Network


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

Hotspot


Dive into the research topics where Hongbing Mei is active.

Publication


Featured researches published by Hongbing Mei.


PLOS ONE | 2014

CMTM3 Inhibits Human Testicular Cancer Cell Growth through Inducing Cell-Cycle Arrest and Apoptosis

Zesong Li; Jun Xie; Jianting Wu; Wenjie Li; Liping Nie; Xiaojuan Sun; Aifa Tang; Xianxin Li; Ren Liu; Hongbing Mei; Feng Wang; Zhiping Wang; Yaoting Gui; Zhiming Cai

Human CMTM3 has been proposed as a putative tumor suppressor gene. The loss of CMTM3 has been found in several carcinomas. However, the regulation of CMTM3 expression and its function in tumor progression remain largely unknown. Here, we investigated the regulation of CMTM3 expression, function and molecular mechanism in human testicular cancer cells. CMTM3 was frequently downregulated or silenced in testicular cancer cell lines and tumor tissues but highly expressed in normal testis tissues. The re-expression of CMTM3 significantly suppressed the colony formation, proliferation, and migration capacity of testicular cancer cells by inducing a G2 cell cycle arrest and apoptosis. Moreover, the re-expression of CMTM3 activated the transcription of p53, induced p53 accumulation, up-regulated the expression of p21, and increased the cleavage of caspase 9, 8, 3, and PARP. The downregulation of CMTM3 in clinical tumor tissues was associated with the methylation of a single CpG site located within the Sp1/Sp3-responsive region of the core promoter. These results indicate that CMTM3 can function as tumor suppressor through the induction of a G2 cell cycle arrest and apoptosis. CMTM3 is thus involved in testicular cancer pathogenesis, and it is frequently at least partially silenced by the methylation of a single, specific CpG site in tumor tissues.


Oncotarget | 2017

Role of long noncoding RNA UCA1 as a common molecular marker for lymph node metastasis and prognosis in various cancers: a meta-analysis

Anbang He; Rong Hu; Zhicong Chen; Xinhui Liao; Jianfa Li; Dailian Wang; Zhaojie Lv; Yuchen Liu; Feng Wang; Hongbing Mei

Accumulating evidences indicated that UCA1 expression was up-regulated in various cancers, and high UCA1 expression was correlated with metastasis and prognosis. This meta-analysis collected all eligible studies and explored the relationships between UCA1 expression and lymph node metastasis (LNM) or overall survival (OS). Literature collection was performed by using electronic databases PubMed, Cochrane Library, and Web of Science (up to June 13, 2016). According to the inclusion and exclusion criteria, twelve studies were included in the meta-analysis. The result showed that high UCA1 expression was correlated with more LNM (OR=2.50, 95 %CI: 1.58-3.96, p<0.0001) in a random-effects model (I2=45 %, p=0.08) and could predict poor OS in cancer patients, with pooled hazard ratio (HR) of 1.65 [95% confidence interval (CI) 1.44-1.88, p<0.00001] indicated by a fixed-effects model (I2=35%, p=0.11). In conclusion, the present meta-analysis demonstrated that high expression of UCA1 might serve as a common molecular marker for predicting lymph node metastasis and prognosis in various cancers.


Cancer Biomarkers | 2016

Decreased expression of LncRNA MIR31HG in human bladder cancer

Anbang He; Zhicong Chen; Hongbing Mei; Yuchen Liu

OBJECTIVE In this study, we examined the relationships between the expression level of long non-coding RNA MIR31HG in bladder cancer and the clinical characteristics. METHODS A total of 55 tissue samples from patients with bladder cancer were collected, and the lncRNA MIR31HG levels in cancer, paired non-cancer tissues and BC cell lines were detected by real-time quantitative RT-PCR (qRT-PCR). The relationships between MIR31HG level and the clinical characteristics were evaluated. RESULTS MIR31HG expression was remarkably decreased in bladder cancer tissues compared with adjacent noncancerous tissues (P < 0.05). MIR31HG expression was also significantly down-regulated in four bladder cancer cell lines (P < 0.001). Clinicopathologic analysis revealed that MIR31HG expression was negatively associated with TNM stage (P = 0.010), but not with other clinicopathological characteristics. CONCLUSIONS These findings revealed that MIR31HG may function as a cancer-suppressor gene to participate in the bladder cancer carcinogenesis and development.


Oncotarget | 2017

Hsa-miR-429 promotes bladder cancer cell proliferation via inhibiting CDKN2B

Jiangeng Yang; Yuchen Liu; Anbang He; Yuhan Liu; Jianting Wu; Xinhui Liao; Zhaojie Lv; Feng Wang; Hongbing Mei

Background and Objectives Hsa-miR-429 is increased in bladder cancer. Its roles in bladder cancer are poorly understood. Methods The expression levels of hsa-miR-429 and cyclin-dependent kinase inhibitor 2B (CDKN2B) were determined using Real-Time qPCR in a total of 50 patients with bladder cancer. Bladder cancer T24 and 5637 cells were transfected CDKN2B siRNA or hsa-miR-429 mimic. CDKN2B expression levels after transfection were detected by Real-Time qPCR and Western blot assay respectively. Binding sites between hsa-miR-429 and 3’-untranslated region of CDKN2B were confirmed by Dual luciferase reporter assay. Cell proliferation was evaluated using MTT and EdU assays. Cell apoptosis was determined using ELISA assay. Results Higher hsa-miR-429 expression levels were associated with higher tumor grade and stage. All patients with low hsa-miR-429 expression survived 5 years, while with high hsa-miR-429 expression, only 58% survived. Hsa-miR-429 and CDKN2B were inversely expressed in bladder cancer. Hsa-miR-429 mimic decreased the expression of CDKN2B at both mRNA and protein levels. The binding site was confirmed between hsa-miR-429 and 3’-untranslated region of CDKN2B. Up-regulation of hsa-miR-429 or down-regulation of CDKN2B promoted cell growth and decreased apoptosis. Conclusions Our data suggest a mechanism for hsa-miR-429 to play oncogenic roles via inhibiting CDKN2B.


OncoTargets and Therapy | 2016

Association of polymorphisms in interleukin-8 gene with cancer risk: a meta-analysis of 22 case-control studies.

Meng Zhang; Tingting Fang; Kai Wang; Hongbing Mei; Zhaojie Lv; Feng Wang; Zhiming Cai; Chaozhao Liang

Interleukin-8 (IL-8) is a kind of chemokine that plays an important role in the development and progression of many human malignancies. Previous studies have uncovered that polymorphisms in IL-8 is associated with the risk of many cancer types, but the results were inconsistent and inconclusive. In the present study, we aimed to explore the roles of IL-8 polymorphisms (rs2227307, rs2227306, +678T/C, rs1126647, and +1633C/T) and cancer risk through a systematic review and meta-analysis. Potential source of heterogeneity was sought out through sensitivity analysis. Desirable data were extracted and registered into databases. Finally, a total of ten publications comprising of 22 case–control studies, including 4,259 cases and 7,006 controls were ultimately eligible for the meta-analysis. No significant association was uncovered for all the five polymorphisms and the overall cancer risk. However, in the stratification analysis by cancer type, a significantly decreased risk of hepatocellular carcinoma was identified for rs2227306 polymorphism (T vs C: odds ratio [OR] =0.721, 95% confidence interval [CI] =0.567–0.916, Pz=0.007; TT vs CC: OR =0.447, 95% CI =0.274–0.728, Pz=0.001; TT vs TC + CC: OR =0.480, 95% CI =0.304–0.760, Pz=0.002). In conclusion, our data shows that rs2227306 polymorphism plays a protective role in hepatocellular carcinoma risk. Future well-designed studies with a larger sample size are warranted to verify our findings.


Biomedicine & Pharmacotherapy | 2018

Overexpression of CRNDE promotes the progression of bladder cancer

Jianli Cheng; Jieqing Chen; Xintao Zhang; Hongbing Mei; Feng Wang; Zhiming Cai

Accumulating evidences indicate that long non-coding RNAs (lncRNAs) are indispensable in cancer initiation and progression. Dysregulation of functional lncRNAs can promote the development of cancers. Previous research have revealed that augmented expression of CRNDE caused poor prognosis of cancer patients and facilitate the tumor progress in various cancers. Nevertheless, the underlying roles of CRNDE in bladder cancer progression are not entirely clear. To further identify the effects CRNDE in bladder cancer progression, we performed the gain and loss of function assay. In this work, we have presented evidence that CRNDE was significantly increased in bladder cancer, and overexpressed expression of CRNDE was positively related with advanced TNM stage of bladder cancer patients. In addition, in vitro experiments showed that CRNDE strengthened cell migration/proliferation and inhibited cell apoptosis in bladder cancer. To sum up, our results exhibited new understand into the role of lncRNA CRNDE in the development of bladder cancer.


Cancer Biomarkers | 2017

Knockdown of long noncoding RNA FGFR3- AS1 induces cell proliferation inhibition, apoptosis and motility reduction in bladder cancer

Xinhui Liao; Jieqing Chen; Yuchen Liu; Anbang He; Jianting Wu; Jianli Cheng; Xintao Zhang; Zhaojie Lv; Feng Wang; Hongbing Mei

OBJECTIVES To study the expression pattern of long non-coding RNA FGFR3 antisense transcript 1(FGFR3-AS1) and the cell proliferation inhibition, apoptosis, and motility changes induced by silencing FGFR3-AS1 in bladder cancer. METHODS The differential expression levels of FGFR3-AS1 and FGFR3 in tumor tissues and paired normal tissues were determined using Real-Time qPCR in a total of 36 patients diagnosed with bladder cancer (urothelial carcinoma). Pearsons coefficient correlation was used for expression correlation assay. Expression differences of FGFR3-AS1 were analyzed according to grading and staging. FGFR3 protein was detected by western blot assay. Human bladder cancer T24 and 5637 cell lines were transiently transfected with FGFR3-AS1-specific siRNA or negative control siRNA. The cell proliferation changes of transfected bladder cancer cells were determined using CCK-8 assay. Apoptosis caused by knockdown of FGFR3-AS1 was evaluated using ELISA assay. Motility changes induced by knockdown of FGFR3-AS1 were measured using wound healing assay and transwell assay. RESULTS Both FGFR3-AS1 and FGFR3 were overexpressed in bladder cancer tissues compared to matched normal tissues. They were also positively expressed in bladder cancer. FGFR3-AS1 expression levels were higher in high grade tumors than those in low grade tumors. FGFR3-AS1 expression levels were higher in invasive tumors than those in non-invasive tumors. Cell proliferation inhibition, increased apoptosis, and decreased motility were observed in FGFR3-AS1 siRNA-transfected T24 and 5637 cell lines. CONCLUSIONS FGFR3-AS1 plays an oncogenic role in human bladder cancer. Knockdown of FGFR3-AS1 may provide a potential new therapeutic approach to this disease.


OncoTargets and Therapy | 2016

Association between two interleukin-2 gene polymorphisms and cancer susceptibility: a meta-analysis.

Meng Zhang; Xiuxiu Tan; Junjie Huang; Lijuan Xie; Hao Wang; Jizhou Shi; Wei Lu; Zhaojie Lv; Hongbing Mei; Chaozhao Liang

Background Several epidemiological studies have illustrated that polymorphisms in interleukin-2 (IL-2) were associated with diverse cancer types. However, recently published statistics were inconsistent and inconclusive. Therefore, the current meta-analysis was performed to elaborate the effects of IL-2 polymorphisms (rs2069762 and rs2069763) on cancer susceptibility. Material and methods A total of 5,601 cancer cases and 7,809 controls from 21 published case–control studies were enrolled in our meta-analysis. Odds ratios (ORs) with 95% confidence intervals (CIs) were estimated to assess the association between IL-2 polymorphisms and cancer susceptibility. Results Our study demonstrated an increased susceptibility to cancer in rs2069762 (G vs T: OR =1.268, 95% CI =1.113–1.445; GG vs TT: OR =1.801, 95% CI =1.289–2.516; GT vs TT: OR =1.250, 95% CI =1.061–1.473; GG + GT vs TT: OR =1.329, 95% CI =1.118–1.579; GG vs GT + TT: OR =1.536, 95% CI =1.162–2.030). In the subgroup analysis, increased susceptibility to cancer was identified in the hospital-based group and PHWE<0.05 (P-value of the Hardy–Weinberg equilibrium [HWE]) group. In addition, a positive association with cancer susceptibility was observed among both Chinese and non-Chinese. However, no relationship was detected between the rs2069763 polymorphism of IL-2 and cancer susceptibility. Conclusion To conclude, rs2069762 polymorphism of IL-2 contributed to an increased susceptibility to cancer, whereas no association was identified between rs2069763 polymorphism and cancer susceptibility. Further detailed studies are warranted to confirm our findings.


OncoTargets and Therapy | 2018

rs11614913 polymorphism in miRNA-196a2 and cancer risk: an updated meta-analysis

Yuhan Liu; Anbang He; Baoer Liu; Yucheng Zhong; Xinhui Liao; Jiangeng Yang; Jieqing Chen; Jianting Wu; Hongbing Mei

Several epidemiological studies have reported that polymorphisms in microRNA-196a2 (miR-196a2) were associated with various cancers. However, the results remained unverified and were inconsistent in different cancers. Therefore, we carried out an updated meta-analysis to elaborate the effects of rs11614913 polymorphism on cancer susceptibility. A total of 84 articles with 35,802 cases and 41,541 controls were included to evaluate the association between the miR-196a2 rs11614913 and cancer risk by pooled odds ratios (ORs) and 95% confidence intervals (CIs). The results showed that miR-196a2 rs11614913 polymorphism is associated with cancer susceptibility, especially in lung cancer (homozygote comparison, OR =0.840, 95% CI =0.734–0.961; recessive model, OR =0.858, 95% CI =0.771–0.955), hepatocellular carcinoma (allelic contrast, OR =0.894, 95% CI =0.800–0.998; homozygote comparison, OR =0.900, 95% CI =0.813–0.997; recessive model, OR =0.800, 95% CI =0.678–0.944), and head and neck cancer (allelic contrast, OR =1.076, 95% CI =1.006–1.152; homozygote comparison, OR =1.214, 95% CI =1.043–1.413). In addition, significant association was found among Asian populations (allele model, OR =0.847, 95% CI =0.899–0.997, P=0.038; homozygote model, OR =0.878, 95% CI =0.788–0.977, P=0.017; recessive model, OR =0.895, 95% CI =0.824–0.972, P=0.008) but not in Caucasians. The updated meta-analysis confirmed the previous results that miR-196a2 rs11614913 polymorphism may serve as a risk factor for patients with cancers.


OncoTargets and Therapy | 2018

Lentivirus-mediated shRNA targeting MUTYH inhibits malignant phenotypes of bladder cancer SW780 cells

Qunjun Gao; Yuhan Liu; Haibiao Xie; Yucheng Zhong; Xinhui Liao; Hengji Zhan; Qun Zhou; Mengting Ding; Kang Yang; Aolin Li; Yuchen Liu; Hongbing Mei; Zhiming Cai

Objectives MUTYH is a protein-coding gene that takes part in base excision repair. Many previous studies have reported that MUTYH is directly related to hereditary adenomatous polyposis and colorectal cancer and is also associated with other cancers. However, the relationship between MUTYH and bladder cancer (BC) is unknown. Materials and methods The expression of MUTYH and clinical characteristics of BC were collected from databases including The Cancer Genome Atlas and Cancer Cell Line Encyclopedia. RNA sequencing and quantitative real-time PCR were used to detect MUTYH expression in SW780 BC cells. The level of MUTYH was stably downregulated by lentivirus-mediated vector in SW780 cells. Cell proliferation was evaluated using Cell Counting Kit-8 assay and 5-ethynyl-20-deoxyuridine assay, migration was detected using scratch assay and Transwell assay, and apoptosis was determined using ELISA. Results MUTYH was upregulated in BC tissues and SW780 cells and its expression level was positively associated with the stage and grade of carcinomas. MUTYH was successfully downregulated in SW780 cells by transducing with a lentivirus-mediated shRNA targeting MUTYH. MUTYH knockdown inhibited the proliferation and migration and induced apoptosis in SW780 cells. Conclusion Our data suggest that MUTYH is a new participant in bladder urothelial carcinoma. MUTYH may play a role as a biomarker and therapeutic target in BC.

Collaboration


Dive into the Hongbing Mei's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge