Network


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

Hotspot


Dive into the research topics where Meiying Zhang is active.

Publication


Featured researches published by Meiying Zhang.


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.


Epigenomics | 2017

Methylation of SLFN11 is a marker of poor prognosis and cisplatin resistance in colorectal cancer

Tao He; Meiying Zhang; Ruipan Zheng; Shufang Zheng; Enqiang Linghu; James G Herman; Mingzhou Guo

AIM The expression of human SLFN11 was reported to sensitize cancer cells to DNA damaging agents. This study is to explore the epigenetic change and the function of SLFN11 in human colorectal cancer (CRC). MATERIALS & METHODS Six CRC cell lines and 128 primary CRC samples were used. RESULTS SLFN11 was methylated in 55.47% (71/128) of primary CRC. The expression of SLFN11 was regulated by promoter region methylation. Methylation of SLFN11 was significantly associated with age, poor 5-year overall survival and 5-year relapse-free survival (all p < 0.05). SLFN11 suppressed CRC cell growth both in vitro and in vivo and sensitized CRC cells to cisplatin. CONCLUSION SLFN11 is frequently methylated in human CRC, and the expression of SLFN11 is regulated by promoter region methylation. Methylation of SLFN11 reduced the sensitivity of CRC cells to cisplatin.


Clinical Epigenetics | 2017

Methylation of DIRAS1 promotes colorectal cancer progression and may serve as a marker for poor prognosis

Ruipan Zheng; Dan Gao; Tao He; Meiying Zhang; Xiaomei Zhang; Enqiang Linghu; Lixin Wei; Mingzhou Guo

BackgroundDIRAS1 is a new member of the Ras gene family. It was described as a potential tumor suppressor in human glioblastomas and esophageal cancer. The role of DIRAS1 in colorectal cancer remains unclear.MethodsTo explore the epigenetic changes and function of DIRAS1 in human colorectal cancer, we studied ten colorectal cancer cell lines and 146 primary colorectal cancer samples and 50 matched adjacent samples using semi-quantitative reverse transcription PCR, immunohistochemistry, methylation-specific PCR and bisulfite sequencing, western blot, flow cytometry, and transwell assays.ResultsDIRAS1 expression was found in DKO and HCT116 cells, while reduced expression was detected in LoVo, SW48, LS180, and SW620 cells, and there was no expression detected in DLD1, HT29, RKO, and SW480 cells. Complete methylation was found in the promoter region of DLD1, HT29, RKO, and SW480 cells. Partial methylation was detected in LoVo, LS180, SW48, and SW620 cells, and unmethylation was found in DKO and HCT116 cells. These results indicate that promoter region methylation correlated with loss of/reduced expression of DIRAS1. Re-expression of DIRAS1 was induced by 5-aza-2′-deoxycytidine, suggesting that the expression of DIRAS1 is regulated by promoter region methylation. DIRAS1 was methylated in 47.3% (69/146) of primary colorectal cancer samples, no methylation was found in non-cancerous colonic tissue samples. Methylation of DIRAS1 was significantly associated with TNM stage (P < 0.05) and short survival time (P = 0.0121). DIRAS1 induced apoptosis and inhibited cell proliferation, migration, and invasion in colorectal cancer. Finally, DIRAS1 suppressed colorectal cancer cell xenograft growth in nude mice.ConclusionsDIRAS1 is frequently methylated in human colorectal cancer and the expression of DIRAS1 is regulated by promoter region methylation. Methylation of DIRAS1 is a marker of poor prognosis in human colorectal cancer.


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.


Scientific Reports | 2017

Methylation of DACT2 promotes breast cancer development by activating Wnt signaling

Jingyi Li; Meiying Zhang; Tao He; Hongxia Li; Tingting Cao; Lili Zheng; Mingzhou Guo

Breast cancer is the most common malignant tumor in women worldwide. To explore the role of DACT2 in breast cancer, 5 cell lines and 153 cases of primary cancer were studied. The expression of DACT2 was detected in BT474, MDA-MB-231 and BT549 cells, while no expression was found in MDA-MB-468 and HBL100 cells. Complete methylation of DACT2 was found in MDA-MB-468 and HBL100 cells, partial methylation was observed in BT474 and BT549 cells, and no methylation was detected in MDA-MB-231 cells. Restoration of DACT2 expression was induced by 5-Aza in MDA-MB-468 and HBL100 cells. DACT2 was methylated in 49.7% (76/153) of primary breast cancer samples. Methylation of DACT2 was significantly associated with tumor size (P < 0.05). Reduced DACT2 expression was significantly associated with promoter region methylation in primary breast cancer (P < 0.05). DACT2 suppressed breast cancer cell growth and induced G1/S phase arrest in breast cancer cells. DACT2 inhibited Wnt/β-catenin signaling in human breast cancer cells and suppressed breast cancer cell tumor growth in xenograft mice. In conclusion, our results demonstrate that DACT2 is frequently methylated in human breast cancer, methylation of DACT2 activates Wnt signaling, and DACT2 suppresses breast cancer cell growth both in vitro and in vivo.


Oncotarget | 2015

Epigenetic silencing of NKD2, a major component of Wnt signaling, promotes breast cancer growth

Yang Dong; Baoping Cao; Meiying Zhang; Weidong Han; James G. Herman; François Fuks; Yali Zhao; 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 | 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 | 2017

Epigenetic regulation of voltage-gated potassium ion channel molecule Kv1.3 in mechanisms of colorectal cancer.

He T; Wang C; Meiying Zhang; Zhang X; Zheng S; Linghu E; Mingzhou Guo


Clinical Epigenetics | 2017

Silencing HOXD10 by promoter region hypermethylation activates ERK signaling in hepatocellular carcinoma

Yulin Guo; Yaojun Peng; Dan Gao; Meiying Zhang; Weili Yang; Enqiang Linghu; James G. Herman; François Fuks; Guanglong Dong; Mingzhou Guo

Collaboration


Dive into the Meiying Zhang's collaboration.

Top Co-Authors

Avatar

Mingzhou Guo

Johns Hopkins 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

Tao He

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
Top Co-Authors

Avatar

Guanglin Zhong

Chinese PLA General Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge