Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 33(5): 599-610, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-23334326

ABSTRACT

In this study, we demonstrate that constitutive activation of Raf-1 oncogenic signaling induces stabilization and accumulation of Aurora-A mitotic kinase that ultimately drives the transition from an epithelial to a highly invasive mesenchymal phenotype in estrogen receptor α-positive (ERα(+)) breast cancer cells. The transition from an epithelial- to a mesenchymal-like phenotype was characterized by reduced expression of ERα, HER-2/Neu overexpression and loss of CD24 surface receptor (CD24(-/low)). Importantly, expression of key epithelial-to-mesenchymal transition (EMT) markers and upregulation of the stemness gene SOX2 was linked to acquisition of stem cell-like properties such as the ability to form mammospheres in vitro and tumor self-renewal in vivo. Moreover, aberrant Aurora-A kinase activity induced phosphorylation and nuclear translocation of SMAD5, indicating a novel interplay between Aurora-A and SMAD5 signaling pathways in the development of EMT, stemness and ultimately tumor progression. Importantly, pharmacological and molecular inhibition of Aurora-A kinase activity restored a CD24(+) epithelial phenotype that was coupled to ERα expression, downregulation of HER-2/Neu, inhibition of EMT and impaired self-renewal ability, resulting in the suppression of distant metastases. Taken together, our findings show for the first time the causal role of Aurora-A kinase in the activation of EMT pathway responsible for the development of distant metastases in ERα(+) breast cancer cells. Moreover, this study has important translational implications because it highlights the mitotic kinase Aurora-A as a novel promising therapeutic target to selectively eliminate highly invasive cancer cells and improve the disease-free and overall survival of ERα(+) breast cancer patients resistant to conventional endocrine therapy.


Subject(s)
Aurora Kinase A/metabolism , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Estrogen Receptor alpha/metabolism , Active Transport, Cell Nucleus , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Breast Neoplasms/enzymology , CD24 Antigen/genetics , Cell Line, Tumor , Cell Movement/genetics , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System/genetics , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Phosphorylation/genetics , Proto-Oncogene Proteins c-raf/metabolism , RNA Interference , RNA, Small Interfering , Receptor, ErbB-2/biosynthesis , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Smad5 Protein/metabolism , Xenograft Model Antitumor Assays
2.
Cancer Gene Ther ; 20(11): 630-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24157925

ABSTRACT

RhoA and its downstream effector Rho-associated coiled-coil-forming kinase (ROCK) are known regulators of the formation of actin cytoskeleton in cells. Actin cytoskeleton is involved in paramyxovirus infection; we, therefore, examined the effect of ROCK inhibition on measles virus (MV) cytopathic effect and replication. Treatment with the ROCK inhibitor, Y27632, significantly increased syncytia size in tumor cell lines following MV infection, associated with cytoskeleton disruption as demonstrated by actin staining. Treatment of prostate cancer, breast cancer and glioblastoma tumor cell lines with Y27632 following MV infection resulted in increased cytopathic effect, as assessed by trypan blue exclusion assays. In addition, there was a significant increase in viral proliferation by at least one log or more as tested in one-step viral growth curves. Increased viral replication was also observed in athymic nude mice bearing MDA-MB-231 xenografts following combination treatment with MV and Y27632. In summary, inhibition of the ROCK kinase by Y27632 enhanced the oncolytic effect of MV and viral proliferation; this approach merits further translational investigation.


Subject(s)
Breast Neoplasms/therapy , Measles virus/physiology , Oncolytic Virotherapy/methods , Prostatic Neoplasms/therapy , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/virology , Cell Line, Tumor , Chlorocebus aethiops , Combined Modality Therapy , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/virology , Pyridines/pharmacology , Random Allocation , Tumor Cells, Cultured , Vero Cells , Xenograft Model Antitumor Assays , rho-Associated Kinases/metabolism
3.
Gene Ther ; 20(4): 444-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22914495

ABSTRACT

Glioblastoma (GBM) is the most common primary brain tumor in adults and has a dismal prognosis despite multimodality treatment. Given the resistance of glioma stem cells (GSC) to chemotherapy and radiation therapy, their eradication could prevent tumor recurrence. We sought to evaluate the antitumor activity of measles virus (MV) derivatives against GSC. We generated neurosphere cultures from patient-derived primary tumor GBM xenografts, and we characterized them for the GSC markers CD133, SOX2, Nestin, ATF5 and OLIG2. Using the MV-strains MV-GFP, MV-CEA and MV-NIS we demonstrated infection, viral replication and significant cytopathic effect in vitro against GSC lines. In tumorigenicity experiments, GBM44 GSC were infected with MV in vitro and subsequently implanted into the right caudate nucleus of nude mice: significant prolongation of survival in mice implanted with infected GSC was observed, compared with mock-infected controls (P=0.0483). In therapy experiments in GBM6 and GBM12 GSC xenograft models, there was significant prolongation of survival in MV-GFP-treated animals compared with inactivated virus-treated controls (GBM6 P=0.0021, GBM12 P=0.0416). Abundant syncytia and viral replication was demonstrated in tumors of MV-treated mice. Measles virus derivatives have significant antitumor activity against glioma-derived stem cells in vitro and in vivo.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Measles virus/genetics , Neoplastic Stem Cells/virology , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Giant Cells/pathology , Giant Cells/virology , Humans , Measles virus/physiology , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oncolytic Viruses/physiology , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...