Share this post on:

Taining 5 milk and 0.05 Tween-20, 2 hours), probed overnight with antibodies specific for PKM1 (Proteintech, 1:1000), PKM2 (Cell Signaling, 1:1000) or b-actin (Cell Signaling, 1:20,000), washed, then incubated with appropriate horseradish peroxidase-conjugated secondary antibodies (Santa Cruz Biotechnology). Antibody binding was detected by incubation with ECL reagents (Amersham SMER 28 site Pharmacia Biotech). Intracranial tumor formation. Immunodeficient mice (nu/ nu; Charles River) were injected intracranially with 46105 luciferase-expressing U87-Scr-Luc (N = 5) or U87-shPK-M2-Luc (N = 5) cells 25033180 as described [25]. Tumor growth was monitored weekly by treating mice with D-luciferin (150 mg/kg IP, GoldBiotechnology) and measuring bioluminescence using a Xenogen IVIS Bioluminescence imaging station (Caliper). Tumor growth was calculated by normalizing luminescence measurements to day1 post injection values. Animals were monitored daily until they developed signs of neurological deficit, at which time they were sacrificed. Statistical analysis. When two groups were compared, the unpaired Student’s t test was applied (P-value). When multiplePyruvate Kinase Modulation in Brain TumorsFigure 1. PKM1 and PKM2 mRNA expression in a series of human normal brain (NB), neural progenitor cells (NSC), and WHO grade I-IV human astrocytoma specimens.A, RNA was isolated from fixed or frozen normal brain (NB) and grade (Gr)- I, II, III, and IV (primary, P and secondary, S) astrocytoma samples, reverse transcribed, then subjected to triplicate qPCR analysis using primers specific for the PKM1 or PKM2 transcript. All values are the mean normalized to HPRT1 expression. B, mean group PKM1 and PKM2 mRNA expression values from panel A and from NSC and established GBM cell lines. C, cDNAs from representative samples in panel A were subjected to PCR amplification using primers amplifying a 442 bp exon 8?1 region common to PKM1 and PKM2. Following incubation with PstI, the uncleaved (PKM1, 442 bp) and cleaved (PKM2, 246 and 196 bp) amplification products were separated by electrophoresis and quantitated, with total signal (PKM1+PKM2) set at 100 for each lane. P, PCR control (Gr-IV amplification products prior to PstI digestion); R, duplicate restriction enzyme controls (amplification products derived using a PKM2 cDNA template post-PstI digestion). doi:10.1371/journal.pone.0057610.gPyruvate Kinase Modulation in Brain Tumorsprotein than brain tumor samples or commonly used GBM cell lines, consistent with previously reported data [21]. These results were also consistent with immunohistochemical analyses of fixed tissue (Fig 2C), which showed that as noted at the RNA level, normal brain expresses higher MedChemExpress INCB039110 levels of PKM1 protein than all gliomas. Consistent with the RNA analysis, levels of PKM1 protein expression were not significantly between the various classes of glioma. In contrast, and consistent with the RNA analyses presented, GBM and GBM cell lines expressed significantly more PKM2 protein than the other lower grade tumors or normal brain (Fig. 2A ). These results therefore show that at both the RNA and protein levels, GBM appear different from lower grade glioma in their high level expression of PKM2. Given the differences in PKM expression and aggressiveness of GBM relative to lower grade tumors, and the link between PKM isoform expression and metabolism, we also determined if changes in PK activity were noted across glioma grades. Consistent with the Western blot and i.Taining 5 milk and 0.05 Tween-20, 2 hours), probed overnight with antibodies specific for PKM1 (Proteintech, 1:1000), PKM2 (Cell Signaling, 1:1000) or b-actin (Cell Signaling, 1:20,000), washed, then incubated with appropriate horseradish peroxidase-conjugated secondary antibodies (Santa Cruz Biotechnology). Antibody binding was detected by incubation with ECL reagents (Amersham Pharmacia Biotech). Intracranial tumor formation. Immunodeficient mice (nu/ nu; Charles River) were injected intracranially with 46105 luciferase-expressing U87-Scr-Luc (N = 5) or U87-shPK-M2-Luc (N = 5) cells 25033180 as described [25]. Tumor growth was monitored weekly by treating mice with D-luciferin (150 mg/kg IP, GoldBiotechnology) and measuring bioluminescence using a Xenogen IVIS Bioluminescence imaging station (Caliper). Tumor growth was calculated by normalizing luminescence measurements to day1 post injection values. Animals were monitored daily until they developed signs of neurological deficit, at which time they were sacrificed. Statistical analysis. When two groups were compared, the unpaired Student’s t test was applied (P-value). When multiplePyruvate Kinase Modulation in Brain TumorsFigure 1. PKM1 and PKM2 mRNA expression in a series of human normal brain (NB), neural progenitor cells (NSC), and WHO grade I-IV human astrocytoma specimens.A, RNA was isolated from fixed or frozen normal brain (NB) and grade (Gr)- I, II, III, and IV (primary, P and secondary, S) astrocytoma samples, reverse transcribed, then subjected to triplicate qPCR analysis using primers specific for the PKM1 or PKM2 transcript. All values are the mean normalized to HPRT1 expression. B, mean group PKM1 and PKM2 mRNA expression values from panel A and from NSC and established GBM cell lines. C, cDNAs from representative samples in panel A were subjected to PCR amplification using primers amplifying a 442 bp exon 8?1 region common to PKM1 and PKM2. Following incubation with PstI, the uncleaved (PKM1, 442 bp) and cleaved (PKM2, 246 and 196 bp) amplification products were separated by electrophoresis and quantitated, with total signal (PKM1+PKM2) set at 100 for each lane. P, PCR control (Gr-IV amplification products prior to PstI digestion); R, duplicate restriction enzyme controls (amplification products derived using a PKM2 cDNA template post-PstI digestion). doi:10.1371/journal.pone.0057610.gPyruvate Kinase Modulation in Brain Tumorsprotein than brain tumor samples or commonly used GBM cell lines, consistent with previously reported data [21]. These results were also consistent with immunohistochemical analyses of fixed tissue (Fig 2C), which showed that as noted at the RNA level, normal brain expresses higher levels of PKM1 protein than all gliomas. Consistent with the RNA analysis, levels of PKM1 protein expression were not significantly between the various classes of glioma. In contrast, and consistent with the RNA analyses presented, GBM and GBM cell lines expressed significantly more PKM2 protein than the other lower grade tumors or normal brain (Fig. 2A ). These results therefore show that at both the RNA and protein levels, GBM appear different from lower grade glioma in their high level expression of PKM2. Given the differences in PKM expression and aggressiveness of GBM relative to lower grade tumors, and the link between PKM isoform expression and metabolism, we also determined if changes in PK activity were noted across glioma grades. Consistent with the Western blot and i.

Share this post on: