3 research outputs found

    Genetic Analysis of PI3k and mTOR Inhibition in U87mg Glioblastoma Cell Line

    Get PDF
    NVP-BEZ235 is a Glioblastoma Multiform chemotherapeutic dual PI3K/mTOR pathway inhibitor created in 2008 and has since been proven experimentally to induce pluripotentcy in oncological cell populations. The inhibition of PI3K and mTOR has shown to coerce phenotypes associated with stem cell markers, most notably OCT4. It is necessary to understand the genetic composure of how PI3K/mTOR inhibited tumor cells are bypassing the canonical pathway for proliferation and growth and utilizing other parallel sources for tumor invasion into other neural regions. Taking a genetic approach with RNA-sequencing allowed us to gain insight into how glioblastoma interact with cytoskeleton factors MAPK4 and GAP43 to bring up novel phenotypes allowing tumors cells to not only grow but also signal other cells to pick-up a stem- like signature

    CRISPR Cas9 Genome Editing in Human Cell Lines with DONOR Vector Made by Gibson Assembly

    Get PDF
    CRISPR Cas9 genome editing allows researchers to modify genesin a multitude of ways including to obtain deletions, epitope-tagged loci, and knock-in mutations. Within six years of its initial application, CRISPR Cas9 genome editing has become widely employed, but disadvantages to this method, such as low modification efficiencies and off-target effects,need careful consideration. Obtaining custom donor vectors can also be expensive and time consuming. This chapter details strategies to overcome barriers to CRISPR Cas9 genome editing as well as recent developments in employing this technique

    PI3K Pathway Inhibition with NVP-BEZ235 Hinders Glycolytic Metabolism in Glioblastoma Multiforme Cells

    Get PDF
    Glioblastoma (GBM) is the most lethal primary brain cancer that lacks effective molecular targeted therapies. The PI3K/AKT/mTOR pathway is activated in 90% of all Glioblastoma multiforme (GBM) tumors. To gain insight into the impact of the PI3K pathway on GBM metabolism, we treated U87MG GBM cells with NVP-BEZ235 (PI3K and mTOR a dual inhibitor) and identified differentially expressed genes with RNA-seq analysis. RNA-seq identified 7803 differentially regulated genes in response to NVP-BEZ235. Gene Set Enrichment Analysis (GSEA) identified two glycolysis-related gene sets that were significantly enriched (p \u3c 0.05) in control samples compared to NVP-BEZ235-treated samples. We validated the inhibition of glycolytic genes by NVP-BEZ235 and examined the impact of the FOXO1 inhibitor (AS1842856) on these genes in a set of GBM cell lines. FOXO1 inhibition alone was associated with reduced LDHA expression, but not ENO1 or PKM2. Bioinformatics analyses revealed that PI3K-impacted glycolytic genes were over-expressed and co-expressed in GBM clinical samples. The elevated expression of PI3K-impacted glycolytic genes was associated with poor prognosis in GBM based on Kaplan–Meier survival analyses. Our results suggest novel insights into hallmark metabolic reprogramming associated with the PI3K-mTOR dual inhibition
    corecore