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.
Mol Pharmacol ; 87(2): 150-61, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25391374

ABSTRACT

Resistance to the human epidermal growth factor receptor (HER2)-targeted antibody trastuzumab is a major clinical concern in the treatment of HER2-positive metastatic breast cancer. Increased expression or signaling from the insulin-like growth factor-1 receptor (IGF-1R) has been reported to be associated with trastuzumab resistance. However, the specific molecular and biologic mechanisms through which IGF-1R promotes resistance or disease progression remain poorly defined. In this study, we found that the major biologic effect promoted by IGF-1R was invasion, which was mediated by both Src-focal adhesion kinase (FAK) signaling and Forkhead box protein M1 (FoxM1). Cotargeting IGF-1R and HER2 using either IGF-1R antibodies or IGF-1R short hairpin RNA in combination with trastuzumab resulted in significant but modest growth inhibition. Reduced invasion was the most significant biologic effect achieved by cotargeting IGF-1R and HER2 in trastuzumab-resistant cells. Constitutively active Src blocked the anti-invasive effect of IGF-1R/HER2 cotargeted therapy. Furthermore, knockdown of FoxM1 blocked IGF-1-mediated invasion, and dual targeting of IGF-1R and HER2 reduced expression of FoxM1. Re-expression of FoxM1 restored the invasive potential of IGF-1R knockdown cells treated with trastuzumab. Overall, our results strongly indicate that therapeutic combinations that cotarget IGF-1R and HER2 may reduce the invasive potential of cancer cells that are resistant to trastuzumab through mechanisms that depend in part on Src and FoxM1.


Subject(s)
Breast Neoplasms/metabolism , Focal Adhesion Kinase 1/biosynthesis , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation, Neoplastic , Receptor, ErbB-2/biosynthesis , Receptor, IGF Type 1/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Forkhead Box Protein M1 , Genes, src/physiology , Humans , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Signal Transduction/physiology
2.
Neuro Oncol ; 14(4): 440-58, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22379189

ABSTRACT

Medulloblastoma is the most common malignant childhood brain tumor. The protein phosphatase and oncogene WIP1 is over-expressed or amplified in a significant number of primary human medulloblastomas and cell lines. In the present study, we examine an important mechanism by which WIP1 promotes medulloblastoma growth using in vitro and in vivo models. Human cell lines and intracerebellar xenografted animal models were used to study the role of WIP1 and the major TP53 regulator, HDM2, in medulloblastoma growth. Stable expression of WIP1 enhances growth of TP53 wild-type medulloblastoma cells, compared with cells with stable expression of an empty-vector or mutant WIP1. In an animal model, WIP1 enhances proliferation and reduces the survival of immunodeficient mice bearing intracerebellar xenografted human medulloblastoma cells. Cells with increased WIP1 expression also exhibit increased expression of HDM2. HDM2 knockdown or treatment with the HDM2 inhibitor Nutlin-3a, the active enantomer of Nutlin-3, specifically inhibits the growth of medulloblastoma cells with increased WIP1 expression. Nutlin-3a does not affect growth of medulloblastoma cells with stable expression of an empty vector or of mutant WIP1. Knockdown of WIP1 or treatment with the WIP1 inhibitor CCT007093 results in increased phosphorylation of known WIP1 targets, reduced HDM2 expression, and reduced growth specifically in WIP1 wild-type and high-expressing medulloblastoma cells. Combined WIP1 and HDM2 inhibition is more effective than WIP1 inhibition alone in blocking growth of WIP1 high-expressing medulloblastoma cells. Our preclinical study supports a role for therapies that target WIP1 and HDM2 in the treatment of medulloblastoma.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/metabolism , Phosphoprotein Phosphatases/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cerebellar Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Medulloblastoma/genetics , Mice , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2C , Proto-Oncogene Proteins c-mdm2/genetics , Transplantation, Heterologous , Tumor Suppressor Protein p53/genetics
3.
PLoS One ; 5(5): e10849, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20520772

ABSTRACT

BACKGROUND: Recent publications have described an important role for cross talk between PI-3 kinase and sonic hedgehog signaling pathways in the pathogenesis of medulloblastoma. METHODOLOGY/PRINCIPAL FINDINGS: We crossed mice with constitutive activation of Smoothened, SmoA1, with Pten deficient mice. Both constitutive and conditional Pten deficiency doubled the incidence of mice with symptoms of medulloblastoma and resulted in decreased survival. Analysis revealed a clear separation of gene signatures, with up-regulation of genes in the PI-3 kinase signaling pathway, including downstream activation of angiogenesis in SmoA1+/-; Pten +/- medulloblastomas. Western blotting and immunohistochemistry confirmed reduced or absent Pten, Akt activation, and increased angiogenesis in Pten deficient tumors. Down-regulated genes included genes in the sonic hedgehog pathway and tumor suppressor genes. SmoA1+/-; Pten +/+ medulloblastomas appeared classic in histology with increased proliferation and diffuse staining for apoptosis. In contrast, Pten deficient tumors exhibited extensive nodularity with neuronal differentiation separated by focal areas of intense staining for proliferation and virtually absent apoptosis. Examination of human medulloblastomas revealed low to absent PTEN expression in over half of the tumors. Kaplan-Meier analysis confirmed worse overall survival in patients whose tumor exhibited low to absent PTEN expression. CONCLUSIONS/SIGNIFICANCE: This suggests that PTEN expression is a marker of favorable prognosis and mouse models with activation of PI-3 kinase pathways may be important tools for preclinical evaluation of promising agents for the treatment of medulloblastoma.


Subject(s)
Heterozygote , Medulloblastoma/enzymology , Medulloblastoma/pathology , PTEN Phosphohydrolase/genetics , Alleles , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Down-Regulation , Hedgehog Proteins/metabolism , Humans , Medulloblastoma/blood supply , Medulloblastoma/diagnosis , Mice , Neovascularization, Pathologic/enzymology , Neurons/enzymology , Neurons/pathology , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Signal Transduction , Survival Analysis , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...