Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Pathol Oncol Res ; 22(2): 287-92, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26253838

ABSTRACT

BACKGROUND: Protein phosphatase 1 α (PP1A) is an enzyme intimately associated with cell cycle, the over expression of which has been demonstrated in glioblastoma (GBM). Further, the nuclear expression of PP1A has been shown to be highly specific to GBM. In addition, PP1A has been shown to be a connecting molecule in the p53 containing GBM sub network. In view of these, we evaluated the prognostic relevance of PP1A. METHODS: GBM tissues were examined for protein expression of PP1A by immunohistochemistry (IHC). Nuclear expression of PP1A was scored in all tumor tissue samples. Survival analyses were performed by Cox-Regression and Kaplan-Meier survival analysis with Log Rank tests. IDH1, ATRX and p53 IHC and stratification of all GBM cases were performed and subgroup specific evaluation of nuclear PP1A correlation with overall and progression free survival was performed. RESULTS: PP1A protein expression showed no correlation with prognosis in all cases of GBM or on stratification based on IDH1 or ATRX expression. However on p53 stratification nuclear PP1A expression emerged as strong independent predictor of poor overall survival only in p53 positive GBMs both in univariate and multivariate analysis. CONCLUSIONS: While PP1A expression uniquely associates with poor prognosis only in p53 expressing GBMs, there is a notable absence of such correlation in p53 negative GBMs; thus skewing the overall relation of this molecule with prognosis in GBM. PP1A emerging as a strong prognostic marker in p53 expressing GBMs, enables us to foresee this molecule as a potential therapeutic target.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Glioblastoma/pathology , Protein Phosphatase 1/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Brain Neoplasms/enzymology , Cohort Studies , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Glioblastoma/enzymology , Humans , Immunoenzyme Techniques , Male , Neoplasm Staging , Prognosis , Survival Rate
2.
Neurol India ; 63(3): 399-404, 2015.
Article in English | MEDLINE | ID: mdl-26053814

ABSTRACT

BACKGROUND: Despite the conventional acceptance of the matrix metalloproteinases (MMP)-2 and MMP-9, as markers of invasion in glioblastoma (GBM), there is no large body of evidence supporting their role as prognostic markers. Since the co-expression of MMPs with p53 was noted to be prognostic in other cancers, we evaluated the protein expression of MMP-2 and MMP-9 in GBM and explored their prognostic relevance with respect to p53 expression. MATERIALS AND METHODS: Tumor tissues from a uniformly treated cohort of 132 GBM patients were examined for MMP-2, MMP-9, and p53 protein expression by immunohistochemistry (IHC). Survival analyses were performed by Cox-regression and Kaplan-Meier (KM) survival analysis. P53 IHC-based stratification of all GBM cases was performed, and subgroup-specific expression of MMP-2 and MMP-9 was correlated with survival. RESULTS: MMP-2 and MMP-9 were expressed in p53 positive as well as p53 negative GBM tumors. MMP-2 and MMP-9 protein expressions had no correlation with prognosis. MMP-9 expression, however, emerged as a strong independent predictor of poor survival in p53 positive GBMs on both Cox-regression analysis (P = 0.036) and KM survival analysis (P = 0.008). Further, even on multivariate analysis, MMP-9 remained strongly associated with poor prognosis (P = 0.010). CONCLUSIONS: MMP-9 expression strongly associates with poor prognosis in p53 positive GBMs, but the absence of such correlation in p53 negative GBMs, skews the overall relation of this molecule with prognosis. The study highlights that the dual positivity of MMP-9 and p53 is of prognostic relevance in GBM.

3.
J Neurosurg ; 121(2): 374-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24878287

ABSTRACT

OBJECT: Insulin-like growth factor binding proteins (IGFBPs) have been implicated in the pathogenesis of glioma. In a previous study the authors demonstrated that IGFBP-3 is a novel glioblastoma biomarker associated with poor survival. Since signal transducer and activator of transcription 1 (STAT-1) has been shown to be regulated by IGFBP-3 during chondrogenesis and is a prosurvival and radioresistant molecule in different tumors, the aim in the present study was to explore the functional significance of IGFBP-3 in malignant glioma cells, to determine if STAT-1 is indeed regulated by IGFBP-3, and to study the potential of STAT-1 as a biomarker in glioblastoma. METHODS: The functional significance of IGFBP-3 was investigated using the short hairpin (sh)RNA gene knockdown approach on U251MG cells. STAT-1 regulation by IGFBP-3 was tested on U251MG and U87MG cells by shRNA gene knockdown and exogenous treatment with recombinant IGFBP-3 protein. Subsequently, the expression of STAT-1 was analyzed with real-time reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC) in glioblastoma and control brain tissues. Survival analyses were done on a uniformly treated prospective cohort of adults with newly diagnosed glioblastoma (136 patients) using Kaplan-Meier and Cox regression models. RESULTS: IGFBP-3 knockdown significantly impaired proliferation, motility, migration, and invasive capacity of U251MG cells in vitro (p < 0.005). Exogenous overexpression of IGFBP-3 in U251MG and U87MG cells demonstrated STAT-1 regulation. The mean transcript levels (by real-time RT-PCR) and the mean labeling index of STAT-1 (by IHC) were significantly higher in glioblastoma than in control brain tissues (p = 0.0239 and p < 0.001, respectively). Multivariate survival analysis revealed that STAT-1 protein expression (HR 1.015, p = 0.033, 95% CI 1.001-1.029) along with patient age (HR 1.025, p = 0.005, 95% CI 1.008-1.042) were significant predictors of shorter survival in patients with glioblastoma. CONCLUSIONS: IGFBP-3 influences tumor cell proliferation, migration, and invasion and regulates STAT-1 expression in malignant glioma cells. STAT-1 is overexpressed in human glioblastoma tissues and emerges as a novel prognostic biomarker.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioblastoma/genetics , Glioma/genetics , Glioma/metabolism , Insulin-Like Growth Factor Binding Protein 3/genetics , STAT1 Transcription Factor/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Female , Glioblastoma/mortality , Humans , Insulin-Like Growth Factor Binding Protein 3/physiology , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Predictive Value of Tests , STAT1 Transcription Factor/biosynthesis , Survival Analysis
4.
PLoS One ; 8(4): e62042, 2013.
Article in English | MEDLINE | ID: mdl-23646114

ABSTRACT

BACKGROUND: Recent research on glioblastoma (GBM) has focused on deducing gene signatures predicting prognosis. The present study evaluated the mRNA expression of selected genes and correlated with outcome to arrive at a prognostic gene signature. METHODS: Patients with GBM (n = 123) were prospectively recruited, treated with a uniform protocol and followed up. Expression of 175 genes in GBM tissue was determined using qRT-PCR. A supervised principal component analysis followed by derivation of gene signature was performed. Independent validation of the signature was done using TCGA data. Gene Ontology and KEGG pathway analysis was carried out among patients from TCGA cohort. RESULTS: A 14 gene signature was identified that predicted outcome in GBM. A weighted gene (WG) score was found to be an independent predictor of survival in multivariate analysis in the present cohort (HR = 2.507; B = 0.919; p<0.001) and in TCGA cohort. Risk stratification by standardized WG score classified patients into low and high risk predicting survival both in our cohort (p = <0.001) and TCGA cohort (p = 0.001). Pathway analysis using the most differentially regulated genes (n = 76) between the low and high risk groups revealed association of activated inflammatory/immune response pathways and mesenchymal subtype in the high risk group. CONCLUSION: We have identified a 14 gene expression signature that can predict survival in GBM patients. A network analysis revealed activation of inflammatory response pathway specifically in high risk group. These findings may have implications in understanding of gliomagenesis, development of targeted therapies and selection of high risk cancer patients for alternate adjuvant therapies.


Subject(s)
Gene Expression Profiling , Glioblastoma/genetics , Glioblastoma/immunology , Transcriptome , Computational Biology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Molecular Sequence Annotation , Prognosis , Proteome , Reproducibility of Results , Risk , Signal Transduction
5.
Pediatr Neurosurg ; 49(6): 325-30, 2013.
Article in English | MEDLINE | ID: mdl-25427587

ABSTRACT

BACKGROUND: Insulin-like growth factor binding proteins (IGFBPs) are known to be differentially expressed in brain tumours. The role of some IGFBPs in malignant CNS tumours, except glioblastoma, is unknown. We evaluated the protein expression of 3 IGFBP isoforms (IGFBP-2, -3, -5) in medulloblastoma and correlated them with histological subtypes and clinical parameters. METHODS: The expression of IGFBP-2, -3 and -5 was analysed in 67 samples of medulloblastoma by immunohistochemistry and correlated with histological subtypes and patient prognosis. RESULTS: IGFBP-3 expression was seen in 37.3% of cases and IGFBP-5 expression in 80.6% of cases. IGFBP-2 expression was totally absent in medulloblastoma. The extent of IGFBP-3 expression was higher in anaplastic when compared to classical and desmoplastic subtypes (p < 0.05). IGFBP-5 expression was significantly higher in classical and anaplastic subtypes when compared to desmoplastic medulloblastoma (p < 0.05). No influence of IGFBPs on survival was noted. CONCLUSIONS: This is the first study to report expression of 3 cancer-related biomarkers - IGFBP-2, -3, -5 in medulloblastoma. Significantly higher extents of expression of IGFBP-3 in large cell variant and IGFBP-5 in classical and anaplastic subtypes suggest a plausible role of these molecules in specific molecular pathways of medulloblastoma.


Subject(s)
Biomarkers, Tumor/metabolism , Cerebellar Neoplasms/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Medulloblastoma/metabolism , Adolescent , Adult , Cerebellar Neoplasms/classification , Child , Child, Preschool , Disease Progression , Disease-Free Survival , Female , Humans , Immunohistochemistry , Male , Medulloblastoma/classification , Middle Aged , Protein Isoforms , Young Adult
6.
Am J Clin Pathol ; 138(2): 177-84, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22904127

ABSTRACT

IDH1 mutations are frequent genetic alterations in low-grade diffuse gliomas and secondary glioblastoma (GBM). To validate mutation frequency, IDH1 gene at codon 132 was sequenced in 74 diffusely infiltrating astrocytomas: diffuse astrocytoma (DA; World Health Organization [WHO] grade II), anaplastic astrocytoma (AA; WHO grade III), and GBM (WHO grade IV). All cases were immunostained with IDH1-R132H monoclonal antibody. Mutational status was correlated with mutant protein expression, patient age, duration of symptoms, and prognosis of patients with GBM. We detected 31 (41.9%) heterozygous IDH1 mutations resulting in arginine-to-histidine substitution (R132H;CGT-CAT). All 12 DAs (100%), 13 of 14 AAs (92.9%), and 6 of 48 GBMs (12.5%) (5/6 [83.3%] secondary, and 1/42 [2.4%] primary) harbored IDH1 mutations. The correlation between mutational status and protein expression was significant (P < . 001). IDH1 mutation status, though not associated with prognosis of patients with GBM, showed significant association with younger age and longer duration of symptoms in the whole cohort (P < .001). Our study validates IDH1 mutant protein expression across various grades of astrocytoma, and demonstrates a high incidence of IDH1 mutations in DA, AA, and secondary GBM.


Subject(s)
Amino Acid Substitution , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Isocitrate Dehydrogenase/genetics , Astrocytoma/mortality , Brain Neoplasms/mortality , Female , Humans , Immunohistochemistry , Male , Mutation , Neoplasm Grading , Neoplasm Invasiveness , Survival Analysis
7.
J Neurooncol ; 107(2): 289-97, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22102081

ABSTRACT

The search for molecular markers which predict response to chemotherapy is an important aspect of current neuro-oncology research. MGMT promoter methylation is the only proved marker of glioblastoma. The purpose of this study was to assess the effect of topoisomerase expression on glioblastoma survival and study the mechanisms involved. The transcript levels of all isoforms of the topoisomerase family in all grades of diffuse astrocytoma were assessed. A prospective study of patients with glioblastoma treated by a uniform treatment procedure was performed with the objective of correlating outcome with gene expression. The ability of TOP2A enzyme to relax the super coiled plasmid DNA in the presence of temozolomide was evaluated to assess its effect on TOP2A. The temozolomide cyctotoxicity of TOP2A-silenced U251 cells was assessed. The transcript levels of TOP2A, TOP2B, and TOP3A are upregulated significantly in GBM in comparison with lower grades of astrocytoma and normal brain samples. mRNA levels of TOP2A correlated significantly with survival of the patients. Higher TOP2A transcript levels in GBM patients predicted better prognosis (P = 0.043; HR = 0.889). Interestingly, we noted that temozolomide inhibited TOP2A activity in in-vitro enzyme assays. We also noted that siRNA knock down of TOP2A rendered a glioma cell line resistant to temozolomide chemotherapy. We demonstrated for the first time that temozolomide is also a TOP2A inhibitor and established that TOP2A transcript levels determine the chemosensitivity of glioblastoma to temozolomide therapy. Very high levels of TOP2A are a good prognostic indicator in GBM patients receiving temozolomide chemotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Brain Neoplasms/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Dacarbazine/analogs & derivatives , Gene Expression Regulation, Enzymologic/drug effects , Glioblastoma/metabolism , Topoisomerase II Inhibitors/pharmacology , Antigens, Neoplasm/genetics , Antineoplastic Agents/pharmacology , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cohort Studies , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Glioblastoma/diagnosis , Glioblastoma/mortality , Humans , Male , Poly-ADP-Ribose Binding Proteins , Prognosis , RNA, Messenger/metabolism , Temozolomide
8.
J Biol Chem ; 286(29): 25882-90, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21613208

ABSTRACT

Glioblastoma is the most common and malignant form of primary astrocytoma. Upon investigation of the insulin-like growth factor (IGF) pathway, we found the IGF2BP3/IMP3 transcript and protein to be up-regulated in GBMs but not in lower grade astrocytomas (p < 0.0001). IMP3 is an RNA binding protein known to bind to the 5'-untranslated region of IGF-2 mRNA, thereby activating its translation. Overexpression- and knockdown-based studies establish a role for IMP3 in promoting proliferation, anchorage-independent growth, invasion, and chemoresistance. IMP3 overexpressing B16F10 cells also showed increased tumor growth, angiogenesis, and metastasis, resulting in poor survival in a mouse model. Additionally, the infiltrating front, perivascular, and subpial regions in a majority of the GBMs stained positive for IMP3. Furthermore, two different murine glioma models were used to substantiate the above findings. In agreement with the translation activation functions of IMP3, we also found increased IGF-2 protein in the GBM tumor samples without a corresponding increase in its transcript levels. Also, in vitro IMP3 overexpression/knockdown modulated the IGF-2 protein levels without altering its transcript levels. Additionally, IGF-2 neutralization and supplementation studies established that the proproliferative effects of IMP3 were indeed mediated through IGF-2. Concordantly, PI3K and MAPK, the downstream effectors of IGF-2, are activated by IMP3 and are found to be essential for IMP3-induced cell proliferation. Thus, we have identified IMP3 as a GBM-specific proproliferative and proinvasive marker acting through IGF-2 resulting in the activation of oncogenic PI3K and MAPK pathways.


Subject(s)
Biomarkers, Tumor/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Insulin-Like Growth Factor II/metabolism , MAP Kinase Signaling System , RNA-Binding Proteins/metabolism , Adolescent , Adult , Aged , Animals , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Glioblastoma/genetics , Glioblastoma/physiopathology , Humans , Insulin-Like Growth Factor II/biosynthesis , Insulin-Like Growth Factor II/genetics , Mice , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Protein Biosynthesis , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Survival Analysis , Up-Regulation , Young Adult
9.
Neuropathology ; 31(4): 376-83, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21134002

ABSTRACT

Tumors from a prospective cohort of adult patients with newly diagnosed glioblastoma (n=73), treated uniformly with radiochemotherapy, were examined for 10q23/PTEN deletion by fluorescence in situ hybridization (FISH). Statistical methods were employed to evaluate the degree of association between 10q23/PTEN deletion status and patient age. Survival analysis was performed using Kaplan-Meier log-rank test and multivariable Cox models to assess the prognostic value of 10q23/PTEN deletion. Interestingly, 10q23/PTEN homozygous deletion was frequent in patients >45 years of age (P=0.034) and the median age of patients harboring PTEN homozygous deletions was significantly higher than those with the retained status (P=0.019). 10q23/PTEN homozygous deletion was associated with shorter survival in the entire cohort as well in patients >45 years (P<0.05), indicating that loss of 10q23/PTEN showed clinical importance in elderly patients. Our study highlights the independent prognostic/predictive value of 10q23/PTEN deletion status as identified by FISH, particularly in glioblastoma patients aged >45 years.


Subject(s)
Brain Neoplasms/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 10 , Glioblastoma/genetics , PTEN Phosphohydrolase/genetics , Adult , Brain Neoplasms/mortality , Female , Gene Dosage , Glioblastoma/mortality , Humans , Male , Predictive Value of Tests , Prognosis , Prospective Studies , Survival Analysis
10.
J Proteome Res ; 9(11): 5557-67, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20822092

ABSTRACT

Glioblastoma (GBM; grade IV astrocytoma) is the most malignant and common primary brain tumor in adults. Using combination of 2-DE and MALDI-TOF MS, we analyzed 14 GBM and 6 normal control sera and identified haptoglobin α2 chain as an up-regulated serum protein in GBM patients. GBM-specific up-regulation was confirmed by ELISA based quantitation of haptoglobin (Hp) in the serum of 99 GBM patients as against lower grades (49 grade III/AA; 26 grade II/DA) and 26 normal individuals (p = 0.0001). Further validation using RT-qPCR on an independent set (n = 78) of tumor and normal brain (n = 4) samples and immunohistochemcial staining on a subset (n = 42) of above samples showed increasing levels of transcript and protein with tumor grade and were highest in GBM (p = <0.0001 and <0.0001, respectively). Overexpression of Hp either by stable integration of Hp cDNA or exogenous addition of purified Hp to immortalized astrocytes resulted in increased cell migration. RNAi-mediated silencing of Hp in glioma cells decreased cell migration. Further, we demonstrate that both human glioma and mouse melanoma cells overexpressing Hp showed increased tumor growth. Thus, we have identified haptoglobin as a GBM-specific serum marker with a role on glioma tumor growth and migration.


Subject(s)
Glioblastoma/diagnosis , Haptoglobins/analysis , Haptoglobins/physiology , Proteomics/methods , Animals , Astrocytes/chemistry , Astrocytes/pathology , Biomarkers, Tumor/blood , Case-Control Studies , Cell Line, Tumor , Cell Movement , Cell Proliferation , Haptoglobins/genetics , Humans , Melanoma/chemistry , Melanoma/pathology , Mice , Up-Regulation
11.
J Clin Pathol ; 63(8): 687-91, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20702468

ABSTRACT

AIMS: To assess the prognostic influence of EGFR amplification/overexpression, p53 immunoreactivity and their age-dependent prognostic effects in a large prospective cohort of uniformly treated adult patients with newly diagnosed glioblastoma. METHODS: Tumours from a uniformly treated prospective cohort of adult patients with newly diagnosed glioblastoma (n=140) were examined for EGFR amplification by fluorescence in situ hybridisation and EGFR/p53 expression by immunohistochemistry. Statistical methods were employed to assess the degree of association between EGFR amplification/overexpression and p53 immunopositivity. Survival analyses were performed by employing Cox proportional hazard models to assess the independent prognostic value of EGFR/p53 alterations and test the propensity for risk with age by assessing their interaction with patient age. RESULTS: A strong positive correlation between EGFR amplification and EGFR overexpression (rho=0.5157; p<0.0001; CI 0.3783 to 0.6309) and a negative association of EGFR amplification (rho=-0.3417; p<0.0001; CI -0.4842 to -0.1816) and EGFR overexpression (rho=-0.3095; p<0.001; CI -0.4561 to -0.1465) with p53 immunopositivity was observed. Only patient age (HR: 1.029; p=0.004; CI 1.009 to 1.049) was associated with shorter survival by univariate Cox regression analysis. Multivariable Cox proportional hazards models revealed a statistically significant interaction between EGFR overexpression and age to be associated with shorter survival (HR: 1.001; p<0.0001; CI 1.000 to 1.002), thus predicting a higher hazard with increasing age. No age interaction of EGFR amplification status (HR: 1.001; p=0.642; CI 0.995 to 1.008) and p53 immunopositivity (HR: 1.000; p=0.841; CI 0.999 to 1.001) was noted in this cohort. CONCLUSIONS: The prognostic value of EGFR overexpression is age-dependent, and there is a propensity for a higher hazard with increasing patient age. Identifying such groups of patients with more aggressive disease becomes mandatory, since they would benefit from intense therapeutic protocols targeting EGFR.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnosis , ErbB Receptors/metabolism , Glioblastoma/diagnosis , Tumor Suppressor Protein p53/metabolism , Adolescent , Adult , Age Factors , Aged , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , Epidemiologic Methods , ErbB Receptors/genetics , Genes, p53 , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/therapy , Humans , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prognosis , Treatment Outcome , Young Adult
12.
Cancer Res ; 70(16): 6437-47, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20663907

ABSTRACT

Glioblastoma (GBM; grade IV astrocytoma) is a very aggressive form of brain cancer with a poor survival and few qualified predictive markers. This study integrates experimentally validated genes that showed specific upregulation in GBM along with their protein-protein interaction information. A system level analysis was used to construct GBM-specific network. Computation of topological parameters of networks showed scale-free pattern and hierarchical organization. From the large network involving 1,447 proteins, we synthesized subnetworks and annotated them with highly enriched biological processes. A careful dissection of the functional modules, important nodes, and their connections identified two novel intermediary molecules CSK21 and protein phosphatase 1 alpha (PP1A) connecting the two subnetworks CDC2-PTEN-TOP2A-CAV1-P53 and CDC2-CAV1-RB-P53-PTEN, respectively. Real-time quantitative reverse transcription-PCR analysis revealed CSK21 to be moderately upregulated and PP1A to be overexpressed by 20-fold in GBM tumor samples. Immunohistochemical staining revealed nuclear expression of PP1A only in GBM samples. Thus, CSK21 and PP1A, whose functions are intimately associated with cell cycle regulation, might play key role in gliomagenesis.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Casein Kinase II/genetics , Genes, cdc , Glioblastoma/genetics , Glioblastoma/metabolism , Protein Phosphatase 1/genetics , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Casein Kinase II/biosynthesis , Casein Kinase II/metabolism , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Immunohistochemistry , Protein Interaction Mapping , Protein Phosphatase 1/biosynthesis , Protein Phosphatase 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
13.
Cancer Epidemiol Biomarkers Prev ; 19(6): 1399-408, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20501753

ABSTRACT

BACKGROUND: Insulin-like growth factor (IGF)-binding protein (IGFBP) isoforms have been implicated in the pathogenesis of human neoplasms including glioma. In view of this, we evaluated the expression of IGFBP isoforms (IGFBP-2, -3, and -5) during malignant progression of astrocytoma and their prognostic significance in glioblastoma. METHODS: The expression of IGFBP isoforms was analyzed in diffusely infiltrating astrocytomas by real-time quantitative PCR (n = 203) and immunohistochemistry (n = 256). Statistical methods were used to assess their grade-specific expression pattern and mRNA-protein intercorrelation. Survival analyses were done on a uniformly treated, prospective cohort of adult patients with newly diagnosed glioblastoma (n = 136) by using Cox regression models. RESULTS: The mean transcript levels of IGFBP-2 and -3 were significantly higher in glioblastomas (GBM) relative to anaplastic astrocytoma (AA), diffuse astrocytoma (DA), and controls whereas IGFBP-5 mRNA was higher in GBM relative to AA and controls (P < 0.05). By immunohistochemistry, the mean labeling index of all isoforms was significantly higher in GBM compared with AA, DA, and control (P < 0.05). A strong positive correlation was observed between their respective mRNA and protein expressions (P < 0.01). Multivariate analysis revealed IGFBP-3 expression (hazard ratio, 1.021; P = 0.030) and patient age (hazard ratio, 1.027; P = 0.007) to be associated with shorter survival in glioblastoma. CONCLUSIONS: This study shows the associations of IGFBP-2, -3, and -5 expression with increasing grades of malignancy in astrocytomas. IGFBP-3 is identified as a novel prognostic glioblastoma biomarker. The strong correlation between their mRNA and protein expression patterns suggests their role in the pathogenesis of these tumors. IMPACT: IGFBP isoforms have emerged as biomarkers with diagnostic and prognostic utility in astrocytomas.


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Insulin-Like Growth Factor Binding Protein 2/biosynthesis , Insulin-Like Growth Factor Binding Protein 5/biosynthesis , Insulin-Like Growth Factor Binding Proteins/biosynthesis , Adolescent , Adult , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/pathology , Cohort Studies , Disease Progression , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunohistochemistry , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Proteins/genetics , Middle Aged , Polymerase Chain Reaction/methods , Prognosis , Prospective Studies , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Survival Analysis , Young Adult
14.
Cancer Biol Ther ; 7(5): 663-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18728403

ABSTRACT

Malignant astrocytomas comprise anaplastic astrocytoma (AA; grade III) and Glioblastoma (GBM; grade IV). GBM is the most malignant with a median survival of 10-12 months in patients. Using cDNA microarray based expression profiling of different grades of astrocytomas, we identified several fold increased levels of PBEF1 transcripts in GBM samples. Pre-B-cell colony enhancing factor 1 gene (PBEF1) encodes Nicotinamide phosphoribosyltransferase (NAmPRTase), which catalyses the rate limiting step in the salvage pathway of NAD metabolism in mammalian cells. Further validation using real time RT-qPCR on an independent set of tumor samples (n=91) and normal brain samples (n=9), GBM specific higher expression of PBEF1 was confirmed. Immunohistochemical staining for PBEF1 on a subset of the above samples largely reinforced our finding. We carried out ELISA analysis on serum samples of astrocytoma patients to determine whether this protein levels would correlate with the presence of tumor and tumor grade. PBEF1 serum levels were substantially elevated in many of the AA and GBM patients. Statistical analysis of these data indicates that in patients with astrocytoma, serum PBEF1 levels correlate with tumor grade and is highest in GBM. Immunohistochemical analysis of an independent set of 51 retrospective GBM cases with known survival data revealed that PBEF1 expression in the tumor tissue along with its co-expression with p53 was associated with poor survival. Thus, we have identified PBEF1 as a potential malignant astrocytoma serum marker and prognostic indicator among GBMs.


Subject(s)
Astrocytoma/metabolism , Biomarkers, Tumor , Brain Neoplasms/metabolism , Brain/metabolism , Cytokines/physiology , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Astrocytoma/blood , Astrocytoma/mortality , Brain Neoplasms/blood , Brain Neoplasms/mortality , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Glioblastoma/blood , Glioblastoma/mortality , Humans , Immunohistochemistry/methods , Nicotinamide Phosphoribosyltransferase/physiology , Oligonucleotide Array Sequence Analysis , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism
15.
Clin Cancer Res ; 14(10): 2978-87, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483363

ABSTRACT

PURPOSE: Current methods of classification of astrocytoma based on histopathologic methods are often subjective and less accurate. Although patients with glioblastoma have grave prognosis, significant variability in patient outcome is observed. Therefore, the aim of this study was to identify glioblastoma diagnostic and prognostic markers through microarray analysis. EXPERIMENTAL DESIGN: We carried out transcriptome analysis of 25 diffusely infiltrating astrocytoma samples [WHO grade II--diffuse astrocytoma, grade III--anaplastic astrocytoma, and grade IV--glioblastoma (GBM)] using cDNA microarrays containing 18,981 genes. Several of the markers identified were also validated by real-time reverse transcription quantitative PCR and immunohistochemical analysis on an independent set of tumor samples (n = 100). Survival analysis was carried out for two markers on another independent set of retrospective cases (n = 51). RESULTS: We identified several differentially regulated grade-specific genes. Independent validation by real-time reverse transcription quantitative PCR analysis found growth arrest and DNA-damage-inducible alpha (GADD45alpha) and follistatin-like 1 (FSTL1) to be up-regulated in most GBMs (both primary and secondary), whereas superoxide dismutase 2 and adipocyte enhancer binding protein 1 were up-regulated in the majority of primary GBM. Further, identification of the grade-specific expression of GADD45alpha and FSTL1 by immunohistochemical staining reinforced our findings. Analysis of retrospective GBM cases with known survival data revealed that cytoplasmic overexpression of GADD45alpha conferred better survival while the coexpression of FSTL1 with p53 was associated with poor survival. CONCLUSIONS: Our study reveals that GADD45alpha and FSTLI are GBM-specific whereas superoxide dismutase 2 and adipocyte enhancer binding protein 1 are primary GBM-specific diagnostic markers. Whereas GADD45alpha overexpression confers a favorable prognosis, FSTL1 overexpression is a hallmark of poor prognosis in GBM patients.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Gene Expression Profiling , Glioblastoma/diagnosis , Glioblastoma/genetics , Brain Neoplasms/mortality , Glioblastoma/mortality , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Oligonucleotide Array Sequence Analysis , Prognosis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL