Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 7(45): 72431-72442, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27626492

ABSTRACT

PROX1 is a transcription factor with an essential role in embryonic development and determination of cell fate. In addition, PROX1 has been ascribed suppressive as well as oncogenic roles in several human cancers, including brain tumors. In this study we explored the correlation between PROX1 expression and patient survival in high-grade astrocytomas. For this purpose, we analyzed protein expression in tissue microarrays of tumor samples stratified by patient age and IDH mutation status. We initially screened 86 unselected high-grade astrocytomas, followed by 174 IDH1-R132H1 immunonegative glioblastomas derived from patients aged 60 years and older enrolled in the Nordic phase III trial of elderly patients with newly diagnosed glioblastoma. Representing the younger population of glioblastomas, we studied 80 IDH-wildtype glioblastomas from patients aged 18-60 years. There was no correlation between PROX1 protein and survival for patients with primary glioblastomas included in these cohorts. In contrast, high expression of PROX1 protein predicted shorter survival in the group of patients with IDH-mutant anaplastic astrocytomas and secondary glioblastomas. The prognostic impact of PROX1 in IDH-mutant 1p19q non-codeleted high-grade astrocytomas, as well as the negative findings in primary glioblastomas, was corroborated by gene expression data extracted from the Cancer Genome Atlas. We conclude that PROX1 is a new prognostic biomarker for 1p19q non-codeleted high-grade astrocytomas that have progressed from pre-existing low-grade tumors and harbor IDH mutations.


Subject(s)
Biomarkers, Tumor/metabolism , Glioblastoma/metabolism , Homeodomain Proteins/metabolism , Isocitrate Dehydrogenase/genetics , Tumor Suppressor Proteins/metabolism , Age Factors , Aged , Astrocytoma/genetics , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cohort Studies , Female , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Isocitrate Dehydrogenase/metabolism , Male , Middle Aged , Mutation , Neoplasm Grading , Prognosis
2.
Sci Rep ; 5: 16495, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26559910

ABSTRACT

Glioblastoma (grade IV glioma) is the most common and aggressive adult brain tumor. A better understanding of the biology of glioblastoma cells is crucial to identify molecular targets stimulating cell death. NPM1 (nucleophosmin) is a multifunctional chaperone that plays an important role in cancer development. Herein, NPM1 was analyzed by immunohistochemistry in human astrocytic gliomas. NPM1 was detected in all tumors but with a significantly higher staining intensity in grade IV than in low grade tumors. Depletion of NPM1 had only modest effects on the viability of U251MG, U1242MG, and U343MGa Cl2:6 glioma cells, despite alterations in nucleolar morphology. Glioma cell cultures depleted of NPM1 exposed to micromolar levels of actinomycin D were more prone to cell death (apoptosis) compared to cultures retaining NPM1. We had previously found that NPM1 binds to linker histone H1.5. Here we could show that silencing of H1.5 triggered glioma cell apoptosis as evidenced by a marked increase in both the numbers of cleaved caspase-3(+) cells and in the amounts of cleaved PARP. Enforced expression of NPM1 suppressed apoptosis in H1.5 depleted glioma cells. Although our studies would suggest little effectiveness of targeting NPM1 alone there could be potential using it as a combination treatment.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Glioblastoma/metabolism , Glioblastoma/mortality , Histone Chaperones/metabolism , Nuclear Proteins/metabolism , Animals , Apoptosis/genetics , Astrocytes/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Nucleolus/pathology , Cell Survival/drug effects , Cell Survival/genetics , Dactinomycin/pharmacology , Disease Models, Animal , Gene Expression , Gene Knockdown Techniques , Gene Silencing , Glioblastoma/genetics , Glioblastoma/pathology , Histone Chaperones/genetics , Humans , Immunohistochemistry , Mice, Knockout , Neoplasm Grading , Neural Stem Cells/metabolism , Nuclear Proteins/genetics , Nucleophosmin , Prognosis , Protein Transport
3.
Acta Oncol ; 54(3): 385-94, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25263081

ABSTRACT

BACKGROUND: The survival of high-grade glioma patients is poor and the treatment of these patients can cause severe side effects. This fosters the necessity to identify prognostic biomarkers, in order to optimize treatment and diminish unnecessary suffering of patients. The aim of this study was to identify prognostic biomarkers for high-grade glioma patients. METHODS: Eleven proteins were selected for analysis due to their suggested importance for survival of patients with other types of cancers and due to a high variation in protein levels between glioma patients (according to the Human Protein Atlas, www.proteinatlas.org). Protein expression patterns of these 11 proteins were analyzed by immunohistochemistry in tumor samples from 97 high-grade glioma patients. The prognostic values of the proteins were analyzed with univariate and multivariate Cox regression analyses for the high-grade glioma patients, including subgroup analyses of histological subtypes and immunohistochemically defined molecular subtypes. RESULTS: The proteins with the most significant (univariate and multivariate p<0.05) correlations were analyzed further with cross-validated Kaplan-Meier analyses for the possibility of predicting survival based on the protein expression pattern of the corresponding candidate. Random Forest classification with variable subset selection was used to analyze if a protein signature consisting of any combination of the 11 proteins could predict survival for the high-grade glioma patients and the subgroup with glioblastoma patients. The proteins which correlated most significantly (univariate and multivariate p<0.05) to survival in the Cox regression analyses were Myc for all high-grade gliomas and FGF2, CA9 and CD44 for the subgroup of proneural gliomas, with FGF2 having a strong negative predictive value for survival. No prognostic signature of the proteins could be found. CONCLUSION: FGF2 is a potential prognostic biomarker for proneural glioma patients, and warrants further investigation.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Fibroblast Growth Factor 2/metabolism , Glioma/metabolism , Glioma/mortality , Neoplasm Proteins/metabolism , Adult , Aged , Aged, 80 and over , Analysis of Variance , Antigens, Neoplasm/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Carbonic Anhydrase IX , Carbonic Anhydrases/metabolism , Female , Glioma/pathology , Glioma/therapy , Humans , Hyaluronan Receptors/metabolism , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Proto-Oncogene Proteins c-myc/metabolism , Regression Analysis , Retrospective Studies , Tissue Array Analysis
4.
Int J Cancer ; 134(5): 1123-31, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24037901

ABSTRACT

Recent studies suggest that the regulatory networks controlling the functions of stem cells during development may be abnormally active in human cancers. An embryonic stem cell (ESC) gene signature was found to correlate with a more undifferentiated phenotype of several human cancer types including gliomas, and associated with poor prognosis in breast cancer. In the present study, we used tissue microarrays of 80 low-grade (WHO Grade II) and 98 high-grade human gliomas (WHO Grades III and IV) to investigate the presence of the ESC-related proteins Nanog, Klf4, Oct4, Sox2 and c-Myc by immunohistochemistry. While similar patterns of co-expressed proteins between low- and high-grade gliomas were present, we found up-regulated protein levels of Nanog, Klf4, Oct4 and Sox2 in high-grade gliomas. Survival analysis by Kaplan-Meier analysis revealed a significant shorter survival in the subgroups of low-grade astrocytomas (n = 42) with high levels of Nanog protein (p = 0.0067) and of Klf4 protein (p = 0.0368), in high-grade astrocytomas (n = 85) with high levels of Nanog (p = 0.0042), Klf4 (p = 0.0447), and c-Myc (p = 0.0078) and in glioblastomas only (n = 71) with high levels of Nanog (p = 0.0422) and of c-Myc (p = 0.0256). In the multivariate model, Nanog was identified as an independent prognostic factor in the subgroups of low-grade astrocytomas (p = 0.0039), high-grade astrocytomas (p = 0.0124) and glioblastomas only (p = 0.0544), together with established clinical variables in these tumors. These findings provide further evidence for the joint regulatory pathways of ESC-related proteins in gliomas and identify Nanog as one of the key players in determining clinical outcome of human astrocytomas.


Subject(s)
Astrocytoma/chemistry , Brain Neoplasms/chemistry , Embryonic Stem Cells/chemistry , Homeodomain Proteins/analysis , Adult , Aged , Aged, 80 and over , Astrocytoma/mortality , Brain Neoplasms/mortality , Female , Humans , Immunohistochemistry , Isocitrate Dehydrogenase/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/analysis , Male , Middle Aged , Nanog Homeobox Protein , Proto-Oncogene Proteins c-myc/analysis , Tissue Array Analysis
5.
Cancer Metastasis Rev ; 31(3-4): 793-805, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22733308

ABSTRACT

The homeobox gene PROX1 is critical for organ development during embryogenesis. The Drosophila homologue, known as prospero has been shown to act as a tumor suppressor by controlling asymmetric cell division of neuroblasts. Likewise, alterations in PROX1 expression and function are associated with a number of human cancers including hematological malignancies, carcinomas of the pancreas, liver and the biliary system, sporadic breast cancer, Kaposiform hemangioendothelioma, colon cancer, and brain tumors. PROX1 is involved in cancer development and progression and has been ascribed both tumor suppressive and oncogenic properties in a variety of different cancer types. However, the exact mechanisms through which PROX1 regulates proliferation, migration, and invasion of cancer cells are by large unknown. This review provides an update on the role of PROX1 in organ development and on its emerging functions in cancer, with special emphasis on the central nervous system and glial brain tumors.


Subject(s)
Embryonic Development , Homeodomain Proteins/physiology , Neoplasms/etiology , Tumor Suppressor Proteins/physiology , Amino Acid Sequence , Animals , Drosophila Proteins/physiology , Eye/embryology , Homeodomain Proteins/chemistry , Humans , Liver/embryology , Molecular Sequence Data , Neoplasm Metastasis , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/chemistry
6.
PLoS One ; 7(5): e37041, 2012.
Article in English | MEDLINE | ID: mdl-22615883

ABSTRACT

GABA (γ-aminobutyric acid) is the main inhibitory neurotransmitter in the CNS and is present in high concentrations in presynaptic terminals of neuronal cells. More recently, GABA has been ascribed a more widespread role in the control of cell proliferation during development where low concentrations of extrasynaptic GABA induce a tonic activation of GABA receptors. The GABA-A receptor consists of a ligand-gated chloride channel, formed by five subunits that are selected from 19 different subunit isoforms. The functional and pharmacological properties of the GABA-A channels are dictated by their subunit composition. Here we used qRT-PCR to compare mRNA levels of all 19 GABA-A channel subunits in samples of human glioma (n = 29) and peri-tumoral tissue (n = 5). All subunits except the ρ1 and ρ3 subunit were consistently detected. Lowest mRNA levels were found in glioblastoma compared to gliomas of lower malignancy, except for the θ subunit. The expression and cellular distribution of the α1, γ1, ρ2 and θ subunit proteins was investigated by immunohistochemistry on tissue microarrays containing 87 gliomas grade II. We found a strong co-expression of ρ2 and θ subunits in both astrocytomas (r = 0.86, p<0.0001) and oligodendroglial tumors (r = 0.66, p<0.0001). Kaplan-Meier analysis and Cox proportional hazards modeling to estimate the impact of GABA-A channel subunit expression on survival identified the ρ2 subunit (p = 0.043) but not the θ subunit (p = 0.64) as an independent predictor of improved survival in astrocytomas, together with established prognostic factors. Our data give support for the presence of distinct GABA-A channel subtypes in gliomas and provide the first link between specific composition of the A-channel and patient survival.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Receptors, GABA/biosynthesis , gamma-Aminobutyric Acid/biosynthesis , Adult , Brain Neoplasms/genetics , Chloride Channels/genetics , Chloride Channels/metabolism , Female , Glioma/genetics , Humans , Kaplan-Meier Estimate , Male , Neoplasm Staging , Prognosis , Protein Isoforms , Protein Subunits , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, GABA/genetics , Retrospective Studies , Survival Rate , gamma-Aminobutyric Acid/genetics
7.
J Neuropathol Exp Neurol ; 69(2): 129-38, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20084020

ABSTRACT

PROX1 is a prospero-related transcription factor that plays a critical role in the development of various organs including the mammalian lymphatic and central nervous systems; it controls cell proliferation and differentiation through different transcription pathwaysand has both oncogenic and tumor-suppressive functions. We investigated PROX1 expression patterns in 56 human astrocytic gliomas of different grades using immunohistochemistry. An average of 79% of cells in World Health Organization Grade IV (glioblastoma, n = 15) and 57% of cells in World Health Organization Grade III (anaplastic astrocytoma, n = 13) were strongly PROX1 positive; low-grade diffuse astrocytomas (Grade II, n = 13) had 21% of cells that were strongly positive; Grade I tumors (n = 15) had 1.5%; and non-neoplastic brain tissue (n = 15) had 3.7% of cells that were PROX1 positive. Double immunolabeling showed that PROX1+ cells in glioblastomas frequently coexpressed early neuronal proteins MAP2 and betaIII-tubulin but not the mature neuronal marker protein NeuN. Analyses of coexpression with proliferation markers suggest that PROX1+ cells have a marginally lower rate of proliferation than other tumor cells but are still mitotically active. We conclude that PROX1 may constitute a useful tool for the diagnosis and grading ofastrocytic gliomas and for distinguishing Grade III and Grade IV tumors from Grade I and Grade II tumors.


Subject(s)
Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Homeodomain Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Antigens, Nuclear/metabolism , Astrocytoma/blood supply , Biomarkers/metabolism , Brain Diseases/metabolism , Brain Neoplasms/blood supply , Cell Proliferation , Humans , Immunohistochemistry , Microtubule-Associated Proteins/metabolism , Microvessels/metabolism , Mitosis , Nerve Tissue Proteins/metabolism , Tubulin/metabolism
8.
J Pathol ; 219(2): 263-74, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19718706

ABSTRACT

Transcriptional regulation by p53 is critical for p53-mediated tumour suppression; however, p53-mediated transactivation has been dissociated from p53-mediated biological processes including apoptosis, DNA repair, and differentiation. We compared the effects of a mutant allele, p53(QS - val135), containing a double mutation in the amino-terminus abrogating transactivation activity and a modification at amino acid 135 partially affecting DNA binding, to complete loss of p53. We applied in vitro endpoints correlated with epithelial tumourigenesis and an in vivo assay of tumour phenotype to assess whether loss of p53-mediated transcriptional regulation underlies the malignant phenotype of p53(-/-)/v-ras(Ha)-overexpressing keratinocytes. Transactivation deficiency of p53QS-val135 was confirmed by reporter gene assays in fibroblasts and differentiating keratinocytes. Ras oncogene-induced senescence was lost in both p53(QS - val135/QS - val135) and p53(-/-) keratinocytes. Similarly, p53(QS - val135/QS - val135), like p53(-/-), cooperated with v-ras(Ha) to enhance malignant conversion. The tumours arising in p53(QS - val135/QS - val135) keratinocytes displayed strong nuclear p53 expression; thus, the p53(QS - val135) allele was maintained and the deficient transactivation function of the expressed p53QS mutant protein was supported by absence of p21(waf1) in these tumours. The p53(QS - val135) allele did not confer a dominant-negative phenotype, as p53(+/QS - val135) keratinocytes senesced normally in response to v-ras(Ha) expression and formed benign tumours. While p53(-/-) keratinocytes displayed diminished response to TGF-beta, p53(QS - val135/QS - val135) and p53(+/+) keratinocytes responded equivalently, indicating that the requirement for p53 in maximizing TGF-beta-mediated growth regulation is independent of its transactivation domain and that the ability of keratinocytes to respond to TGF-beta is insufficient to suppress the malignant phenotype in this model. Furthermore, TGF-beta enhances p53QS-induced activation of a dual p53-TGF-beta responsive reporter in a keratinocyte cell line. These findings support an essential role for p53-mediated transcriptional regulation in suppressing malignancies arising from ras-induced skin tumours, consistent with previous findings in spontaneous carcinogenesis in other organs, and highlight the potential importance of senescence for tumour suppression in vivo.


Subject(s)
Carcinoma, Squamous Cell/prevention & control , Cell Transformation, Neoplastic/genetics , Genes, p53/genetics , Skin Neoplasms/prevention & control , Transforming Growth Factor beta/physiology , Animals , Carcinoma, Squamous Cell/genetics , Cells, Cultured , Cellular Senescence/genetics , Genotype , Keratinocytes/transplantation , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Skin Neoplasms/genetics , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...