Network


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

Hotspot


Dive into the research topics where Changwei Lin is active.

Publication


Featured researches published by Changwei Lin.


Oncology Research | 2016

Knockdown of Long Noncoding RNA GHET1 Inhibits Cell Proliferation and Invasion of Colorectal Cancer.

Jianyu Zhou; Xiaorong Li; Meirong Wu; Changwei Lin; Yihang Guo; Buning Tian

Emerging evidence has identified the vital role of long noncoding RNAs (lncRNAs) in the development of colorectal cancer. In this study, we aimed to investigate the role of lncRNA gastric carcinoma highly expressed transcript 1 (GHET1) in colorectal cancer. We analyzed the expression of GHET1 in colorectal cancer (CRC) tissues by using ISH. We found that GHET1 expression was significantly increased in the CRC samples compared with adjacent tissues. Furthermore, the cancer tissues had higher GHET1 mRNA levels than their matched adjacent tissues. GHET1 expression was also significantly increased in the CRC cell lines compared with human normal colon epithelial cells. Downregulation of GHET1 mediated by shRNA suppressed the proliferation, cell cycle arrest, migration, and invasion of colorectal cancer cells in vitro. In addition, inhibition of GHET1 reversed the epithelial-mesenchymal transition in colorectal cancer cell lines. Taken together, our results suggest the potential use of GHET1 as a therapeutic target of colorectal cancer.


Chinese Journal of Cancer Research | 2014

Downregulation of serum miR-17 and miR-106b levels in gastric cancer and benign gastric diseases

Qinghai Zeng; Cuihong Jin; Wenhang Chen; Fang Xia; Qi Wang; Fan Fan; Juan Du; Yihang Guo; Changwei Lin; Kaiyan Yang; Jingjing Li; Xiaowei Peng; Xiaorong Li; Ke Cao

Altered microRNA (miRNA) associated with gastric cancer (GC) development and miR-17 and miR-106b were differentially expressed in GC tissues. This study detected serum levels of miR-17 and miR-106b expression in GC, benign gastric disease (BGD) and healthy controls to assess them as tumor markers for GC. Serum samples from 40 GC, 32 BGD (10 gastric ulcer, 14 gastric polyps, and 8 gastric ulcer with polyps) and 36 healthy individuals were subjected to quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis of miR-17 and miR-106b expression. The data showed that the serum levels of miR-17 and miR-106b were significantly reduced in healthy individuals and BGD patients compared to GC patients. There was a significant association of miR-17 and miR-106b expression with age, but not with other clinicopathological features, such as gender, tumor differentiation, stage and lymphatic metastasis. Further analysis showed that, in discriminating GC patients from healthy controls, miR-17 could yield a receiver-operating characteristic (ROC) area under the curve (AUC) of 0.879 with 80.6% sensitivity and 87.5% specificity and miR-106b could yield an AUC of 0.856 with 75.0% sensitivity and 92.5% specificity. The combined AUC of miR-17 and miR-106b was 0.913 with 83.3% sensitivity and 87.5% specificity. Collectively, these data suggest that detection of serum miR-17 and miR-106b levels should be further evaluated as novel non-invasive biomarkers in early GC detection and surveillance of disease progression.


Molecular Medicine Reports | 2015

Combinational treatment with microRNA‑133b and cetuximab has increased inhibitory effects on the growth and invasion of colorectal cancer cells by regulating EGFR.

Jianyu Zhou; Lv Lv; Changwei Lin; Gui Hu; Yihang Guo; Meirong Wu; Buning Tian; Xiaorong Li

Colorectal cancer (CRC) is the third most common cancer with a very poor prognosis predominantly due to its high rate of tumor invasion and migration, and its resistance to anti‑epidermal growth factor receptor (EGFR) therapy. Although CRC has been widely studied, the underlying molecular mechanism remains to be elucidated. MicroRNA (miR)‑133b has been demonstrated to act as a tumor suppressor in several human cancer types by regulating EGFR. However, the detailed involvement of miR‑133b and EGFR in CRC cells remain to be elucidated. The present study used reverse transcription quantitative polymerase chain reaction and characterized the downregulation of the expression levels of miR‑133b in CRC tissues and cell lines. Cell functional assays demonstrated that restored expression of miR‑133b inhibited the growth and invasion of CRC cells. In addition, a luciferase reporter assay revealed that miR‑133b directly targeted EGFR and repressed its expression levels in CRC cells. Additionally, combination treatment with miR‑133b mimics and the monoclonal anti‑EGFR antibody, cetuximab, which is approved and frequently used for treating patients with CRC, exhibited improved inhibitory effects on the growth and invasion of CRC cells compared with treatment with either alone. Taken together, the present study characterized the role of the miR‑133b/EGFR interaction in CRC cells and this suggested the combinational therapy with cetuximab and miR‑133b was positive and may be a potential novel treatment for patients with CRC in the future.


Oncotarget | 2017

Comprehensive analysis of differentially expressed profiles of lncRNAs and construction of miR-133b mediated ceRNA network in colorectal cancer

Hao Wu; Runliu Wu; Miao Chen; Daojiang Li; Jing Dai; Yi Zhang; Kai Gao; Jun Yu; Gui Hu; Yihang Guo; Changwei Lin; Xiaorong Li

Background Growing evidence suggests that long non-coding RNAs (lncRNAs) play a key role in tumorigenesis. However, the mechanism remains largely unknown. Results Thousands of significantly dysregulated lncRNAs and mRNAs were identified by microarray. Furthermore, a miR-133b-meditated lncRNA-mRNA ceRNA network was revealed, a subset of which was validated in 14 paired CRC patient tumor/non-tumor samples. Gene set enrichment analysis (GSEA) results demonstrated that lncRNAs ENST00000520055 and ENST00000535511 shared KEGG pathways with miR-133b target genes. Materials and Methods We used microarrays to survey the lncRNA and mRNA expression profiles of colorectal cancer and para-cancer tissues. Gene Ontology (GO) and KEGG pathway enrichment analyses were performed to explore the functions of the significantly dysregulated genes. An innovate method was employed that combined analyses of two microarray data sets to construct a miR-133b-mediated lncRNA-mRNA competing endogenous RNAs (ceRNA) network. Quantitative RT-PCR analysis was used to validate part of this network. GSEA was used to predict the potential functions of these lncRNAs. Conclusions This study identifies and validates a new method to investigate the miR-133b-mediated lncRNA-mRNA ceRNA network and lays the foundation for future investigation into the role of lncRNAs in colorectal cancer.


Oncotarget | 2016

Negative feedback between TAp63 and Mir-133b mediates colorectal cancer suppression

Jing Dai; Hao Wu; Yi Zhang; Kai Gao; Gui Hu; Yihang Guo; Changwei Lin; Xiaorong Li

Background TAp63 is known as the most potent transcription activator and tumor suppressor. microRNAs (miRNAs) are increasingly recognized as essential components of the p63 pathway, mediating downstream post-transcriptional gene repression. The aim of present study was to investigate a negative feedback loop between TAp63 and miR-133b. Results Overexpression of TAp63 inhibited HCT-116 cell proliferation, apoptosis and invasion via miR-133b. Accordingly, miR-133b inhibited TAp63 expression through RhoA and its downstream pathways. Moreover, we demonstrated that TAp63/miR-133b could inhibit colorectal cancer proliferation and metastasis in vivo and vitro. Materials and Methods We evaluated the correlation between TAp63 and miR-133b in HCT-116 cells and investigated the roles of the TAp63/miR-133b feedback loop in cell proliferation, apoptosis and metastasis via MTT, flow cytometry, Transwell, and nude mouse xenograft experiments. The expression of TAp63, miR-133b, RhoA, α-tubulin and Akt was assessed via qRT-PCR, western blot and immunofluorescence analyses. miR-133b target genes were identified through luciferase reporter assays. Conclusions miR-133b plays an important role in the anti-tumor effects of TAp63 in colorectal cancer. miR-133b may represent a tiemolecule between TAp63 and RhoA, forming a TAp63/miR-133b/RhoA negative feedback loop, which could significantly inhibit proliferation, apoptosis and metastasis.


Molecular Medicine Reports | 2015

MicroRNA‑133b inhibits connective tissue growth factor in colorectal cancer and correlates with the clinical stage of the disease.

Yihang Guo; Xiaorong Li; Changwei Lin; Yi Zhang; Gui Hu; Jianyu Zhou; Juan Du; Kai Gao; Yi Gan; Hao Deng

Accumulating evidence indicates that dysregulation of microRNA‑133b (miR‑133b) is an important step in the development of certain types of human cancer and contributes to tumorigenesis. Altered expression of miR‑133b has been reported in colon carcinoma, but its association with clinical stage in colorectal cancer (CRC) has remained elusive. Connective tissue growth factor (CTGF), a potentially promising candidate gene for interaction with miR‑133b, was screened using microarray analysis. The expression of miR‑133b and CTGF was evaluated using reverse transcription‑quantitative polymerase chain reaction and western blot analysis. The regulatory effects of miR‑133b on CTGF were evaluated using a dual‑luciferase reporter assay. CTGF was identified as a functional target of miR‑133b. The results demonstrated low expression of miR‑133b in CRC specimens with poor cell differentiation (P=0.011), lymph node metastasis (P=0.037) and advanced clinical stages (stage III or IV vs. I or II; P=0.036). Furthermore, there was a significant association between a high level of expression of CTGF mRNA and an advanced clinical stage (stage III or IV vs. I or II; P=0.015) and lymph node metastasis (P=0.034). CTGF expression was negatively regulated by miR‑133b in the human colorectum, suggesting that miR‑133b and CTGF may be candidate therapeutic targets in colorectal cancer.


Cancer Cell International | 2018

Long noncoding RNA LINC01510 promotes the growth of colorectal cancer cells by modulating MET expression

Chaoqun Cen; Jian Li; Jingjing Liu; Mingshi Yang; Tianyi Zhang; Yu Zuo; Changwei Lin; Xiaorong Li

BackgroundAbnormal expression of long non-coding RNA (lncRNAs) often facilitates unrestricted growth of cancer cells. Long intergenic non-protein coding RNA 1510, an enhancer lncRNA (LINC01510), a lncRNA enhancer is upregulated in colorectal cancer (CRC), and its expression might relate to MET as revealed by lncRNA microarray data. However, the potential biological role of LINC01510 and its regulatory mechanism in CRC remain unclear. Therefore, we investigated the involvement of LINC01510 in the proliferation of CRC cells.MethodsMicroarray analysis, In situ hybridization, colony formation assay, MTT assay, Western blotting, quantitative RT-PCR and flow cytometry were applied. The two-tailed Student’s t test and analysis of variance or general linear model of single factor variable was used for statistical analyse.ResultsIn the present study, we found that LINC01510 was significantly upregulated in CRC tissues and cell lines. The LINC01510 expression level were associated with the clinicopathological grade and stage. Meanwhile, gain- and loss-of-function assays demonstrated that LINC01510 overexpression increased CRC cell proliferation, and promoted cell cycle progression from the G1 phase to the S phase. Further study indicated that LINC01510 was positively correlated with the expression of MET, and its effects were most likely at the transcriptional level.ConclusionsTaken together, our findings suggested that upregulation of LINC01510 contributes to the proliferation of CRC cells, at least in part, through the regulation of MET protein. LINC01510 could be a candidate prognostic biomarker and a target for new therapies in CRC patients.


Oncotarget | 2017

miR-133b, a particular member of myomiRs, coming into playing its unique pathological role in human cancer

Daojiang Li; Lu Xia; Miao Chen; Changwei Lin; Hao Wu; Yi Zhang; Songqing Pan; Xiaorong Li

MicroRNAs, a family of single-stranded and non-coding RNAs, play a crucial role in regulating gene expression at posttranscriptional level, by which it can mediate various types of physiological and pathological process in normal developmental progress and human disease, including cancer. The microRNA-133b originally defined as canonical muscle-specific microRNAs considering their function to the development and health of mammalian skeletal and cardiac muscles, but new findings coming from our group and others revealed that miR-133b have frequently abnormal expression in various kinds of human cancer and its complex complicated regulatory networks affects the tumorigenicity and development of malignant tumors. Very few existing reviews on miR-133b, until now, are principally about its role in homologous cluster (miR-1, −133 and -206s), however, most of constantly emerging new researches now are focused mainly on one of them, so In this article, to highlight the unique pathological role of miR-133b playing in tumor, we conduct a review to summarize the current understanding about one of the muscle-specific microRNAs, namely miR-133b, acting in human cancer. The review focused on the following four aspects: the overview of miR-133b, the target genes of miR-133b involved in human cancer, the expression of miR-133b and regulatory mechanisms leading to abnormal expression of miR-133b.


Oncotarget | 2017

miR-133b down-regulates ABCC1 and enhances the sensitivity of CRC to anti-tumor drugs

Miao Chen; Daojiang Li; Ni Gong; Hao Wu; Chen Su; Canbin Xie; Hong Xiang; Changwei Lin; Xiaorong Li

Multidrug resistance (MDR) is the main cause of failed chemotherapy treatments. Therefore, preventing MDR is pivotal in treating colorectal cancer (CRC). In a previous study miR-133b was shown to be a tumor suppressor. Additionally, in CRC cells transfected with miR-133b, ATP-binding cassette (ABC) subfamily C member 1(ABCC1) was shown to be significantly down regulated. Whether miR-133b also enhances the chemosensitivity of drugs used to treat CRC by targeting ABCC1 is still unclear. Here, we utilized flow cytometry and high-performance liquid chromatography (HPLC) analysis to identify the ability of miR-133b to reserve MDR in CRC. We then used a dual-luciferase reporter assay to validate that miR-133b targets ABCC1. Further in vivo experiments were designed to validate the method in which miR-133b reversed MDR in CRC cells. The results demonstrated that the level of miR-133b was down-regulated and the expression of ABCC1 was up-regulated in drug-resistant CRC cells compared to non-drug-resistant CRC cells. The restoration of miR-133b expression in CRC drug-resistant cells in vitro resulted in reduced IC50s to chemotherapeutic drugs, significantly induced G1 accumulation, inhibited growth and promoted necrosis in combination with either 5-fluorouracil (5-FU) or vincristine (VCR), and decreased the expression of ABCC1. The dual-luciferase assay demonstrated that miR-133b directly targets ABCC1. The combination of agomiRNA-133b with chemotherapeutic drugs in vivo inhibited tumor growth induced by CRC drug-resistant cells. A xenograft from the in vivo model resulted in up-regulated levels of miR-133b and down-regulated levels of ABCC1. Therefore, miR-133b enhances the chemosensitivity of CRC cells to anti-tumor drugs by directly down-regulating ABCC1. This discovery provides a therapeutic strategy in which miR-133b is used as a potential sensitizer for drug-resistant CRC.Multidrug resistance (MDR) is the main cause of failed chemotherapy treatments. Therefore, preventing MDR is pivotal in treating colorectal cancer (CRC). In a previous study miR-133b was shown to be a tumor suppressor. Additionally, in CRC cells transfected with miR-133b, ATP-binding cassette (ABC) subfamily C member 1(ABCC1) was shown to be significantly down regulated. Whether miR-133b also enhances the chemosensitivity of drugs used to treat CRC by targeting ABCC1 is still unclear. Here, we utilized flow cytometry and high-performance liquid chromatography (HPLC) analysis to identify the ability of miR-133b to reserve MDR in CRC. We then used a dual-luciferase reporter assay to validate that miR-133b targets ABCC1. Further in vivo experiments were designed to validate the method in which miR-133b reversed MDR in CRC cells. The results demonstrated that the level of miR-133b was down-regulated and the expression of ABCC1 was up-regulated in drug-resistant CRC cells compared to non-drug-resistant CRC cells. The restoration of miR-133b expression in CRC drug-resistant cells in vitro resulted in reduced IC50s to chemotherapeutic drugs, significantly induced G1 accumulation, inhibited growth and promoted necrosis in combination with either 5-fluorouracil (5-FU) or vincristine (VCR), and decreased the expression of ABCC1. The dual-luciferase assay demonstrated that miR-133b directly targets ABCC1. The combination of agomiRNA-133b with chemotherapeutic drugs in vivo inhibited tumor growth induced by CRC drug-resistant cells. A xenograft from the in vivo model resulted in up-regulated levels of miR-133b and down-regulated levels of ABCC1. Therefore, miR-133b enhances the chemosensitivity of CRC cells to anti-tumor drugs by directly down-regulating ABCC1. This discovery provides a therapeutic strategy in which miR-133b is used as a potential sensitizer for drug-resistant CRC.


Oncotarget | 2017

Comparative study of joint bioinformatics analysis of underlying potential of ‘neurimmiR’, miR-212-3P/miR-132-3P, being involved in epilepsy and its emerging role in human cancer

Lu Xia; Daojiang Li; Changwei Lin; Shuchun Ou; Xiaorong Li; Songqing Pan

Considering the critical roles of miR-132/212 participated in central nervous system, many researches started to explored the contributions of miR-132/212 to epilepsy and achieve something worthwhile. Further illuminates all the genes targeted by miR-132/212 may be a valuable means for us to completely understand the working mechanism playing in epilepsy, by which it can influence diverse biological process. This study attempts to establish macrocontrol regulation system and knowledge that miR-212-3p/132-3p effected the epilepsy, for this literature search, miRbase, Vienna RNAfold webserver, Human miRNA tissue atlas, DIANA-TarBase, miRtarbase, STRING, TargetScanhuman, Cytoscape plugin ClueGO + Cluepedia+STRING, DAVID Bioinformatics Resources, Starbase, GeneCards suite and GEO database are comprehensive employed, miR-132-3p/212-3p and its target gene were found have highly expressed in brain and lots of molecular function and metabolic pathways associated with epilepsy may be intervened by it. Meanwhile, the emerging role of miR-132-3p/212-3p being involved in human cancer also been analyzed by several webtools for TCGA data integrative analysis, most remarkably and well worth exploring in our research conclusion that showed miR-132-3p/212-3p may be the core molecular underlying tumor-induced epileptogenesis.

Collaboration


Dive into the Changwei Lin's collaboration.

Top Co-Authors

Avatar

Xiaorong Li

Central South University

View shared research outputs
Top Co-Authors

Avatar

Gui Hu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Yi Zhang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Yihang Guo

Central South University

View shared research outputs
Top Co-Authors

Avatar

Daojiang Li

Central South University

View shared research outputs
Top Co-Authors

Avatar

Jianyu Zhou

Central South University

View shared research outputs
Top Co-Authors

Avatar

Kai Gao

Central South University

View shared research outputs
Top Co-Authors

Avatar

Juan Du

Central South University

View shared research outputs
Top Co-Authors

Avatar

Miao Chen

Central South University

View shared research outputs
Top Co-Authors

Avatar

Chen Su

Central South University

View shared research outputs
Researchain Logo
Decentralizing Knowledge