Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 229
Filter
1.
Leukemia ; 29(3): 677-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25189415

ABSTRACT

To decipher the mutational pattern of primary CNS lymphoma (PCNSL), we performed whole-exome sequencing to a median coverage of 103 × followed by mutation verification in 9 PCNSL and validation using Sanger sequencing in 22 PCNSL. We identified a median of 202 (range: 139-251) potentially somatic single nucleotide variants (SNV) and 14 small indels (range: 7-22) with potentially protein-changing features per PCNSL. Mutations affected the B-cell receptor, toll-like receptor, and NF-κB and genes involved in chromatin structure and modifications, cell-cycle regulation, and immune recognition. A median of 22.2% (range: 20.0-24.7%) of somatic SNVs in 9 PCNSL overlaps with the RGYW motif targeted by somatic hypermutation (SHM); a median of 7.9% (range: 6.2-12.6%) affects its hotspot position suggesting a major impact of SHM on PCNSL pathogenesis. In addition to the well-known targets of aberrant SHM (aSHM) (PIM1), our data suggest new targets of aSHM (KLHL14, OSBPL10, and SUSD2). Among the four most frequently mutated genes was ODZ4 showing protein-changing mutations in 4/9 PCNSL. Together with mutations affecting CSMD2, CSMD3, and PTPRD, these findings may suggest that alterations in genes having a role in CNS development may facilitate diffuse large B-cell lymphoma manifestation in the CNS. This may point to intriguing mechanisms of CNS tropism in PCNSL.


Subject(s)
Central Nervous System Neoplasms/genetics , Exome , Lymphoma, Large B-Cell, Diffuse/genetics , Polymorphism, Genetic , Somatic Hypermutation, Immunoglobulin , Adult , Aged , Central Nervous System Neoplasms/pathology , Female , Genetic Loci , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin Heavy Chains/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Middle Aged , Proto-Oncogene Proteins c-pim-1/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptors, Steroid/genetics , Retrospective Studies
2.
Oncogene ; 31(44): 4698-708, 2012 11 01.
Article in English | MEDLINE | ID: mdl-22249262

ABSTRACT

The Notch1-mediated signaling pathway has a central role in the maintenance of neural stem cells and contributes to growth and progression of glioblastomas, the most frequent malignant brain tumors in adults. Here, we demonstrate that the Notch1 receptor promotes survival of glioblastoma cells by regulation of the anti-apoptotic Mcl-1 protein. Notch1-dependent regulation of Mcl-1 occurs cell type dependent at a transcriptional or post-translational level and is mediated by the induction of epidermal growth factor receptor (EGFR). Inhibition of the Notch1 pathway overcomes apoptosis resistance and sensitizes glioblastoma cells to apoptosis induced by ionizing radiation, the death ligand TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) or the Bcl-2/Bcl-XL inhibitor ABT-737. In conclusion, targeting Notch1 might represent a promising novel strategy in the treatment of glioblastomas.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Receptor, Notch1/metabolism , Signal Transduction , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-raf/metabolism , RNA Processing, Post-Transcriptional , Receptor, Notch1/genetics , Transcription, Genetic
3.
Leukemia ; 25(12): 1797-807, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21818113

ABSTRACT

Recent studies addressing the molecular characteristics of PCNSL, which is defined as malignant B-cell lymphoma with morphological features of DLBCL, have significantly improved our understanding of the pathogenesis of this lymphoma entity, which is associated with an inferior prognosis as compared with DLBCL outside the CNS. This unfavorable prognosis stimulated intense efforts to improve therapy and induced recent series of clinical studies, which addressed the role of radiotherapy and various chemotherapeutic regimens. This review combines the discussion of diagnosis, differential diagnosis and recent progress in studies addressing the molecular pathogenesis as well as therapeutic options in PCNSL.


Subject(s)
Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/therapy , Humans
4.
Leukemia ; 23(10): 1875-84, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19494841

ABSTRACT

To determine the pattern of genetic alterations in primary central nervous system lymphomas (PCNSL), 19 PCNSL were studied by high-density single-nucleotide polymorphism arrays. Recurrent losses involved 6p21.32, 6q21, 8q12-12.2, 9p21.3, 3p14.2, 4q35.2, 10q23.21 and 12p13.2, whereas gains involved 18q21-23, 19q13.31, 19q13.43 and the entire chromosomes X and 12. Partial uniparental disomies (pUPDs) were identified in 6p and 9p21.3. These genomic alterations affected the HLA locus, the CDKN2A/p16, CDKN2B/p15 and MTAP, as well as the PRDM1, FAS, MALT1, and BCL2 genes. Increased methylation values of the CDKN2A/p16 promoter region were detected in 75% (6/8) PCNSL. Gene expression profiling showed 4/21 (20%) minimal common regions of imbalances to be associated with a differential mRNA expression affecting the FAS, STAT6, CD27, ARHGEF6 and SEPT6 genes. Collectively, this study unraveled novel genomic imbalances and pUPD with a high resolution in PCNSL and identified target genes of potential relevance in the pathogenesis of this lymphoma entity.


Subject(s)
Brain Neoplasms/genetics , Chromosome Aberrations , Chromosomes, Human/genetics , Uniparental Disomy , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , DNA Methylation , Female , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide/genetics
5.
Oncogene ; 27(52): 6646-56, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18663354

ABSTRACT

Defects in the apoptotic signaling cascades contribute to the poor therapeutic response of malignant gliomas. As glioblastomas are characterized by high expression levels of anti-apoptotic Bcl-2 family proteins, we studied the effects of the novel Bcl-2 inhibitor, ABT-737, on malignant glioma cells. ABT-737 treatment released the pro-apoptotic Bax protein from its binding partner Bcl-2 and potently induced apoptotic cell death in glioblastoma cells in vitro and in vivo. The local administration of ABT-737 prolonged the survival in an intracranial glioma xenograft model. Downregulation of Mcl-1 and overexpression of Bcl-2 sensitized the cells to ABT-737-mediated apoptosis. Moreover, ABT-737 potentiated the cytotoxicity of the chemotherapeutic drugs vincristine and etoposide, and of the death ligand TRAIL. As glioma stem cells may play a crucial role for the tumor progression and the resistance to treatment in glioblastomas, we investigated the effects of ABT-737 on the subpopulation of glioma cells exhibiting stem cell characteristics. Inhibition of proliferation and induction of apoptosis by ABT-737 were less efficient in glioma stem cells than in non-stem cell-like glioma cells. As the resistance of glioma stem cells was associated with high Mcl-1 expression levels, ABT-737 treatment combined with downregulation of Mcl-1 could represent a promising novel approach in glioblastoma treatment.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Glioblastoma/genetics , Humans , Mice , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Piperazines/pharmacology , Protein Binding , Proto-Oncogene Proteins c-bcl-2/classification , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Survival Rate , TNF-Related Apoptosis-Inducing Ligand/metabolism , Xenograft Model Antitumor Assays
6.
Oncogene ; 27(8): 1155-66, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-17700518

ABSTRACT

PEA-15 (phosphoprotein enriched in astrocytes 15 kDa) is a death effector domain-containing protein, which is involved in the regulation of apoptotic cell death. Since PEA-15 is highly expressed in cells of glial origin, we studied the role of PEA-15 in human malignant brain tumors. Immunohistochemical analysis of PEA-15 expression shows strong immunoreactivity in astrocytomas and glioblastomas. Phosphorylation of PEA-15 at Ser(116) is found in vivo in perinecrotic areas in glioblastomas and in vitro after glucose deprivation of glioblastoma cells. Overexpression of PEA-15 induces a marked resistance against glucose deprivation-induced apoptosis, whereas small interfering RNA (siRNA)-mediated downregulation of endogenous PEA-15 results in the sensitization to glucose withdrawal-mediated cell death. This antiapoptotic activity of PEA-15 under low glucose conditions depends on its phosphorylation at Ser(116). Moreover, siRNA-mediated knockdown of PEA-15 abolishes the tumorigenicity of U87MG glioblastoma cells in vivo. PEA-15 regulates the level of phosphorylated extracellular-regulated kinase (ERK)1/2 in glioblastoma cells and the PEA-15-dependent protection from glucose deprivation-induced cell death requires ERK1/2 signaling. PEA-15 transcriptionally upregulates the Glucose Transporter 3, which is abrogated by the inhibition of ERK1/2 phosphorylation. Taken together, our findings suggest that Ser(116)-phosphorylated PEA-15 renders glioma cells resistant to glucose deprivation-mediated cell death as encountered in poor microenvironments, for example in perinecrotic areas of glioblastomas.


Subject(s)
Apoptosis/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Glioblastoma/enzymology , Glucose/deficiency , Intracellular Signaling Peptides and Proteins/physiology , MAP Kinase Signaling System/physiology , Phosphoproteins/physiology , Animals , Apoptosis Regulatory Proteins , Brain Neoplasms/enzymology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Glioblastoma/metabolism , Glioblastoma/pathology , Glucose/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Nude , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Phosphorylation
7.
Leukemia ; 22(2): 400-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17989719

ABSTRACT

To characterize the molecular origin of primary lymphomas of the central nervous system (PCNSL), 21 PCNSLs of immunocompetent patients were investigated by microarray-based gene expression profiling. Comparison of the transcriptional profile of PCNSL with various normal and neoplastic B-cell subsets demonstrated PCNSL (i) to display gene expression patterns most closely related to late germinal center B cells, (ii) to display a gene expression profile similar to systemic diffuse large B-cell lymphomas (DLBCLs) and (iii) to be in part assigned to the activated B-cell-like (ABC) or the germinal center B-cell-like (GCB) subtype of DLBCL.


Subject(s)
B-Lymphocytes/pathology , Central Nervous System Neoplasms/genetics , Gene Expression Profiling , Germinal Center/pathology , Lymphoma/genetics , Aged , Aged, 80 and over , Central Nervous System Neoplasms/pathology , Female , Humans , Immunocompetence , Lymphoma/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Microarray Analysis , Middle Aged
8.
Oncogene ; 26(38): 5662-8, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17334394

ABSTRACT

In a genome-wide screen using differential methylation hybridization (DMH), we have identified a CpG island within the 5' region and untranslated first exon of the secretory granule neuroendocrine protein 1 gene (SGNE1/7B2) that showed hypermethylation in medulloblastomas compared to fetal cerebellum. Bisulfite sequencing and combined bisulfite restriction assay were performed to confirm the methylation status of this CpG island in primary medulloblastomas and medulloblastoma cell lines. Hypermethylation was detected in 16/23 (70%) biopsies and 7/8 (87%) medulloblastoma cell lines, but not in non-neoplastic fetal (n=8) cerebellum. Expression of SGNE1 was investigated by semi-quantitative competitive reverse transcription-polymerase chain reaction and found to be significantly downregulated or absent in all, but one primary medulloblastomas and all cell lines compared to fetal cerebellum. After treatment of medulloblastoma cell lines with 5-aza-2'-deoxycytidine, transcription of SGNE1 was restored. No mutation was found in the coding region of SGNE1 by single-strand conformation polymorphism analysis. Reintroduction of SGNE1 into the medulloblastoma cell line D283Med led to a significant growth suppression and reduced colony formation. In summary, we have identified SGNE1 as a novel epigenetically silenced gene in medulloblastomas. Its frequent inactivation, as well as its inhibitory effect on tumor cell proliferation and focus formation strongly argues for a significant role in medulloblastoma development.


Subject(s)
Cerebellar Neoplasms/pathology , DNA Methylation , Medulloblastoma/pathology , Neuroendocrine Secretory Protein 7B2/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/genetics , CpG Islands/genetics , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Down-Regulation/drug effects , Down-Regulation/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medulloblastoma/genetics , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription, Genetic/drug effects
9.
Oncogene ; 26(7): 1088-97, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-16909113

ABSTRACT

The molecular pathogenesis of pleomorphic xanthoastrocytoma (PXA), a rare astrocytic brain tumor with a relatively favorable prognosis, is still poorly understood. We characterized 50 PXAs by comparative genomic hybridization (CGH) and found the most common imbalance to be loss on chromosome 9 in 50% of tumors. Other recurrent losses affected chromosomes 17 (10%), 8, 18, 22 (4% each). Recurrent gains were identified on chromosomes X (16%), 7, 9q, 20 (8% each), 4, 5, 19 (4% each). Two tumors demonstrated amplifications mapping to 2p23-p25, 4p15, 12q13, 12q21, 21q21 and 21q22. Analysis of 10 PXAs with available high molecular weight DNA by high-resolution array-based CGH indicated homozygous 9p21.3 deletions involving the CDKN2A/p14(ARF)/CDKN2B loci in six tumors (60%). Interphase fluorescence in situ hybridization to tissue sections confirmed the presence of tumor cells with homozygous 9p21.3 deletions. Mutational analysis of candidate genes on 9q, PTCH and TSC1, revealed no mutations in PXAs with 9q loss and no evidence of TSC1 promoter methylation. However, PXAs consistently showed low TSC1 transcript levels. Taken together, our study identifies loss of chromosome 9 as the most common chromosomal imbalance in PXAs and suggests important roles for homozygous CDKN2A/p14(ARF)/CDKN2B deletion as well as low TSC1 mRNA expression in these tumors.


Subject(s)
Astrocytoma/genetics , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Deletion , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Proteins/deficiency , Adolescent , Adult , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p15/deficiency , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Female , Homozygote , Humans , Male , Middle Aged , RNA, Messenger/biosynthesis , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
10.
Verh Dtsch Ges Pathol ; 91: 294-301, 2007.
Article in German | MEDLINE | ID: mdl-18314627

ABSTRACT

AIMS: Induction of apoptosis by the death ligand TRAIL might be a promising therapeutic approach in colorectal cancer therapy. However, some colon cancer cells are resistant to TRAIL because of the expression of anti-apoptotic proteins, such as FLIP. We studied the role of FLIP for apoptosis resistance in colon cancer and developed an approach to overcome the resistance to TRAIL. METHODS: The mechanisms of TRAIL-induced cell death in colon cancer cells were studied by Western blot analysis, apoptosis assays, transient and stable transfections, siRNA-mediated knockdown, and FACS analysis. RESULTS: The anti-apoptotic protein FLIP is expressed in the majority of colon carcinoma. Stable over-expression of FLIP renders colon carcinoma cells resistant to the death ligand, TRAIL. siRNA-mediated down-regulation of FLIP sensitizes the cells to TRAIL-induced apoptosis. FLIP-expressing colon cancer cells can be sensitized to TRAIL-induced apoptosis by the anti-diabetic drug troglitazone. Troglitazone induces a pronounced reduction in protein expression levels of FLIP. The troglitazone-dependent down-regulation of FLIP occurs on a post-translational level and involves the accelerated FLIP degradation by the proteasome. Moreover, troglitazone suppresses the expression of the anti-apoptotic protein, survivin, and induces the cell surface expression of the TRAIL receptor 2. CONCLUSIONS: The anti-apoptotic FLIP protein plays an important role in apoptosis resistance of colon carcinoma cells. Troglitazone down-regulates FLIP and sensitizes the cells to TRAIL-induced apoptosis. A combined treatment with troglitazone and TRAIL might be a promising experimental therapy for some forms of colorectal cancer because troglitazone sensitizes tumor cells to TRAIL-induced apoptosis via various mechanisms, thereby minimizing the risk of acquired tumor cell resistance.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Chromans/pharmacology , Colonic Neoplasms/pathology , Hypoglycemic Agents/pharmacology , TNF-Related Apoptosis-Inducing Ligand/physiology , Thiazolidinediones/pharmacology , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/drug effects , Cell Death/drug effects , Cell Line, Tumor , Flow Cytometry , Humans , TNF-Related Apoptosis-Inducing Ligand/drug effects , Troglitazone
11.
Verh Dtsch Ges Pathol ; 91: 343-50, 2007.
Article in German | MEDLINE | ID: mdl-18314633

ABSTRACT

PEA-15 (Phosphoprotein enriched in astrocytes 15 kD) is a death effector domain-containing protein, which is involved in the regulation of apoptotic cell death. Since PEA-15 is highly expressed in cells of glial origin, we studied the role of PEA-15 in human malignant brain tumors. Immunohistochemical analysis of PEA-15 expression shows strong immunoreactivity in astrocytomas and glioblastomas. Phosphorylation of PEA-15 at Ser116 is found in vivo in perinecrotic areas in glioblastomas and in vitro after glucose deprivation of glioblastoma cells. Overexpression of PEA-15 induces a marked resistance against glucose deprivation-induced apoptosis, whereas siRNA-mediated down-regulation of endogenous PEA-15 results in the sensitization to glucose withdrawal-mediated cell death. This anti-apoptotic activity of PEA-15 under low glucose conditions depends on its phosphorylation at Ser116 Moreover, siRNA-mediated knockdown of PEA-15 abolishes the tumorigenicity of U87MG glioblastoma cells in vivo. PEA-15 regulates the level of phosphorylated ERK1/2 in glioblastoma cells and the PEA-15-dependent protection from glucose deprivation-induced cell death requires ERK1/2 signaling. PEA-15 transcriptionally up-regulates the glucose transporter 3, which is abrogated by the inhibition of ERK1/2 phosphorylation. Taken together, our findings suggest that Ser116-phosphorylated PEA-15 renders glioma cells resistant to glucose deprivation-mediated cell death as encountered in poor microenvironments, e.g. in perinecrotic areas of glioblastomas.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Glucose/physiology , Intracellular Signaling Peptides and Proteins/physiology , Phosphoproteins/physiology , Apoptosis Regulatory Proteins , Astrocytes/cytology , Astrocytes/pathology , Astrocytoma/pathology , Brain Neoplasms/pathology , Cell Death , Cell Line, Tumor , Glioblastoma/pathology , Humans
12.
Neuropathol Appl Neurobiol ; 32(5): 517-24, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16972885

ABSTRACT

Cytogenetic and molecular genetic studies have shown frequent losses on the long arm of chromosome 14 in different types of human gliomas. Using differential methylation hybridization as a genome-wide screening approach to determine DNA methylation patterns in gliomas, we recently identified two DNA fragments in 14q23.1 (CGI-clone musical sharp396) and 14q32.12 (CGI-clone musical sharp519) that were differentially methylated between astrocytic gliomas and mixed oligoastrocytomas. To validate this observation, we examined these 14q32.12 locus for methylation in an extended series of 43 astrocytic and oligodendroglial gliomas. All tumours were additionally investigated for loss of heterozygosity (LOH). Microsatellite analysis showed LOH in seven of 28 (25%) oligodendroglial tumours and three of 15 (20%) astrocytic tumours. Seven tumours demonstrated LOH at all informative 14q loci whereas three tumours carried partial deletions defining a commonly deleted region at 14q22.3-q32.1 between the microsatellite markers D14S282 and D14S995. Methylation-specific PCR analysis of the 14q32.12 locus revealed hypermethylation in 12 of 43 gliomas (28%). Hypermethylation was restricted to tumours with oligodendroglial differentiation (12 of 28 tumours, 43%). However, none of the hypermethylated tumours demonstrated LOH on 14q and vice versa. In total, 19 of 28 oligodendroglial tumours (68%) showed either hypermethylation at the 14q32.12 locus or LOH at 14q22.3-q32.2. Taken together, our data lend further support for the location of one or more yet to be identified glioma-associated tumour suppressor gene(s) on 14q. In addition, the restriction of 14q32.12 methylation to oligodendroglial tumours suggests a role for epigenetic DNA modifications in these particular gliomas.


Subject(s)
Brain Neoplasms/pathology , Chromosomes, Human, Pair 14/genetics , DNA Methylation , Oligodendroglia/pathology , Adolescent , Adult , Aged , Alleles , Child , Child, Preschool , DNA, Neoplasm/drug effects , Female , Humans , Male , Microsatellite Repeats/genetics , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Sulfites/pharmacology
13.
Neuropathol Appl Neurobiol ; 31(3): 270-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15885064

ABSTRACT

Argyrophilic grain disease (AGD) constitutes a neurodegenerative disorder that occurs in the brains of the elderly and affects 5% of all patients with dementia. Tau protein-containing lesions known as argyrophilic grains and located predominantly in limbic regions of the brain characterize this disease. Dementia is encountered in only a subset of cases that display the morphological pattern of AGD. The aim of this study is to determine the role of concurrent Alzheimer's disease (AD)-related pathology for the development of dementia in AGD patients. A total of 204 post-mortem brains from 30 demented and 49 nondemented AGD patients, 39 AD patients, and from 86 nondemented controls without AGD were staged for AD-related neurofibrillary tangles (NFTs) as well as amyloid beta-protein (Abeta) deposition. To identify differences in AD-related pathology between demented and nondemented AGD cases, and to differentiate the pattern of AD-related changes in demented and nondemented AGD cases from that seen in AD and nondemented controls, we statistically compared the stages of Abeta and NFT distribution among these groups. Using a logistic regression model, we showed that AGD has a significant effect on the development of dementia beyond that attributable to AD-related pathology (P < 0.005). Demented AGD cases showed lower stages of AD-related pathology than did pure AD cases but higher stages than nondemented AGD patients. AGD associated dementia was seen in the presence of NFT (Braak)-stages II-IV and Abeta-phases 2-3, whereas those stages were not associated with dementia in the absence of AGD. In conclusion, AGD is a clinically relevant neurodegenerative entity that significantly contributes to the development of dementia by lowering the threshold for cognitive deficits in the presence of moderate amounts of AD-related pathology.


Subject(s)
Alzheimer Disease/complications , Brain/pathology , Inclusion Bodies/pathology , Neurodegenerative Diseases/complications , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Female , Humans , Logistic Models , Male , Middle Aged , Neurodegenerative Diseases/pathology , Neurofibrillary Tangles/pathology
14.
Neuropediatrics ; 36(2): 71-7, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15822019

ABSTRACT

PURPOSE: The aim of the present study was to evaluate survival and factors influencing long-term outcome of patients with AFP/beta-HCG secreting (non-seminomatous) central nervous system germ cell tumors (secCNSGCT), who were prospectively collected in the cooperative MAKEI (German: maligne Keimzelltumoren) 89 protocol. PATIENTS AND METHODS: Between January 1989 and January 1994, 28 patients with secCNS GCT were registered and treated according to the MAKEI 89 protocol. The protocol recommended, after a clinically or histologically proven diagnosis and cisplatin-based chemotherapy, a resection of residual tumor and craniospinal irradiation (30 Gy) with a tumor boost (20 Gy). RESULTS: The estimated (Kaplan-Meier) event-free survival (EFS) of protocol patients is 0.57 +/- 0.09 (n = 28) and the relapse-free survival (RFS) is 0.67 +/- 0.10 (at five and ten years). With respect to long-term survival, the combination of marked neurological symptoms at diagnosis along with primary tumor resection seem to be the main negative prognostic risk factors (Fisher exact test p < 0.05). CNS dissemination at diagnosis can also be considered as a negative risk factor as 3 of 5 patients with primary dissemination died of the disease. CONCLUSION: Cisplatin-based three agent chemotherapy followed by resection of the residual tumor and craniospinal irradiation (CSI) with tumor boost is a successful and well-tolerated treatment for secCNSGCTs. The possibility of a clinical diagnosis based on MRI and tumor markers together with the use of modern neurosurgical techniques gives us the chance to postpone or even avoid major surgery. This gives an additional chance to reduce acute morbidity and further decrease late effects.


Subject(s)
Central Nervous System Neoplasms/mortality , Central Nervous System Neoplasms/surgery , Evaluation Studies as Topic , Germinoma/mortality , Germinoma/surgery , Adolescent , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Biomarkers, Tumor , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/physiopathology , Child , Child, Preschool , Cisplatin/therapeutic use , Cognition/physiology , Combined Modality Therapy/methods , Cranial Irradiation , Female , Germinoma/drug therapy , Germinoma/physiopathology , Hexachlorocyclohexane/metabolism , Humans , Longitudinal Studies , Male , Prognosis , Prospective Studies , Retrospective Studies , Survival Rate , Time , Time Factors , Treatment Outcome , alpha-Fetoproteins/metabolism
15.
Neuropathol Appl Neurobiol ; 30(5): 532-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15488029

ABSTRACT

The sonic hedgehog (Shh) and the Wnt signalling pathways are involved in the development of medulloblastomas (MBs), the most frequent malignant brain tumours in children. Components of these two developmental and cancer-associated pathways, including (Patched) PTCH, SMOH, adenomatous polyposis coli (APC), beta-catenin and AXIN1 show somatic mutations in sporadic MBs. In this study we analysed SUFU (human Suppressor of Fused), which acts as a negative regulator of both the Shh and Wnt signalling pathways and therefore represents a putative tumour suppressor gene, to find out if it is also involved in the pathogenesis of sporadic MBs. We screened 145 primitive neuroectodermal tumours (PNETs) including 90 classic MBs, 42 of the desmoplastic variant and two medullomyoblastomas as well as 11 MB cell lines for mutations using single-strand conformational polymorphism (SSCP) and sequencing analysis. 18% of the MBs exhibited allelic losses on chromosome 10q. In contrast to a previous report, in which truncating mutations of SUFU have been identified in 9% of MBs, we were not able to identify somatic mutations of SUFU in our large tumour panel. We uncovered single nucleotide polymorphisms (SNPs) in exon 4, 8, 11 and in intron 2 in the SUFU gene. Expression analysis by competitive reverse transcription-polymerase chain reaction (RT-PCR) revealed no difference in SUFU mRNA levels of both MB subtypes and normal foetal or adult cerebellar tissues. Our results indicate that genetic alterations of the SUFU gene, do not contribute significantly to the molecular pathogenesis of MBs.


Subject(s)
Brain Neoplasms/genetics , DNA, Neoplasm/genetics , Neuroectodermal Tumors, Primitive/genetics , Repressor Proteins/genetics , Adolescent , Adult , Base Sequence , Cell Line, Tumor , Child , Child, Preschool , DNA Mutational Analysis , DNA Primers , Humans , Infant , Infant, Newborn , Middle Aged , Mutation , Polymorphism, Single-Stranded Conformational , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Neurology ; 63(1): 167-9, 2004 Jul 13.
Article in English | MEDLINE | ID: mdl-15249632

ABSTRACT

Comparative investigation of immunoglobulin (Ig) heavy chain gene rearrangements and DNA sequence analyses of a primary lymphoma of the CNS (PCNSL) and its recurrence revealed that both tumors used the same Ig gene segment. In addition to shared somatic mutations, the primary and the recurrent PCNSLs harbored somatic mutations unique to each tumor. Clonal evolution rather than subclone selection appears to underlie the development of tumor recurrence in this case.


Subject(s)
B-Lymphocytes/pathology , Clone Cells/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Supratentorial Neoplasms/pathology , Temporal Lobe/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Base Sequence , Breast Neoplasms/surgery , Combined Modality Therapy , Cytarabine/administration & dosage , DNA, Neoplasm/genetics , Embryonal Carcinoma Stem Cells , Female , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin , Germinal Center/pathology , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/surgery , Magnetic Resonance Imaging , Methotrexate/administration & dosage , Molecular Sequence Data , Neoplasm Recurrence, Local/drug therapy , Neoplasms, Second Primary/surgery , Parietal Lobe/pathology , Remission Induction , Sequence Alignment , Sequence Homology, Nucleic Acid , Supratentorial Neoplasms/drug therapy , Supratentorial Neoplasms/surgery , Temporal Lobe/surgery
17.
Klin Padiatr ; 216(3): 141-9, 2004.
Article in English | MEDLINE | ID: mdl-15175958

ABSTRACT

UNLABELLED: Malignant non-germinomatous intracranial germ cell tumors (MNGGCTs) are a heterogenous group of neoplastic lesions. Their treatment concept follows a multimodal concept that may include tumor resection for local tumor control, craniospinal irradiation to cover leptomenigeal tumor spread and chemotherapy to eliminate systemic tumor dissemination. A Platinum-based chemotherapy proven to be highly effective in testicular and non-testicular malignant germ cell tumors in adults as well as in children has also been chosen for intracranial sites. While therapeutic concepts have been thoroughly evaluated for children and adolescents with extracranial nongonadal GCTs, no such detailed long term follow-up data are available for intracranial MNGGCTs. This paper reports on the long-term outcome of 41 patients with intracranial malignant non-germinomatous GCTs enrolled into the German prospective protocol MAKEI 89. The analysis focuses on the impact of surgery, radio- and chemotherapy. PATIENTS AND METHODS: Between January 1989 and January 1994, 41 patients with malignant intracranial non-germinomatous GCTs were registered. Patients were compared in respect to protocol (n = 27) and non-protocol treatment (n = 14). Estimated were with chi (2) and Fisher exact test the impact of surgery, chemotherapy and irradiation on outcome. RESULTS: The estimated (Kaplan-Meier) 5-year event free survival (EFS) of patients treated according to protocol recommendations was 0.59 +/- 0.06 (n = 27), compared to an EFS of 0.37 +/- 0.33 for patients with different treatments (n = 14) (p = 0.70, log-rank). The 5-year relapse-free survival rate (RFS) was 0.74 +/- 0.06 in protocol patients and 0.38 +/- 0.33 in non-protocol patients (median observation time of 112 months after diagnosis for surviving patients) (p = 0.14, log-rank). Surgery, complete or incomplete had no significant impact on survival (p = 0.12). Radiotherapy, in terms of craniospinal irradiation had a significant influence on survival (p = 0.035) as well as a cumulative cisplatin dose >/= 400 mg/m (2) (p = 0.002). CONCLUSION: Cisplatin chemotherapy and craniospinal irradiation with tumor boost are of significant influence on long term survival in patients with MNGGCTs. The exclusion of major surgery at diagnosis using modern advances in neurosurgery or related tumor resection after neoadjuvant chemotherapy will allow a further reduction of treatment related mortality and long lasting morbidity. The analysis reveals that, given effective treatment, intracranial malignant non-germinomatous GCTs should not longer carry a poor prognosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/surgery , Cisplatin/administration & dosage , Cranial Irradiation , Neoadjuvant Therapy , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/surgery , Pinealoma/surgery , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Chemotherapy, Adjuvant , Child , Child, Preschool , Cisplatin/adverse effects , Combined Modality Therapy , Disease-Free Survival , Female , Follow-Up Studies , Germany , Humans , Male , Neoplasms, Germ Cell and Embryonal/mortality , Neoplasms, Germ Cell and Embryonal/radiotherapy , Pinealoma/drug therapy , Pinealoma/mortality , Pinealoma/radiotherapy , Prognosis , Prospective Studies , Radiotherapy, Adjuvant , Survival Analysis
18.
Neuropathol Appl Neurobiol ; 30(3): 225-32, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15175076

ABSTRACT

Focal epilepsies in young patients are frequently associated with differentiated glioneuronal tumours. Dysplastic neurones represent a characteristic neuropathological feature of gangliogliomas, the most common entity encountered in this group. Here, we have analysed two major components of the reelin pathway involved in neuronal migration and cortical development, that is, p35 and disabled-1 (dab1), in gangliogliomas. Genomic structures of human dab1 and p35 were identified 'in silico' using the HTGS databank, NCBI BLAST 2.1. DNA sequence analysis was carried out in gangliogliomas obtained from 29 epilepsy patients vs. peripheral blood DNA from non-affected control individuals (n = 100). Gene expression of dab1 and p35 was determined by real-time RT-PCR (reverse transcriptase polymerase chain reaction) in gangliogliomas (n = 14) vs. non-neoplastic central nervous system tissue (n = 20). The human dab1 gene contains 13 coding exons and is located on chromosome 1p31-32. A single coding exon constitutes the human p35 gene, which is located on chromosome 17q11.2. A novel homologueous genomic region on chromosome 2 has to be taken into account for future studies on p35. One ganglioglioma patient showed a unique polymorphism in the p35 gene. The single base exchange (C to A) at nucleotide 904 of the p35 cDNA (GenBank X80343, start ATG, codon 302) results in a leucine-isoleucine amino acid substitution. No mutations of the dab1 and p35 genes in gangliogliomas were observed. However, significantly lower levels of dab1 and p35 gene transcripts were detected in gangliogliomas compared to controls (dab1 28.24%, t-test P < 0.001; p35 21.28%, t-test P < 0.001, in gangliogliomas vs. controls). Our data suggest that mutational events of dab1 and p35 are not involved in the molecular pathogenesis of gangliogliomas. A potential functional role of these developmentally regulated genes for the formation of epileptogenic glioneuronal lesions remains to be elucidated.


Subject(s)
Brain Neoplasms/genetics , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Ganglioglioma/genetics , Gene Expression Regulation, Neoplastic/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Adaptor Proteins, Signal Transducing , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion Molecules, Neuronal/metabolism , DNA Primers , Extracellular Matrix Proteins/metabolism , Ganglioglioma/metabolism , Ganglioglioma/pathology , Humans , Lasers , Polymorphism, Single-Stranded Conformational , Reelin Protein , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases
19.
Acta Neuropathol ; 105(4): 358-64, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12624789

ABSTRACT

Pleomorphic xanthoastrocytoma (PXA) is a rare, superficially located astrocytic glioma of children and young adults, which is associated with a relatively favorable prognosis. Here we report on the expression of the hematopoietic progenitor cell and vascular endothelial cell associated antigen CD34 in PXAs. We found CD34 immunoreactivity in various fractions of tumor cells in 44 of 60 PXAs investigated (73%). CD34 expression was more common in PXAs of WHO grade II (37 of 44 tumors, 84%) than in PXAs with anaplastic features (7 of 16 tumors, 44%). Immunoreactivity for CD34 was also commonly detected in single or clustered dysplastic neural cells within the cerebral cortex adjacent to the PXAs. Reverse transcription-PCR revealed that PXAs express the full-length CD34 transcript and a known splice variant encoding a truncated form of CD34. Both transcripts were detectable at higher levels in PXAs as compared to diffuse astrocytomas and non-neoplastic brain tissue. Taken together, our findings demonstrate that PXAs frequently express CD34 not only in vascular endothelial cells but also in tumor cells and in dysplastic cells of the adjacent cortex. Therefore, immunostaining for CD34 may be a helpful tool for the histological differential diagnosis of PXAs.


Subject(s)
Antigens, CD34/biosynthesis , Astrocytoma/metabolism , Brain Neoplasms/metabolism , Adolescent , Adult , Antigens, CD34/genetics , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Child , Child, Preschool , Diagnosis, Differential , Female , Glioma/pathology , Humans , Immunohistochemistry , Male , Middle Aged , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
20.
Pathologe ; 23(4): 260-83, 2002 Jul.
Article in German | MEDLINE | ID: mdl-12185780

ABSTRACT

New developments in neuro-oncology have prompted an update of the World Health Organization (WHO) classification of tumors of the nervous system. Major changes include the addition of new entities and the refinement of criteria for the diagnosis and grading of various neoplasms, in particular the meningiomas. As novel clinico-pathological entities, the chordoid glioma of the third ventricle, the atypical teratoid/rhabdoid tumor (AT/RT), the solitary fibrous tumor, and the perineurioma have been listed. The former lipomatous medulloblastoma of the cerebellum, previously incorporated in the family of embryonal tumors, is now classified as cerebellar liponeurocytoma. The term mixed pineocytoma/pineoblastoma has been replaced by pineal parenchymal tumor of intermediate differentiation. Furthermore, the large cell medulloblastoma and the tanycytic ependymoma were established as novel tumor variants. A separate chapter on the peripheral neuroblastic tumors has now been included in the classification. Substantial revisions were introduced in the meningioma chapter. For both atypical meningioma WHO grade II and anaplastic meningioma WHO grade III, histopathological criteria are now precisely defined. An important new addition to the WHO 2000 classification of nervous system tumors is the inclusion of molecular pathology findings. With this combination of pathology and genetics it has set the stage for a new format of the WHO tumor classification series.


Subject(s)
Nervous System Neoplasms/classification , Astrocytoma/pathology , Brain Neoplasms/pathology , Diagnosis, Differential , Humans , Meningioma/pathology , Nervous System Neoplasms/genetics , Nervous System Neoplasms/pathology , Peripheral Nervous System Neoplasms/pathology , World Health Organization
SELECTION OF CITATIONS
SEARCH DETAIL
...