Network


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

Hotspot


Dive into the research topics where Weiming Duan is active.

Publication


Featured researches published by Weiming Duan.


European Journal of Cancer | 2011

Growth of the pancreatic cancer cell line PANC-1 is inhibited by protein phosphatase 2A inhibitors through overactivation of the c-Jun N-terminal kinase pathway

Wei Li; Zheng Chen; Feiran Gong; Yang Zong; Kai Chen; Dao-Ming Li; Hong Yin; Weiming Duan; Yi Miao; Min Tao; Xiao Han; Zekuan Xu

Protein phosphatase 2A (PP2A) is a multimeric serine/threonine phosphatase that can dephosphorylate multiple kinases. It is generally considered to be a cancer suppressor as its inhibition can induce phosphorylation and activation of substrate kinases that mainly accelerate growth. We previously reported that cantharidin, an active constituent of a traditional Chinese medicine, potently and selectively inhibited PP2A, yet efficiently repressed the growth of pancreatic cancer cells through activation of the c-Jun N-terminal kinase (JNK) pathway. This suggested that activation of kinase pathways might also be a potential strategy for cancer therapy. In this study, we have confirmed that the basal activity of the phospatidylinositol 3-kinase (PI3K)/JNK/activator protein 1 (AP-1) pathway promoted pancreatic cancer cell growth when stimulated by growth factors. Interestingly, although treatment with the PP2A inhibitors, cantharidin or okadaic acid (OA), amplified the PI3K-dependent activation of JNK, cell growth was repressed. We therefore hypothesised that a specific level of activity of the JNK pathway might be required to maintain the promitogenic function, as both repression and overactivation of JNK could inhibit cell proliferation. It was found that the JNK-dependent growth inhibition was independent of the activation of AP-1, but dependent on the repression of Akt. Although the PP2A inhibitors triggered overactivation of JNK and inhibited cell growth, excessively activated protein kinase C (PKC) improved cell survival. Combined treatment with a PP2A inhibitor and a PKC inhibitor produced a synergistic effect, which indicates a potentially promising therapeutic approach to pancreatic cancer treatment.


Anti-Cancer Drugs | 2014

Artesunate induces G2/M cell cycle arrest through autophagy induction in breast cancer cells.

Kai Chen; Liu-Mei Shou; Fang Lin; Weiming Duan; Meng-Yao Wu; Xin Xie; Yufeng Xie; Wei Li; Min Tao

We found that artesunate (ART) inhibited the growth of MCF-7 and MDA-MB-231 breast cancer cells. ART arrested the cell cycle in the G2/M phase, which was accompanied by an upregulation of p21. ART upregulated the expression of Beclin1, an initiator of autophagy (type II programmed cell death). In addition, ART stimulated the aggregation of LC3, which is considered to be a marker of autophagosome formation. We further verified the transformation of LC3 from type I into type II. 3-MA, a classical autophagy inhibitor, attenuated ART-induced autophagosome formation, cell growth repression, G2/M arrest, and p21 upregulation. Autophagy induction and p21 upregulation were also repressed by knockdown of Beclin1. Furthermore, ART sensitized breast cancer cells to the chemotherapeutic agent epirubicin through an autophagy-dependent cascade. Our study showed that ART induced autophagy in breast cancer cells and indicated that the anticancer effects of ART were exerted through an autophagy pathway. Moreover, ART sensitized breast cancer cells to epirubicin chemotherapy. Our results provide a basis for further development of ART as a novel therapeutic agent for the treatment of breast cancer.


Oncology Letters | 2013

Inhibition of MCF-7 breast cancer cell-induced platelet aggregation using a combination of antiplatelet drugs

Lian Lian; Wei Li; Zhen-Yu Li; Yixiang Mao; You-Tao Zhang; Yi-Ming Zhao; Kai Chen; Weiming Duan; Min Tao

Cancer metastasis is a highly coordinated and dynamic multistep process in which cancer cells interact with a variety of host cells. Morphological studies have documented the association of circulating tumor cells with host platelets. Tumor cell-induced platelet aggregation (TCIPA) contributes significantly to hematogenous metastasis; however, the molecular mechanisms involved in breast cancer TCIPA are poorly characterized. In this study, MCF-7 metastatic human breast cancer cells induced dose-dependent aggregation of washed platelets. Four major platelet activation pathways, glycoprotein (GP)-Ib-IX, GPIIb/IIIa, thromboxane (TX)-A2 and adenosine diphosphate (ADP) were activated during TCIPA and were inhibited by their respective inhibitors, 7E3, SZ-1, aspirin and apyrase. Pretreatment of platelets with 7E3, SZ-1 or apyrase significantly inhibited TCIPA, while pretreatment with aspirin had no effect. Moreover, combined pretreatment of platelets with 7E3, SZ-1 and apyrase significantly inhibited TCIPA, compared to single inhibitors. Combinations of antiplatelet drugs may represent a promising strategy to prevent cancer metastasis.


Oncology Reports | 2015

Participation of autophagy in the cytotoxicity against breast cancer cells by cisplatin.

Meng Shen; Weiming Duan; Meng-Yao Wu; Wenjie Wang; Lu Liu; Meng-Dan Xu; Jie Zhu; Dao-Ming Li; Qi Gui; Lian Lian; Fei-Ran Gong; Kai Chen; Wei Li; Min Tao

Breast cancer is one of the most common cancers affecting women worldwide. Conventional chemotherapy is still one of the major approaches to the treatment of breast cancer. Autophagy, also termed as type II programmed cell death (PCD), exhibits either a protumorigenic or antitumorigenic function. In the present study, we investigated whether autophagy could be involved in the effect of chemotherapy against breast cancer. Epirubicin, docetaxel, methotrexate, cyclophosphamide, fluorouracil (5-FU) and cisplatin were applied in the present investigation. All of these chemotherapeutics presented cytotoxicity against breast cancer cells. DsRed-LC3 reporter assay revealed that only docetaxel and cisplatin induced autophagy. Autophagy inhibitor 3-methyladenine (3-MA) strengthened the cytotoxicity of docetaxel, yet impaired the cytotoxicity of cisplatin, suggesting that docetaxel stimulates protumorigenic autophagy, while cisplatin-induced autophagy could be antitumorigenic. Real-time PCR revealed that cisplatin upregulated multiple autophagy-related genes, including AMBRA1, ATG3, ATG4C, ATG4D, ATG5, ATG7, ATG13, ATG14, ATG16L2, Beclin1, DRAM1, GABARAP, GABARAPL1, GABARAPL2, HDAC6, IRGM, MAP1LC3B and ULK1, indicating that cisplatin induced autophagy through a multiple mechanism involved manner.


Cancer Cell International | 2015

The association between expressions of Ras and CD68 in the angiogenesis of breast cancers

Wei Li; Rong-Rui Liang; Chong Zhou; Meng-Yao Wu; Lian Lian; Gao-Feng Yuan; Ming-Yun Wang; Xin Xie; Liu-Mei Shou; Fei-Ran Gong; Kai Chen; Weiming Duan; Min Tao

ObjectiveAngiogenesis is a critical step of breast cancer metastasis. Oncogenic Ras promotes the remodeling of cancer microenviroment. Tumor-associated macrophages (TAMs) are a prominent inflammatory cell population emerging in the microenviroment and facilitating the angiogenesis and metastasis. In the present study, we tried to investigate the relationship between the expression of Ras and infiltration of TAM, both of which could further promote angiogenesis.MethodsExpressions of Ras, CD68 and CD34 were assessed by immunohistochemistry. The infiltration of macrophages was evaluated by counting the number of CD68+ cells. Vessel endothelial cells were defined as CD34+ cells. Angiogenesis vascularity was defined by microvessel density (MVD) assay through counting the number of vessels per field counted in the area of highest vascular density. The Kaplan–Meier survival analysis was used to estimate the overall survival (OS). Macrophages were derived from monocytes in the presence of macrophage colony-stimulating-factor (MCSF). Breast cancer cells were treated with macrophage-conditioned medium (MCM) and tested the expressions of K-, H- and N-Ras by using realtime-PCR.ResultsRas positive status was correlated with ER, PR and Her-2 positivity, larger tumour size and lymph node metastasis, as well as higher TNM stages. A higher number of CD68+ cells was correlated with larger tumour size, higher TNM stages and Her-2 positivity. Both Ras positivity and infiltration of CD68+ macrophages correlated with poor OS. The number of CD68+ cells was positively correlated with the expression of Ras. Treatment with MCM did not up-regulate but repressed the expression of Ras. Both up-regulation of Ras and infiltration of TAMs correlated with increased MVD.ConclusionExpression of Ras and infiltration of TAM were positively correlated, and both participated in angiogenesis. Elevated Ras could be responsible for the infiltration of TAM.


Oncotarget | 2016

FH535, a β-catenin pathway inhibitor, represses pancreatic cancer xenograft growth and angiogenesis.

Lu Liu; Qiaoming Zhi; Meng Shen; Fei-Ran Gong; Binhua P. Zhou; Lian Lian; Bairong Shen; Kai Chen; Weiming Duan; Meng-Yao Wu; Min Tao; Wei Li

The WNT/β-catenin pathway plays an important role in pancreatic cancer carcinogenesis. We evaluated the correlation between aberrant β-catenin pathway activation and the prognosis pancreatic cancer, and the potential of applying the β-catenin pathway inhibitor FH535 to pancreatic cancer treatment. Meta-analysis and immunohistochemistry showed that abnormal β-catenin pathway activation was associated with unfavorable outcome. FH535 repressed pancreatic cancer xenograft growth in vivo. Gene Ontology (GO) analysis of microarray data indicated that target genes responding to FH535 participated in stemness maintenance. Real-time PCR and flow cytometry confirmed that FH535 downregulated CD24 and CD44, pancreatic cancer stem cell (CSC) markers, suggesting FH535 impairs pancreatic CSC stemness. GO analysis of β-catenin chromatin immunoprecipitation sequencing data identified angiogenesis-related gene regulation. Immunohistochemistry showed that higher microvessel density correlated with elevated nuclear β-catenin expression and unfavorable outcome. FH535 repressed the secretion of the proangiogenic cytokines vascular endothelial growth factor (VEGF), interleukin (IL)-6, IL-8, and tumor necrosis factor-α, and also inhibited angiogenesis in vitro and in vivo. Protein and mRNA microarrays revealed that FH535 downregulated the proangiogenic genes ANGPT2, VEGFR3, IFN-γ, PLAUR, THPO, TIMP1, and VEGF. FH535 not only represses pancreatic CSC stemness in vitro, but also remodels the tumor microenvironment by repressing angiogenesis, warranting further clinical investigation.


Cancer Cell International | 2017

TNF-α sensitizes chemotherapy and radiotherapy against breast cancer cells

Xiao Wu; Meng-Yao Wu; Min Jiang; Qiaoming Zhi; Xiaojie Bian; Meng-Dan Xu; Fei-Ran Gong; Juan Hou; Min Tao; Liu-Mei Shou; Weiming Duan; Kai Chen; Meng Shen; Wei Li

PurposeDespite new developments in cancer therapy, chemotherapy and radiotherapy remain the cornerstone of breast cancer treatment. Therefore, finding ways to reduce the toxicity and increase sensitivity is particularly important. Tumor necrosis factor alpha (TNF-α) exerts multiple functions in cell proliferation, differentiation and apoptosis. In the present study, we investigated whether TNF-α could enhance the effect of chemotherapy and radiotherapy against breast cancer cells.MethodsCell growth was determined by MTT assay in vitro, and by using nude mouse tumor xenograft model in vivo. Cell cycle and apoptosis/necrosis were evaluated by flow cytometry. DNA damage was visualized by phospho-Histone H2A.X staining. mRNA expression was assessed by using real-time PCR. Protein expression was tested by Western blot assay.ResultsTNF-α strengthened the cytotoxicity of docetaxel, 5-FU and cisplatin against breast cancer cells both in vitro and in vivo. TNF-α activated NF-κB pathway and dependently up-regulated expressions of CyclinD1, CyclinD2, CyclinE, CDK2, CDK4 and CDK6, the key regulators participating in G1→S phase transition. As a result, TNF-α drove cells out of quiescent G0/G1 phase, entering vulnerable proliferating phases. Treatment of TNF-α brought more DNA damage after Cs137-irradiation and strengthened G2/M and S phase cell cycle arrest induced by docetaxel and cisplatin respectively. Moreover, the up-regulation of RIP3 (a necroptosis marker) by 5-FU, and the activation of RIP3 by TNF-α, synergistically triggered necroptosis (programmed necrosis). Knockdown of RIP3 attenuated the synergetic effect of TNF-α and 5-FU.ConclusionTNF-α presented radiotherapy- and chemotherapy-sensitizing effects against breast cancer cells.


OncoTargets and Therapy | 2015

FH535 inhibited metastasis and growth of pancreatic cancer cells.

Meng-Yao Wu; Rong-Rui Liang; Kai Chen; Meng Shen; Yali Tian; Dao-Ming Li; Weiming Duan; Qi Gui; Fei-Ran Gong; Lian Lian; Wei Li; Min Tao

FH535 is a small-molecule inhibitor of the Wnt/β-catenin signaling pathway, which a substantial body of evidence has proven is activated in various cancers, including pancreatic cancer. Activation of the Wnt/β-catenin pathway plays an important role in tumor progression and metastasis. We investigated the inhibitory effect of FH535 on the metastasis and growth of pancreatic cancer cells. Western blotting and luciferase reporter gene assay indicated that FH535 markedly inhibited Wnt/β-catenin pathway viability in pancreatic cancer cells. In vitro wound healing, invasion, and adhesion assays revealed that FH535 significantly inhibited pancreatic cancer cell metastasis. We also observed the inhibitory effect of FH535 on pancreatic cancer cell growth via the tetrazolium and plate clone formation assays. Microarray analyses suggested that changes in the expression of multiple genes could be involved in the anti-cancer effect of FH535 on pancreatic cancer cells. Our results indicate for the first time that FH535 inhibits pancreatic cancer cell metastasis and growth, providing new insight into therapy of pancreatic cancer.


Oncotarget | 2017

Genomic characteristics of pancreatic squamous cell carcinoma, an investigation by using high throughput sequencing after in-solution hybrid capture

Meng-Dan Xu; Shu-Ling Liu; Yi-Zhong Feng; Qiang Liu; Meng Shen; Qiaoming Zhi; Zeyi Liu; Dong-Mei Gu; Jie Yu; Liu-Mei Shou; Fei-Ran Gong; Qi Zhu; Weiming Duan; Kai Chen; Junning Zhang; Meng-Yao Wu; Min Tao; Wei Li

Squamous cell carcinoma (SCC) of pancreas is a rare histotype of pancreatic ductal carcinoma which is distinct from pancreatic adenocarcinoma (AC). Although there are standard treatments for pancreatic AC, no precise therapies exist for pancreatic SCC. Here, we screened 1033 cases of pancreatic cancer and identified 2 cases of pure SCC, which were pathologically diagnosed on the basis of finding definite intercellular bridges and/or focal keratin peal formation in the tumor cells. Immunohistochemistry assay confirmed the positive expression of CK5/6 and p63 in pancreatic SCC. To verify the genomic characteristics of pancreatic SCC, we employed in-solution hybrid capture targeting 137 cancer-related genes accompanied by high throughput sequencing (HTS) to compare the different genetic variants in SCC and AC of pancreas. We compared the genetic alterations of known biomarkers of pancreatic adenocarcinoma in different pancreatic cancer tissues, and identified nine mutated genes in SCC of pancreas: C7orf70, DNHD1, KPRP, MDM4, MUC6, OR51Q1, PTPRD, TCF4, TET2, and nine genes (ABCB1, CSF1R, CYP2C18, FBXW7, ITPA, KIAA0748, SOD2, SULT1A2, ZNF142) that are mutated in pancreatic AC. This study may have taken one step forward on the discovery of potential biomarkers for the targeted treatment of SCC of the pancreas.


Cancer Research | 2014

Abstract 5199: Distinct expression pattern of microRNAs bioinformatically predicted to target SATB1 in human breast cancer

Weiming Duan; Zixing Chen; Li Yao; Min Tao; Kai Chen; Wei Liu; Jiannong Cen

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Recently, genome organizer protein special AT-rich sequence-binding protein 1(SATB1) has been reported to promote a metastatic phenotype and correlate with poor prognosis in breast cancer. The mechanism of SATB1 elevation in breast cancer remains elusive. This study was to investigate whether microRNAs (miRNAs) play a role in modulating SATB1 expression in ductual breast cancer by using archival formalin-fixed and paraffin-embedded (FFPE) clinical samples. Firstly, four canonical target prediction databases, including TargetScan, PicTar, miRanda and miRDB, were utilized to predict the interaction between miRNA and the 3′-UTR of SATB1 mRNA. The results of the prediction software programs were then integrated by displaying Venn diagram. Secondly, the Qiagen miRNeasy FFPE kit was used to isolate total RNA from 93 specimens with primary invasive ductual breast cancer. Expression of candidate miRNA transcriptional levels was measured using TaqMan MicroRNA Assays with miRNA-specific stem loop primers by relative real-time quantitative RT-PCR. The relative levels of miRNA expression were calculated from the relevant signals by normalization with the signal for RNU6B miRNA expression in each sample. Meanwhile, SATB1 protein expression levels were assessed by immunohistochemistry(IHC). Finally, correlations between the expression levels of these miRNAs and SATB1 were analyzed. Overall 22 miRNAs, 24 miRNAs, 76 miRNAs and 75 miRNAs were found by TargetScan, PicTar, miRanda and miRDB, to target SATB1 respectively, whereas only miR-7, miR-21, miR-23a, miR-23b, miR-34a and miR-155 were found by all four programs to be predicted to regulate SATB1. Quantitative RT-PCR detection analysis showed that expression levels of miR-34a were much higher in SATB1-negative than in SATB1-positive tumors (p<0.001). whereas lower miR-7 expression showed a strong trend toward an association with elevated SATB1 expression (P = 0.056). We then extended the analysis of miR-34a expression in breast cancer tissues to include all scores for SATB1 protein expression, assessed semiquantitatively by IHC. miR-34a expression levels were gradually decreased as SATB1 protein scores increased. Moreove, our quantitative RT-PCR results showed no differences in miR-21, miR-23a, miR-23b, and miR-155 expression between SATB1-negative and SATB1-positive tumors. We thus conclude that SATB1 maybe a novel target of miR-34a in human breast cancer. Downregulation of miR-34a promotes human breast cancer to progress through reduced repression of SATB1. Restoration of miR-34a may prove therapeutic in breast cancer patients in which aberrant SATB1 expression plays a role. Note: This abstract was not presented at the meeting. Citation Format: Weiming Duan, Zixing Chen, Li Yao, Min Tao, Kai Chen, Wei Liu, Jiannong Cen. Distinct expression pattern of microRNAs bioinformatically predicted to target SATB1 in human breast cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5199. doi:10.1158/1538-7445.AM2014-5199

Collaboration


Dive into the Weiming Duan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhen-Yu Li

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar

Feiran Gong

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiao Han

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Yang Zong

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Yi Miao

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Zekuan Xu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Zheng Chen

Nanjing Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge