Network


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

Hotspot


Dive into the research topics where Jun Mao is active.

Publication


Featured researches published by Jun Mao.


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.


Cancer Science | 2015

MicroRNA-34a suppresses the breast cancer stem cell-like characteristics by downregulating Notch1 pathway

Le Kang; Jun Mao; Yajun Tao; Bo Song; Wei Ma; Ying Lu; Lijing Zhao; Jiazhi Li; Baoxue Yang; Lianhong Li

MicroRNAs play pivotal roles in cancer stem cell regulation. Previous studies have shown that microRNA‐34a (miR‐34a) is downregulated in human breast cancer. However, it is unknown whether and how miR‐34a regulates breast cancer stem cells. Notch signaling is one of the most important pathways in stem cell maintenance and function. In this study, we verified that miR‐34a directly and functionally targeted Notch1 in MCF‐7 cells. We reported that miR‐34a negatively regulated cell proliferation, migration, and invasion and breast cancer stem cell propagation by downregulating Notch1. The expression of miR‐34a was negatively correlated with tumor stages, metastasis, and Notch1 expression in breast cancer tissues. Furthermore, overexpression of miR‐34a increased chemosensitivity of breast cancer cells to paclitaxel (PTX) by downregulating the Notch1 pathway. Mammosphere formation and expression of the stemness factor ALDH1 were also reduced in the cells treated with miR‐34a and PTX compared to those treated with PTX alone. Taken together, our results indicate that miR‐34a inhibited breast cancer stemness and increased the chemosensitivity to PTX partially by downregulating the Notch1 pathway, suggesting that miR‐34a/Notch1 play an important role in regulating breast cancer stem cells. Thus miR‐34a is a potential target for prevention and therapy of breast cancer.


Biomedicine & Pharmacotherapy | 2016

MicroRNA-138 modulates metastasis and EMT in breast cancer cells by targeting vimentin

Jun Zhang; Dan Liu; Zhuo Feng; Jun Mao; Chunying Zhang; Ying Lu; Jiazhi Li; Qingqing Zhang; Qing Li; Lianhong Li

Increasing evidence indicates that dysregulation of microRNAs (miRNAs) plays critical roles in malignant transformation and tumor progression. In this study, in order to investigate the association of miR-138 with breast cancer we investigated the role of miR-138 in breast cancer metastasis. Levels of miR-138 were determined by qRT-PCR in 45 breast cancer samples. Cell migration and invasion assays were performed in a stably expressing miRNA-138 breast cancer cell line established using a lentivirus expression system. Epithelial-mesenchymal transition (EMT) was evaluated using qRT-PCR and Western Blots to detect epithelial marker E-cadherin and mesenchymal marker, vimentin. Luciferase reporter assays were used to identify downstream targets and biological function of miR-138. Breast cancer tissues had significantly lower expression of miR-138 compared to non-tumor tissues. Low miR-138 levels were associated with lymph node metastasis and invasion. miR-138 overexpression inhibited metastasis of breast cancer cells. miR-138 overexpression also down-regulated vimentin expression and upregulated E-cadherin expression, suggesting that miR-138 inhibited EMT. Our results support the involvement of miR-138 in breast tumorigenesis, especially lymph node metastasis. We propose that miR-138 might be used as therapeutic agent for breast cancer.


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

BACKGROUNDnThe 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.nnnMETHODSnHuman 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.nnnRESULTSnPTEN, 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.nnnCONCLUSIONSnOur 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.


Biomedicine & Pharmacotherapy | 2014

Annexin A7 and its binding protein galectin-3 influence mouse hepatocellular carcinoma cell line in vitro

Lin Song; Jun Mao; Jun Zhang; Mohammed Mohammed Ibrahim; Lianhong Li; Jianwu Tang

Lymph node metastasis is recognized as an important mode of liver cancer metastasis. Our previous study has built two hepatocarcinoma cell lines, Hca-F with high (75%) and Hca-P with low (25%) incidences of lymph node metastasis, and has indicated that annexin A7 is an important factor in the lymphatic metastasis of tumors. There is evidence that galectin-3 is the binding protein of annexin A7 and works in protein complexes. Our current study shows that both annexin A7 and galectin-3 express higher in Hca-F than Hca-P. Annexin A7 was successfully down-regulated in Hca-P by RNA interference, and this resulted in concomitant reduction of galactin 3 expression in annexin A7 down regulated compared to the control and N-control cells. Using CCK-8 assay, the expression level of annexin A7 and galectin-3 were found to have correlation with the proliferation ability; Transwell assay showed annexin A7 and galectin-3 are involved in cell migration and invasion regulation in mouse hepatocellular carcinoma cell lines, immunofluorescence assay indicate annexin A7 and galectin-3 were co-located annexin A7 and galectin-3 played roles in DNA damage and cell proliferation cycle checkpoint arrest pathway. Those phenomena indicated that annexin A7 influences lymphatic metastasis of tumors by interacting with galectin-3 through the regulation of tumor cell proliferation, attachment, migration and invasion.


Biomedicine & Pharmacotherapy | 2016

The mechanism between epithelial mesenchymal transition in breast cancer and hypoxia microenvironment

Tong Gao; Jiazhi Li; Ying Lu; Chunying Zhang; Qing Li; Jun Mao; Lianhong Li

Hypoxia microenvironment widely exists in solid tumor tissues, which is mainly due to the rapid growth of cells within the tumor more than the speed of capillary in neoplasm, resulting in tumor tissue hypoxia. In hypoxia, hypoxia inducible factor 1 (HIF-1) is activated and regulate the expression of a series of hypoxia inducible genes, resulting in a series of hypoxia adaptation reaction. Researchs proved that, HIF-1 is closely related to the invasion, metastasis, prognosis of the tumor, and the expression of HIF-1 is higher in metastatic tissues compared with primary cancer tissues. In the evolution process of breast cancer, epithelial mesenchymal transition (EMT) define the characteristics of migration and invasion of breast cancer cells, which can also allow cancer cells to acquire the ability of self-renewing and stemness, so as to promote the generation of breast cancer stem cells. The incidence of EMT cancer stem cells are higher within the resistant to conventional treatment. This review focuses on breast cancer (stem cells), targeting the mechanism between hypoxia and EMT in tumor (stem cells), with the purpose of finding the new therapy to 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.

Collaboration


Dive into the Jun Mao's collaboration.

Top Co-Authors

Avatar

Lianhong Li

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Ying Lu

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Bo Wang

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 Song

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Jianwu Tang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Jiazhi Li

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Jun Zhang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaotang Yu

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Ma

Dalian Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge