23 research outputs found

    The CDKN2A G500 Allele Is More Frequent in GBM Patients with No Defined Telomere Maintenance Mechanism Tumors and Is Associated with Poorer Survival

    Get PDF
    Prognostic markers for glioblastoma multiforme (GBM) are important for patient management. Recent advances have identified prognostic markers for GBMs that use telomerase or the alternative lengthening of telomeres (ALT) mechanism for telomere maintenance. Approximately 40% of GBMs have no defined telomere maintenance mechanism (NDTMM), with a mixed survival for affected individuals. This study examined genetic variants in the cyclin-dependent kinase inhibitor 2A (CDKN2A) gene that encodes the p16INK4a and p14ARF tumor suppressors, and the isocitrate dehydrogenase 1 (IDH1) gene as potential markers of survival for 40 individuals with NDTMM GBMs (telomerase negative and ALT negative by standard assays), 50 individuals with telomerase, and 17 individuals with ALT positive tumors. The analysis of CDKN2A showed NDTMM GBMs had an increased minor allele frequency for the C500G (rs11515) polymorphism compared to those with telomerase and ALT positive GBMs (p = 0.002). Patients with the G500 allele had reduced survival that was independent of age, extent of surgery, and treatment. In the NDTMM group G500 allele carriers had increased loss of CDKN2A gene dosage compared to C500 homozygotes. An analysis of IDH1 mutations showed the R132H mutation was associated with ALT positive tumors, and was largely absent in NDTMM and telomerase positive tumors. In the ALT positive tumors cohort, IDH1 mutations were associated with a younger age for the affected individual. In conclusion, the G500 CDKN2A allele was associated with NDTMM GBMs from older individuals with poorer survival. Mutations in IDH1 were not associated with NDTMM GBMs, and instead were a marker for ALT positive tumors in younger individuals

    Alpha-enolase is upregulated on the cell surface and responds to plasminogen activation in mice expressing a ∆133p53α mimic.

    No full text
    The p53 protein is a master regulator of the stress response. It acts as a tumor suppressor by inducing transcriptional activation of p53 target genes, with roles in apoptosis, cell cycle arrest and metabolism. The discovery of at least 12 isoforms of p53, some of which have tumor-promoting properties, has opened new avenues of research. Our previous work studied tumor phenotypes in four mouse models with different p53 backgrounds: wild-type p53, p53 null, mutant p53 lacking the proline domain (mΔpro), and a mimic for the human Δ133p53α p53 isoform (Δ122p53). To identify the major proteins affected by p53 function early in the response to DNA damage, the current study investigated the entire proteome of bone marrow, thymus, and lung in the four p53 models. Protein extracts from untreated controls and those treated with amsacrine were analyzed using two-dimensional fluorescence difference gel electrophoresis. In the bone marrow, reactive proteins were universally decreased by wild-type p53, including α-enolase. Further analysis of α-enolase in the p53 models revealed that it was instead increased in Δ122p53 hematopoietic and tumor cell cytosol and on the cell surface. Alpha-enolase on the surface of Δ122p53 cells acted as a plasminogen receptor, with tumor necrosis factor alpha induced upon plasminogen stimulation. Taken together, these data identified new proteins associated with p53 function. One of these proteins, α-enolase, is regulated differently by wild-type p53 and Δ122p53 cells, with reduced abundance as part of a wild-type p53 response and increased abundance with Δ122p53 function. Increased cell surface α-enolase on Δ122p53 cells provides a possible explanation for the model's pro-inflammatory features and suggests that p53 isoforms may direct an inflammatory response by increasing the amount of α-enolase on the cell surface

    Placental Hypomethylation Is More Pronounced in Genomic Loci Devoid of Retroelements

    No full text
    The human placenta is hypomethylated compared to somatic tissues. However, the degree and specificity of placental hypomethylation across the genome is unclear. We assessed genome-wide methylation of the human placenta and compared it to that of the neutrophil, a representative homogeneous somatic cell. We observed global hypomethylation in placenta (relative reduction of 22%) compared to neutrophils. Placental hypomethylation was pronounced in intergenic regions and gene bodies, while the unmethylated state of the promoter remained conserved in both tissues. For every class of repeat elements, the placenta showed lower methylation but the degree of hypomethylation differed substantially between these classes. However, some retroelements, especially the evolutionarily younger Alu elements, retained high levels of placental methylation. Surprisingly, nonretrotransposon-containing sequences showed a greater degree of placental hypomethylation than retrotransposons in every genomic element (intergenic, introns, and exons) except promoters. The differentially methylated fragments (DMFs) in placenta and neutrophils were enriched in gene-poor and CpG-poor regions. The placentally hypomethylated DMFs were enriched in genomic regions that are usually inactive, whereas hypermethylated DMFs were enriched in active regions. Hypomethylation of the human placenta is not specific to retroelements, indicating that the evolutionary advantages of placental hypomethylation go beyond those provided by expression of retrotransposons and retrogenes

    Increased α-enolase function on the Δ122p53 cell membrane.

    No full text
    <p><b>A.</b> Alpha-enolase was not the only plasminogen receptor increased on the Δ122p53 cell membrane. The cell membrane and cytosolic fractions of untreated PBMCs from p53+ and Δ122p53 mice were separated and subjected to western blotting with an antibody to histone H2B and alpha-enolase. β-actin, FAK, and CD18 were used as loading controls for total protein, cytosolic, and cell membrane fractions, respectively. <b>B.</b> LPS was added to enhance the amount of alpha-enolase on the Δ122p53 PBMCs cell membrane. The cell membrane and cytosolic fractions of untreated Δ122p53 PBMCs or those incubated with LPS (5 μg/mL for 6 hours) were separated and subjected to western blotting with an antibody to alpha-enolase. FAK and CD18 were used as loading controls for cytosolic and cell membrane fractions, respectively. <b>C.</b> TNF-alpha released from Δ122p53 following pre-incubation with lys-plasminogen and LPS (5 μg/mL for 6 hours) with or without two anti-alpha-enolase antibodies (each at 15 μg/mL) to block plasminogen binding or rabbit IgG as a control (30 μg/mL). Prior to TNF-alpha measurement, tissue plasminogen activator (3 nM) was added and the amount of TNF-alpha secreted in culture media was measured by ELISA. The results represent the mean ± SD (n = 3 for each measurement) ***, <i>P</i> < 0.001, **, <i>P</i> < 0.01.</p

    Increased α-enolase on the Δ122p53 tumor cell membrane.

    No full text
    <p>Tumors from Δ122p53 mice had increased alpha-enolase at the cell surface compared to tumors from p53- mice. Sarcomas were dissected from Δ122p53 and p53- mice at necropsy, the cytosolic and cell membrane fractions were separated, and these fractions were subjected to western blotting using an antibody to alpha-enolase. The cell membrane results from two tumors per genotype (1 and 2) are shown. Western blotting for vimentin was used as a loading control for the mesenchymal cell membrane fraction and FAK was included to control for cytosolic contamination in cell membrane preparations.</p

    Altered abundance of alpha-enolase in p53+ and Δ122p53 bone marrow.

    No full text
    <p><b>A.</b> Alpha-enolase is decreased in wild-type p53 (p53+) bone marrow upon DNA damage with amsacrine treatment and increased in Δ122p53 bone marrow cells irrespective of amsacrine treatment compared with p53 null (p53-) cells by western blotting. Amsacrine treated (T) or untreated (U). <b>B.</b> Network analysis of the differentially expressed proteins in p53+ bone marrow untreated and p53+ bone marrow treated with amsacrine from two-dimensional fluorescence difference gel electrophoresis using Ingenuity Pathways Analysis software. Gray shading indicates proteins identified in the current study. Solid and dashed lines indicate direct and indirect interactions, respectively. <b>C.</b> The decrease in α-enolase in p53+ treated bone marrow was not due to reduced expression of <i>ENO1</i>. No significant differences were found in the amounts of <i>ENO1</i> transcript expressed in p53+ and Δ122p53 bone marrow and peripheral blood mononuclear cells (PBMCs) in untreated cells (U) and cells treated with amsacrine (T) using real-time PCR. The results are expressed as the mean ± SD, from 3 mice per genotype and represent the fold increase in <i>ENO1</i> expression, normalized for <i>beta2-M</i> expression. <b>D.</b> The percentage of alpha-enolase positive cells was reduced in p53+ bone marrow and peripheral blood mononuclear cells (PBMCs) treated with amsacrine. Bone marrow and PBMCs from five mice per genotype were extracted and treated with amsacrine (T) or left untreated (U). Following short-term culture, cells were fixed and cell clots sectioned. Alpha-enolase was detected using immunohistochemistry. Positive cells were identified by light microscopy and the percentage of positive cells per total cell count (500 cells) was compared between treated and untreated cells; the results are expressed as the mean ± SD (n = 6 mice per genotype), ****, <i>P</i> < 0.0001. <b>E.</b> The ubiquitin-associated proteasome inhibitor MG132 was added to p53+ and p53- bone marrow treated with amsacrine for 4 or 6 hours. Post-treatment, the amount of alpha-enolase in cell lysates was compared between cells treated with amsacrine or left untreated, by western blotting. <b>F.</b> MG132 treatment did not lead to a statistically significant increase in the percentage of apoptotic cells in p53+ bone marrow at 4 and 6 hours post-treatment with MG132 alone or with MG132 and amsacrine. Apoptotic cells were determined from counting the percentage of active caspase-3-positive cells in bone marrow from immunohistochemistry-stained sections and light microscopy. The percentage of positive cells per total cell count (500 cells) was compared between treated and untreated cells; the results are expressed as the mean ± SD (n = 3).</p

    Human Papillomavirus E6/E7 Expression in Preeclampsia-Affected Placentae

    No full text
    Whether HPV is causative of pregnancy complications is uncertain. E6 and E7 affect functions underling preeclampsia (PET) in cultured trophoblasts, but whether E6 and E7 is produced in the placenta is uncertain. Here, we investigated whether E6/E7 was expressed in the placentae from pregnancies with PET, other pregnancy complications (fetal growth restriction (FGR) and diabetes mellitus), and uncomplicated pregnancies. Placental tissues collected from two geographical locations were subjected to RNAscope analyses of high- and low- risk E6/E7, and individual HPV types identified using an HPV array. High-risk E6/E7 expression was increased in both PET cohorts, (81% and 86% of patients positive for high-risk HPV DNA compared to 13% of control patients). Various HPV types were identified. Although HPV 18 was the most frequent in all cohorts, the majority of individuals had multiple HPV types (55% of the PET compared to 25% of the control cohort). Further evidence that E6 and E7 is present early when placental pathology underlying preeclampsia is established, is provided with the finding of high-risk E6/E7 in the first-trimester placenta anchoring trophoblast. In conclusion, E6/E7 expression and multiple HPV types were frequent in placentae from preeclampsia-complicated pregnancies
    corecore