Network


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

Hotspot


Dive into the research topics where Sen-lin Xu is active.

Publication


Featured researches published by Sen-lin Xu.


Journal of Immunology | 2012

Tumor-Associated Microglia/Macrophages Enhance the Invasion of Glioma Stem-like Cells via TGF-β1 Signaling Pathway

Xian-zong Ye; Sen-lin Xu; Yan-hong Xin; Shi-cang Yu; Yi-Fang Ping; Lu Chen; Hualiang Xiao; Bin Wang; Liang Yi; Qing-liang Wang; Xue-feng Jiang; Lang Yang; Peng Zhang; Cheng Qian; You-Hong Cui; Xia Zhang; Xiu-wu Bian

The invasion of malignant glioma cells into the surrounding normal brain tissues is crucial for causing the poor outcome of this tumor type. Recent studies suggest that glioma stem-like cells (GSLCs) mediate tumor invasion. However, it is not clear whether microenvironment factors, such as tumor-associated microglia/macrophages (TAM/Ms), also play important roles in promoting GSLC invasion. In this study, we found that in primary human gliomas and orthotopical transplanted syngeneic glioma, the number of TAM/Ms at the invasive front was correlated with the presence of CD133+ GSLCs, and these TAM/Ms produced high levels of TGF-β1. CD133+ GSLCs isolated from murine transplanted gliomas exhibited higher invasive potential after being cocultured with TAM/Ms, and the invasiveness was inhibited by neutralization of TGF-β1. We also found that human glioma-derived CD133+ GSLCs became more invasive upon treatment with TGF-β1. In addition, compared with CD133− committed tumor cells, CD133+ GSLCs expressed higher levels of type II TGF-β receptor (TGFBR2) mRNA and protein, and downregulation of TGFBR2 with short hairpin RNA inhibited the invasiveness of GSLCs. Mechanism studies revealed that TGF-β1 released by TAM/Ms promoted the expression of MMP-9 by GSLCs, and TGFBR2 knockdown reduced the invasiveness of these cells in vivo. These results demonstrate that TAM/Ms enhance the invasiveness of CD133+ GSLCs via the release of TGF-β1, which increases the production of MMP-9 by GSLCs. Therefore, the TGF-β1 signaling pathway is a potential therapeutic target for limiting the invasiveness of GSLCs.


Cancer Research | 2014

Metastatic consequences of immune escape from NK cell cytotoxicity by human breast cancer stem cells

Bin Wang; Qiang Wang; Zhe Wang; Jun Jiang; Shi-cang Yu; Yi-Fang Ping; Jing Yang; Sen-lin Xu; Xian-zong Ye; Chuan Xu; Lang Yang; Cheng Qian; Ji Ming Wang; You-Hong Cui; Xia Zhang; Xiu-wu Bian

Breast cancer stem-like cells (BCSC) are crucial for metastasis but the underlying mechanisms remain elusive. Here, we report that tumor-infiltrating natural killer (NK) cells failed to limit metastasis and were not associated with improved therapeutic outcome of BCSC-rich breast cancer. Primary BCSCs were resistant to cytotoxicity mediated by autologous/allogeneic NK cells due to reduced expression of MICA and MICB, two ligands for the stimulatory NK cell receptor NKG2D. Furthermore, the downregulation of MICA/MICB in BCSCs was mediated by aberrantly expressed oncogenic miR20a, which promoted the resistance of BCSC to NK cell cytotoxicity and resultant lung metastasis. The breast cancer cell differentiation-inducing agent, all-trans retinoic acid, restored the miR20a-MICA/MICB axis and sensitized BCSC to NK cell-mediated killing, thereby reducing immune escape-associated BCSC metastasis. Together, our findings reveal a novel mechanism for immune escape of human BCSC and identify the miR20a-MICA/MICB signaling axis as a therapeutic target to limit metastatic breast cancer.


Stem Cells | 2012

Connexin 43 Reverses Malignant Phenotypes of Glioma Stem Cells by Modulating E-Cadherin†‡§

Shi-cang Yu; Hualiang Xiao; Xue-feng Jiang; Qing-liang Wang; Yan Li; Xiao-jun Yang; Yi-Fang Ping; Jiang Jie Duan; Jian-yong Jiang; Xian-zong Ye; Sen-lin Xu; Yang‐Hong Xin; Xiao-hong Yao; Jian-hong Chen; Wei‐Hua Chu; Wei Sun; Bing Wang; Ji Ming Wang; Xia Zhang; Xiu-wu Bian

Malfunctioned gap junctional intercellular communication (GJIC) has been thought associated with malignant transformation of normal cells. However, the role of GJIC‐related proteins such as connexins in sustaining the malignant behavior of cancer stem cells remains unclear. In this study, we obtained tumorspheres formed by glioma stem cells (GSCs) and adherent GSCs and then examined their GJIC. All GSCs showed reduced GJIC, and differentiated glioma cells had more gap junction‐like structures than GSCs. GSCs expressed very low level of connexins, Cx43 in particular, which are key components of gap junction. We observed hypermethylation in the promoter of gap junction protein α1, which encodes Cx43 in GSCs. Reconstitution of Cx43 in GSCs inhibited their capacity of self‐renewal, invasiveness, and tumorigenicity via influencing E‐cadherin and its coding protein, which leads to changes in the expression of Wnt/β‐catenin targeting genes. Our results suggest that GSCs require the low expression of Cx43 for maintaining their malignant phenotype, and upregulation of Cx43 might be a potential strategy for treatment of malignant glioma. STEM CELLS 2012; 30:108‐120.


BMC Cancer | 2014

ALDH1A1 expression correlates with clinicopathologic features and poor prognosis of breast cancer patients: a systematic review and meta-analysis.

Ying Liu; Donglai Lv; Jiang-Jie Duan; Sen-lin Xu; Jing-fang Zhang; Xiao-jun Yang; Xia Zhang; You-Hong Cui; Xiu-wu Bian; Shi-cang Yu

BackgroundAldehyde dehydrogenase 1 family member A1 (ALDH1A1) has been identified as a putative cancer stem cell (CSC) marker in breast cancer. However, the clinicopathological and prognostic significance of this protein in breast cancer patients remains controversial.MethodsThis meta-analysis was conducted to address the above issues using 15 publications covering 921 ALDH1A1+ cases and 2353 controls. The overall and subcategory analyses were performed to detect the association between ALDH1A1 expression and clinicopathological/prognostic parameters in breast cancer patients.ResultsThe overall analysis showed that higher expression of ALDH1A1 is associated with larger tumor size, higher histological grade, greater possibility of lymph node metastasis (LNM), higher level expression of epidermal growth factor receptor 2 (HER2), and lower level expression of estrogen receptor (ER)/progesterone receptor (PR). The prognosis of breast cancer patients with ALDH1A1+ tumors was poorer than that of the ALDH1A1- patients. Although the relationships between ALDH1A1 expression and some clinicopathological parameters (tumor size, LNM, and the expression of HER2) was not definitive to some degree when we performed a subcategory analysis, the predictive values of ALDH1A1 expression for histological grade and survival of breast cancer patients were significant regardless of the different cutoff values of ALDH1A1 expression, the different districts where the patients were located, the different clinical stages of the patients, the difference in antibodies used in the studies, and the surgery status.ConclusionsOur results indicate that ALDH1A1 is a biomarker to predict tumor progression and poor survival of breast cancer patients. This marker should be taken into consideration in the development of new diagnostic and therapeutic program for breast cancer.


Modern Pathology | 2014

ALDH1A1 defines invasive cancer stem-like cells and predicts poor prognosis in patients with esophageal squamous cell carcinoma

Lang Yang; Yong Ren; Xi Yu; Feng Qian; Bai-shi-jiao Bian; Hualiang Xiao; Wei-Guang Wang; Sen-lin Xu; Jing Yang; Qiang Liu; Zhe Wang; Wei Guo; Gang Xiong; Kang Yang; Cheng Qian; Xia Zhang; Peng Zhang; You-Hong Cui; Xiu-wu Bian

Invasion and metastasis are the major cause of deaths in patients with esophageal cancer. In this study, we isolated cancer stem-like cells from an esophageal squamous cell carcinoma cell line EC109 based on aldehyde dehydrogenase 1A1 (ALDH1A1), and found that ALDH1A1high cells possessed the capacities of self-renewal, differentiation and tumor initiation, indications of stem cell properties. To support their stemness, ALDH1A1high cells exhibited increased potential of invasion and metastasis as compared with ALDH1A1low cells. ALDH1A1high esophageal squamous cell carcinoma cells expressed increased levels of mRNA for vimentin, matrix metalloproteinase 2, 7 and 9 (MMP2, MMP7 and MMP9), but decreased the level of E-cadherin mRNA, suggesting that epithelial–mesenchymal transition and secretary MMPs may be attributed to the high invasive and metastatic capabilities of ALDH1A1high cells. Furthermore, we examined esophageal squamous cell carcinoma specimens from 165 patients and found that ALDH1A1high cells were associated with esophageal squamous dysplasia and the grades, differentiation and invasion depth, lymph node metastasis and UICC stage of esophageal squamous cell carcinoma, as well as poor prognosis of patients. Our results provide the strong evidence that ALDH1A1high cancer stem-like cells contribute to the invasion, metastasis and poor outcome of human esophageal squamous cell carcinoma.


American Journal of Pathology | 2011

Annexin 1 Released by Necrotic Human Glioblastoma Cells Stimulates Tumor Cell Growth through the Formyl Peptide Receptor 1

Yan Yang; Ying Liu; Xiao-hong Yao; Yi-Fang Ping; Tao Jiang; Qin Liu; Sen-lin Xu; Jian Huang; Haiwei Mou; Wanghua Gong; Keqiang Chen; Xiu-wu Bian; Ji Ming Wang

Highly malignant human gliomas overexpress the G-protein-coupled chemoattractant receptor formyl peptide receptor (FPR1), which promotes tumor progression when activated. Our previous studies demonstrated that necrotic glioblastoma cells release chemotactic agonist(s) that activate FPR1 on viable tumor cells. In the present study, we identified an FPR1 agonist released by necrotic human glioblastoma cells. Necrotic tumor cell supernatant (NecSup) contained Annexin 1 (Anx A1), a chemotatic polypeptide agonist for FPR1. Immunoabsorption of Anx A1 with a specific antibody markedly reduced the chemotactic activity of NecSup for tumor cells and diminished its capacity to promote tumor cell growth, invasion, and colony formation on soft agar. In addition, Anx A1 was present in tumor xenografts formed by human glioblastoma cells in nude mice. Anx A1 knockdown significantly reduced the tumorigenicity of glioblastoma cells in nude mice, but FPR1/Anx A1 double knockdown diminished tumor growth even further. The clinical relevance of Anx A1 in gliomas was supported by the observation that Anx A1 was more highly expressed in poorly differentiated human primary gliomas compared with lower grade tumors. Our study implicates Anx A1 as a major component in necrotic tumor cell-derived stimulants of the growth of glioblastoma via the activation of FPR1.


Stem Cells and Development | 2013

Strategies for Isolating and Enriching Cancer Stem Cells: Well Begun Is Half Done

Jiang-Jie Duan; Wen Qiu; Sen-lin Xu; Bin Wang; Xian-zong Ye; Yi-Fang Ping; Xia Zhang; Xiu-wu Bian; Shi-cang Yu

Cancer stem cells (CSCs) constitute a subpopulation of cancer cells that have the potential for self-renewal, multipotent differentiation, and tumorigenicity. Studies on CSC biology and CSC-targeted therapies depend on CSC isolation and/or enrichment methodologies. Scientists have conducted extensive research in this field since John Dicks group successfully isolated CSCs based on the expression of the CD34 and CD38 surface markers. Progress in CSC research has been greatly facilitated by the enrichment and isolation of these cells. In this review, we summarize the current strategies used in our and other laboratories for CSC isolation and enrichment, including methods based on stem cell surface markers, intracellular enzyme activity, the concentration of reactive oxygen species, the mitochondrial membrane potential, promoter-driven fluorescent protein expression, autofluorescence, suspension/adherent culture, cell division, the identification of side population cells, resistance to cytotoxic compounds or hypoxia, invasiveness/adhesion, immunoselection, and physical property. Although many challenges remain to be overcome, it is reasonable to believe that more reliable, efficient, and convenient methods will be developed in the near future.


Cancer Letters | 2015

Semaphorin-3F suppresses the stemness of colorectal cancer cells by inactivating Rac1

Jun Rao; Zhi-hang Zhou; Jing Yang; Yu Shi; Sen-lin Xu; Bin Wang; Yi-Fang Ping; Lu Chen; You-Hong Cui; Xia Zhang; Feng Wu; Xiu-wu Bian

Tumor cell stemness has been recognized as a key contributor to tumor initiation, progression and recurrence. Our previous studies have found that semaphorin-3F (SEMA3F), an axon guidance molecule in the development of central nervous system, inhibited the growth and metastasis of colorectal cancer (CRC). However, a possible role for SEMA3F in regulating cancer cell stemness remains unknown. Here, we report a novel mechanism of the acquirement of stemness of CRC cells regulated by SEMA3F. Knockdown of SEMA3F significantly promoted the self-renewal and tumorigenicity of CRC cells, and increased the expression of stemness-associated genes, while overexpressing SEMA3F reduced the stemness of CRC cells. Mechanistically, GTP-Rac1 was involved in SEMA3F mediated regulation of CRC cell stemness by targeting the Wnt/β-catenin pathway. Clinically, GTP-Rac1 expression was inversely correlated with SEMA3F levels in CRC samples and patients with SEMA3F(low)/GTP-Rac1(high) CRC showed poorer prognosis. Our findings demonstrate the ability of SEMA3F to inhibit the stemness of human CRC cells by suppressing Rac1 activation, which suggests a novel therapeutic approach for CRC.


The Journal of Pathology | 2015

SEMA3F prevents metastasis of colorectal cancer by PI3K–AKT‐dependent down‐regulation of the ASCL2–CXCR4 axis

Zhi-hang Zhou; Jun Rao; Jing Yang; Feng Wu; Juan Tan; Sen-lin Xu; Yanqing Ding; Na Zhan; Xu-Gang Hu; You-Hong Cui; Xia Zhang; Weiguo Dong; Xindong Liu; Xiu-wu Bian

Semaphorin‐3F (SEMA3F), an axonal repulsant in nerve development, has been shown to inhibit the progression of human colorectal cancer (CRC); however, the underlying mechanism remains elusive. In this study we found a negative correlation between the levels of SEMA3F and CXCR4 in CRC specimens from 85 patients, confirmed by bioinformatics analysis of gene expression in 229 CRC samples from the Cancer Genome Atlas. SEMA3Fhigh/CXCR4low patients showed the lowest frequency of lymph node and distant metastasis and the longest survival. Mechanistically, SEMA3F inhibited the invasion and metastasis of CRC cells through PI3K–AKT‐dependent down‐regulation of the ASCL2–CXCR4 axis. Specifically, ASCL2 enhanced the invasion and metastasis of CRC cells in vitro and expression of ASCL2 correlated with distant metastasis, tumour size and poor overall survival in CRC patients. Treatment of CRC cells with the CXCR4 antagonist AMD3100 attenuated SEMA3F knockdown‐induced invasion and metastasis of CRC cells in vitro and in vivo. Our study thus demonstrates that SEMA3F functions as a suppressor of CRC metastasis via down‐regulating the ASCL2–CXCR4 axis. Copyright


Cell Death & Differentiation | 2017

Tetraspanin CD9 stabilizes gp130 by preventing its ubiquitin-dependent lysosomal degradation to promote STAT3 activation in glioma stem cells

Yu Shi; Wenchao Zhou; Lin Cheng; Cong Chen; Zhi Huang; Xiaoguang Fang; Qiulian Wu; Zhi-Cheng He; Sen-lin Xu; Justin D. Lathia; Yi-Fang Ping; Jeremy N. Rich; Xiu Wu Bian; Shideng Bao

Glioblastoma (GBM) is the most malignant and lethal brain tumor harboring glioma stem cells (GSCs) that promote tumor propagation and therapeutic resistance. GSCs preferentially express several critical cell surface molecules that regulate the pro-survival signaling for maintaining the stem cell-like phenotype. Tetraspanin CD9 has recently been reported as a GSC biomarker that is relevant to the GSC maintenance. However, the underlying molecular mechanisms of CD9 in maintaining GSC property remain elusive. Herein, we report that CD9 stabilizes the IL-6 receptor glycoprotein 130 (gp130) by preventing its ubiquitin-dependent lysosomal degradation to facilitate the STAT3 activation in GSCs. CD9 is preferentially expressed in GSCs of human GBM tumors. Mass spectrometry analysis identified gp130 as an interacting protein of CD9 in GSCs, which was confirmed by immunoprecipitation and immunofluorescent analyses. Disrupting CD9 or gp130 by shRNA significantly inhibited the self-renewal and promoted the differentiation of GSCs. Moreover, CD9 disruption markedly reduced gp130 protein levels and STAT3 activating phosphorylation in GSCs. CD9 stabilized gp130 by preventing its ubiquitin-dependent lysosomal degradation to promote the BMX-STAT3 signaling in GSCs. Importantly, targeting CD9 potently inhibited GSC tumor growth in vivo, while ectopic expression of the constitutively activated STAT3 (STAT3-C) restored the tumor growth impaired by CD9 disruption. Collectively, we uncovered a critical regulatory mechanism mediated by tetraspanin CD9 to maintain the stem cell-like property and tumorigenic potential of GSCs.

Collaboration


Dive into the Sen-lin Xu's collaboration.

Top Co-Authors

Avatar

Xiu-wu Bian

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xia Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

You-Hong Cui

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Shi-cang Yu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yi-Fang Ping

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jiang-Jie Duan

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jing Yang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Lang Yang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yu Shi

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xian-zong Ye

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge