Network


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

Hotspot


Dive into the research topics where You-Hong Cui is active.

Publication


Featured researches published by You-Hong Cui.


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.


Hepatology | 2012

Nanog regulates self‐renewal of cancer stem cells through the insulin‐like growth factor pathway in human hepatocellular carcinoma

Juanjuan Shan; Junjie Shen; Limei Liu; Feng Xia; Chuan Xu; Guangjie Duan; Yanmin Xu; Qinghua Ma; Zhi Yang; Qianzhen Zhang; Leina Ma; Jia Liu; Senlin Xu; Xiaochu Yan; Ping Bie; You-Hong Cui; Xiu-wu Bian; Cheng Qian

Hepatocellular carcinoma (HCC) exhibits cellular heterogeneity and embryonic stem‐cell–related genes are preferentially overexpressed in a fraction of cancer cells of poorly differentiated tumors. However, it is not known whether or how these cancer cells contribute to tumor initiation and progression. Here, our data showed that increased expression of pluripotency transcription factor Nanog in cancer cells correlates with a worse clinical outcome in HCC. Using the Nanog promoter as a reporter system, we could successfully isolate a small subpopulation of Nanog‐positive cells. We demonstrate that Nanog‐positive cells exhibited enhanced ability of self‐renewal, clonogenicity, and initiation of tumors, which are consistent with crucial hallmarks in the definition of cancer stem cells (CSCs). NanogPos CSCs could differentiate into mature cancer cells in in vitro and in vivo conditions. In addition, we found that NanogPos CSCs exhibited resistance to therapeutic agents (e.g., sorafenib and cisplatin) and have a high capacity for tumor invasion and metastasis. Knock‐down expression of Nanog in NanogPos CSCs could decrease self‐renewal accompanied with decreased expression of stem‐cell–related genes and increased expression of mature hepatocyte‐related genes. Overexpression of Nanog in NanogNeg cells could restore self‐renewal. Furthermore, we found that insulin‐like growth factor (IGF)2 and IGF receptor (IGF1R) were up‐regulated in NanogPos CSCs. Knock‐down expression of Nanog in NanogPos CSCs inhibited the expression of IGF1R, and overexpression of Nanog in NanogNeg cells increased the expression of IGF1R. A specific inhibitor of IGF1R signaling could significantly inhibit self‐renewal and Nanog expression, indicating that IGF1R signaling participated in Nanog‐mediated self‐renewal. Conclusion: These data indicate that Nanog could be a novel biomarker for CSCs in HCC, and that Nanog could play a crucial role in maintaining the self‐renewal of CSCs through the IGF1R‐signaling pathway. (HEPATOLOGY 2012;56:1004–1014)


The Journal of Pathology | 2011

The chemokine CXCL12 and its receptor CXCR4 promote glioma stem cell‐mediated VEGF production and tumour angiogenesis via PI3K/AKT signalling

Yi-Fang Ping; Xiao-hong Yao; Jian-yong Jiang; Lin-tao Zhao; Shi-cang Yu; Tao Jiang; Marie Cm Lin; Jian-hong Chen; Bin Wang; Rong Zhang; You-Hong Cui; Cheng Qian; Ji Ming Wang; Xiu-wu Bian

Chemokines and their receptors are actively involved in inflammation, immune responses, and cancer development. Here we report the detection of CD133+ glioma stem‐like cells (GSCs) co‐expressing a chemokine receptor CXCR4 in human primary glioma tissues. These GSCs were located in areas adjacent to tumour vascular capillaries, suggesting an association between GSCs and tumour angiogenesis. To test this hypothesis, we isolated CD133+ GSCs from surgical specimens of human primary gliomas and glioma cell lines. As compared to CD133− cells, CD133+ GSCs expressed significantly higher levels of CXCR4 mRNA and protein, and migrated more efficiently in response to the CXCR4 ligand CXCL12. In addition, CXCL12 induced vascular endothelial growth factor (VEGF) production by CD133+ GSCs via activation of the PI3K/AKT signalling pathway. Furthermore, knocking down of CXCR4 using RNA interference or inhibition of CXCR4 function by an antagonist AMD3100 not only reduced VEGF production by CD133+ GSCs in vitro, but also attenuated the growth and angiogenesis of tumour xenografts in vivo formed by CD133+ GSCs in SCID mice. These results indicate that CXCL12 and its receptor CXCR4 promote GSC‐initiated glioma growth and angiogenesis by stimulating VEGF production. Copyright


Cancer Letters | 2011

MicroRNA-122 sensitizes HCC cancer cells to adriamycin and vincristine through modulating expression of MDR and inducing cell cycle arrest.

Yanmin Xu; Feng Xia; Leina Ma; Juanjuan Shan; Junjie Shen; Zhi Yang; Jia Liu; You-Hong Cui; Xiu-wu Bian; Ping Bie; Cheng Qian

Hepatocellular carcinoma (HCC) is a hypervascular cancer characterized by rapid progression as well as resistance to conventional chemotherapy. It has been shown that microRNAs play critical roles in pathogenesis of HCC. MicroRNA-122 (miR-122) is a liver-specific microRNA and is frequently downregulated in HCC. In the present study, we investigated whether restoration of miR-122 in HCC cells could render cells sensitive to chemotherapeutic agents adriamycin (ADM) or vincristine (VCR). Our data showed that overexpression of miR-122 in HCC cells induced by adenovirus expressing miR-122 could render cell sensitive to ADM or VCR. Analysis of cell cycle distribution showed that the anti-proliferative effect of miR-122 is associated with increase of cell number in the G2/M phase. Moreover, treatment with Ad-miR122 and ADM or VCR resulted in high accumulation of HCC cells in G2/M phase. We further demonstrated that overexpression of miR-122 could modulate the sensitivity of the HCC cells to chemotherapeutic drugs through downregulating MDR related genes MDR-1, GST-π, and MRP, antiapoptotic gene Bcl-w and cell cycle related gene cyclin B1. Taken together, our findings demonstrated that combination of Ad-miR122 with chemotherapeutic agents inhibited HCC cell growth by inducing G2/M arrest and that this arrest is associated, at least in part, with reduced expression of MDR related genes and Cyclin B1.


PLOS ONE | 2013

Decrease of 5-Hydroxymethylcytosine Is Associated with Progression of Hepatocellular Carcinoma through Downregulation of TET1

Chungang Liu; Limei Liu; Xuejiao Chen; Junjie Shen; Juanjuan Shan; Yanmin Xu; Zhi Yang; Lin Wu; Feng Xia; Ping Bie; You-Hong Cui; Xiu-wu Bian; Cheng Qian

DNA methylation is an important epigenetic modification and is frequently altered in cancer. Convert of 5-methylcytosine (5 mC) to 5-hydroxymethylcytosine (5 hmC) by ten-eleven translocation (TET) family enzymes plays important biological functions in embryonic stem cells, development, aging and disease. Recent reports showed that level of 5 hmC was altered in various types of cancers. However, the change of 5 hmC level in hepatocellular carcinoma (HCC) and association with clinical outcome were not well defined. Here, we reported that level of 5 hmC was decreased in HCC tissues, as compared with non-tumor tissues. Clincopathological analysis showed the decreased level of 5 hmC in HCC was associated with tumor size, AFP level and poor overall survival. We also found that the decreased level of 5 hmC in non-tumor tissues was associated with tumor recurrence in the first year after surgical resection. In an animal model with carcinogen DEN-induced HCC, we found that the level of 5 hmC was gradually decreased in the livers during the period of induction. There was further reduction of 5 hmC in tumor tissues when tumors were developed. In contrast, level of 5 mC was increased in HCC tissues and the increased 5 mC level was associated with capsular invasion, vascular thrombosis, tumor recurrence and overall survival. Furthermore, our data showed that expression of TET1, but not TET2 and TET3, was downregulated in HCC. Taken together, our data indicated 5 hmC may be served as a prognostic marker for HCC and the decreased expression of TET1 is likely one of the mechanisms underlying 5 hmC loss in HCC.


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.


Oncogene | 2015

Oncogenic miR-20a and miR-106a enhance the invasiveness of human glioma stem cells by directly targeting TIMP-2

Z Wang; B Wang; Y Shi; C Xu; H L Xiao; L N Ma; S L Xu; Lang Yang; Q L Wang; W Q Dang; S C Yu; Y F Ping; You-Hong Cui; Hsiang-Fu Kung; C Qian; Xia Zhang; Xiu-wu Bian

Emerging evidence has shown that cancer stem cells (CSCs) are the cellular determinants to promote cancer invasion and metastasis. However, the mechanism underlying CSC invasion remains unknown. MicroRNAs are evolutionally conserved small noncoding RNAs that are critical for the regulation of gene expression, and their expressions are often dysregulated in cancers. In the present study, we demonstrated that two functionally related microRNAs, miR-20a and -106a (miR-20a/106a), were capable of enhancing the invasiveness of CD133+ glioma stem cells (GSCs) isolated from both glioblastoma cell line U87 and primary human glioma specimens. We found that the level of miR-20a/106a in GSCs was significantly higher than that in the committed CD133− glioma cells, and correlated with the invasive capability of GSCs. By bioinformatic analysis, we identified tissue inhibitor of metalloproteinases-2 (TIMP-2) as one of the miR-20a/106a-targeted genes. TIMP-2 level correlated inversely with miR-20/106 expression. Directly targeting by miR-20a/106a on 3′-untranslation region (3′-UTR) of TIMP-2 mRNA was confirmed by 3′-UTR dual-luciferase reporter assay. Knockdown of miR-20a/106a in GSCs increased endogenous TIMP-2 protein abundance, thereby inhibiting GSC invasion. We also found that Nordy, a synthetic lipoxygenase inhibitor, inhibited GSC invasiveness by elevating the expression of TIMP-2 via downregulation of miR-20a/106a. Our results indicate that miR-20a/106a has a key role in GSC invasion and may serve as targets for treatment of glioblastoma.


PLOS ONE | 2013

A Novel Zebrafish Xenotransplantation Model for Study of Glioma Stem Cell Invasion

Xiao-jun Yang; Ai Gu; Chuan Xu; Shi-cang Yu; Ting-ting Li; You-Hong Cui; Xia Zhang; Xiu-wu Bian

Invasion and metastasis of solid tumors are the major causes of death in cancer patients. Cancer stem cells (CSCs) constitute a small fraction of tumor cell population, but play a critical role in tumor invasion and metastasis. The xenograft of tumor cells in immunodeficient mice is one of commonly used in vivo models to study the invasion and metastasis of cancer cells. However, this model is time-consuming and labor intensive. Zebrafish (Danio rerio) and their transparent embryos are emerging as a promising xenograft tumor model system for studies of tumor invasion. In this study, we established a tumor invasion model by using zebrafish embryo xenografted with human glioblastoma cell line U87 and its derived cancer stem cells (CSCs). We found that CSCs-enriched from U87 cells spreaded via the vessels within zebrafish embryos and such cells displayed an extremely high level of invasiveness which was associated with the up-regulated MMP-9 by CSCs. The invasion of glioma CSCs (GSCs) in zebrafish embryos was markedly inhibited by an MMP-9 inhibitor. Thus, our zebrafish embryo model is considered a cost-effective approach tostudies of the mechanisms underlying the invasion of CSCs and suitable for high-throughput screening of novel anti-tumor invasion/metastasis agents.


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.

Collaboration


Dive into the You-Hong Cui'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

Cheng Qian

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

Lang Yang

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

Peng Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Sen-lin Xu

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

Jing Yang

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge