Network


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

Hotspot


Dive into the research topics where Ranran Shi is active.

Publication


Featured researches published by Ranran Shi.


Molecular Neurobiology | 2015

Retraction Note to: CXCR4 Signaling Induced Epithelial-Mesenchymal Transition by PI3K/AKT and ERK Pathways in Glioblastoma.

Baoyu Lv; Xiangshan Yang; Shunzeng Lv; Lei Wang; Kaixi Fan; Ranran Shi; Fengling Wang; Huishu Song; Xiaochen Ma; Xuefen Tan; Kun Xu; Jingjing Xie; Guangmei Wang; Man Feng; Li Zhang

Stromal cell-derived factor 1 (SDF-1) and its receptor, CXCR4, play an important role in tumor progression. Epithelial-mesenchymal transition (EMT) process is linked to disease pathophysiology. This study aimed to investigate the roles and underlying mechanisms of SDF-1/CXCR4 axis in EMT process of glioblastoma. In the present study, CXCR4 activation and inhibition in U87 were induced with exogenous SDF-1 and with CXCR4 small interfering RNA (siRNA), respectively. CXCR4 downstream signal molecules AKT, ERK, and EMT biomarkers (vementin, snail, N-cadherin, and E-cadherin) were tested using the Western blot. Our results showed that SDF-1 can induce AKT and ERK phosphorylation in a dose-dependent manner, and endogenous CXCR4 can be blocked thoroughly by CXCR4 siRNA in U87. Notably SDF-1 alone treatment can induce the upregulation of vementin, snail, and N-cadherin of U87; besides, the downregulation of E-cadherin also occurred. On the contrary, CXCR4 siRNA significantly prohibited SDF-1-induced AKT and ERK phosphorylation, at the same time, EMT biomarker changes were not observed. Function analysis revealed that CXCR4 siRNA obviously interfered with U87 cell migration and proliferation, according to wound healing assay. In conclusion, this study suggested that EMT process can be triggered by the SDF-1/CXCR4 axis in glioblastoma, and then involved in the tumor cell invasion and proliferation via activation of PI3K/AKT and ERK pathway. Our study lays a new foundation for the treatment of glioblastoma through antagonizing CXCR4.


Molecular Neurobiology | 2015

CXCL12/CXCR4 Axis Upregulates Twist to Induce EMT in Human Glioblastoma.

Chengjun Yao; Panpan Li; Huishu Song; Fuxi Song; Yalan Qu; Xiaochen Ma; Ranran Shi; Jinsong Wu

In recent decades, the chemokine receptor CXCR4 and its ligand CXCL12 have been extensively reported to be associated with tumorigenesis. In addition, Twist signaling induces the epithelial-mesenchymal transition (EMT) process in glioblastoma development. In the present study, in vitro assays were used to investigate the role of CXCR4 and Twist in human glioblastoma. We explored the impact of CXCR4 and Twist on human glioblastoma using in vitro protein and gene assays. We found the administration of CXCL12 upregulated the expression of p-ERK, p-AKT, Twist, N-cadherin, and MMP9 in U87 cells, whereas the increase of E-cadherin protein was affected. Subsequently, Twist activity and EMT signaling were directly influenced by PD98059 and LY294002. Most importantly, the genetic silencing of Twist inhibited CXCL12-induced EMT occurrence, including proliferation, migration, and tumor formation of U87 cells. In conclusion, CXCL12/CXCR4 pathway activates ERK and PI3K/AKT signaling to upregulate Twist pathway, leading to the progression of EMT in human glioblastoma. Our study creates a new stage for molecule-targeted therapy of human glioblastoma.


Molecular Neurobiology | 2014

SDF-1/CXCR4 Axis Regulates Cell Cycle Progression and Epithelial-Mesenchymal Transition via Up-regulation of Survivin in Glioblastoma.

Anyan Liao; Ranran Shi; Yuliang Jiang; Suqing Tian; Panpan Li; Fuxi Song; Yalan Qu; Jinna Li; Haiqin Yun; Xiangshan Yang

Stromal cell-derived factor 1 (SDF-1)/CXCR4 ligand-receptor axis is widely recommended as an attractive target for cancer therapy. Meanwhile, epithelial-mesenchymal transition (EMT) process is linked to disease pathophysiology. As one of inhibitors of apoptosis proteins, survivin is implicated in the onset and development of cancer. In the present study, we tried to determine the cause-effect associations between SDF-1/CXCR4 axis and survivin expression in glioblastoma U-251 cell line. Survivin activation and inhibition were induced with exogenous SDF-1 and survivin small interfering RNA (survivin siRNA), respectively. Western blot was used to detect relevant proteins in SDF-1/CXCR4 axis. Western blot analysis revealed that survivin expression in U-251 increased in a dose- and time-dependent manner in response to SDF-1 treatment. However, the interference with MEK/ERK and PI3K/AKT pathway prohibited SDF-1-induced survivin up-regulation. Importantly, survivin knockdown abrogated cell cycle progression and the expression of snail and N-cadherin, compared with non-transfectants. In conclusion, the present study shows that SDF-1 up-regulates survivin via MEK/ERK and PI3K/AKT pathway, leading to cell cycle progression and EMT occurrence dependent on survivin. The blockade of survivin will allow for the treatment of glioblastoma.


Journal of Molecular Neuroscience | 2013

CrkL efficiently mediates cell proliferation, migration, and invasion induced by TGF-β pathway in glioblastoma.

Shunzeng Lv; Juan Qin; Ruiyang Yi; Melody Coreman; Ranran Shi; Huihui Kang; Chengjun Yao

Crk-like (CrkL) is an adapter protein that has crucial roles in cell proliferation, adhesion, and migration. However, the expression pattern and potential mechanism of CrkL protein in glioblastoma multiforme (GBM) have not been fully elucidated. To determine roles of CrkL in cell signaling, proliferation, and migration, small interfering RNAs and plasmids transfection were used to suppress or overexpress CrkL in U87 and U251; soft-agar assay and wound-healing assay were used to observe cell invasiveness, migration, and proliferation. Erk1/2, Smad2, and matrix metalloproteinase 9 (MMP9) were also analyzed by western blot. CrkL was expressed in U87 and U251 cell lines and can be activated by transforming growth factor-beta 1 (TGF-β1) in vitro; CrkL knockdown significantly suppressed the expression of phosph-ERK1/2 and MMP9 but enhanced phosph-Smad2 expression compared with control (p < 0.001). Overexpression of CrkL against control upregulated phosph-ERK1/2 and MMP9 and, at the same time, downregulated phosph-Smad2 (p < 0.01). On the other hand, CrkL knockdown could significantly affect U87 and U251 invasiveness (p < 0.01) and wound closure (p < 0.01) using soft-agar assay and wound-healing assay. These studies suggest that CrkL efficiently mediates cell proliferation, migration, and invasion induced by TGF-β pathway in glioblastoma. Furthermore, CrkL can be used as a potential and efficient therapeutic target of GBM and may also mediate other signaling pathway.


Tumor Biology | 2015

RETRACTED ARTICLE: Bone morphogenetic protein 2 mediates epithelial-mesenchymal transition via AKT and ERK signaling pathways in gastric cancer

Anyan Liao; Weijie Wang; Dawei Sun; Yuliang Jiang; Suqing Tian; Jinna Li; Xiangshan Yang; Ranran Shi

Although deregulation of bone morphogenetic protein 2 (BMP2) signaling has been linked to various types of cancers, the relationships between abnormal activation of these signaling pathways and tumorigenesis are not clear in gastric cancer. We hypothesized that BMP2 might be involved in epithelial-mesenchymal transition (EMT) process of gastric cancer. Here, BMPR-II activation and inhibition in gastric cancer cell line AGS were induced with exogenous BMP2 and with BMPR-II small interfering RNA (siRNA), respectively. BMPR-II downstream signal molecules AKT, ERK phosphorylation, and EMT biomarkers (vimentin, snail, N-cadherin, and E-cadherin) were tested using the Western blot. In the present study, our results showed that BMP2 can induce AKT and ERK phosphorylation in a dose-dependent method, and endogenous BMPR-II can be inhibited completely by BMPR-II siRNA in AGS. Notably BMP2 alone treatment can induce the up-regulation of vimentin, snail, and N-cadherin in AGS cells, besides, the down-regulation of E-cadherin also occurred. On the contrary, BMPR-II siRNA significantly prohibited BMP2-induced AKT and ERK phosphorylation, at the same time, EMT biomarkers changes were not observed. On the other hand, BMPR-II knockdown could significantly affect AGS wound closure and the migration ability (p < 0.001) compared to control siRNA and BMP2 alone. In conclusion, this study suggested that EMT process can be triggered by the BMP2/BMPR axis in gastric cancer and then involved in the tumor cell migration, invasion, and metastasis via the activation of PI3K/AKT and MEK/ERK pathways. Our study lays a new foundation for the treatment of gastric cancer through antagonizing BMP2 system.


Molecular Neurobiology | 2015

The Downregulation of MicroRNA-146a Modulates TGF-β Signaling Pathways Activity in Glioblastoma.

Shunzeng Lv; Bowen Sun; Congxin Dai; Ranran Shi; Xingtong Zhou; Wenyuan Lv; Xiao Zhong; Renzhi Wang; Wenbin Ma

Transforming growth factor-β (TGF-β) is considered to be one of the main factors responsible for glioblastoma tumorigenesis. MicroRNAs have recently been shown to regulate cell proliferation, differentiation, and apoptosis. However, the involvement of miRNA-146a in TGF-β1-induced glioblastoma development remains largely unknown. Here, miRNA-164a transfection was used to overexpress miRNA-164a in U87, and then real-time quantitative PCR and Western blot were applied to detect the gene transcription and protein expression. In addition, MTT and wound healing assay were also used to observe cell proliferation and migration. Our data revealed that miRNA-146a was downregulated by TGF-β1 treatment, but upregulated by miRNA-164a transfection. MiRNA-146a overexpression significantly reduced SMAD4 protein expression instead of p-SMAD2. Besides, miRNA-146a overexpression also decreased the messenger RNA (mRNA) and protein expression of epidermal growth factor receptor (EGFR) and MMP9 as well as the p-ERK1/2 level. Furthermore, the upregulation of miRNA-146a suppressed TGF-β1-mediated U87 proliferation and migration. These results demonstrate that miRNA-146a acts as a novel regulator to modulate the activity and transduction of TGF-β signaling pathways in glioblastoma, and the downregulation of miRNA-146a is required for overexpression of EGFR and MMP9, which can be considered an efficiently therapeutic target and a better understanding of glioblastoma pathogenesis.


Molecular Neurobiology | 2015

Retraction Note to: The Downregulation of MicroRNA-146a Modulates TGF-β Signaling Pathways Activity in Glioblastoma.

Shunzeng Lv; Bowen Sun; Congxin Dai; Ranran Shi; Xingtong Zhou; Wenyuan Lv; Xiao Zhong; Renzhi Wang; Wenbin Ma

Transforming growth factor-β (TGF-β) is considered to be one of the main factors responsible for glioblastoma tumorigenesis. MicroRNAs have recently been shown to regulate cell proliferation, differentiation, and apoptosis. However, the involvement of miRNA-146a in TGF-β1-induced glioblastoma development remains largely unknown. Here, miRNA-164a transfection was used to overexpress miRNA-164a in U87, and then real-time quantitative PCR and Western blot were applied to detect the gene transcription and protein expression. In addition, MTT and wound healing assay were also used to observe cell proliferation and migration. Our data revealed that miRNA-146a was downregulated by TGF-β1 treatment, but upregulated by miRNA-164a transfection. MiRNA-146a overexpression significantly reduced SMAD4 protein expression instead of p-SMAD2. Besides, miRNA-146a overexpression also decreased the messenger RNA (mRNA) and protein expression of epidermal growth factor receptor (EGFR) and MMP9 as well as the p-ERK1/2 level. Furthermore, the upregulation of miRNA-146a suppressed TGF-β1-mediated U87 proliferation and migration. These results demonstrate that miRNA-146a acts as a novel regulator to modulate the activity and transduction of TGF-β signaling pathways in glioblastoma, and the downregulation of miRNA-146a is required for overexpression of EGFR and MMP9, which can be considered an efficiently therapeutic target and a better understanding of glioblastoma pathogenesis.


Molecular Neurobiology | 2016

Retraction Note to “SDF-1/CXCR4 Axis Regulates Cell Cycle Progression and Epithelial-Mesenchymal Transition via Up-regulation of Survivin in Glioblastoma”

Anyan Liao; Ranran Shi; Yuliang Jiang; Suqing Tian; Panpan Li; Fuxi Song; Yalan Qu; Jinna Li; Haiqin Yun; Xiangshan Yang

Stromal cell-derived factor 1 (SDF-1)/CXCR4 ligand-receptor axis is widely recommended as an attractive target for cancer therapy. Meanwhile, epithelial-mesenchymal transition (EMT) process is linked to disease pathophysiology. As one of inhibitors of apoptosis proteins, survivin is implicated in the onset and development of cancer. In the present study, we tried to determine the cause-effect associations between SDF-1/CXCR4 axis and survivin expression in glioblastoma U-251 cell line. Survivin activation and inhibition were induced with exogenous SDF-1 and survivin small interfering RNA (survivin siRNA), respectively. Western blot was used to detect relevant proteins in SDF-1/CXCR4 axis. Western blot analysis revealed that survivin expression in U-251 increased in a dose- and time-dependent manner in response to SDF-1 treatment. However, the interference with MEK/ERK and PI3K/AKT pathway prohibited SDF-1-induced survivin up-regulation. Importantly, survivin knockdown abrogated cell cycle progression and the expression of snail and N-cadherin, compared with non-transfectants. In conclusion, the present study shows that SDF-1 up-regulates survivin via MEK/ERK and PI3K/AKT pathway, leading to cell cycle progression and EMT occurrence dependent on survivin. The blockade of survivin will allow for the treatment of glioblastoma.


Molecular Neurobiology | 2015

Retraction Note to: The Clinical Utility of Matrix Metalloproteinase 9 in Evaluating Pathological Grade and Prognosis of Glioma Patients: A Meta-Analysis

Xiangshan Yang; Shunzeng Lv; Yuting Liu; Daotang Li; Ranran Shi; Zhenyu Tang; Jianzhen Fan; Zhongfa Xu

In the recent years, matrix metalloproteinase 9 (MMP-9) has been focused on as an indicator of glioma grade and prognosis, especially in China. However, all results resulted in many conflicts. So, it is necessary to conduct a meta-analysis to secure a convincing correlation between MMP-9 and grade and prognosis. Eligible studies were included via multiple searches, and then odds ratios (ORs) and hazard ratios (HRs) with 95 % confidence intervals (95 % CIs) were estimated. Funnel plots were available for evaluation of publication bias. In addition, heterogeneity and sensitivity were also analyzed. In the present meta-analysis, 23 articles were allowed for inclusion with total 1,635 patients. Coincidentally, all studies were conducted in Chinese populations. High MMP-9 expression in gliomas was closely associated with high WHO grade (III+IV) (n = 22, OR = 5.25, 95 % CI = 4.09–6.73; p = 0.000), while MMP-9 expression did not correlate to age (n = 4, OR = 1.02, 95 % CI = 0.67–1.54; p = 0.929) and gender (n = 5, OR = 0.91, 95 % CI = 0.63–1.33; p = 0.632). Besides, overall survival analysis from two articles revealed MMP-9 expression significantly predicted 5-year-OS (HR = 6.44, 95 % CI = 3.88–10.70; p = 0.000) in glioma patients. No heterogeneity and publication bias were observed across all studies. To conclude, this meta-analysis suggests MMP-9 is potently associated with high grade and poor 5 years prognosis, and MMP-9 test of glioma tissues should be established in department of pathology as a routine in clinical practice.


Molecular Neurobiology | 2015

RETRACTED ARTICLE: Nuclear Protein C23 on the Cell Surface Plays an Important Role in Activation of CXCR4 Signaling in Glioblastoma

Congxin Dai; Shunzeng Lv; Ranran Shi; Jing Ding; Xiao Zhong; Huishu Song; Xiaochen Ma; Jianzhen Fan; Bowen Sun; Renzhi Wang; Wenbin Ma

The chemokine receptor CXCR4 and its ligand stromal cell-derived factor 1 (SDF-1) plays an important role in tumor progression and are associated with angiogenesis. Meanwhile, the implications of C23 in multiple signaling pathways have been also investigated. However, the effects of C23 on CXCR4 pathway in glioblastoma are not fully characterized. In the present study, C23 and CXCR4 of U87 cell line were inhibited by anti-C23 and anti-CXCR4 antibodies, respectively; and then C23 and CXCR4 siRNAs were used to knock down endogenous C23 and CXCR4, respectively. In addition, MTT assay was also introduced. Our data showed that either anti-C23 or anti-CXCR4 antibodies efficaciously repressed the phosphorylation levels of ERK (p < 0.000) and AKT (p < 0.000) compared with SDF-1 alone and control. As expected, either C23 or CXCR4 siRNAs indeed resulted in C23 and CXCR4 knockdown and further suppressed the expression of p-ERK and p-AKT. Most importantly, immunoprecipitation revealed C23 interacted with CXCR4 once U87 was exposed to SDF-1 treatment. In addition, MTT assay identified that C23 or CXCR4 siRNAs could obviously decreased cell proliferation capacity (p = 0.002). In conclusion, our results suggest that C23 plays a crucial role in activation of SDF-1-induced ERK and PI3K/AKT pathways via interacting with CXCR4. Furthermore, C23 could be recommended as an important element in glioblastoma development and a new target for glioblastoma treatment.

Collaboration


Dive into the Ranran Shi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yuting Liu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Bowen Sun

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Congxin Dai

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Renzhi Wang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xingtong Zhou

Peking Union Medical College Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge