Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Glia ; 65(12): 1914-1926, 2017 12.
Article in English | MEDLINE | ID: mdl-28836293

ABSTRACT

High-grade gliomas (HGG), including glioblastomas, are characterized by invasive growth, resistance to therapy, and high inter- and intra-tumoral heterogeneity. The key histological hallmarks of glioblastoma are pseudopalisading necrosis and microvascular proliferation, which allow pathologists to distinguish glioblastoma from lower-grade gliomas. In addition to being genetically and molecularly heterogeneous, HGG are also heterogeneous with respect to the composition of their microenvironment. The question of whether this microenvironmental heterogeneity is driven by the molecular identity of the tumor remains controversial. However, this question is of utmost importance since microenvironmental, non-neoplastic cells are key components of the most radiotherapy- and chemotherapy-resistant niches of the tumor. Our work demonstrates a versatile, reliable, and reproducible adult HGG mouse model with NF1-silencing as a driver mutation. This model shows significant differences in tumor microenvironment, expression of subtype-specific markers, and response to standard therapy when compared to our established PDGFB-overexpressing HGG mouse model. PDGFB-overexpressing and NF1-silenced murine tumors closely cluster with human proneural and mesenchymal subtypes, as well as PDGFRA-amplified and NF1-deleted/mutant human tumors, respectively, at both the RNA and protein expression levels. These models can be generated in fully immunocompetent mixed or C57BL/6 genetic background mice, and therefore can easily be incorporated into preclinical studies for cancer cell-specific or immune cell-targeting drug discovery studies.


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Glioma/pathology , Mutation/genetics , Proto-Oncogene Proteins c-sis/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation , Cerebral Ventricles/pathology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Disease Models, Animal , Doublecortin Domain Proteins , Gene Expression Regulation, Neoplastic/drug effects , Glioma/diagnostic imaging , Glioma/genetics , Glioma/therapy , Humans , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Neuropeptides/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Temozolomide
2.
Oncogene ; 34(5): 558-67, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-24488013

ABSTRACT

Diffuse infiltrative invasion is a major cause for the dismal prognosis of glioblastoma multiforme (GBM), but the underlying mechanisms remain incompletely understood. Using human glioma stem cells (GSCs) that recapitulate the invasive propensity of primary GBM, we find that EphA2 critically regulates GBM invasion in vivo. EphA2 was expressed in all seven GSC lines examined, and overexpression of EphA2 enhanced intracranial invasion. The effects required Akt-mediated phosphorylation of EphA2 on serine 897. In vitro the Akt-EphA2 signaling axis is maintained in the absence of ephrin-A ligands and is disrupted upon ligand stimulation. To test whether ephrin-As in tumor microenvironment can regulate GSC invasion, the newly established Efna1;Efna3;Efna4 triple knockout mice (TKO) were used in an ex vivo brain slice invasion assay. We observed significantly increased GSC invasion through the brain slices of TKO mice relative to wild-type (WT) littermates. Mechanistically EphA2 knockdown suppressed stem cell properties of GSCs, causing diminished self-renewal, reduced stem marker expression and decreased tumorigenicity. In a subset of GSCs, the reduced stem cell properties were associated with lower Sox2 expression. Overexpression of EphA2 promoted stem cell properties in a kinase-independent manner and increased Sox2 expression. Disruption of Akt-EphA2 cross-talk attenuated stem cell marker expression and neurosphere formation while having minimal effects on tumorigenesis. Taken together, the results show that EphA2 endows invasiveness of GSCs in vivo in cooperation with Akt and regulates glioma stem cell properties.


Subject(s)
Brain Neoplasms/genetics , Carcinogenesis/genetics , Glioblastoma/genetics , Oncogene Protein v-akt/genetics , Receptor, EphA2/genetics , Animals , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Mice , Mice, Knockout , Neoplasm Invasiveness/genetics , Neoplastic Stem Cells/pathology , Oncogene Protein v-akt/metabolism , Phosphorylation/genetics , SOXB1 Transcription Factors/genetics , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL