Network


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

Hotspot


Dive into the research topics where Baoping Cao is active.

Publication


Featured researches published by Baoping Cao.


Epigenomics | 2012

Methylation of TFPI-2 is an early event of esophageal carcinogenesis

Yan Jia; Yunsheng Yang; Malcolm V. Brock; Baoping Cao; Qimin Zhan; Yazhuo Li; Yuanzi Yu; James G. Herman; Mingzhou Guo

AIMS To explore the epigenetic changes and the function of TFPI-2 in esophageal cancer. MATERIALS & METHODS Nine esophageal cancer cell lines, nine normal esophageal mucosa, 60 esophageal dysplasia and 106 advanced esophageal cancer samples were included in this study. TFPI-2 methylation was examined by methylation-specific PCR. TFPI-2 expression was evaluated by immunohistochemistry in tissue samples. The effect of TFPI-2 on proliferation, apoptosis, invasion and migration was analyzed by colony formation assay, western blot assay, transwell assay and flow cytometric analysis. RESULTS TFPI-2 expression was regulated by promoter region hypermethylation in human esophageal cancer cell lines, and TFPI-2 expression is inversely correlated with methylation in primary cancer. Methylation was found in 28.2, 33.3 and 33.3% of grade 1, 2 and 3 esophageal dysplasia, and 67% of primary esophageal cancer, but no methylation was found in normal mucosa. Methylation is significantly related to tumor differentiation. Inhibition of invasion, migration, colony formation and proliferation, and induction of apoptosis occurred with the restoration of TFPI-2 expression in the KYSE70 cell line. CONCLUSION TFPI-2 is frequently methylated in esophageal cancer with a progression tendency. TFPI-2 is a potential tumor suppressor in esophageal cancer.


Tumor Biology | 2012

AKT signaling pathway activated by HIN-1 methylation in non-small cell lung cancer

Yuanzi Yu; Dongtao Yin; Mohammad O. Hoque; Baoping Cao; Yan Jia; Yunsheng Yang; Mingzhou Guo

The purpose of this study is to determine the epigenetic changes and function of High in Normal-1 (HIN-1) in non-small cell lung cancer (NSCLC). HIN-1 expression was examined by semiquantitative RT-PCR before and after 5-aza-2′-deoxycytidine (5-aza) treatment in NSCLC cell lines. Promoter methylation status of HIN-1 was tested by methylation-specific PCR (MSP). Effect of forced expression of HIN-1 on different key molecules of AKT signaling pathway was tested by Western Blot analysis in H157 and H23 cell lines. Promoter methylations are inversely correlated with expression of HIN-1 in eight (H23, H157, 95D, H1299, H358, H1752, H460, A549) of ten NSCLC cell lines and re-expression was observed by 5-aza treatment. We then tested promoter methylation of HIN-1 in primary NSCLC tissues. Methylation was detected in 73 out of 152 (48%) NSCLC cases. Forced expression of HIN-1in NSCLC cell lines inhibited colony formation and induce apoptosis. Furthermore, overexpression of HIN-1 reduces the expression of phosphorated-AKT (p-AKT), c-myc, Bcl-2 and cyclinD1 while Bax was increased. Our data suggest that HIN-1 is a potential tumor suppressor gene in NSCLC, silenced by promoter hypermethylation and negatively regulate AKT signaling pathway.


Oncotarget | 2016

Methylation of DACT2 accelerates esophageal cancer development by activating Wnt signaling

Meiying Zhang; Enqiang Linghu; Qimin Zhan; Tao He; Baoping Cao; Malcolm V. Brock; James G. Herman; Rong Xiang; Mingzhou Guo

Esophageal cancer is one of the most common malignancies worldwide. DACT2 is frequently methylated in human lung, hepatic, gastric and thyroid cancers. The methylation status and function of DACT2 remain to be elucidated in human esophageal cancer. Ten esophageal cancer cell lines, 42 cases of dysplasia and 126 cases of primary esophageal cancer samples were analyzed in this study. The expression of DACT2 was detected in YES2 cells, while reduced DACT2 expression levels were found in TE8 and KYSE70 cells, and complete loss of DACT2 expression was found in KYSE30, KYSE140, KYSE150, KYSE410, KYSE450, TE3 and TE7 cells. Loss of expression or reduced expression of DACT2 correlated with promoter region hypermethylation in esophageal cancer cells. Restoration of DACT2 expression was induced by 5-aza-2′-deoxycytidine. In human primary esophageal squamous carcinoma, 69% (87/126) of samples were methylated. Methylation of DACT2 was significantly associated with tumor stage and metastasis (P < 0.01, P < 0.05). DACT2 suppressed colony formation, cell migration and invasion in esophageal cancer cells, and it also suppressed esophageal cancer cell xenograft growth. DACT2 inhibited Wnt signaling in human esophageal cancer cells. In conclusion, DACT2 is frequently methylated in human esophageal cancer and its expression is regulated by promoter region methylation. DACT2 suppresses esophageal cancer growth by inhibiting Wnt signaling.


Oncotarget | 2015

Silencing NKD2 by promoter region hypermethylation promotes gastric cancer invasion and metastasis by up-regulating SOX18 in human gastric cancer

Yan Jia; Baoping Cao; Yunsheng Yang; Enqiang Linghu; Qimin Zhan; Youyong Lu; Yingyan Yu; James G. Herman; Mingzhou Guo

Naked cuticle homolog2 (NKD2) is located in chromosome 5p15.3, which is frequently loss of heterozygosity in human colorectal and gastric cancers. In order to understand the mechanism of NKD2 in gastric cancer development, 6 gastric cancer cell lines and 196 cases of human primary gastric cancer samples were involved. Methylation specific PCR (MSP), gene expression array, flow cytometry, transwell assay and xenograft mice model were employed in this study. The expression of NKD1 and NKD2 was silenced by promoter region hypermethylation. NKD1 and NKD2 were methylated in 11.7% (23/196) and 53.1% (104/196) in human primary gastric cancer samples. NKD2 methylation is associated with cell differentiation, TNM stage and distant metastasis significantly (all P < 0.05), and the overall survival time is longer in NKD2 unmethylated group compared to NKD2 methylated group (P < 0.05). Restoration of NKD2 expression suppressed cell proliferation, colony formation, cell invasion and migration, induced G2/M phase arrest, and sensitized cancer cells to docetaxel. NKD2 inhibits SOX18 and MMP-2,7,9 expression and suppresses BGC823 cell xenograft growth. In conclusion, NKD2 methylation may serve as a poor prognostic and chemo-sensitive marker in human gastric cancer. NKD2 impedes gastric cancer metastasis by inhibiting SOX18.


Journal of Thoracic Oncology | 2016

Silencing NKD2 by promoter region hypermethylation promotes esophageal cancer progression by activating wnt signaling

Baoping Cao; Weili Yang; Yongshuai Jin; Meiying Zhang; Tao He; Qimin Zhan; James G. Herman; Guanglin Zhong; Mingzhou Guo

Introduction Naked cuticle homolog 2 (NKD2) was found to be frequently methylated in human breast and gastric cancers. However, the epigenetic changes and mechanisms of NKD2 in human esophageal cancer remain unclear. Methods Nine esophageal cancer cell lines and 154 cases of primary esophageal cancer samples were analyzed using methylation‐specific polymerase chain reaction, immunohistochemical analysis, Western blot, and xenograft mouse models. Results Loss of NKD2 expression and complete methylation were found in KYSE150 and TE1 cells. Reduced NKD2 expression and partial methylation of the promoter region were observed in KYSE30, KYSE70, KYSE410, KYSE140, and COLO680 cells. High levels of NKD2 expression and unmethylation were detected in KYSE450 and TE8 cells. Reexpression of NKD2 was induced by 5‐aza‐2′‐deoxycytidine in cells in which NKD2 was not expressed or cells in which NKD2 expression was reduced. NKD2 was methylated in 53.2% of human primary esophageal cancer samples (82 of 154), and promoter region hypermethylation was significantly associated with reduced expression of NKD2 (p < 0.01). NKD2 methylation was associated with tumor, node, and metastasis stage and lymph node metastasis (p < 0.01). Our results suggest that NKD2 is regulated by promoter region methylation and that methylation of NKD2 may serve as a prognostic marker in esophageal cancer. Our further studies demonstrate that NKD2 suppresses cell proliferation, colony formation, cell invasion, and migration and also induces G1/S checkpoint arrest in esophageal cancer cells. NKD2 suppressed xenograft tumor growth and inhibited Wnt signaling in human esophageal cancer cells. Conclusions NKD2 is frequently methylated in human esophageal cancer, and the expression of NKD2 is regulated by promoter region methylation. NKD2 suppresses esophageal cancer progression by inhibiting Wnt signaling both in vitro and in vivo.


Discovery Medicine | 2013

Epigenetic silencing of CXCL14 induced colorectal cancer migration and invasion

Baoping Cao; Yunsheng Yang; Yuanming Pan; Yan Jia; Malcolm V. Brock; James G. Herman; Mingzhou Guo


Genes & Cancer | 2015

RASSF10 suppresses hepatocellular carcinoma growth by activating P53 signaling and methylation of RASSF10 is a docetaxel resistant marker

Yongshuai Jin; Baoping Cao; Meiying Zhang; Qimin Zhan; James G. Herman; Miao Yu; Mingzhou Guo


American Journal of Cancer Research | 2014

DACT2 is frequently methylated in human gastric cancer and methylation of DACT2 activated Wnt signaling.

Wenji Yan; Xuefeng Liu; Yan Jia; Baoping Cao; Yingyan Yu; Youyong Lv; Malcolm V. Brock; Jame G Herman; Julien Licchesi; Yunsheng Yang; Mingzhou Guo


American Journal of Cancer Research | 2015

Epigenetic silencing BCL6B induced colorectal cancer proliferation and metastasis by inhibiting P53 signaling.

Sui Hu; Baoping Cao; Meiying Zhang; Enqiang Linghu; Qimin Zhan; Malcolm V. Brock; James G. Herman; Gaoping Mao; Mingzhou Guo


Discovery Medicine | 2016

Systematic study of novel lncRNAs in different gastrointestinal cancer cells.

Baoping Cao; Song N; Zhang M; Di C; Yunsheng Yang; Lu Y; Chen R; Lu Zj; Mingzhou Guo

Collaboration


Dive into the Baoping Cao's collaboration.

Top Co-Authors

Avatar

Mingzhou Guo

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yunsheng Yang

Chinese PLA General Hospital

View shared research outputs
Top Co-Authors

Avatar

Yan Jia

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Enqiang Linghu

Chinese PLA General Hospital

View shared research outputs
Top Co-Authors

Avatar

Qimin Zhan

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Yingyan Yu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Tao He

Chinese PLA General Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge