Network


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

Hotspot


Dive into the research topics where Zhong-Sheng Xia is active.

Publication


Featured researches published by Zhong-Sheng Xia.


Clinics and Research in Hepatology and Gastroenterology | 2016

Diabetes mellitus increases the risk of colorectal neoplasia: An updated meta-analysis

Su Luo; Jie-Yao Li; Li-Na Zhao; Tao Yu; Wa Zhong; Zhong-Sheng Xia; Ti-Dong Shan; Hui Ouyang; Hong-Sheng Yang; Qi-Kui Chen

OBJECTIVE Recent studies proved that patients with diabetes were at significantly higher risk of developing colorectal cancer. However, the association between diabetes mellitus and the risk of colorectal adenoma remains undefined. Thus we conducted an updated meta-analysis to identify the association between diabetes mellitus and the risk of colorectal neoplasia including adenoma and cancer. METHODS We conducted a search in databases including Pubmed, Web of Science, EMBASE Databases, Cochrane CENTRAL, Wanfang Data, and CNKI database. Case-control and cohort studies were included. All articles were published before January 2015 and the quality of each study was evaluated by the Newcastle-Ottawa Scale. Odds ratios (ORs) or relative risks (RRs) and its corresponding 95% confidence intervals (CIs) for each study were calculated and summary relative risk estimates with corresponding 95% CIs were generated using the random-effects model. Heterogeneity and publication bias were assessed. RESULTS Twenty-nine articles including ten case-control studies and nineteen cohort studies were included in this meta-analysis. In a pooled analysis of all studies, diabetes mellitus was associated with increased risk of colorectal neoplasia (RR=1.35, 95% CI=1.28-1.42). The risk increased significantly for both colorectal cancer (RR=1.37, 95% CI=1.30-1.45) and adenoma (RR=1.26, 95% CI=1.11-1.44). Subgroup analyses on study design, gender, geographical region, and type of diabetes mellitus further evidenced these findings. CONCLUSIONS Diabetes mellitus was associated with an increased risk of colorectal neoplasia. Not only the increased risk of colorectal cancer but also the higher risk of adenoma was identified in patients with diabetes mellitus.


Cell Proliferation | 2016

miRNA‐30e regulates abnormal differentiation of small intestinal epithelial cells in diabetic mice by downregulating Dll4 expression

Ti-Dong Shan; Hui Ouyang; Tao Yu; Jie-Yao Li; Can-Ze Huang; Hong-Sheng Yang; Wa Zhong; Zhong-Sheng Xia; Qi-Kui Chen

Depression of the Notch/Hes1 pathway has been reported to play a role in abnormal differentiation of intestinal epithelial cells (IECs) in diabetes mellitus (DM). However, the mechanism by which this pathway influences IEC differentiation has remained unclear. In this study, we have investigated the role of microRNAs (miRNAs) in regulating the Notch/Hes1 pathway in IECs of DM mice.


Journal of Diabetes and Its Complications | 2014

Enhanced proliferation in colorectal epithelium of patients with type 2 diabetes correlates with β-catenin accumulation

Jie-Yao Li; Tao Yu; Zhong-Sheng Xia; Guang-Cheng Chen; Yu-Hong Yuan; Wa Zhong; Li-Na Zhao; Qi-Kui Chen

AIMS β-Catenin accumulation promotes proliferation. However, the correlation between proliferation of colorectal epithelium and β-catenin in type 2 diabetes mellitus (DM) patients remains unclear. METHODS Colorectal epithelium samples from distal ends of colorectal adenocarcinomas without histological aberrances were divided into two groups: DM patients with type 2 DM for more than 1year (n=27) and non-DM patients without hyperglycemia (n=20). Samples from patients without colorectal epithelial disease or hyperglycemia served as a control group (n=6). Proliferative index was calculated as the percentage of proliferating cell nuclear antigen positive cells. Wnt/β-catenin signaling was assessed immunohistochemically and phosphorylation of β-catenin was assessed by immunofluorescence. RESULTS Compared with the non-DM or control group, the proliferative index and expression of lactate dehydrogenase A and Wnt/β-catenin signaling were significantly higher in the DM group (all p<0.01). The proliferative index correlated positively with β-catenin expression (Spearman correlation coefficient=0.55; p<0.01). Reduced phosphorylation at serine 33/37 and increased phosphorylation at serine 675 of β-catenin were detected in the DM group (all p<0.01). CONCLUSIONS Enhanced proliferation, accompanied by increased aerobic glycolysis, was detected in colorectal epithelium of patients with diabetes. β-Catenin accumulation with altered phosphorylation correlated with the proliferative changes.


Cell and Tissue Research | 2016

Overexpression of miR-429 impairs intestinal barrier function in diabetic mice by down-regulating occludin expression

Tao Yu; Xi-Ji Lu; Jie-Yao Li; Ti-Dong Shan; Can-Ze Huang; Hui Ouyang; Hong-Sheng Yang; Ji-Hao Xu; Wa Zhong; Zhong-Sheng Xia; Qi-Kui Chen

Diabetes mellitus (DM) is a group of metabolic diseases characterised by insulin deficiency/resistance and hyperglycaemia. We previously reported the presence of an impaired tight junction and decreased expression of occludin (Ocln) and zonula occludens-1 (ZO-1) in the intestinal epithelial cells (IECs) of type 1 DM mice, but the exact mechanism remains unclear. In this study, we investigated the role of microRNAs (miRNAs) in impairing the tight junction in IECs of DM mice. Using an integrated comparative miRNA microarray, miR-429 was found to be up-regulated in IECs of type 1 DM mice. Then, miR-429 was confirmed to directly target the 3’-UTR of Ocln, although it did not target ZO-1. Moreover, miR-429 down-regulated the Ocln expression in IEC-6 cells in vitro. Finally, exogenous agomiRNA-429 was shown to down-regulate Ocln and induce intestinal barrier dysfunction in normal mice, while exogenous antagomiRNA-429 up-regulated Ocln in vivo and improved intestinal barrier function in DM mice. In conclusion, increased miR-429 could down-regulate the expression of Ocln by targeting the Ocln 3′-UTR, which impaired intestinal barrier function in DM mice.


PLOS ONE | 2015

Rebamipide Promotes the Regeneration of Aspirin-Induced Small-Intestine Mucosal Injury through Accumulation of β-Catenin

Yu Lai; Wa Zhong; Tao Yu; Zhong-Sheng Xia; Jie-Yao Li; Hui Ouyang; Ti-Dong Shan; Hong-Sheng Yang; Qi-Kui Chen

Background The effect of rebamipide on repairing intestinal mucosal damage induced by nonsteroidal anti-inflammatory drugs and its mechanism remain unclear. In this study, we sought to explore the mechanism whereby rebamipide could promote the regeneration of aspirin-induced intestinal mucosal damage. Methods BALB/c mice were administered aspirin (200 mg/kg/d) for 5 days to induce acute small intestinal injury (SII). Subsequently, SII mice were treated with rebamipide (320 mg/kg/d) for 5 days. The structure of intestinal barrier was observed with transmission electron microscope, and Zo-1 and occludin expressions were detected. The proliferative index was indicated by the percentage of proliferating cell nuclear antigen positive cells. The prostaglandin E2 (PGE2) levels in the small intestine tissues were measured by an enzyme immunoassay. The mRNA and protein expression levels of cyclooxygenase (COX) and β-catenin signal were detected in the small intestine using quantitative PCR and Western blot, respectively. Results COX expression was significantly down-regulated in aspirin induced SII (P < 0.05). In SII mice treated with rebamipide, histopathological findings of aspirin-induced intestinal inflammation were significantly milder and tight junctions between intestinal epithelial cells were improved significantly. The proliferative index increased after rebamipide treatment when compared with that in the control mice. The expressions of COX-2, β-catenin, and c-myc and the PGE2 concentrations in small intestinal tissues were significantly increased in mice with rebamipide treatments (P < 0.05). Conclusion Rebamipide administration in aspirin-induced SII mice could improve the intestinal barrier structure and promote the regeneration of small intestinal epithelial injury through up-regulating COX-2 expression and the accumulation of β-catenin.


Molecular Medicine Reports | 2009

Stable reversal of multidrug resistance in colon cancer cells by RNA interference targeting the MDR1 gene

Zhong-Sheng Xia; Liyong Zhang; Qi-Kui Chen; Crista Royal; Zhong Yu; Zhongmin Liu; Bao-Ling Adam

Colon cancer is one of the most common cancers in the world. Overexpression of MDR1 mRNA and P-gp is associated with the classic multidrug resistance of colon cancer cells. In our previous study, we reported on the transient specific reversal of MDR1/P-gp-dependent multi-drug resistance by RNA interference (RNAi) in colon cancer cells. In this study, RNAi targeting the MDR1 gene stably reversed MDR1/P-gp-dependent multidrug resistance in colon cancer cells. The plasmid vectors pSilencer-#4029, encoding #4029 MDR1 siRNA, and pSilencer-#4123, encoding #4123 MDR1 siRNA, were constructed and then transfected into COLO 320DM, a colon cancer multidrug-resistant cell line. Clone cells were screened by G418 and identified by RT-PCR and Western blot analysis. Cellular viability was measured using the MTT assay. Cell cycle analysis and intracellular adriamycin accumulation were assessed by flow cytometry. MDR1 mRNA and P-gp expression in positive clone cells, those stably transfected with MDR1 siRNA, was inhibited. The IC50 values of the antitumor drugs were significantly decreased in the positive clones compared to the COLO 320DM parent cell line, and the PI/AI values of positive clones treated with antitumor drugs were significantly decreased compared to the parent cells. In addition, the intracellular adriamycin accumulation of positive clones treated with adriamycin was significantly increased compared with COLO 320DM. This demonstrates that the stable transfection of plasmid vectors encoding MDR1 siRNA can stably reverse the MDR1/P-gp-dependent multidrug resistance of colon cancer cells.


Oncology Reports | 2012

The effects of cyclooxygenase-2 gene silencing by siRNA on cell proliferation, cell apoptosis, cell cycle and tumorigenicity of Capan-2 human pancreatic cancer cells

Yingqiang Zhong; Zhong-Sheng Xia; Juan Liu; Ying Lin; Hui Zan

The prognosis of pancreatic cancer is still very poor. No specific effective gene therapy for pancreatic cancer has been found. As a key enzyme of the metabolic process of arachidonic acid, cyclooxygenase-2 (COX-2) has been found to be closely related to the tumorigenesis of epithelial cancers. However, the antitumor effect of small interfering RNA (siRNA) targeting COX-2 in pancreatic cancer has not yet been verified. Therefore, the aim of this study was to investigate the effects of COX-2 gene silencing by siRNA on cell proliferation, cell apoptosis, cell cycle and tumorigenicity of pancreatic cancer cells. COX-2 mRNA was detected by RT-PCR and real-time PCR. COX-2 protein was detected by Western blotting. The cell proliferation was measured by cell counting using microscopy. The cell apoptosis and cell cycle were measured by flow cytometry. The tumorigenicity of Capan-2 pancreatic cancer cells transfected with COX-2 siRNA was evaluated using a nude mouse xenograft model. The expression of COX-2 mRNA as well as COX-2 protein were downregulated after COX-2 siRNA transfection. COX-2 siRNA could inhibit the growth of Capan-2 cells significantly by decreasing the cell proliferation, increasing cell apoptosis and regulating cell cycle as well. In vivo experiments demonstrated that the mean volume and weight of subcutaneous xenografts in nude mice derived from Capan-2 cells transfected with COX-2 siRNA were significantly decreased. COX-2 siRNA could inhibit the growth of Capan-2 pancreatic cancer cells and also decrease the tumorigenicity of Capan-2 cells, implicating a new potential therapeutic target in pancreatic cancer.


Stem Cell Research & Therapy | 2017

Sox9 transcriptionally regulates Wnt signaling in intestinal epithelial stem cells in hypomethylated crypts in the diabetic state

Can-Ze Huang; Ji-Hao Xu; Wa Zhong; Zhong-Sheng Xia; Si-Yi Wang; Di Cheng; Jie-Yao Li; Ting-Feng Wu; Qi-Kui Chen; Tao Yu

BackgroundDistinctive structures called crypts harbor intestinal epithelial stem cells (IESCs) which generate progenitor and terminally differentiated cells in the intestinal epithelium. Mammalian IESCs and their daughter cells require the participation of DNA methylation and the transcription factor Sox9 for proliferation and differentiation. However, the association between Sox9 and DNA methylation in this process remains elusive.MethodsThe DNA methylation of small intestinal epithelial crypts in db/db mice was detected via combining methylated DNA immunoprecipitation with microarray hybridization. DNA methylation of Sox9 promoter in crypts and IESCs was validated using bisulfite sequence analysis. The target sequence of the transcription factor Sox9 in IESCs was investigated via chromatin immunoprecipitation (ChIP) combined with deep sequencing (ChIP-seq).ResultsIncreased Sox9 expression is accompanied by the loss of methylation in its promoter in IESCs. Sox9 targets the enhancers of the Wnt signaling pathway-related genes. Sox9 predominantly acts as a transcriptional activator at proximal enhancers of Wnt4, Tab2, Sox4, and Fzd8, but also functions as a potential transcriptional inhibitor at a distant enhancer of Cdk1. Lack of Sox9 transcriptional activation in specific repressors of the Wnt signaling pathway leads to the loss of intrinsic inhibitory action and ultimately produces overactivation of this pathway in db/db mice.ConclusionsOur study sheds light on the connections among DNA methylation, transcription factor modulation, and Wnt signaling in IESCs in the diabetic state. Hypomethylation in the Sox9 promoter is correlated to increased Sox9 expression in db/db IESCs. Although there is increased expression of Sox9 in db/db IESCs, the loss of Sox9 transcriptional activation in specific repressors of the Wnt signaling pathway might result in abnormalities in this pathway.


Development Growth & Differentiation | 2015

Lgr5 positive stem cells sorted from small intestines of diabetic mice differentiate into higher proportion of absorptive cells and Paneth cells in vitro

Xian‐Yang Zhong; Tao Yu; Wa Zhong; Jie-Yao Li; Zhong-Sheng Xia; Yu-Hong Yuan; Zhong Yu; Qi-Kui Chen

Intestinal epithelial stem cells (IESCs) can differentiate into all types of intestinal epithelial cells (IECs) and Leucine‐rich repeat‐containing G protein‐coupled receptor 5 (Lgr5) is a marker for IESC. Previous studies reported enhanced proliferation of IECs in diabetic mice. In this study, the in vitro differentiation of Lgr5 positive IESCs sorted from diabetic mice was further investigated. The diabetic mouse model was induced by streptozotocin (STZ), and crypt IECs were isolated from small intestines. Subsequently, Lgr5 positive IESCs were detected by flow cytometry (FCM) and sorted by magnetic activated cell sorting (MACS). Differentiation of the sorted IESCs was investigated by detecting the IEC markers in the diabetic mice using immunostaining, quantitative real‐time reverse–transcription polymerase chain reaction (qRT–PCR), and Western blot analysis, which was compared with normal mice. We found that the proportion of Lgr5 positive cells in the crypt IECs of diabetic mice was higher than that of control mice (P < 0.05). Lgr5 positive IESCs could be significantly enriched in Lgr5 positive cell fraction sorted by MACS. Furthermore, the absorptive cell marker sucrase‐isomaltase (SI) and the Paneth cell marker lysozyme 1 (Lyz1) were more highly expressed in the differentiated cells derived from Lgr5 positive IESCs of diabetic mice in vitro (P < 0.05). We demonstrate that the number of Lgr5 positive IESCs is significantly increased in the small intestines of STZ‐induced diabetic mice. Lgr5 positive IESCs sorted from the diabetic mice can differentiate into a higher proportion of absorptive cells and Paneth cells in vitro. We characterized the expression of Lgr5 in the small intestine of diabetic mice, and sorted Lgr5 positive intestinal epithelial stem cells (IESCs) for investigating their differentiation in vitro. We proved that the quantity of Lgr5 positive IESCs was significantly increased in the small intestines of diabetic mice. IESCs sorted from the diabetic mice can differentiate into a higher proportion of absorptive cells and Paneth cells in vitro.


Oncology Reports | 2014

miR-5000-3p, miR-5009-3P and miR-552: Potential microRNA biomarkers of side population cells in colon cancer

Zhong-Sheng Xia; Lin Wang; Tao Yu; Wa Zhong; Guo Da Lian; Di Wu; Hui-Min Zhou; Guang-Cheng Chen

Colon cancer is one of the most common cancers in the world. Multidrug resistance is related to poor prognosis of advanced colon cancer. The side population plays an important role in multiple drug resistance (MDR) of colon cancer. MicroRNA biomarkers of the side population of colon cancer is still unknown. In the present study, we aimed to explore miRNA markers of side population (SP) cells of colon cancer. The side population was sorted by flow cytometry. Cell viability was measured using an MTT assay. MicroRNA profiling analysis was performed to compare microRNA expression levels in the SP cells of colon cancer with levels in the non-SP cells of colon cancer. RT-PCR was applied to verify the result obtained from the microRNA profiling analysis. miR-5000-3p, miR-5009-3P and miR-552 were all found to be upregulated in SP cells of the colon cancer cell lines HCT-15, HT-29 and LoVo. RT-PCR confirmed the result from the microRNA profiling analysis. This implied that miR-5000-3p, miR-5009-3P and miR-552 may be potential microRNA biomarkers of the side population in colon cancer, which may provide new specific targets of the side population for the reversal of MDR of colon cancer.

Collaboration


Dive into the Zhong-Sheng Xia's collaboration.

Top Co-Authors

Avatar

Qi-Kui Chen

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Tao Yu

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Wa Zhong

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Jie-Yao Li

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hui Ouyang

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhong Yu

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Li-Na Zhao

Sun Yat-sen University

View shared research outputs
Researchain Logo
Decentralizing Knowledge