Network


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

Hotspot


Dive into the research topics where Bin Yan is active.

Publication


Featured researches published by Bin Yan.


Molecular Cancer | 2017

Metformin suppresses cancer initiation and progression in genetic mouse models of pancreatic cancer

Ke Chen; Weikun Qian; Zhengdong Jiang; Liang Cheng; Jie Li; Liankang Sun; Cancan Zhou; Luping Gao; Meng Lei; Bin Yan; Junyu Cao; Wanxing Duan; Qingyong Ma

BackgroundPancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-associated mortality worldwide with an overall five-year survival rate less than 7%. Accumulating evidence has revealed the cancer preventive and therapeutic effects of metformin, one of the most widely prescribed medications for type 2 diabetes mellitus. However, its role in pancreatic cancer is not fully elucidated. Herein, we aimed to further study the preventive and therapeutic effects of metformin in genetically engineered mouse models of pancreatic cancer.MethodsLSL-KrasG12D/+; Pdx1-Cre (KC) mouse model was established to investigate the effect of metformin in pancreatic tumorigenesis suppression; LSL-KrasG12D/+; Trp53fl/+; Pdx1-Cre (KPC) mouse model was used to evaluate the therapeutic efficiency of metformin in PDAC. Chronic pancreatitis was induced in KC mice by peritoneal injection of cerulein.ResultsFollowing metformin treatment, pancreatic acinar-to-ductal metaplasia (ADM) and mouse pancreatic intraepithelial neoplasia (mPanIN) were decreased in KC mice. Chronic pancreatitis induced a stroma-rich and duct-like structure and increased the formation of ADM and mPanIN lesions, in line with an increased cytokeratin 19 (CK19)-stained area. Metformin treatment diminished chronic pancreatitis-mediated ADM and mPanIN formation. In addition, it alleviated the percent area of Masson’s trichrome staining, and decreased the number of Ki67-positive cells. In KPC mice, metformin inhibited tumor growth and the incidence of abdominal invasion. More importantly, it prolonged the overall survival.ConclusionsMetformin inhibited pancreatic cancer initiation, suppressed chronic pancreatitis-induced tumorigenesis, and showed promising therapeutic effect in PDAC.


Annals of the New York Academy of Sciences | 2017

Resveratrol and cancer treatment: updates

Zhengdong Jiang; Ke Chen; Liang Cheng; Bin Yan; Weikun Qian; Junyu Cao; Jie Li; Erxi Wu; Qingyong Ma; Wei Yang

Cancer, a growing health problem worldwide, affects millions of people every year. The overall survival rates of most cancers have been prolonged owing to the efforts of clinicians and scientists. However, some tumors develop resistance to chemoradiotherapeutic agents, and the cancer research community continues to search for effective sensitizers. Resveratrol, a natural polyphenolic phytoalexin, has shown promising effects in inhibiting proliferation and cancer progression in several tumor models. However, its molecular mechanisms and applications in chemotherapy and radiotherapy have yet to be fully determined. In this concise review, we highlight the role and related molecular mechanisms of resveratrol in cancer treatment. In particular, we focus on the role of resveratrol in the tumor microenvironment and the sensitization of cancer cells for chemotherapy and radiotherapy. Resveratrol shows promising efficacies in cancer treatment and may be applied in clinical therapy, but it requires further clinical study.


Molecular Oncology | 2017

Loss of AMPK activation promotes the invasion and metastasis of pancreatic cancer through an HSF1‐dependent pathway

Ke Chen; Weikun Qian; Jie Li; Zhengdong Jiang; Liang Cheng; Bin Yan; Junyu Cao; Liankang Sun; Cancan Zhou; Meng Lei; Wanxing Duan; Jiguang Ma; Qingyong Ma; Zhenhua Ma

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with a mortality rate that closely parallels its incidence rate, and a better understanding of the molecular and cellular mechanisms associated with the invasion and distant metastasis is required. Heat shock factor 1 (HSF1) is a very highly conserved factor in eukaryotes that regulates the protective heat shock response. Here, we show that HSF1 is abnormally activated in pancreatic cancer. The knockdown of HSF1 impaired the invasion and migration and epithelial–mesenchymal transition (EMT) of pancreatic cancer cells in vitro; however, the upregulation of HSF1 showed the opposite effects. In vivo, the pharmacological inhibition of HSF1 significantly reduced the tumor burden, decreased the incidence of invasion, and prolonged the overall survival of transgenic mice harboring the spontaneous pancreatic cancer. We suggest that the loss of AMP‐activated protein kinase (AMPK) activation mediates the abnormal activation of HSF1 based on the findings that phospho‐HSF1 (p‐HSF1) was highly expressed in human PDAC tissues with a low expression of p‐AMPK and that in those tissues with a high p‐AMPK expression, the level of p‐HSF1 was decreased. The in vivo and in vitro activation of AMPK impaired the activity of HSF1, and HSF1 mediated the effects of the AMPK knockdown‐induced pancreatic cancer invasion and migration. Our study revealed a novel mechanism by which the loss of AMPK activation amplifies the activity of HSF1 to promote the invasion and metastasis of pancreatic cancer.


Oxidative Medicine and Cellular Longevity | 2018

Resveratrol-Induced Downregulation of NAF-1 Enhances the Sensitivity of Pancreatic Cancer Cells to Gemcitabine via the ROS/Nrf2 Signaling Pathways

Liang Cheng; Bin Yan; Ke Chen; Zhengdong Jiang; Cancan Zhou; Junyu Cao; Weikun Qian; Jie Li; Liankang Sun; Jiguang Ma; Qingyong Ma; Huanchen Sha

NAF-1 (nutrient-deprivation autophagy factor-1), which is an outer mitochondrial membrane protein, is known to play important roles in calcium metabolism, antiapoptosis, and antiautophagy. Resveratrol, a natural polyphenolic compound, is considered as a potent anticancer agent. Nevertheless, the molecular mechanisms underlying the effects of resveratrol and NAF-1 and their mediation of drug resistance in pancreatic cancer remain unclear. Here, we demonstrate that resveratrol suppresses the expression of NAF-1 in pancreatic cancer cells by inducing cellular reactive oxygen species (ROS) accumulation and activating Nrf2 signaling. In addition, the knockdown of NAF-1 activates apoptosis and impedes the proliferation of pancreatic cancer cells. More importantly, the targeting of NAF-1 by resveratrol can improve the sensitivity of pancreatic cancer cells to gemcitabine. These results highlight the significance of strategies that target NAF-1, which may enhance the efficacy of gemcitabine in pancreatic cancer therapy.


Oxidative Medicine and Cellular Longevity | 2018

Resveratrol Inhibits ROS-Promoted Activation and Glycolysis of Pancreatic Stellate Cells via Suppression of miR-21

Bin Yan; Liang Cheng; Zhengdong Jiang; Ke Chen; Cancan Zhou; Liankang Sun; Junyu Cao; Weikun Qian; Jie Li; Tao Shan; Jianjun Lei; Qingyong Ma; Jiguang Ma

Activation of pancreatic stellate cells (PSCs) initiates pancreatic fibrosis in chronic pancreatitis and furnishes a niche that enhances the malignancy of pancreatic cancer cells (PCCs) in pancreatic ductal adenocarcinoma (PDAC). Resveratrol (RSV), a natural polyphenol, exhibits potent antioxidant and anticancer effects. However, whether and how RSV influences the biological properties of activated PSCs and the effects of these changes on tumor remain unknown. In the present study, we found that RSV impeded hydrogen peroxide-driven reactive oxygen species- (ROS-) induced activation, invasion, migration, and glycolysis of PSCs. In addition, miR-21 expression in activated PSCs was downregulated after RSV treatment, whereas the PTEN protein level increased. miR-21 silencing attenuated ROS-induced activation, invasion, migration, and glycolysis of PSCs, whereas the overexpression of miR-21 rescued the responses of PSCs treated with RSV. Moreover, RSV or N-acetyl-L-cysteine (NAC) administration or miR-21 knockdown in PSCs reduced the invasion and migration of PCCs in coculture, and the effects of RSV were partly reversed by miR-21 upregulation. Collectively, RSV inhibits PCC invasion and migration through suppression of ROS/miR-21-mediated activation and glycolysis in PSCs. Therefore, targeting miR-21-mediated glycolysis by RSV in tumor stroma may serve as a new strategy for clinical PDAC prevention or treatment.


Oncology Reports | 2018

Norepinephrine enhances cell viability and invasion, and inhibits apoptosis of pancreatic cancer cells in a Notch‑1‑dependent manner

Weikun Qian; Shifang Lv; Jie Li; Ke Chen; Zhengdong Jiang; Liang Cheng; Cancan Zhou; Bin Yan; Junyu Cao; Qingyong Ma; Wanxing Duan

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive types of cancer, which is associated with a poor prognosis due to complexities in prevention, early diagnosis and effective treatment. The lack of understanding regarding its induction and specific pro‑cancer mechanisms may contribute to its poor prognosis. The Notch‑1 pathway is widely considered to be a critical tumor‑promoting factor in PDAC. Previous studies have indicated that chronic psychological stress may promote the development of PDAC partially via the main downstream stress hormone, norepinephrine (NE); however, to the best of our knowledge, the role of the Notch‑1 pathway in this process has not been studied. Therefore, the present study aimed to explore this process. The expression levels of Notch‑1 pathway‑associated molecules were measured in response to NE using reverse transcription‑quantitative polymerase chain reaction and western blotting. Alongside NE treatment, two Notch‑1 pathway blockers, Notch‑1‑specific small interfering (si)RNA and DAPT (an inhibitor of the Notch‑1 pathway), were used to explore the relationship between NE and the Notch‑1 pathway in the development of pancreatic cell malignant biological behaviors, including cell viability, apoptosis and cell invasion. The results demonstrated that treatment with NE enhanced cell viability and invasion, and inhibited apoptosis of PDAC cells; however, these effects were suppressed following treatment with Notch‑1‑specific siRNA and DAPT. In conclusion, NE may enhance the malignant biological behaviors of PDAC via activating the Notch‑1 pathway.


Oncology Reports | 2018

A novel three‑miRNA signature predicts survival in cholangiocarcinoma based on RNA‑Seq data

Junyu Cao; Liankang Sun; Jie Li; Cancan Zhou; Liang Cheng; Ke Chen; Bin Yan; Weikun Qian; Qingyong Ma; Wanxing Duan

Accumulating evidence illustrates that many microRNAs (miRNAs) are abnormally expressed in cholangiocarcinoma and play important roles in tumorigenesis, tumor progression and metastasis. These miRNAs may serve as prognostic biomarkers and potential therapeutic targets. The aim of the present study was to identify the differentially expressed miRNAs in cholangiocarcinoma tissues vs. normal tissues by analyzing high‑throughput data derived from The Cancer Genome Atlas (TCGA) database. Furthermore, we evaluated the prognostic performance of the differentially expressed miRNAs and developed a novel three‑miRNA signature which predicted survival in cholangiocarcinoma patients. According to the cut‑off criteria of P<0.01 and |log2FC|>1.0, a total of 100xa0miRNAs (54xa0upregulated and 46xa0downregulated) were found to be differentially expressed and some of them were significantly associated with clinical features. Of the above 100xa0miRNAs, we obtained three miRNAs (miR‑10b, miR‑22 and miR‑551b) which were markedly related to patient overall survival (OS). Subsequently, a novel three‑miRNA signature was established and validated to be effective to predict survival. The results demonstrated that the survival rate, as well as the survival time of patients were obviously enhanced in relation to a lower miRNA signature index. Univariate and multivariate Cox regression analyses revealed that the three‑miRNA signature was an independent prognostic factor in cholangiocarcinoma. The reliability of the three‑miRNA signature was validated by an independent cohort from Gene Expression Omnibus (GEO). Furthermore, the functional enrichment analysis emphasized that the target genes of the aforementioned miRNAs may be involved in a variety of pathways and processes associated with cancer. Finally, these three miRNAs were detected for verification of expression using RT‑qPCR, and miR‑551b was selected for the verification of biological functions in cholangiocarcinoma cells. The results revealed that overexpression of miR‑551b decreased cancer cell proliferation and promoted apoptosis. Collectively, the results of the present study indicated that a specific three‑miRNA signature could be considered as an alternative prognostic marker in cholangiocarcinoma.


Life Sciences | 2018

Metformin suppresses tumor angiogenesis and enhances the chemosensitivity of gemcitabine in a genetically engineered mouse model of pancreatic cancer

Weikun Qian; Jie Li; Ke Chen; Zhengdong Jiang; Liang Cheng; Cancan Zhou; Bin Yan; Junyu Cao; Qingyong Ma; Wanxing Duan

Aims: Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant diseases and has few effective and reliable therapeutic strategies. The anti‐tumor effect of metformin is widely known, however, there is only limited evidence regarding the anti‐angiogenesis effect and chemosensitization of metformin and its underlying mechanisms in PDAC. Main methods: In the present study, we adopted a spontaneous PDAC mouse model named LSL‐KrasG12D/+; Trp53fl/+; Pdx1‐Cre (KPC) mice to explore the mechanism of the modulation of tumor angiogenesis and chemosensitization of metformin by treating KPC mice with metformin, gemcitabine or a combination of the two. H&E staining, Masson staining and immunohistochemical staining were adopted to describe the histopathology and biomarkers of the KPC in different groups. Key findings: Metformin plus gemcitabine reduced tumorigenic potential of PDAC. Specifically, metformin showed an anti‐pancreatic stellate cells (PSCs) effect via decreasing the expression of sonic hedgehog (SHH) and then sparked some downstream effects, for example, inhibiting the production of vascular endothelial growth factor (VEGF) in the tumor microenvironment, reducing the formation of tumor neovascularization, attenuating the desmoplastic reaction and enhancing the antitumor effect of gemcitabine. Significance: We concluded that metformin suppressed tumor angiogenesis and enhanced the chemosensitivity of gemcitabine via inactivating PSCs in PDAC of KPC mice.


Journal of Experimental & Clinical Cancer Research | 2018

Inhibiting YAP expression suppresses pancreatic cancer progression by disrupting tumor-stromal interactions

Zhengdong Jiang; Cancan Zhou; Liang Cheng; Bin Yan; Ke Chen; Xin Chen; Liang Zong; Jianjun Lei; Wanxing Duan; Qinhong Xu; Xuqi Li; Zheng Wang; Qingyong Ma; Jiguang Ma

BackgroundHippo/YAP pathway is known to be important for development, growth and organogenesis, and dysregulation of this pathway leads to tumor progression. We and others find that YAP is up-regulated in pancreatic ductal adenocarcinoma (PDAC) and associated with worse prognosis of patients. Activated pancreatic stellate cells (PSCs) forming the components of microenvironment that enhance pancreatic cancer cells (PCs) invasiveness and malignance. However, the role and mechanism of YAP in PDAC tumor-stromal interaction is largely unknown.MethodsThe expression of YAP in Pancreatic cancer cell lines and PDAC samples was examined by Western blot and IHC. The biological role of YAP on cancer cell proliferation, epithelial-mesenchymal transition (EMT) and invasion were evaluated by MTT, Quantitative real-time PCR analysis, Western blot analysis and invasion assay. The effect of YAP on PSC activation was evaluated by PC-PSC co-culture conditions and xenograft PDAC mouse model.ResultsFirstly, knockdown of YAP inhibits PDAC cell proliferation and invasion in vitro. In addition, YAP modulates the PC and PSC interaction via reducing the production of connective tissue growth factor (CTGF) from PCs, inhibits paracrine-mediated PSC activation under PC-PSC co-culture conditions and in turn disrupts TGF-β1-mediated tumor-stromal interactions. Lastly, inhibiting YAP expression prevents tumor growth and suppresses desmoplastic reaction in vivo.ConclusionsThese results demonstrate that YAP contributes to the proliferation and invasion of PC and the activation of PSC via tumor-stromal interactions and that targeting YAP may be a promising therapeutic strategy for PDAC treatment.


International Journal of Oncology | 2018

Overexpression of Gremlin 1 by sonic hedgehog signaling promotes pancreatic cancer progression

Yongtian Yu; Liang Cheng; Bin Yan; Cancan Zhou; Weikun Qian; Ying Xiao; Tao Qin; Junyu Cao; Liang Han; Qingyong Ma; Jiguang Ma

Sonic hedgehog (SHH) signaling is an important promotor of desmoplasia, a critical feature in pancreatic cancer stromal reactions involving the activation of pancreatic stellate cells (PSCs). Gremlin 1 is widely overexpressed in cancer-associated stromal cells, including activated PSCs. In embryonic development, SHH is a potent regulator of Gremlin 1 through an interaction network. This subtle mechanism in the cancer microenvironment remains to be fully elucidated. The present study investigated the association between Gremlin 1 and SHH, and the effect of Gremlin 1 in pancreatic cancer. The expression of Gremlin 1 in different specimens was measured using immunohistochemistry. The correlations among clinico-pathological features and levels of Gremlin 1 were evaluated. Primary human PSCs and pancreatic cancer cell lines were exposed to SHH, cyclopamine, GLI family zinc finger-1 (Gli-1) small interfering RNA (siRNA), and Gremlin 1 siRNA to examine their associations and effects using an MTT assay, reverse transcription-quantitative polymerase chain reaction analysis, western blot analysis, and migration or invasion assays. The results revealed the overexpression of Gremlin 1 in pancreatic cancer tissues, mainly in the stroma. The levels of Gremlin 1 were significantly correlated with survival rate and pT status. In addition, following activation of the PSCs, the expression levels of Gremlin 1 increased substantially. SHH acts as a potent promoter of the expression of Gremlin 1, and cyclopamine and Gli-1 siRNA modulated this effect. In a screen of pancreatic cancer cell lines, AsPC-1 and BxPC-3 cells expressed high levels of Gremlin 1, but only AsPC-1 cells exhibited a high expression level of SHH. The results of the indirect co-culture experiment suggested that paracrine SHH from the AsPC-1 cells induced the expression of Gremlin 1 in the PSCs. Furthermore, Gremlin 1 siRNA negatively regulated the proliferation and migration of PSCs, and the proliferation, invasion and epithelial-mesenchymal transition of AsPC-1 and BxPC-3 cells. Based on the data from the present study, it was concluded that an abnormal expression level of Gremlin 1 in pancreatic cancer was induced by SHH signaling, and that the overexpression of Gremlin 1 enabled pancreatic cancer progression.

Collaboration


Dive into the Bin Yan's collaboration.

Top Co-Authors

Avatar

Liang Cheng

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Qingyong Ma

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Cancan Zhou

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Junyu Cao

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Weikun Qian

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Jie Li

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Ke Chen

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Zhengdong Jiang

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Liankang Sun

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Wanxing Duan

Xi'an Jiaotong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge