Network


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

Hotspot


Dive into the research topics where Rui-Min Chang is active.

Publication


Featured researches published by Rui-Min Chang.


Hepatology | 2013

MicroRNA‐140‐5p suppresses tumor growth and metastasis by targeting transforming growth factor β receptor 1 and fibroblast growth factor 9 in hepatocellular carcinoma

Hao Yang; Feng Fang; Rui-Min Chang; Lian-Yue Yang

By comparing the expression profiles of microRNAs (miRNAs) in different hepatocellular carcinoma (HCC) subtypes, we identified miR‐140‐5p as an HCC‐related miRNA. We found that miR‐140‐5p was significantly decreased in HCC tissues and all of six liver cancer cell lines examined and its expression levels were correlated with multiple nodules, vein invasion, capsular formation, and differentiation, as well as overall and disease‐free survival of HCC. We also found that miR‐140‐5p suppressed HCC cell proliferation and HCC metastasis. Multipathway reporter arrays suggested that miR‐140‐5p inhibited transforming growth factor β (TGF‐β) and mitogen‐activated protein kinase / extracellular signal‐regulated kinase (MAPK/ERK) signaling. TGFB receptor 1 (TGFBR1) and fibroblast growth factor 9 (FGF9) were then characterized as the direct targets for miR‐140‐5p after it was found that ectopic miR‐140‐5p expression suppressed TGFBR1 and FGF9 expression. Silencing TGFBR1 and FGF9 by small interfering RNA (siRNA) resembled the phenotype resulting from ectopic miR‐140‐5p expression, while overexpression of TGFBR1 and FGF9 attenuated the effect of miR‐140‐5p on HCC growth and metastasis. Conclusion: These data elucidated a tumor suppressor role for miR‐140‐5p in HCC development and progression with therapeutic potential. Our correlation studies in clinical HCC samples further suggest that miR‐140‐5p could be a valuable biomarker for HCC prognosis. (Hepatology 2013 )


Hepatology | 2014

MicroRNA‐331‐3p promotes proliferation and metastasis of hepatocellular carcinoma by targeting PH domain and leucine‐rich repeat protein phosphatase

Rui-Min Chang; Hao Yang; Feng Fang; Jiang-Feng Xu; Lian-Yue Yang

Hepatocellular carcinoma (HCC) is a highly invasive tumor with frequent intrahepatic or pulmonary metastasis, which is the main reason for high recurrence and poor survival of HCC after liver resection. However, the mechanisms for metastasis remain incompletely clear. Given that microRNAs (miRNAs) are implicated in HCC progression, we explored a potential role of miRNAs in metastasis by performing miRNA expression profiling in three subtypes of HCC with different metastatic potentials. We discovered miR‐331‐3p as one of most significantly overexpressed miRNAs and highly associated with metastasis of HCC. Increased expression of miR‐331‐3p was correlated with poor long‐term survival of HCC. We provided both in vivo and in vitro evidence demonstrating that miR‐331‐3p promoted proliferation and metastasis of HCC cells. Using an integrated approach, we uncovered that PH domain and leucine‐rich repeat protein phosphatase (PHLPP) was a novel target of miR‐331‐3p. Indeed, the miR‐331‐3p‐mediated effects were antagonized by reexpression of PHLPP or mimicked by silencing of PHLPP. We further showed that miR‐331‐3p‐mediated inhibition of PHLPP resulted in stimulation of protein kinase B (AKT) and subsequent epithelial mesenchymal transition (EMT). Finally, inhibition of miR‐331‐3p through a jetPEI‐mediated delivery of anti‐miR‐331‐3p vector resulted in marked inhibition of proliferation and metastasis of HCC in xenograft mice. Conclusion: miR‐331‐3p promotes proliferation and EMT‐mediated metastasis of HCC through suppression of PHLPP‐mediated dephosphorylation of AKT. Our work implicates miR‐331‐3p as a potential prognostic biomarker and a novel therapeutic target. (Hepatology 2014;60:1251–1263)


Cancer | 2012

Heat shock factor 1 promotes invasion and metastasis of hepatocellular carcinoma in vitro and in vivo

Feng Fang; Rui-Min Chang; Lian‐Yue Yang

Heat shock factor 1 (HSF1) is a powerful, multifaceted modifier of carcinogenesis. However, the clinical significance and biologic function of HSF1 in hepatocellular carcinoma (HCC) remain unknown.


Journal of Hepatology | 2015

MicroRNA-188-5p suppresses tumor cell proliferation and metastasis by directly targeting FGF5 in hepatocellular carcinoma.

Feng Fang; Rui-Min Chang; Lei Yu; Xiong Lei; Shuai Xiao; Hao Yang; Lian-Yue Yang

BACKGROUND & AIMS Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. However, the detailed molecular mechanisms underlying HCC progression are still not completely clear. Given the crucial role of microRNAs (miRNAs) in cancer metastasis, we aimed to analyze the expression and function of a metastasis-associated miRNA named miR-188-5p in HCC. METHODS miRNA array analysis was performed to search for metastasis-associated miRNAs in HCC. miR-188-5p expressions in tumor tissues and adjacent non-tumorous liver tissues of HCC patients and cell lines were evaluated by real-time PCR. The protein expression levels were analyzed by Western blot and immunohistochemistry. Luciferase reporter assays was used to validate the target of miR-188-5p. The effect of miR-188-5p on HCC progression was studied in vitro and in vivo. RESULTS miR-188-5p was significantly decreased in HCC and its expression levels were highly correlated with multiple nodules, microvascular invasion, overall and disease-free survival of HCC. Ectopic expression of miR-188-5p suppressed HCC cell proliferation and metastasis in vitro and in vivo. Fibroblast growth factor 5 (FGF5) was identified as a major target of miR-188-5p. Enforced expression of miR-188-5p inhibited the expression of FGF5 significantly and the restoration of FGF5 expression reversed the inhibitory effects of miR-188-5p on HCC cell proliferation and metastasis. CONCLUSIONS These findings collectively demonstrate a tumor suppressor role of miR-188-5p in HCC progression via targeting FGF5, suggesting that miR-188-5p could serve as a potential prognostic biomarker and therapeutic target for HCC.


Hepatology | 2016

Actin-like 6A predicts poor prognosis of hepatocellular carcinoma and promotes metastasis and epithelial-mesenchymal transition

Shuai Xiao; Rui-Min Chang; Ming‐Yang Yang; Xiong Lei; Xiao Liu; Wen‐Bin Gao; Jing‐Lei Xiao; Lian-Yue Yang

Hepatocellular carcinoma (HCC) is one of the most lethal cancers worldwide because of metastasis. Epithelial‐mesenchymal transition (EMT) is widely considered to be crucial to the invasion‐metastasis cascade during cancer progression. Actin‐like 6A (ACTL6A) is initially verified important for cell proliferation, differentiation, and migration. In this study, we find that ACTL6A plays an essential role in metastasis and EMT of HCC. ACTL6A expression is up‐regulated in HCC cells and tissues. A high level of ACTL6A in HCCs is correlated with aggressive clinicopathological features and is an independent poor prognostic factor for overall and disease‐free survival of HCC patients. Ectopic expression of ACTL6A markedly promotes HCC cells migration, invasion, as well as EMT in vitro and promotes tumor growth and metastasis in the mouse xenograft model. Opposite results are observed when ACTL6A is knocked down. Mechanistically, ACTL6A promotes metastasis and EMT through activating Notch signaling. ACTL6A knockdown has the equal blockage effect as the Notch signaling inhibitor, N‐[N‐(3,5‐difluorophenacetyl)‐L‐alanyl]‐S‐phenylglycine t‐butylester, in HCC cells. Further studies indicate that ACTL6A might manipulate SRY (sex determining region Y)‐box 2 (SOX2) expression and then activate Notch1 signaling. Conclusions: ACTL6A promotes metastasis and EMT by SOX2/Notch1 signaling, indicating a prognostic biomarker candidate and a potential therapeutic target for HCC. (Hepatology 2016;63:1256–1271)


Cancer | 2013

SIN1 promotes invasion and metastasis of hepatocellular carcinoma by facilitating epithelial-mesenchymal transition

Jiang-Feng Xu; Xuedong Li; Hao Yang; Rui-Min Chang; Chenchen Kong; Lian-Yue Yang

Stress‐activated protein kinase (SAPK) interacting protein 1 (SIN1) is essential for early embryonic development and is the key regulator of Akt, which plays an important role in various pathological conditions such as cancer. However, the biological function and clinical significance of SIN1 in hepatocellular carcinoma (HCC) remains unknown.


Oncotarget | 2015

MicroRNA-424 inhibits Akt3/E2F3 axis and tumor growth in hepatocellular carcinoma

Hao Yang; Wei Zheng; Xiao Shuai; Rui-Min Chang; Lei Yu; Feng Fang; Lian-Yue Yang

By comparing the expression profiles of miRNAs in different subtypes of HCC, we identified miR-424 as a HCC related miRNA. We found that the expression of miR-424 was significantly decreased in HCC tissues and six liver cancer cell lines. Significantly, its expression levels were correlated with tumor size, multiple nodules, vein invasion, TNM stage and overall survival of HCC. We showed that up-regulated miR-424 suppressed HCC cell proliferation in vivo and in vitro. Multi-pathway reporter arrays suggested that miR-424 suppressed the pRb-E2F pathway. Consistently, Akt3 and E2F3 were identified as the targets of miR-424 as evidenced by that ectopic miR-424 expression suppressed Akt3 and E2F3 expressions. Silencing Akt3 and E2F3 by siRNA pheno-copied the effect of ectopic miR-424 on HCC growth. Whereas, overexpression of Akt3 and E2F3 attenuated the effect of miR-424 on HCC growth. Together, our data demonstrated a tumor suppressor role for miR-424 in HCC development and progression with therapeutic implications. The strong correlation of miR-424 expression with HCC patient survival suggests that miR-424 could be a valuable biomarker for HCC prognosis.


Tumor Biology | 2016

MicroRNA-130b promotes proliferation and EMT-induced metastasis via PTEN/p-AKT/HIF-1α signaling

Rui-Min Chang; Jiang-Feng Xu; Feng Fang; Hao Yang; Lian-Yue Yang

Hepatocellular carcinoma (HCC) is a major cause of cancer-related deaths owing to its high rate of postoperative recurrence and metastasis. New research is continuously identifying novel metastasis-associated oncogenes and tumor suppressor genes. miRNAs are noncoding RNAs that regulate protein synthesis post-translationally. miR-130b is one of several miRNAs involved in tumor metastasis. However, the role of miR-130b in HCC remains controversial. Here, we demonstrate that miR-130b is highly expressed in HCC and that it correlates with tumor number, vascular invasion, and TNM stage—important predictors of postoperative recurrence and metastases. Moreover, high levels of miR-130b predicted poor overall and disease-free survival of HCC patients, and in vitro and in vivo research revealed that knockdown or overexpression of miR-130b inhibited and promoted proliferation and metastasis of HCC cells, respectively. We identified PTEN as a direct functional target of miR-130b using miRNA databases and a dual luciferase report assay. Next, using a gain and loss assay and epithelial-mesenchymal transition (EMT) relative assays, we show that miR-130b may promote proliferation and EMT-induced metastasis via PTEN/p-AKT/HIF-1α signaling. Collectively, our data suggests that miR-130b may have prognostic value in HCC. Additionally, the miR-130b/PTEN/p-AKT/HIF-1α axis identified in this study provides novel insight into the mechanisms of HCC metastasis, which may facilitate the development of new therapeutics against HCC.


Oncotarget | 2016

JARID2 promotes invasion and metastasis of hepatocellular carcinoma by facilitating epithelial-mesenchymal transition through PTEN/AKT signaling

Xiong Lei; Jiang-Feng Xu; Rui-Min Chang; Feng Fang; Chao-Hui Zuo; Lian-Yue Yang

JARID2 is crucial for maintenance of pluripotency and differentiation of embryonic stem cells. However, little is known about the role of JARID2 in metastasis of hepatocellular carcinoma (HCC). This study found that JARID2 expression was significantly higher in HCC tissues than that in adjacent non-tumor liver tissues (ANLTs), and its expression level correlated with HCC metastasis. High JARID2 expression was significantly correlated with multiple tumor nodules, high Edmondson-Steiner grade, microvascular invasion, advanced TNM stage and advanced BCLC stage (all P < 0.05) and indicated poor prognosis of HCC in training and validation cohorts (all P < 0.05) totaling 182 patients. High JARID2 expression was an independent and significant risk factor for disease-free survival (DFS; P = 0.017) and overall survival (OS; P = 0.041) after curative liver resection in training cohort, and also validated as an independent and significant risk factor for DFS (P = 0.033) and OS (P = 0.031) in validation cohort. Moreover, down-regulation of JARID2 dramatically inhibited HCC cell migration, invasion, proliferation in vitro and metastasis in vivo, whereas overexpression of JARID2 significantly increased migration, invasion, proliferation in vitro and metastasis in vivo. Mechanistically, the data showed that JARID2 exerted its function by repressing PTEN expression through increasing H3K27 trimethylation (H3K27me3) at PTEN promoter region, which subsequently resulted in activation of protein kinase B (AKT) and enhanced epithelial-mesenchymal transition (EMT). In conclusion, this study revealed that JARID2 promotes invasion and metastasis of HCC by facilitating EMT through PTEN/AKT signaling.


Clinical Cancer Research | 2017

miRNA-487a Promotes Proliferation and Metastasis in Hepatocellular Carcinoma

Rui-Min Chang; Shuai Xiao; Xiong Lei; Hao Yang; Feng Fang; Lian-Yue Yang

Purpose: Hepatocellular carcinoma (HCC) harbors highly metastatic properties, accounting for postoperative recurrence and metastasis. However, the mechanisms for metastasis and recurrence remain incompletely clear. This study aimed to investigate the role of hsa-miR-487a (miR-487a) in promoting the proliferation and metastasis of HCC and to elucidate the underlying molecular mechanisms. Experimental Design: 198 HCC samples were analyzed for association between miR-487a expression and patient clinicopathological features and prognosis. The roles of miR-487a in proliferation and metastasis were validated both in vivo and in vitro. The upstream regulator and downstream targets of miR-487a were determined using a dual luciferase reporter assay, chromatin immunoprecipitation and immunohistochemistry. Results: Our results demonstrate that upregulated miR-487a correlates with a poor prognosis for HCC patients. miR-487a enhances proliferation and metastasis of HCC cells by directly binding to sprouty-related EVH1 domain containing 2 (SPRED2) or phosphoinositide-3-Kinase regulatory subunit 1 (PIK3R1). Interestingly, miR-487a mainly promotes metastasis via SPRED2 induced mitogen activated protein kinase signaling and promotes proliferation via PIK3R1 mediated AKT signaling. Transcription of miR-487a was found to be activated by up-regulated heat shock factor 1, which we previously demonstrated to be an important metastasis-associated transcription factor in a previous study. Phosphorodiamidate morpholino oligomers effectively silenced miR-487a and inhibited HCC tumor progression in mouse models. Conclusions: Our findings show that miR-487a, mediated by heat shock factor 1, promotes proliferation and metastasis of HCC by PIK3R1 and SPRED2 binding, respectively. Our study provides a rationale for developing miR-487a as a potential prognostic marker or a potential therapeutic target against HCC. Clin Cancer Res; 23(10); 2593–604. ©2016 AACR.

Collaboration


Dive into the Rui-Min Chang's collaboration.

Top Co-Authors

Avatar

Lian-Yue Yang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Feng Fang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Hao Yang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Jiang-Feng Xu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Shuai Xiao

Central South University

View shared research outputs
Top Co-Authors

Avatar

Xiong Lei

Central South University

View shared research outputs
Top Co-Authors

Avatar

Lei Yu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Chenchen Kong

Central South University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xuedong Li

Central South University

View shared research outputs
Researchain Logo
Decentralizing Knowledge