Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Clin Genet ; 79(6): 546-53, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20618351

ABSTRACT

Beckwith­Wiedemann syndrome (BWS) is one of the most prevalent congenital disorders predominantly caused by epigenetic alterations. Here we present an extensive case study of a monozygotic monochorionic male twin pair discordant for BWS. Our analysis allows to correlate BWS symptoms, like a protruding tongue, indented ears and transient neonatal hypoglycaemia, to an abnormal methylation at the KvDMR1. DNAs extracted from peripheral blood, skin fibroblasts, saliva and buccal swab of both twins, their sister and parents were analysed at 11 differentially methylated regions (DMRs) including all four relevant DMRs of the BWS region. The KvDMR1 was exclusively found to be hypomethylated in all cell types of the affected BWS twin, while the unaffected twin and the relatives showed normal methylation in fibroblasts, buccal swab and saliva DNA. Interestingly, the twins share a common blood-specific hypomethylation phenotype most probably caused by a feto-fetal transfusion between both twins. Because microsatellite analysis furthermore revealed a normal biparental karyotype for chromosome 11, our results point to an exclusive correlation of the observed BWS symptoms to locally restricted epimutations at the KvDMR1 of the maternal chromosome.


Subject(s)
Beckwith-Wiedemann Syndrome/genetics , DNA Methylation , Genetic Loci , Genomic Imprinting , Twins, Monozygotic/genetics , Adult , Beckwith-Wiedemann Syndrome/metabolism , Child, Preschool , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/metabolism , Female , Genetic Association Studies , Genetic Markers , Humans , Infant, Newborn , Male , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Uniparental Disomy/genetics
2.
Neurology ; 60(8): 1348-50, 2003 Apr 22.
Article in English | MEDLINE | ID: mdl-12707441

ABSTRACT

Of 85 patients with ALS, the authors identified 3 patients with balanced translocations and 2 patients with pericentric inversions, all affecting distinct chromosomal loci. The high rate of constitutional aberrations (5.9%) suggests that ALS is, in part, associated with recombination-based rearrangements of genomic sequences.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Chromosome Disorders/genetics , Chromosome Inversion , Translocation, Genetic , Adult , Age of Onset , Aged , Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/epidemiology , Cells, Cultured/ultrastructure , Chromosome Banding , Chromosome Disorders/epidemiology , Dementia/complications , Dementia/genetics , Female , Germany/epidemiology , Humans , Karyotyping , Lymphocytes/ultrastructure , Male , Middle Aged , Phenotype
3.
Cytogenet Cell Genet ; 94(1-2): 1-8, 2001.
Article in English | MEDLINE | ID: mdl-11701945

ABSTRACT

Glioblastoma multiforme (GBM) is characterized by intratumoral heterogeneity as to both histomorphology and genetic changes, displaying a wide variety of numerical chromosome aberrations the most common of which are monosomy 10 and trisomy 7. Moreover, GBM in vitro are known to have variable karyotypes within a given tumor cell culture leading to rapid karyotype evolution through a high incidence of secondary numerical chromosome aberrations. The aim of our study was to investigate to what extent this mitotic instability of glioblastoma cells is also present in vivo. We assessed the spatial distribution patterns of numerical chromosome aberrations in vivo in a series of 24 GBM using two-color in situ hybridization for chromosomes 7/10, 8/17, and 12/18 on consecutive 6-microm paraffin-embedded tissue slides. The chromosome aberration patterns were compared with the histomorphology of the investigated tumor assessed from a consecutive HE-stained section, and with the in vitro karyotype of cell cultures established from the tumors. All investigated chromosomes showed mitotic instability, i.e., numerical aberrations within significant amounts of tumor cells in a scattered distribution through the tumor tissue. As to chromosomes 10 and 17, only monosomy occurred, as to chromosome 7 only trisomy/polysomy, apparently as a result of selection in favor of the respective aberration. Conversely, chromosomes 8, 12, and 18 displayed scattered patterns of monosomy as well as trisomy within a given tumor reflecting a high mitotic error rate without selective effects. The karyotypes of the tumor cell cultures showed less variability of numerical aberrations apparently due to clonal adaptation to in vitro conditions. We conclude that glioblastoma cells in vivo are characterized by an extensive tendency to mitotic errors. The resulting clonal diversity of chromosomally aberrant cells may be an important biological constituent of the well-known ability of glioblastomas to preserve viable tumor cell clones under adaptive stress in vivo, in clinical terms to rapidly recur after antitumoral therapy including radio- or chemotherapy.


Subject(s)
Chromosome Aberrations , Glioblastoma/genetics , Glioblastoma/pathology , Mitosis , Mutagenesis/genetics , Adult , Aged , Cell Size , Chromosome Banding , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Middle Aged , Monosomy/genetics , Trisomy/genetics , Tumor Cells, Cultured
4.
J Neurosurg ; 95(4): 601-7, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11596954

ABSTRACT

OBJECT: The goal of this study was to determine whether in meningiomas cytogenetic findings are suitable as a predictive parameter relevant to prognosis. METHODS: Between 1992 and 1998 at the Department of Neurosurgery, Saarland University, 198 patients underwent surgery to resect meningiomas. The meningiomas were investigated cytogenetically and the patients were followed up for a mean period of 33 months. On the basis of the cytogenetic findings, the meningiomas were subdivided into four groups: Group 0 meningiomas displayed a normal diploid chromosome set; Group 1 tumors were found to have monosomy 22 as the sole cytogenetic aberration; Group 2 tumors were markedly hypodiploid meningiomas with loss of additional autosomes in addition to monosomy 22; and Group 3 meningiomas had deletions of the short arm of a chromosome 1, as well as additional chromosomal aberrations including loss of one chromosome 22. One hundred ninety-eight patients in whom tumor resections were determined to be Simpson Grade I or II could be followed up after complete tumor extirpation. In 20 patients, one or several recurrences were documented during the period of observation. The tumors were classified according to their different, but mostly uniform chromosomal aberrations. Recurrences were found in six (4.3%) of 139 tumors in Groups 0 and 1 and in two (10.5%) of 19 tumors in Group 2; the highest rate of recurrence was found in 12 (30%) of 40 tumors in Group 3. This supports the notion that the deletion of the short arm of one chromosome 1 is an important prognostic factor in meningiomas. The results of this study document a significant correlation between histological grade (p < 0.0001), location (p < 0.0001), and recurrences of meningiomas (p < 0.0001) (significance determined using chi-square tests). CONCLUSIONS: The cytogenetic classification of meningiomas provides a significant contribution to the predictability of tumor recurrence and is, therefore, a valuable criterion for the neurosurgeon's postoperative management protocol.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 22 , Meningeal Neoplasms/genetics , Meningioma/genetics , Aged , Cytogenetic Analysis , Disease Progression , Female , Follow-Up Studies , Gene Deletion , Humans , Male , Meningeal Neoplasms/surgery , Meningioma/surgery , Middle Aged , Monosomy , Neoplasm Recurrence, Local/genetics , Predictive Value of Tests , Retrospective Studies
5.
Cytogenet Cell Genet ; 93(3-4): 207-20, 2001.
Article in English | MEDLINE | ID: mdl-11528114

ABSTRACT

Meningioma is the most frequent tumor of neuroectodermal origin in humans. It is usually benign. Only a minority of cases shows progression to an anaplastic tumor (WHO grade II and III). Meningioma is generally a sporadic tumor. Multiple and familial cases are rare and mostly associated with (hereditary) neurofibromatosis 2 (NF2). Meningiomas show an unexpectedly high recurrence rate. Also, completely removed low-grade tumors can recur. Recurrence and multiplicity are correlated with the formation of a peritumoral edema. On the cytogenetic level, meningioma is the best-studied tumor in humans. Grade I tumors show either uniform monosomy 22 or a diploid karyotype. The majority of high-grade, but only a minority of low-grade, meningiomas show loss of merlin, a cytoskeleton-cytoplasm-linker protein. Merlin is the product of the NF2 gene located on chromosome 22. A second tumor suppressor gene on chromosome 22 has not yet been detected. In contrast to other solid tumors, progression of meningiomas is correlated with increasing hypodiploidy, showing characteristic clonal evolutions that mostly include chromosomes 14, 18, and 19 and, more rarely, 6 and 10. Structural aberrations are infrequent, except for the loss of the short arm of chromosome 1, which appears to be the decisive step for anaplastic growth. Comparative histochemical and molecular cytogenetic studies point to the alkaline phosphatase gene (ALPL, liver-bone-kidney type) located on 1p36.1-->p34 as a candidate tumor suppressor gene. A model is proposed that tries to explain - with a minimum number of essential steps - the origin, progression, infiltration, and recurrence of meningiomas.


Subject(s)
Chromosome Aberrations/genetics , Meningioma/genetics , Meningioma/pathology , Models, Genetic , Alkaline Phosphatase/genetics , Biopsy , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 22/genetics , Clone Cells/metabolism , Clone Cells/pathology , Disease Progression , Genes, Tumor Suppressor/genetics , Genotype , Humans , Karyotyping , Membrane Proteins/genetics , Meningioma/enzymology , Meningioma/physiopathology , Monosomy/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local , Neurilemmoma/genetics , Neurilemmoma/pathology , Neurofibromin 2 , Phenotype
6.
Pathol Res Pract ; 197(6): 427-31, 2001.
Article in English | MEDLINE | ID: mdl-11432670

ABSTRACT

In a recent study, 23 microdissected areas of 10 glioblastoma multiforme (GBM) were investigated for quantitative genomic aberrations using comparative genomic hybridization (CGH). To validate the chromosomal aberrations, as revealed by CGH after microdissection, parallel tissue sections were stained immunohistochemically with an antibody that detects both wild-type epidermal growth factor receptor (EGFR) and the deletion mutant form of the receptor (EGFRvIII). Immunostaining was correlated with CGH data of chromosome 7, because chromosome 7 is the most frequently aberrant chromosome in GBM (here four of 10 tumors), and this aberration often indicates an abnormality of EGFR. Nine of nine areas that showed gain in or amplification (2 areas) of chromosome 7 with CGH contained EGFR-immunoreactive cells. Only three of 14 areas without abnormality of chromosome 7 in CGH contained EGFR-immunoreactive cells; eleven of 14 areas were immunonegative. Our findings demonstrate a strong correlation between immunohistochemistry of EGFR and the copy numbers of chromosome 7, as revealed by CGH after microdissection in glioblastoma multiforme.


Subject(s)
Brain Neoplasms/metabolism , Chromosome Aberrations , Chromosome Disorders , Chromosomes, Human, Pair 7 , ErbB Receptors/metabolism , Glioblastoma/metabolism , Adult , Aged , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA, Neoplasm/analysis , Dissection , Female , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunohistochemistry , Karyotyping , Male , Micromanipulation , Middle Aged , Nucleic Acid Hybridization
7.
Cancer Genet Cytogenet ; 121(2): 124-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11063794

ABSTRACT

We examined homogenized tissue samples of biopsies from 19 astrocytomas of different grades for genetic imbalances using comparative genomic hybridization (CGH): three astrocytomas grade II, and 16 astrocytomas grade IV (glioblastoma multiforme), one of the glioblastomas representing the recurrence of a benign oligoastrocytoma. In two of three cases of astrocytoma grade II, a gain of chromosome 7 was found. The alterations in the glioblastomas were complex, and most frequently showed the characteristic gain of chromosome 7 and loss of chromosome 10. The single analyzed case of recurrence of an oligoastrocytoma was characterized by a unique CGH pattern. This tumor showed two distinct alterations: apart from an amplification on 15q24q26, we found a distinct amplification of a small region on 20p11.2p12, which has not been previously described in brain tumors. Partial or complete gains of chromosome 20 arose in six other tumors; we conclude that chromosome 20 in particular 20p11. 2p12, may harbor relevant genes for glioma progression.


Subject(s)
Brain Neoplasms/genetics , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 20 , Glioblastoma/genetics , Nucleic Acid Hybridization/methods , Adult , Aged , Aged, 80 and over , Female , Gene Amplification , Humans , Karyotyping , Male , Middle Aged
8.
J Biol Chem ; 275(48): 37469-73, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-10984493

ABSTRACT

The tumor suppresser protein p53 is critical for guarding the genome from incorporation of damaged DNA (Lane, D. P. (1992) Nature 358, 15-16). A relevant stress that activates p53 function is UV light (Noda, A., Toma-Aiba, Y., and Fujiwara, Y. (2000) Oncogene 19, 21-31). Another well known component of the mammalian UV response is the transcription factor c-Jun (Angel, P., and Karin, M. (1991) Biochim. Biophys. Acta 1072, 129-157). We show here that upon UV irradiation p53 activates transcription of the human mismatch repair gene MSH2. Interestingly, this up-regulation critically depends on functional interaction with c-Jun. Hence, the synergistic interaction of a proto-oncogene with a tumor suppresser gene is required for the regulation of the mammalian stress response through activation of expression of MSH2.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins , Gene Expression Regulation/radiation effects , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Base Sequence , Cell Line , DNA , Humans , MutS Homolog 2 Protein , Promoter Regions, Genetic , Proto-Oncogene Mas , Tumor Cells, Cultured
9.
Clin Cancer Res ; 5(11): 3569-77, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10589773

ABSTRACT

Meningiomas are cytogenetically characterized by loss of one chromosome 22 as a typical primary aberration and progression-associated secondary chromosome changes, of which monosomy 1p is the most common. The aim of this study was to evaluate the significance of monosomy 1p and enzyme activity loss of tissue nonspecific alkaline phosphatase (ALPL), whose gene maps to chromosome 1p36.1-p34, as parameters for the diagnosis of progression-prone meningiomas. We analyzed smear preparations of 56 meningiomas and additional paraffin sections of 17 of the cases by two-color fluorescence in situ hybridization (FISH) using the D1Z1 and D1Z2 probes and by a metaphase cytogenetic analysis of 30 of these tumors. The results were compared to clinical and morphological parameters and the expression of ALPL. Smear preparations showed deletion of 1p36 in 27% of common-type, 70% of atypical (intermediate-type), and 100% of anaplastic meningiomas. Monosomy 1p, as detected by FISH or the karyotype, was strongly associated with complete loss of ALPL activity. Intermediate-type and anaplastic meningiomas of younger patients displayed an increasing rate of cells with trisomy 1q and relative loss of 1p. The highly significant correlation of FISH results and ALPL histochemistry with clinical parameters gives evidence of their strong prognostic relevance. The complete activity loss of ALPL and the immunologically detected loss of ALPL protein in areas of meningiomas with monosomy 1p indicate a cytogenetically undetectable inactivation of the homologous Alpl allele. The apparently homozygous loss of expression of ALPL supports the notion that Alpl is a candidate tumor suppressor gene in meningiomas.


Subject(s)
Alkaline Phosphatase/genetics , Chromosome Deletion , Chromosomes, Human, Pair 1 , Meningeal Neoplasms/enzymology , Meningeal Neoplasms/genetics , Meningioma/genetics , Monosomy , Biopsy , Chromosome Mapping , Disease Progression , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Meningeal Neoplasms/pathology , Meningioma/enzymology , Meningioma/pathology , Trisomy
10.
J Neuropathol Exp Neurol ; 58(9): 993-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499441

ABSTRACT

The term "multiforme" in glioblastoma multiforme (GBM) indicates the highly variable histomorphology that cannot be addressed by studies on homogenized tissue probes. In order to relate genetic findings with histomorphologically distinct areas we used microdissection to procure defined cell populations from microscopic tissue sections under direct visualization. Formalin-fixed and paraffin-embedded tissue sections of 10 GBM were evaluated for intratumoral genetic heterogeneity by microdissection of multiple areas of 20-50 tumor cells and DOP-PCR of DNA isolated from the dissected cell groups, followed by comparative genomic hybridization (CGH). Microdissected cells from histomorphologically normal extratumoral blood vessels from the same slides served as controls. The individual tumors showed variable combinations of primary chromosomal gains and losses common to all studied areas of a given case along with secondary, area-specific additional aberrations. CGH displayed a wider variety of chromosomal aberrations than metaphase cytogenetics of cell cultures from the same tumors. The most frequent aberrations observed were previously unperceived gains on chromosomes 4q (8/10) and 5q (5/10). Other nonrandom aberrations were gains on 12q (6/10), 13q (6/10), and 7 (5/10), and losses of 22 (5/10). Amplifications on 7p were intratumorally heterogeneous and only found in single areas of 2 tumors. In contrast to normal extratumoral vessels, vascular proliferates in most cases demonstrated chromosomal aberrations (CGH) which were partially different from the aberrations observed in the tumor itself. The described method gives evidence of considerable intratumoral genetic heterogeneity in GBM and provides a sensitive tool for the detection of quantitative chromosomal changes that are present only regionally within a given tumor.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Adult , Aged , Blood Vessels/cytology , Blood Vessels/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cerebrovascular Circulation , Chromosome Aberrations , Dissection , Endothelium, Vascular/pathology , Female , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Male , Middle Aged , Nucleic Acid Hybridization , Oligonucleotide Probes , Polymerase Chain Reaction , Reference Values
11.
Genes Chromosomes Cancer ; 26(3): 270-2, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10502328

ABSTRACT

Chordoid meningioma is a rare histological subtype of meningiomas. Cytogenetic analysis of three cases revealed the unique feature of an unbalanced translocation der(1)t(1;3)(p12-13;q11) that was ascertained by chromosome microdissection and reverse fluorescence in situ hybridization. As the t(1;3) has not been observed in other subtypes of meningioma, it may represent a specific cytogenetic marker of chordoid meningiomas. It is not yet clear whether a fusion gene or the combined loss of genes from chromosome arms 1p and 3p is the pathogenetically important outcome of the translocation.


Subject(s)
Chordoma/genetics , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 3 , Meningioma/genetics , Translocation, Genetic , Aged , Cell Transformation, Neoplastic/genetics , Chordoma/pathology , Female , Humans , Karyotyping , Male , Middle Aged
12.
Cancer Genet Cytogenet ; 113(2): 115-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10484976

ABSTRACT

Smear preparations of 23 fresh astrocytoma biopsies were analyzed by two-color fluorescence in situ hybridization with cosmids specific for the P16 and the TP53 genes. Additionally, tissue sections of the same tumors were immunostained with the use of a monoclonal antibody that recognizes both wild-type and mutant TP53 protein. In 21 astrocytomas, loss of P16 was observed in a significant proportion of cells. Cells with homozygous P16 loss were present in 13 astrocytomas; 14 astrocytomas showed cells with heterozygous loss of P16. Remarkably, 5 astrocytomas showed a scattered mosaic pattern of cells with homozygous and, respectively, heterozygous p16 loss. Homozygous deletion of TP53 was not observed. Cells with heterozygous TP53 loss were detected in 12 tumors, in 7 of them in association with P16 loss. One tumor showed aberrant cells for neither TP53 nor P16 but strong immunostaining for TP53. Positive TP53 immunostaining was found in 16 astrocytomas. Heterozygous loss of TP53 was significantly correlated with TP53 protein expression. We conclude that, unlike typical tumor suppressor genes, P16 might enhance cellular proliferation after heterozygous loss through a dosage effect and that the distribution of cells with homozygous loss of P16 speaks in favor of a polyclonal loss of the second copy of this gene.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Tumor Suppressor Protein p53/genetics , Astrocytoma/metabolism , Astrocytoma/pathology , Biopsy , Brain/pathology , Brain/physiology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Gene Deletion , Genetic Heterogeneity , Homozygote , Humans , In Situ Hybridization, Fluorescence , Neoplasm Staging , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
13.
Arch Virol ; 144(3): 621-6, 1999.
Article in English | MEDLINE | ID: mdl-10226625

ABSTRACT

SV40 and/or DNA sequences indistinguishable from SV40 have been detected in several types of human tumours. The oncoprotein of Simian virus 40, SV40 large T-antigen (Tag), is known to bind and inactivate tumour suppressor proteins, such as members of the retinoblastoma family and p53, thereby promoting cell transformation. In this study, we used the yeast two-hybrid system to investigate whether the Simian virus 40 (SV40) large T-antigen is able to interact with p73, a noval discovered putative tumour suppressor, that is homologous both structurally and functionally to p53. The yeast two-hybrid system is a genetic method to detect protein-protein-interactions in vivo. Our results suggest that the SV40 large T-antigen is not able to bind p73 in yeast although both proteins are expressed in the transformed yeast strain as was shown by western blot analysis.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , DNA-Binding Proteins/metabolism , Genes, Tumor Suppressor , Nuclear Proteins/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Cloning, Molecular , DNA-Binding Proteins/genetics , Humans , Nuclear Proteins/genetics , Tumor Protein p73 , Tumor Suppressor Proteins , Yeasts
14.
Int J Cancer ; 84(1): 6-9, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-9988224

ABSTRACT

Calpain, also named CANP (for calcium-activated neutral protease), is an intracellular cytoplasmatic non-lysosomal cysteine endopeptidase that requires calcium ions for activity. Many substrates of the calpain isoenzymes, such as the transcription factors c-Fos and c-Jun, the tumor supressor protein p53, protein kinase C, pp60c-src and the adhesion molecule integrin, have been implicated in the pathogenesis of different human tumors, suggesting an important role of the calpains in malignant diseases. We now report differential expression of the calpain I gene (CL I) in a variety of tumors, extending our study to a larger series of renal cell carcinomas. Using Northern-blot analysis, we studied calpain I expression in 30 renal cell carcinomas as compared with matched healthy tissues. Tumor samples were classified according to their histological type: 21 clear cell carcinomas, 4 chromophobe carcinomas, 3 papillary carcinomas and 2 oncocytomas. In renal tumor samples, calpain I gene mRNA was expressed at highly variable levels, significantly depending on the different histological types. Moreover, there was a correlation of higher calpain I expression with increased malignancy: within the clear cell carcinoma subset, tumor samples with advanced nodal status (N1 and N2) showed a significantly higher calpain I expression than tumors without metastasis to regional lymph nodes. Our data suggest an important role of calpain isoenzymes in carcinogenesis and tumor progression.


Subject(s)
Calpain/genetics , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Blotting, Northern , Calpain/biosynthesis , Carcinoma, Renal Cell/secondary , Gene Expression , Humans , Kidney Neoplasms/pathology , Lymphatic Metastasis , RNA, Messenger/biosynthesis
16.
Hum Mol Genet ; 7(12): 1859-72, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9811929

ABSTRACT

By screening a meningioma expression library with autologous serum we identified four cDNA clones representing a novel gene with striking homology to Caenorhabditis elegans hyaluronidase as indicated by BLASTP analysis. In humans hyaluronidase has been implicated in cancer development and three human genes are known to encode proteins with hyaluronidase activity. None of the human genes, however, showed any homology at the nucleotide or amino acid sequence level to the newly isolated antigen we termed meningioma expressed antigen 5 (MGEA5). Somatic cell hybrid mapping and fluorescence in situ hybridization mapped the gene for MGEA5 to chromosomal band 10q24.1-q24.3. Reverse transcription (RT)-PCR and northern blot hybridization revealed expression of the gene encoding MGEA5 in several meningioma and additional human tissues. Expression analysis also indicated an alternative splicing event giving rise to a shorter and altered transcript termed MGEA5s. The expression of MGEA5 and MGEA5s as fusion proteins revealed an approximate molecular weight of 92 and 54 kDa, respectively. Using heterologous sera we found antibodies against MGEA5s in five out of 23 meningioma patients, whereas no immune reaction was detected in 12 control sera from healthy individuals. Confirmation of hyaluronidase activity was independently achieved by turbidometric analysis and a gel matrix assay. A model for involvement of the novel hyaluronidase gene in meningioma development is proposed.


Subject(s)
Hyaluronoglucosaminidase/genetics , Meningioma/enzymology , Meningioma/genetics , Neoplasm Proteins/genetics , Acetylglucosaminidase , Alternative Splicing , Amino Acid Sequence , Antigens, Neoplasm , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 10/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Evolution, Molecular , Genetic Variation , Histone Acetyltransferases , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Open Reading Frames , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , beta-N-Acetylhexosaminidases
18.
Hum Genet ; 101(2): 190-7, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9402967

ABSTRACT

Gene amplification, which occurs in more than 50% of malignant gliomas, is considered to play a pivotal role in tumorigenesis. There are, however, few studies aimed toward the isolation of novel genes from amplified sequences. Previously, we reported amplification of the protooncogene MET (hepatocyte growth factor receptor; 7q31) in more than 20% of glioblastomas. For an approximate size estimation of the amplification unit we analyzed three glioblastomas all of which carried an amplified MET gene, by Southern blot analysis and/or competitive polymerase chain reaction using eight DNA markers. Although the extent of the amplified domain varied, the close vicinity of the MET gene was the only region consistently amplified in these glioblastomas. A yeast artificial chromosome (YAC) contig of 900 kb was refined spanning the amplified region flanking the MET gene. The YAC inserts were subcloned into 59 cosmids, which were used for exon trapping. Eight sequences were identical to parts of the genes MET and CAPZA2 (human actin capping protein alpha-subunit). Two newly identified exons and the CAPZA2 exons were amplified in tumor TX3095, which retains an amplified MET gene. The new exons were localized close to MET and CAPZA2. Characterization of the clones, which were termed glioma-amplified sequence (GAS)7-1 and GAS7-2, showed an open reading frame and a different expression pattern in multiple human tissues. This study reports the identification of a cluster of amplified genes including two novel genes in a region amplified in more than 20% of glioblastomas.


Subject(s)
Chromosomes, Human, Pair 7 , Exons/genetics , Gene Amplification , Glioma/genetics , Multigene Family , Actin Capping Proteins , Actin Depolymerizing Factors , CapZ Actin Capping Protein , Cosmids , Destrin , Humans , In Situ Hybridization, Fluorescence , Microfilament Proteins/genetics , Molecular Sequence Data , Proto-Oncogene Proteins c-met/genetics
19.
Hum Mol Genet ; 6(12): 2031-41, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9356211

ABSTRACT

There is increasing evidence that tumor expressed genes induce immune responses in cancer patients. To identify meningioma expressed antigens, we established a meningioma expression library which was screened with autologous serum. Out of 20 positive cDNA clones eight share high sequence homologies as determined by sequence analysis. These eight clones can be grouped into three classes which differ in length and which are characterized by specific sequence variations. The longest open reading frame was found to be 2412 bp encoding an immunoreactive antigen termed meningioma expressed antigen 6 (MEA6). Using five sequence specific primer pairs, somatic hybrid panel mapping revealed locations of the three classes on several human chromosomes including chromosomes 2, 3, 6, 7, 9, 13 and 14. The mapping results were confirmed by fluorescence in situ hybridization. RT-PCR showed consistent expression of all classes in several meningiomas and additional tissues using the same set of primer pairs as for chromosomal mapping. The expression data were confirmed by northern blot analysis. For the predicted amino acid sequence BLASTX revealed a homology to a human C219-reactive peptide which was previously isolated by an antibody directed against p-glycoprotein. Sequence properties of the MEA protein include an acidic activation domain, a proline-rich region and two coiled-coil domains indicating protein binding and activation functions.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Chromosome Mapping , DNA, Complementary/isolation & purification , Meningioma/genetics , Meningioma/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Amino Acid Sequence , Antigens, Neoplasm/isolation & purification , Base Sequence , Cloning, Molecular , Humans , Meningioma/chemistry , Molecular Sequence Data , Neoplasm Proteins/isolation & purification , Protein Structure, Secondary , Protein Structure, Tertiary
20.
J Neuropathol Exp Neurol ; 56(8): 879-86, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9258258

ABSTRACT

Apart from defined histomorphologic features, increased Ki-67 indices and various numeric and structural chromosome aberrations, meningiomas of the intermediate (WHO grade II, atypical meningioma) and anaplastic type (WHO grade III) are cytogenetically distinguished from common-type meningiomas (WHO grade I) by frequent loss of the distal part of the short arm of one chromosome 1 (1p-), which formerly proved to be an independent predictor of shorter recurrence-free intervals. Histochemically, loss of alkaline phosphatase activity (ALPL, liver/bone/kidney type, EC 3.1.3.1) was another frequent, specific finding in meningiomas with signs of dedifferentiation. In a prospective study including 66 meningiomas, all common-type meningiomas except one case (18/19) were reactive for ALPL, whereas 75% (30/39) of intermediate type and all anaplastic meningiomas (8/8) showed loss of enzyme activity in large areas of the tumor. Exclusively, the ALPL negative phenotype was associated with 1p loss (15/19). Our data suggest that ALPL, which is coded as a single copy gene on chromosome 1p36.1-p34, is a useful marker enzyme for the loss of a putative regulatory (tumor suppressor) gene on chromosome 1p, or that ALPL itself represents a new tumor suppressor gene homozygously inactivated in meningiomas.


Subject(s)
Alkaline Phosphatase/deficiency , Chromosomes, Human, Pair 1 , Gene Deletion , Genes, Tumor Suppressor , Meningeal Neoplasms/enzymology , Meningioma/enzymology , Disease Progression , Histocytochemistry/methods , Humans , Immunohistochemistry , Prospective Studies , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...