6 research outputs found

    Akt1 expression in muscle samples according to the <i>AKT1-rs1130233</i> polymorphism.

    No full text
    <p>Bar graphs illustrating the mean±SD expression of total Akt1 (<b>A</b>) and phospho-Akt1 (<b>B</b>) in muscle samples from patients with differential <i>AKT1-rs1130233</i> genotypes (N = 9 samples in each group) *<i>p</i><0.05.</p

    <i>AKT1</i> and <i>SELP</i> Polymorphisms Predict the Risk of Developing Cachexia in Pancreatic Cancer Patients

    No full text
    <div><p>Pancreatic ductal adenocarcinoma (PDAC) patients have the highest risk of developing cachexia, which is a direct cause of reduced quality of life and shorter survival. Novel biomarkers to identify patients at risk of cachexia are needed and might have a substantial impact on clinical management. Here we investigated the prognostic value and association of <i>SELP-rs6136</i>, <i>IL6-rs1800796</i> and <i>AKT1-rs1130233</i> polymorphisms with cachexia in PDAC. Genotyping was performed in DNA from blood samples of a test and validation cohorts of 151 and 152 chemo-naive locally-advanced/metastatic PDAC patients, respectively. The association of <i>SELP-rs6136</i>, <i>IL6-rs1800796</i> and <i>AKT1-rs1130233</i> polymorphisms with cachexia as well as the correlation between cachexia and the candidate polymorphisms and overall survival were analyzed. Akt expression and phosphorylation in muscle biopsies were evaluated by specific ELISA assays. <i>SELP-rs6136-AA</i> and <i>AKT1-rs1130233-AA/GA</i> genotypes were associated with increased risk of developing cachexia in both cohorts (<i>SELP: p</i> = 0.011 and <i>p</i> = 0.045; <i>AKT1: p</i> = 0.004 and <i>p</i> = 0.019 for the first and second cohorts, respectively), while patients carrying <i>AKT1-rs1130233-GG</i> survived significantly longer (<i>p</i> = 0.002 and <i>p</i> = 0.004 for the first and second cohorts, respectively). In the multivariate analysis <i>AKT1-rs1130233-AA/GA</i> genotypes were significant predictors for shorter survival, with an increased risk of death of 1.7 (<i>p</i> = 0.002) and 1.6 (<i>p</i> = 0.004), in the first and second cohorts, respectively. This might be explained by the reduced phosphorylation of Akt1 in muscle biopsies from patients harboring <i>AKT1-rs1130233-AA/GA</i> (<i>p</i> = 0.003), favoring apoptosis induction. In conclusion, <i>SELP</i> and <i>AKT1</i> polymorphisms may play a role in the risk of cachexia and death in PDAC patients, and should be further evaluated in larger prospective studies.</p></div

    Correlation between cachexia and candidate <i>SELP</i>, <i>AKT1</i> and <i>IL-6</i> SNPs.

    No full text
    <p><i>*p-values were calculated with Fisher's exact test.</i></p><p><i>SNPs, single nucleotide polymorphisms.</i></p><p>Note: <i>SELP</i> and <i>IL6</i> SNPs was detectable in all the samples, while the <i>AKT1</i> genotype could not be determined in 2 of the patients of the second cohort.</p><p>Correlation between cachexia and candidate <i>SELP</i>, <i>AKT1</i> and <i>IL-6</i> SNPs.</p

    Clinical outcome according to cachexia and <i>AKT1-rs1130233</i> polymorphism.

    No full text
    <p>(<b>A</b> and <b>B</b>) Kaplan–Meier survival curves according to cachexia in the first and second cohort of patients. (<b>C</b> and <b>D</b>) Kaplan–Meier survival curves according to the <i>AKT1-rs1130233</i> polymorphism in the first and second cohort of patients. <i>p</i>-values were calculated with the log-rank test. OS: Overall survival.</p

    Additional file 1: Figure S1. of Phospho-Akt overexpression is prognostic and can be used to tailor the synergistic interaction of Akt inhibitors with gemcitabine in pancreatic cancer

    No full text
    PFS curves according to phospho-Akt expression in radically-resected PDACs, showing that patients with high and “very high” phospho-Akt (right panel) had a significantly worse PFS. Figure S2. Growth inhibitory effects after MK-2206 exposure in LPC006 (72-hours). Figure S3. Growth inhibitory effects after 72 hours exposure to perifosine, gemcitabine or their combination at a fixed ratio based on IC50 values in CFPAC-1 and PANC-1 cells. On the X-axis the drug concentrations for the combination are referred to gemcitabine. Figure S4. Phospho-Akt (serine residue-473) expression, normalized to total Akt, after 4-hour exposure, as determined by ELISA. Figure S5. A Phospho-Akt (serine residue-473) expression, normalized to total Akt, after 24-hour exposure. B Phospho-Akt (threonine residue-308) expression, normalized to total Akt, after 24-hour exposure, as determined by ELISA. Figure S6. Expression of gemcitabine determinants in PANC-1 cells treated with perifosine, as determined by qRT-PCR. Dashed line, values in untreated samples. Figure S7. Wound-healing assay in CFPAC-1 and PANC-1 exposed to perifosine, gemcitabine or their combination (IC50 values, 24 hours). Figure S8. Wound-healing assay in LPC006 exposed to MK-2206, gemcitabine or to their combination (IC50 values, 24 hours). Figure S9. Annexin-V assay in LPC028 and LPC006. Figure S10: Modulation of caspase-3, caspase-6/-8/ and caspase-9 in CFPAC-1 and PANC-1, as determined by a specific fluorometric assay. Figure S11. Cell growth inhibition in LPC006 cells after 72-hour exposure to MK-2205, NVP-BEZ235 at IC50 values, together with DMSO or with the Glut1 inhibitor PGL13, at 30 μM. Points, or Columns, mean values obtained from three independent experiments; bars, SEM. *Significantly different from controls. (PPTX 328 kb
    corecore