Network


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

Hotspot


Dive into the research topics where An-Gang Yang is active.

Publication


Featured researches published by An-Gang Yang.


PLOS ONE | 2010

MicroRNA Let-7a Inhibits Proliferation of Human Prostate Cancer Cells In Vitro and In Vivo by Targeting E2F2 and CCND2

Qingchuan Dong; Ping Meng; Tao Wang; Weiwei Qin; Weijun Qin; Fuli Wang; Jianlin Yuan; Zhinan Chen; An-Gang Yang; He-Yao Wang

Background Previous work has shown reduced expression levels of let-7 in lung tumors. But little is known about the expression or mechanisms of let-7a in prostate cancer. In this study, we used in vitro and in vivo approaches to investigate whether E2F2 and CCND2 are direct targets of let-7a, and if let-7a acts as a tumor suppressor in prostate cancer by down-regulating E2F2 and CCND2. Methodology/Principal Findings Real-time RT-PCR demonstrated that decreased levels of let-7a are present in resected prostate cancer samples and prostate cancer cell lines. Cellular proliferation was inhibited in PC3 cells and LNCaP cells after transfection with let-7a. Cell cycle analysis showed that let-7a induced cell cycle arrest at the G1/S phase. A dual-luciferase reporter assay demonstrated that the 3′UTR of E2F2 and CCND2 were directly bound to let-7a and western blotting analysis further indicated that let-7a down-regulated the expression of E2F2 and CCND2. Our xenograft models of prostate cancer confirmed the capability of let-7a to inhibit prostate tumor development in vivo. Conclusions/Significance These findings help to unravel the anti-proliferative mechanisms of let-7a in prostate cancer. Let-7a may also be novel therapeutic candidate for prostate cancer given its ability to induce cell-cycle arrest and inhibit cell growth, especially in hormone-refractory prostate cancer.


Molecular and Cellular Biochemistry | 2007

Notch activation promotes cell proliferation and the formation of neural stem cell-like colonies in human glioma cells

Xueping Zhang; Gang Zheng; Lian Zou; Hui-Ling Liu; Li-Hong Hou; Peng Zhou; Dan-Dan Yin; Qi-Jun Zheng; Liang Liang; Su-Zhen Zhang; Lei Feng; Libo Yao; An-Gang Yang; Hua Han; Jingyuan Chen

Since Notch signaling plays a critical role in stem cells and oncogenesis, we hypothesized that Notch signaling might play roles in cancer stem cells and cancer cells with a stem cell phenotype. In this study, we accessed potential functions of the Notch pathway in the formation of cancer stem cells using human glioma. Using RT-PCR, we found that most human astrogliomas of different grades expressed moderate to high level of Notch receptors and ligands. mRNA of Hes5 but not Hes1, both of which are major downstream molecules of the Notch pathway, was also detected. In human glioma cell lines BT325, U251, SHG-44, and U87, mRNA encoding different types of Notch receptors were detected, but active form of Notch1 (NIC) was only detected in SHG-44 and U87 by Western blot. Interestingly, proliferation of these two glioma cell lines appeared faster than that of the other two lines in which NIC was not detected. We have over-expressed NIC of Notch1 in SHG-44 cells by constitutive transfection to evaluate the effects of Notch signaling on glioma cells. Our results showed that over-expression of NIC in SHG-44 cells promoted the growth and the colony-forming activity of SHG-44 cells. Interestingly, over-expression of NIC increased the formation neurosphere-like colonies in the presence of growth factors. These colonies expressed nestin, and could be induced to cells expressing neuron-, astrocyte-, or oligodendrocyte-specific markers, consistent with phenotypes of neural stem cells. These data suggest that Notch signaling promote the formation of cancer stem cell-like cells in human glioma.


PLOS ONE | 2013

Overexpression of YAP and TAZ is an independent predictor of prognosis in colorectal cancer and related to the proliferation and metastasis of colon cancer cells.

Lijuan Wang; Shengjia Shi; Zhang-Yan Guo; Xiang Po Zhang; Suxia Han; An-Gang Yang; Wei-Hong Wen; Qing Zhu

Background and Objective Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are nuclear effectors of the Hippo pathway. Although they are abundantly expressed in the cytoplasm and nuclei of human colorectal cancer (CRC), and related to tumor proliferation status, there have been few studies on the predictive role of YAP and TAZ expression on the overall survival of patients with CRC. This study investigated YAP and TAZ expression in both CRC patients and colon cancer cell lines, and assessed their prognostic value. Methods Paraffin-embedded specimens from 168 eligible patients were used to investigate YAP and TAZ expression by immunohistochemistry, and compared with experimental results in colon cancer HCT116 cell line to explore their clinical significance in CRC. Results Statistically significant positive correlations were found between protein expression of YAP and TAZ in CRC tissues. Patients with higher YAP or TAZ expression showed a trend of shorter survival times; more importantly, our cohort study indicated that patients with both YAP and TAZ overexpression presented the worst outcomes. This was supported by multivariate analysis. In HCT116 colon cancer cells, the capacity for proliferation, metastasis, and invasion was dramatically reduced by knockdown of YAP and TAZ expressions by siRNA. Conclusions Co-overexpression of YAP and TAZ is an independent predictor of prognosis for patients with CRC, and may account for the higher proliferation, metastasis, and poor survival outcome of these patients.


Hepatology | 2014

c‐Myc‐mediated epigenetic silencing of MicroRNA‐101 contributes to dysregulation of multiple pathways in hepatocellular carcinoma

Lei Wang; Xiang Zhang; Lin-Tao Jia; Sijun Hu; Jing Zhao; Jiandong Yang; Weihong Wen; Zhe Wang; Tao Wang; Jun Zhao; Ruian Wang; Yan-Ling Meng; Yongzhan Nie; Kefeng Dou; Si-Yi Chen; Libo Yao; Daiming Fan; Rui Zhang; An-Gang Yang

The MYC oncogene is overexpressed in hepatocellular carcinoma (HCC) and has been associated with widespread microRNA (miRNA) repression; however, the underlying mechanisms are largely unknown. Here, we report that the c‐Myc oncogenic transcription factor physically interacts with enhancer of zeste homolog 2 (EZH2), a core enzymatic unit of polycomb repressive complex 2 (PRC2). Furthermore, miR‐101, an important tumor‐suppressive miRNA in human hepatocarcinomas, is epigenetically repressed by PRC2 complex in a c‐Myc‐mediated manner. miR‐101, in turn, inhibits the expression of two subunits of PRC2 (EZH2 and EED), thus creating a double‐negative feedback loop that regulates the process of hepatocarcinogenesis. Restoration of miR‐101 expression suppresses multiple malignant phenotypes of HCC cells by coordinate repression of a cohort of oncogenes, including STMN1, JUNB, and CXCR7, and further increases expression of endogenous miR‐101 by inhibition of PRC2 activation. In addition, co‐overexpression of c‐Myc and EZH2 in HCC samples was closely associated with lower expression of miR‐101 (P < 0.0001) and poorer prognosis of HCC patients (P < 0.01). Conclusions: c‐Myc collaborates with EZH2‐containing PRC2 complex in silencing tumor‐suppressive miRNAs during hepatocarcinogenesis and provides promising therapeutic candidates for human HCC. (Hepatology 2014;59:1850–1863)


Gastroenterology | 2011

HER2 Interacts With CD44 to Up-regulate CXCR4 via Epigenetic Silencing of microRNA-139 in Gastric Cancer Cells

Wei Bao; Hai–Jing Fu; Qiao–Sheng Xie; Lei Wang; Rui Zhang; Zhang–Yan Guo; Jing Zhao; Yan–Ling Meng; Xin–Ling Ren; Tao Wang; Qing Li; Bo–Quan Jin; Li–Bo Yao; Rui–An Wang; Dai–Ming Fan; Si-Yi Chen; Lin Tao Jia; An-Gang Yang

BACKGROUND & AIMS Human epidermal growth factor receptor 2 (HER2) (neu/ERBB2) is overexpressed on many types of cancer cells, including gastric cancer cells; HER2 overexpression has been associated with metastasis and poor prognosis. We investigated the mechanisms by which HER2 regulates cell migration and invasion. METHODS HER2 expression or activity was reduced in gastric cancer cell lines using small interfering RNAs or the monoclonal antibody, trastuzumab. We identified proteins that interact with HER2 or microRNAs (miRNAs) involved in HER2 signaling. We used various software programs to identify miRNAs that regulate factors in the HER2 signaling pathway. We analyzed expression patterns of these miRNAs in gastric cancer cell lines and tumor samples from patients. RESULTS We found that CD44 binds directly to HER2, which up-regulates the expression of metastasis-associated protein-1, induces deacetylation of histone H3 lysine 9, and suppresses transcription of microRNA139 (miR-139) to inhibit expression of its target gene, C-X-C chemokine receptor type 4 (CXCR4). Knockdown of HER2 and CD44 reduced invasive activity of cultured gastric cancer cells and suppressed tumor growth in nude mice. Lymph node metastasis was associated with high levels of HER2, CD44, and CXCR4, and reduced levels of miR-139 in human metastatic gastric tumors. Cultures of different types of metastatic cancer cells with histone deacetylase inhibitors and/or DNA methyltransferase resulted in up-regulation of miR-139. CONCLUSIONS HER2 interaction with CD44 up-regulates CXCR4 by inhibiting expression of miR-139, at the epigenetic level, in gastric cancer cells. These findings indicate how HER2 signaling might promote gastric tumor progression and metastasis.


The FASEB Journal | 2008

RBP-J, the transcription factor downstream of Notch receptors, is essential for the maintenance of vascular homeostasis in adult mice

Guo-Rui Dou; Yao-Chun Wang; Xing-Bin Hu; Lihong Hou; Chun-Mei Wang; Jian-Feng Xu; Yusheng Wang; Yingmin Liang; Libo Yao; An-Gang Yang; Hua Han

In adults, angiogenic abnormalities are involved in not only tumor growth but several human inherited diseases as well. It is unclear, however, concerning how the normal vascular structure is maintained and how angiogenesis is initiated in normal adults. Using the Cre‐LoxP‐mediated conditional gene deletion, we show in the present study that in adult mice disruption of the transcription factor recombination signal‐binding protein Jκ (RBP‐J) in endothelial cells strikingly induced spontaneous angiogenesis in multiple tissues, including retina and cornea, as well as in internal organs, such as liver and lung. In a choroidal neovascularization model, which mimics the angiogenic process in tumor growth and age‐related macular degeneration, RBP‐J deficiency induced a more intensive angiogenic response to injury. This could be transmitted by bone marrow, indicating that RBP‐J could modulate bone marrow‐derived endothelial progenitor cells in adult angiogenesis. In addition, in the absence of RBP‐J, proliferation of endothelial cells increased significantly, leading to accumulative vessel outgrowth. These findings suggest that in adults RBP‐J‐mediated Notch signaling may play an essential role in the maintenance of vascular homeostasis by repressing endothelial cell proliferation.—Dou, G.‐R., Wang, Y.‐C., Hu, X.‐B., Hou, L.‐H., Wang, C.‐M., Xu, J.‐F., Wang, Y.‐S., Liang, Y.‐M., Yao, L.‐B., Yang, A.‐G., Han, H. RBP‐J, the transcription factor downstream of Notch receptors, is essential for the maintenance of vascular homeostasis in adult mice. FASEB J. 22, 1606–1617 (2008)


Biochemical and Biophysical Research Communications | 2012

MicroRNA-320a suppresses human colon cancer cell proliferation by directly targeting β-catenin

Jianyong Sun; Yi Huang; Jipeng Li; Xiang Zhang; Lei Wang; Yan-Ling Meng; Bo Yan; Yongqian Bian; Jing Zhao; Weizhong Wang; An-Gang Yang; Rui Zhang

Recent profile studies of microRNA (miRNA) expression have documented a deregulation of miRNA (miR-320a) in human colorectal carcinoma. However, its expression pattern and underlying mechanisms in the development and progression of colorectal carcinoma has not been elucidated clearly. Here, we performed real-time PCR to examine the expression levels of miR-320a in colon cancer cell lines and tumor tissues. And then, we investigated its biological functions in colon cancer cells by a gain of functional strategy. Further more, by the combinational approaches of bioinformatics and experimental validation, we confirmed target associations of miR-320a in colorectal carcinoma. Our results showed that miR-320a was frequently downregulated in cancer cell lines and colon cancer tissues. And we demonstrated that miR-320a restoration inhibited colon cancer cell proliferation and β-catenin, a functionally oncogenic molecule was a direct target gene of miR-320a. Finally, the data of real-time PCR showed the reciprocal relationship between miR-320a and β-catenins downstream genes in colon cancer tissues. These findings indicate that miR-320a suppresses the growth of colon cancer cells by directly targeting β-catenin, suggesting its application in prognosis prediction and cancer treatment.


Journal of Biological Chemistry | 2006

The Repression of Human Differentiation-related Gene NDRG2 Expression by Myc via Miz-1-dependent Interaction with the NDRG2 Core Promoter

Jian Zhang; Fuyang Li; Xinping Liu; Lan Shen; Junye Liu; Jin Su; Wei Zhang; Yanchun Deng; Li-Feng Wang; Na Liu; Wei Han; Jing Zhang; Shaoping Ji; An-Gang Yang; Hua Han; Libo Yao

The N-myc downstream-regulated gene 1 (ndrg1) is highly expressed in N-myc knock-out mice through an unknown regulatory mechanism. As one member of the human NDRG gene family, NDRG2 encodes a protein highly homologous to Ndrg1. However, it is uncertain whether the expression of human NDRG2 is regulated by Myc because mouse ndrg2 and -3 are not affected by Myc. In this study, we provide the novel evidence that the expression of human NDRG2 is down-regulated by Myc via transcriptional repression. A high level of NDRG2 was observed as Myc expression was reduced in differentiated cells, whereas a low level of NDRG2 was shown following increased Myc expression upon serum stimulation. The ectopic expression of c-Myc dramatically reduces the cellular Ndrg2 protein and mRNA level. We further identified the core promoter region of NDRG2 that is required for Myc repression on NDRG2 transcription, and we verified the interaction of Myc with the core promoter region both in vitro and in vivo. Moreover, the c-Myc-mediated repression of NDRG2 requires association with Miz-1, and possibly the recruitment of other epigenetic factors, such as histone deacetylases, to the promoter. The regulatory function of Myc on NDRG2 gene expression implicated the role of the Ndrg2 in regulating cell differentiation.


Carcinogenesis | 2013

Pseudogene OCT4-pg4 functions as a natural micro RNA sponge to regulate OCT4 expression by competing for miR-145 in hepatocellular carcinoma

Lei Wang; Zhang-Yan Guo; Rui Zhang; Bo Xin; Rui Chen; Jing Zhao; Tao Wang; Weihong Wen; Lin-Tao Jia; Libo Yao; An-Gang Yang

The POU transcription factor OCT4 is a pleiotropic regulator of gene expression in embryonic stem cells. Recent studies demonstrated that OCT4 is aberrantly expressed in multiple types of human cancer; however, the underlying molecular mechanism remains largely unknown. In this study, we report that OCT4-pg4, a pseudogene of OCT4, is abnormally activated in hepatocellular carcinoma (HCC). The expression level of OCT4-pg4 is positively correlated with that of OCT4, and both gene transcripts can be directly targeted by a tumor-suppressive micro RNA miR-145. We find that the non-coding RNA OCT4-pg4 is biologically active, as it can upregulate OCT4 protein level in HCC. Mechanistic analysis revealed that OCT4-pg4 functions as a natural micro RNA sponge to protect OCT4 transcript from being inhibited by miR-145. In addition, our study also showed that OCT4-pg4 can promote growth and tumorigenicity of HCC cells, thus exerting an oncogenic role in hepatocarcinogenesis. Furthermore, survival analysis suggests that high OCT4-pg4 level is significantly correlated with poor prognosis of HCC patients. Taken together, our finding adds a new layer of post-transcriptional regulation of OCT4 and sheds new light on the treatment of human HCC.


Nucleic Acids Research | 2008

N-Myc downstream-regulated gene 2 is involved in p53-mediated apoptosis

Na Liu; Li-Feng Wang; Xia Li; Qi Yang; Xinping Liu; Jing Zhang; Jian Zhang; Yousheng Wu; Shaoping Ji; Yingqi Zhang; An-Gang Yang; Hua Han; Libo Yao

The tumor suppressor, p53, is a transcription factor which can modulate the transcription of a number of target genes that are involved in cell-cycle arrest and apoptosis. However, the apoptotic pathway mediated by p53 is not fully understood. Here, we showed that N-Myc downstream-regulated gene 2 (NDRG2) is a new target gene that is regulated by p53. NDRG2 mRNA and protein levels can be upregulated in a p53-dependent manner. The first intron of the NDRG2 gene contains a site that binds p53 directly and mediates wild-type p53-dependent transactivation. In addition, silencing of NDRG2 attenuates p53-mediated apoptosis, whereas overexpression of NDRG2 suppresses tumor cell growth, regardless of the presence or absence of p53. Our results indicate that NDRG2 is a novel p53-inducible target that is involved in the p53-mediated apoptosis pathway.

Collaboration


Dive into the An-Gang Yang's collaboration.

Top Co-Authors

Avatar

Lin-Tao Jia

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Boquan Jin

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jing Zhao

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Rui Zhang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Tao Wang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Libo Yao

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Si-Yi Chen

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Kun Yang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhuwei Xu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yan-Ling Meng

Fourth Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge