhenhuan Hou

Dalian Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge