Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Ther Oncolytics ; 12: 214-223, 2019 Mar 29.
Article in English | MEDLINE | ID: mdl-30847385

ABSTRACT

Arsenic trioxide (ATO) has been found to exert its anti-cancer activity in various human malignancies. In our previous report, we have shown that ATO inhibited cell growth and invasion via downregulation of Skp2 in pancreatic cancer (PC) cells. It has been extensively demonstrated that microRNAs (miRNAs) play a pivotal role in tumorigenesis. ATO might induce PC cell apoptosis and regulate Skp2 downregulation through the regulation of miRNAs. One study has demonstrated that miR-330-5p exerts a tumor-suppressive function in PC cell lines. Here, we investigated the role of miRNA-330-5p in ATO-mediated anti-tumor activity and explored whether ATO could regulate miR-330-5p in PC cells. We found that ATO treatment upregulated the expression of miR-330-5p. Moreover, miR-330-5p inhibitor rescued the ATO-mediated tumor-suppressive function. The combination of miR-330-5p mimic with ATO reduced cell growth, motility, and invasion, and enhanced apoptosis to a greater degree in PC cells. This study suggests that the combination of miR-330-5p mimic with ATO may be a potential therapeutic strategy for the treatment of PC.

2.
Am J Transl Res ; 11(2): 991-997, 2019.
Article in English | MEDLINE | ID: mdl-30899398

ABSTRACT

The S-phase kinase associated protein 2 (Skp2), a member of the F-box protein family, regulates cell cycle progression and is highly expressed in pancreatic cancer (PC). Recently, we reported that arsenic trioxide (ATO) inhibited cell growth and invasion via downregulation of Skp2 in PC cells. Emerging evidence has revealed that Skp2 plays a crucial role in drug resistance in several kinds of cancers. Here, we determined whether ATO enhanced the sensitivity of PC cell lines to gemcitabine (GEM). We found that the combined treatment of ATO and GEM demonstrated strong antitumor effects in Patu8988 and Panc-1 PC cells. In addition, ATO potentiated the effects of GEM via downregulation of the Skp2 pathway in PC cells. Together, these findings suggested that Skp2 may be a promising therapeutic target to overcome resistance to GEM in PC.

3.
Am J Cancer Res ; 7(2): 173-186, 2017.
Article in English | MEDLINE | ID: mdl-28337369

ABSTRACT

Pancreatic cancer (PC) is one of the highly aggressive malignancies in the United States. It has been shown that multiple signaling pathways are involved in the pathogenesis of PC, such as JNK, PI3K/AKT, Rho GTPase, Hedgehog (Hh) and Skp2. In recent years, accumulated evidence has demonstrated that Notch signaling pathway plays critical roles in the development and progression of PC. Therefore, in this review we discuss the recent literature regarding the function and regulation of Notch in the pathogenesis of PC. Moreover, we describe that Notch signaling pathway could be down-regulated by its inhibitors or natural compounds, which could be a novel approach for the treatment of PC patients.

4.
Asian Pac J Cancer Prev ; 16(9): 3805-10, 2015.
Article in English | MEDLINE | ID: mdl-25987041

ABSTRACT

Arsenic trioxide (ATO) has been found to exert anti-cancer activity in various human malignancies. However, the molecular mechanisms by which ATO inhibits tumorigenesis are not fully elucidated. In the current study, we explored the molecular basis of ATO-mediated tumor growth inhibition in pancreatic cancer cells. We used multiple approaches such as MTT assay, wound healing assay, Transwell invasion assay, annexin V-FITC, cell cycle analysis, RT-PCR and Western blotting to achieve our goal. We found that ATO treatment effectively caused cell growth inhibition, suppressed clonogenic potential and induced G2-M cell cycle arrest and apoptosis in pancreatic cancer cells. Moreover, we observed a significant down-regulation of Skp2 after treatment with ATO. Furthermore, we revealed that ATO regulated Skp2 downstream genes such as FOXO1 and p53. These findings demonstrate that inhibition of Skp2 could be a novel strategy for the treatment of pancreatic cancer by ATO.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Oxides/pharmacology , Pancreatic Neoplasms/pathology , S-Phase Kinase-Associated Proteins/metabolism , Apoptosis/drug effects , Arsenic Trioxide , Blotting, Western , Cell Cycle/drug effects , Flow Cytometry , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Signal Transduction , Tumor Cells, Cultured , Wound Healing/drug effects
5.
Oncotarget ; 5(11): 3444-54, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24899509

ABSTRACT

Chromosome maintenance region 1 (CRM1) also called Exportin 1 (Xpo1), a protein found elevated in pancreatic ductal adenocarcinoma (PDAC), blocks tumor suppressor protein (TSP) function through constant nuclear export. Earlier we had shown that targeting CRM1 by our newly developed specific inhibitors of nuclear export (SINE) leads to inhibition of pancreatic cancer cell proliferation and tumor growth arrest. In this paper we define the mechanism of SINE action. Our lead SINE KPT-185 inhibits PDAC cell growth, cell migration, tumor invasion and induces apoptosis and G2-M cell cycle arrest in low nano molar range (IC50s~150 nM). Mechanistically we demonstrate that the activity of KPT-185 is associated with nuclear retention of Fbw7; which degrades nuclear Notch-1 leading to decreased tumor promoting markers such as C-Myc, Cyclin-D1, Hes1 and VEGF. The orally bioavailable SINE (KPT-251) showed potent anti-tumor activity in a Colo-357 PDAC xenografts model; residual tumor analysis showed activation of Fbw7 concomitant with attenuation of Notch1 and its downstream genes. These results suggest that the antitumor activity of KPT-185 is in part due to nuclear retention of Fbw7 and consequent Notch1 degradation. The new CRM1 inhibitors, therefore, hold strong potential and warrant further clinical investigations for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Karyopherins/metabolism , Pancreatic Neoplasms/drug therapy , Receptor, Notch1/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Ubiquitin-Protein Ligases/metabolism , Acrylates/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Female , Humans , Karyopherins/genetics , Mice , Mice, Inbred ICR , Mice, SCID , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Prognosis , Receptor, Notch1/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Triazoles/pharmacology , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays , Exportin 1 Protein
6.
Curr Drug Discov Technol ; 10(2): 147-54, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23237677

ABSTRACT

Transcription factors (TFs) play central role in normal cellular physiology and their aberrant expression is linked to different diseases. Hepatocyte Nuclear Factors (HNFs) are TFs that have been recognized to play multiple roles in liver physiology. Emerging research has highlighted their function in the sustenance of solid tumors, indicating that HNFs could serve as possible therapeutic targets in cancer. Although, there have been many attempts to develop HNF targeted drugs, the myriad downstream targets associated with these transcription factors, some of which are critical for normal cell homeostasis, led to the realization that HNFs are not easily druggable. Therefore, identifying and optimizing drugs that can selectively inactivate HNFs is a challenge to the pharmaceutical industry. To achieve this, a more in-depth understanding is required of the HNFs binding partners, the protein interaction networks it regulates and the resulting phenotype. This calls for network analysis of the pathways regulated by HNFs and how chemical perturbations can selectively activate or suppress their functions. Network biology is an emerging field of research that is finding applications in cancer drug discovery. Specifically, network pharmacology is cementing its position in cancer research and has various applications such as biomarker identification, in determining synergistic drug pairs and in drug repurposing. Developing a network understanding of HNFs, the target it hits and responses thereof can enhance our ability to design drugs against these TFs. This article reviews how network pharmacology can help in the identification of druggable avenues in TFs and also allow the selection of drugs and their synergistic pairs against HNFs for cancer therapy.


Subject(s)
Hepatocyte Nuclear Factors/metabolism , Neoplasms/metabolism , Animals , Biomarkers, Tumor/metabolism , Humans , Neoplasms/drug therapy , Systems Biology
7.
PLoS One ; 6(6): e20537, 2011.
Article in English | MEDLINE | ID: mdl-21673986

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related death in the United States, suggesting that novel strategies for the prevention and treatment of PDAC are urgently needed. K-ras mutations are observed in >90% of pancreatic cancer, suggesting its role in the initiation and early developmental stages of PDAC. In order to gain mechanistic insight as to the role of mutated K-ras, several mouse models have been developed by targeting a conditionally mutated K-ras(G12D) for recapitulating PDAC. A significant co-operativity has been shown in tumor development and metastasis in a compound mouse model with activated K-ras and Ink4a/Arf deficiency. However, the molecular mechanism(s) by which K-ras and Ink4a/Arf deficiency contribute to PDAC has not been fully elucidated. METHODOLOGY/PRINCIPAL FINDINGS: To assess the molecular mechanism(s) that are involved in the development of PDAC in the compound transgenic mice with activated K-ras and Ink4a/Arf deficiency, we used multiple methods, such as Real-time RT-PCR, western blotting assay, immunohistochemistry, MTT assay, invasion, EMSA and ELISA. We found that the deletion of Ink4a/Arf in K-ras(G12D) expressing mice leads to PDAC, which is in part mediated through the activation of Notch and NF-κB signaling pathways. Moreover, we found down-regulation of miR-200 family, which could also play important roles in tumor development and progression of PDAC in the compound transgenic mice. CONCLUSIONS/SIGNIFICANCE: Our results suggest that the activation of Notch and NF-κB together with the loss of miR-200 family is mechanistically linked with the development and progression of PDAC in the compound K-ras(G12D) and Ink4a/Arf deficient transgenic mice.


Subject(s)
Carcinoma, Ductal/pathology , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Genes, ras/genetics , NF-kappa B/metabolism , Pancreatic Neoplasms/pathology , Receptors, Notch/metabolism , Signal Transduction , Animals , Apoptosis/genetics , Calcium-Binding Proteins/metabolism , Carcinoma, Ductal/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA/metabolism , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/genetics , Integrases/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Membrane Proteins/metabolism , Mice , Mice, Transgenic , MicroRNAs/genetics , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Serrate-Jagged Proteins , Signal Transduction/genetics , Trans-Activators/genetics
8.
Sci China B Chem ; 51(2): 166-170, 2008.
Article in English | MEDLINE | ID: mdl-38624277

ABSTRACT

Total 200 properties related to structural characteristics were employed to represent structures of 400 HA coded proteins of influenza virus as training samples. Some recognition models for HA proteins of avian influenza virus (AIV) were developed using support vector machine (SVM) and linear discriminant analysis (LDA). The results obtained from LDA are as follows: the identification accuracy (R ia) for training samples is 99.8% and R ia by leave one out cross validation is 99.5%. Both R ia of 99.8% for training samples and R ia of 99.3% by leave one out cross validation are obtained using SVM model, respectively. External 200 HA proteins of influenza virus were used to validate the external predictive power of the resulting model. The external R ia for them is 95.5% by LDA and 96.5% by SVM, respectively, which shows that HA proteins of AIVs are preferably recognized by SVM and LDA, and the performances by SVM are superior to those by LDA.

SELECTION OF CITATIONS
SEARCH DETAIL
...