Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Neoplasia ; 3(2): 105-9, 2001.
Article in English | MEDLINE | ID: mdl-11420745

ABSTRACT

To assess the utility of fluorescence in situ hybridization (FISH) for analysis of MYCN gene amplification in neuroblastoma, we compared this assay with Southern blot analysis using tumor specimens collected from 232 patients with presenting characteristics typical of this disease. The FISH technique identified MYCN amplification in 47 cases, compared with 39 by Southern blotting, thus increasing the total number of positive cases by 21%. The major cause of discordancy was a low fraction of tumor cells (< or =30% replacement) in clinical specimens, which prevented an accurate estimate of MYCN copy number by Southern blotting. With FISH, by contrast, it was possible to analyze multiple interphase nuclei of tumor cells, regardless of the proportion of normal peripheral blood, bone marrow, or stromal cells in clinical samples. Thus, FISH could be performed accurately with very small numbers of tumor cells from touch preparations of needle biopsies. Moreover, this procedure allowed us to discern the heterogeneous pattern of MYCN amplification that is characteristic of neuroblastoma. We conclude that FISH improves the detection of MYCN gene amplification in childhood neuroblastomas in a clinical setting, thus facilitating therapeutic decisions based on the presence or absence of this prognostically important biologic marker.


Subject(s)
Blotting, Southern/methods , Genes, myc/genetics , In Situ Hybridization, Fluorescence/methods , Neuroblastoma/genetics , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Child, Preschool , Female , Humans , Infant , Male , Neuroblastoma/diagnosis
2.
Nat Med ; 6(5): 529-35, 2000 May.
Article in English | MEDLINE | ID: mdl-10802708

ABSTRACT

Caspase 8 is a cysteine protease regulated in both a death-receptor-dependent and -independent manner during apoptosis. Here, we report that the gene for caspase 8 is frequently inactivated in neuroblastoma, a childhood tumor of the peripheral nervous system. The gene is silenced through DNA methylation as well as through gene deletion. Complete inactivation of CASP8 occurred almost exclusively in neuroblastomas with amplification of the oncogene MYCN. Caspase 8-null neuroblastoma cells were resistant to death receptor- and doxorubicin-mediated apoptosis, deficits that were corrected by programmed expression of the enzyme. Thus, caspase 8 acts as a tumor suppressor in neuroblastomas with amplification of MYCN.


Subject(s)
Caspases/genetics , Gene Amplification , Gene Silencing , Genes, myc , Neuroblastoma/genetics , Antineoplastic Agents/pharmacology , Apoptosis , Caspase 8 , Caspase 9 , Caspases/biosynthesis , Child , DNA Methylation , Doxorubicin/pharmacology , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Recombinant Proteins/biosynthesis , Retroviridae/genetics , Signal Transduction , Tumor Cells, Cultured
3.
Cancer Genet Cytogenet ; 115(2): 100-5, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10598141

ABSTRACT

Loss of heterozygosity (LOH) for markers on chromosome arm 16q in Wilms tumor has been linked to an increased risk of treatment failure. We therefore postulated that fluorescence in situ hybridization (FISH) with probes from this region might enhance current strategies for identifying high-risk patients at diagnosis. In a blinded comparative pilot study of 19 Wilms tumor samples from 18 patients with favorable histology, FISH and DNA polymorphism analysis yielded concordant results in 14 cases, either retention (n = 6) or loss (n = 8) of chromosome arm 16q markers. Discordant findings in 4 of the 5 remaining cases resulted from detection of LOH, but no loss by FISH. Two of these cases, directly comparable at marker D16S422, appeared to have tumor-specific uniparental disomy, in that 2 copies of D16S422 and the 16 centromere were evident, despite LOH. In 2 other cases, the discrepancies could be explained by LOH confined to loci distal to the D16S422 locus. In the fifth case, FISH detected 2 distinct populations of tumor cells, one characterized by normal diploidy and the other by monosomy 16, whereas DNA polymorphism analysis failed to indicate LOH altogether. Thus, FISH confirmed the presence of allelic loss (hence, the possible location of biologically important tumor suppressor genes) on the distal long arm of chromosome 16 in cases of favorable-histology Wilms tumor, with the advantages of technical simplicity, successful analysis of samples that were otherwise uninformative by analysis of DNA polymorphisms, and the addition of internal controls for chromosomal aneusomy. We suggest that combined analysis of the chromosome 16q region in Wilms tumor by FISH and DNA polymorphism analysis would improve evaluations to identify high-risk patients who might benefit from alternative therapy.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 16 , In Situ Hybridization, Fluorescence , Kidney Neoplasms/genetics , Wilms Tumor/genetics , Child , Child, Preschool , DNA, Neoplasm/analysis , Female , Homozygote , Humans , Infant , Loss of Heterozygosity , Male , Polymorphism, Genetic
4.
Gene ; 229(1-2): 223-8, 1999 Mar 18.
Article in English | MEDLINE | ID: mdl-10095122

ABSTRACT

The murine transcription factor murine cyclin D-binding Myb-like protein (mDmp1) arrests the cell cycle in G1 phase, through an activity that can be overridden by direct interaction with the D-type cyclins. Here, we describe the identification, sequence, chromosomal localization, and expression of the human cognate, hDMP1. The hDMP1 cDNA contains a 2280bp open reading frame that shares a high degree of identity with the mDmp1 coding region. The 4.4kb hDMP1 messenger RNA is ubiquitously expressed in normal human tissues, with highest levels in testis and substructures within the brain. By use of fluorescence in situ hybridization with a human genomic P1 probe, we assigned hDMP1 to chromosome 7, band q21. This chromosomal region is frequently deleted as part of the 7q-minus and monosomy 7 abnormalities of human acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). We analyzed hDMP1 copy number by fluorescence in situ hybridization in leukemic blasts from nine patients with abnormalities of the long arm of chromosome 7, and in each case one allele of the hDMP1 gene was deleted. Functional analysis of the mDmp1 protein has shown that it negatively regulates cell proliferation, which suggests that this gene is a candidate suppressor of malignant transformation. Further study will be needed to determine whether gene-specific mutations implicate hDMP1 as a tumor suppressor in acute leukemias with deletions of the long arm of chromosome 7 or in other types of human malignancy.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Cell Cycle/genetics , Cell Division/genetics , Chromosome Mapping , Cloning, Molecular , Cyclin D , Cyclins/metabolism , Gene Deletion , Genes, Tumor Suppressor , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Leukemia/genetics , Molecular Sequence Data , Protein Binding/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Tumor Cells, Cultured
5.
Neoplasia ; 1(1): 80-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10935473

ABSTRACT

We examined 18 neuroblastoma cell lines and 32 primary single-copy MYCN tumor specimens to determine whether mutations of p73, a novel p53-related gene located in chromosome band 1p36.33, contribute to the genesis or progression of childhood neuroblastoma. By fluorescence in situ hybridization, 16 of the 18 cell lines, but only 3 of the 32 primary tumors, had evidence of a deleted p73 allele. Sequence analysis of the p73 coding region in the mRNAs expressed by these cell lines and tumors did not reveal inactivating mutations, suggesting that p73 is not homozygously inactivated in neuroblastoma. However, several novel splice forms of p73 mRNAs were identified, including one without exon 11 that predominated in multiple MYCN-amplified cell lines. Its encoded p73 protein differed from other splice forms in that the C-terminus was derived from an alternative reading frame. Further study of the functional properties of the protein encoded by this splice form of p73 will be needed to determine whether it contributes to the pathogenesis of childhood neuroblastoma with MYCN gene amplification.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Neuroblastoma/genetics , Nuclear Proteins/genetics , Adolescent , Amino Acid Sequence , Child , Child, Preschool , Female , Gene Dosage , Humans , Infant , Infant, Newborn , Male , Molecular Sequence Data , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Proteins
6.
Neoplasia ; 1(3): 253-61, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10935480

ABSTRACT

The p27KIP1 gene, which encodes a cyclin-dependent kinase (CDK) inhibitor, has been assigned to chromosome band 12p12, a region often affected by cytogenetically apparent deletions or translocations in childhood acute lymphoblastic leukemia (ALL). As described here, fluorescence in situ hybridization (FISH) analysis of 35 primary ALL samples with cytogenetic evidence of 12p abnormalities revealed hemizygous deletions of p27KIP1 in 29 cases. Further analysis of 19 of these cases with two additional gene-specific probes from the 12p region (hematopoietic cell phosphatase, HCP and cyclin D2, CCND2) showed that p27KIP1 is located more proximally on the short arm of chromosome 12 and is deleted more frequently than either HCP or CCND2. Of 16 of these cases with hemizygous deletion of p27KIP1, only eight showed loss of HCP or CCND2, whereas loss of either of the latter two loci was uniformly associated with loss of p27KIP1. Missense mutations or mutations leading to premature termination codons were not detected in the coding sequences of the retained p27KIP1 alleles in any of the 16 ALL cases examined, indicating a lack of homozygous inactivation. By Southern blot analysis, one case of primary T-cell ALL had hemizygous loss of a single p27KIP1 allele and a 34.5-kb deletion, including the second coding exon of the other allele. Despite homozygous inactivation of p27KP1 in this case, our data suggest that haploinsufficiency for p27KIP1 is the primary consequence of 12p chromosomal deletions in childhood ALL. The oncogenic role of reduced, but not absent, levels of p27KIP1 is supported by recent studies in murine models and evidence that this protein not only inhibits the activity of complexes containing CDK2 and cyclin E, but also promotes the assembly and catalytic activity of CDK4 or CDK6 in complexes with cyclin D.


Subject(s)
Cell Cycle Proteins , Gene Deletion , Microtubule-Associated Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Repressor Proteins , Tumor Suppressor Proteins , Adolescent , Base Sequence , Blotting, Southern , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p27 , DNA-Binding Proteins/genetics , Female , HL-60 Cells , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Molecular Sequence Data , Proto-Oncogene Proteins c-ets , Restriction Mapping , Transcription Factors/genetics , ETS Translocation Variant 6 Protein
7.
J Pediatr Surg ; 33(11): 1695-8, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9856898

ABSTRACT

BACKGROUND/PURPOSE: Deletion of the short arm of chromosome 1 (1p) is one of the poor prognostic factors in human neuroblastomas. Recent studies have suggested that one or more of the neuroblastoma tumor suppressor genes reside in this region and have identified the shortest region of overlap (SRO) on 1p36. The purpose of this study was to examine deletions of 1p in human neuroblastomas by fluorescence in situ hybridization (FISH). METHODS: Two-color FISH analysis was performed to detect chromosome 1p36 abnormalities in 42 MYCN-amplified neuroblastomas. Four different probes from the 1p36 region, the E2F2, NPPA, D1S160, and CDC2L1 loci were used for detection of 1p abnormalities. A repeat sequence probe, which is specific for the heterochromatic region of chromosome 1 (pUC1.77), was used as a control. RESULTS: Large deletions of 1p36 were observed in 31 (73.8%) of 42 tumors, whereas the remaining 11 (26.2%) showed no deletion. In these 11 tumors, a translocation of 1p was found in one and a duplication of 1p was detected in another. CONCLUSIONS: A strong correlation between 1p abnormalities and MYCN amplification was found in this study. MYCN-amplified neuroblastomas were found to show large deletions of 1p encompassing the SRO. FISH provided a rapid and reliable method to detect hemizygous deletions of 1p.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Genes, myc , Neuroblastoma/genetics , Child, Preschool , Culture Techniques , Female , Genetic Markers , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Sensitivity and Specificity
8.
Proc Natl Acad Sci U S A ; 95(23): 13765-70, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9811875

ABSTRACT

X-linked lymphoproliferative syndrome (XLP) is an inherited immunodeficiency characterized by increased susceptibility to Epstein-Barr virus (EBV). In affected males, primary EBV infection leads to the uncontrolled proliferation of virus-containing B cells and reactive cytotoxic T cells, often culminating in the development of high-grade lymphoma. The XLP gene has been mapped to chromosome band Xq25 through linkage analysis and the discovery of patients harboring large constitutional genomic deletions. We describe here the presence of small deletions and intragenic mutations that specifically disrupt a gene named DSHP in 6 of 10 unrelated patients with XLP. This gene encodes a predicted protein of 128 amino acids composing a single SH2 domain with extensive homology to the SH2 domain of SHIP, an inositol polyphosphate 5-phosphatase that functions as a negative regulator of lymphocyte activation. DSHP is expressed in transformed T cell lines and is induced following in vitro activation of peripheral blood T lymphocytes. Expression of DSHP is restricted in vivo to lymphoid tissues, and RNA in situ hybridization demonstrates DSHP expression in activated T and B cell regions of reactive lymph nodes and in both T and B cell neoplasms. These observations confirm the identity of DSHP as the gene responsible for XLP, and suggest a role in the regulation of lymphocyte activation and proliferation. Induction of DSHP may sustain the immune response by interfering with SHIP-mediated inhibition of lymphocyte activation, while its inactivation in XLP patients results in a selective immunodeficiency to EBV.


Subject(s)
Lymphoproliferative Disorders/genetics , Mutation , X Chromosome , src Homology Domains/genetics , Amino Acid Sequence , Chromosome Mapping , Genetic Linkage , Genetic Markers , Humans , Molecular Sequence Data
9.
Cancer Res ; 57(18): 4042-7, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9307291

ABSTRACT

We analyzed 27 samples of primary medulloblastoma, using comparative genomic hybridization and a novel statistical approach to evaluate chromosomal regions for significant gain or loss of genomic DNA. An array of nonrandom changes was found in most samples. Two discrete regions of high-level DNA amplification of chromosome bands 5p15.3 and 11q22.3 were observed in 3 of 27 tumors. Nonrandom genomic losses were most frequent in regions on chromosomes 10q (41% of samples), 11 (41%), 16q (37%), 17p (37%), and 8p (33%). Regions of DNA gain most often involved chromosomes 17q (48%) and 7 (44%). These findings suggest a greater degree of genomic imbalance in medulloblastoma than has been recognized previously and highlight chromosomal loci likely to contain oncogenes or tumor suppressor genes that may contribute to the molecular pathogenesis of this tumor.


Subject(s)
Chromosome Aberrations/genetics , DNA, Neoplasm/genetics , Medulloblastoma/genetics , Adolescent , Child , Child, Preschool , Chromosome Disorders , Chromosome Mapping , Female , Humans , In Situ Hybridization, Fluorescence/methods , Infant , Male , Nucleic Acid Hybridization/methods
10.
Genes Chromosomes Cancer ; 16(1): 15-20, 1996 May.
Article in English | MEDLINE | ID: mdl-9162192

ABSTRACT

Childhood endodermal sinus tumors (CESTs) are a unique category of germ cell tumors involving the testis and extragonadal region in children less than 4 years of age. Recent studies of CEST have shown recurrent cytogenetic abnormalities involving the short arm of chromosome 1, most commonly, a deletion of distal 1p. Experience with neuroblastomas has shown that cytogenetic analyses may underestimate the frequency of 1p deletion. To determine the frequency of deletion of Ip in CEST and to verify that 1p is, in fact, deleted and not translocated, we analyzed ten tumors by two-color fluorescence in situ hybridization on single-cell suspensions of interphase nuclei by using a cosmid probe from the PITSLRE kinase (p58) locus (previously mapped to 1p36) cohybridized with plasmid probe pUC1.77 (which recognizes the 1q heterochromatic region) to determine the copy number of chromosome 1. Eight of the ten tumors examined showed evidence of deletion of 1p36. Five of the eight tumors exhibited multiple subdones, and all subdones showed deletion of at least one copy of 1p36, indicating that the deletion probably occurred before the development of chromosome 1 aneusomy. We conclude that deletions of the short arm of chromosome 1, specifically 1p36, do occur in CEST and probably occur at a, higher incidence than that found in neuroblastoma Further studies are needed to determine the degree of overlap of the common area of deletion in CEST with that of neuroblastoma and to determine whether 1p deletion in CEST has prognostic significance.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1 , Endodermal Sinus Tumor/genetics , Germinoma/genetics , Testicular Neoplasms/genetics , Child, Preschool , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Sacrococcygeal Region/pathology
11.
Oncogene ; 12(2): 265-75, 1996 Jan 18.
Article in English | MEDLINE | ID: mdl-8570204

ABSTRACT

A t(3;5)(q25.1;q34) chromosomal translocation associated with myelodysplastic syndrome and acute myeloid leukemia (AML) was found to rearrange part of the nucleophosmin (NPM) gene on chromosome 5 with sequences from a novel gene on chromosome 3. Chimeric transcripts expressed by these cells contain 5' NPM coding sequences fused in-frame to those of the new gene, which we named myelodysplasia/myeloid leukemia factor 1 (MLF1). RNA-based polymerase chain reaction analysis revealed identical NPM-MLF1 mRNA fusions in each of the three t(3;5)-positive cases of AML examined. The predicted MLF1 amino acid sequence lacked homology to previously characterized proteins and did not contain known functional motifs. Normal MLF1 transcripts were expressed in a variety of tissues, most abundantly in testis, ovary, skeletal muscle, heart, kidney and colon. Anti-MLF1 antibodies detected the wild-type 31 kDa protein in K562 and HEL erythroleukemia cell lines, but not in HL-60, U937 or KG-1 myeloid leukemia lines. By contrast, t(3;5)-positive leukemia cells expressed a 54 kDa NPM-MLF1 protein, but not normal MLF1. Immunostaining experiments indicated that MLF1 is normally located in the cytoplasm, whereas NPM-MLF1 is targeted to the nucleus, with highest levels in the nucleolus. The nuclear/nucleolar localization of NPM-MLF1 mirrors that of NPM, indicating that NPM trafficking signals direct MLF1 to an inappropriate cellular compartment in myeloid leukemia cells.


Subject(s)
Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 5 , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Nuclear Proteins/genetics , Recombinant Fusion Proteins/genetics , Translocation, Genetic , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Gene Rearrangement , Humans , Molecular Sequence Data , Nucleophosmin , RNA, Messenger/analysis , Recombinant Fusion Proteins/analysis
12.
Proc Natl Acad Sci U S A ; 92(12): 5520-4, 1995 Jun 06.
Article in English | MEDLINE | ID: mdl-7777541

ABSTRACT

Deletion of the short arm of human chromosome 1 is the most common cytogenetic abnormality observed in neuroblastoma. To characterize the region of consistent deletion, we performed loss of heterozygosity (LOH) studies on 122 neuroblastoma tumor samples with 30 distal chromosome 1p polymorphisms. LOH was detected in 32 of the 122 tumors (26%). A single region of LOH, marked distally by D1Z2 and proximally by D1S228, was detected in all tumors demonstrating loss. Also, cells from a patient with a constitutional deletion of 1p36, and from a neuroblastoma cell line with a small 1p36 deletion, were analyzed by fluorescence in situ hybridization. Cells from both sources had interstitial deletions of 1p36.2-36.3 which overlapped the consensus region of LOH defined by the tumors. Interstitial deletion in the constitutional case was confirmed by allelic loss studies using the panel of polymorphic markers. Four proposed candidate genes--DAN, ID3 (heir-1), CDC2L1 (p58), and TNFR2--were shown to lie outside of the consensus region of allelic loss, as defined by the above deletions. These results more precisely define the location of a neuroblastoma suppressor gene within 1p36.2-36.3, eliminating 33 centimorgans of proximal 1p36 from consideration. Furthermore, a consensus region of loss, which excludes the four leading candidate genes, was found in all tumors with 1p36 LOH.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1 , Neuroblastoma/genetics , Base Sequence , Chromosome Mapping , DNA Primers , Heterozygote , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data
13.
Blood ; 85(9): 2321-30, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7727766

ABSTRACT

The tandemly linked p16INK4aMTS1 and p15INK4b/MTS2 genes on chromosome 9, band p21 encode proteins that function as specific inhibitors of the cyclin D-dependent kinases CDK4 and CDK6. This locus undergoes frequent bi-allelic deletion in human cancer cell lines, suggesting that the encoded proteins may function as tumor suppressors. However, more recent analysis of primary tumor samples has shown a much lower frequency of abnormalities affecting this region, raising doubt over the importance of these proteins in human malignancies. Hemizygous deletions and rearrangements of chromosome 9, band p21, are among the most frequent cytogenetic abnormalities detected in pediatric acute lymphoblastic leukemia (ALL), occurring in approximately 10% of cases. To determine if the p16INK4a/p15INK4b locus might be the target of these chromosomal lesions, we analyzed both genes in primary clinical samples from 43 pediatric ALL patients using interphase fluorescence in situ hybridization, Southern blot analysis, and the polymerase chain reaction. Deletions of p16INK4a/p15INK4b were identified in 18 of 20 cases with cytogenetically observed abnormalities of 9p and 5 of 23 with apparently normal chromosomes 9p, with the majority containing bi-allelic deletions (16 homozygous/7 hemizygous). Although most homozygous deletions involved both genes, Southern blot analysis showed an interstitial deletion in a single case that was confined to p16INK4a, suggesting that p15INK4b was not the critical target gene in this case. Sequence analysis of both p16INK4a and p15INK4b in all seven cases with hemizygous deletions failed to show mutations within the coding regions of the retained alleles. In this select group of patients, deletion of p16INK4a/p15INK4b was associated with T-cell phenotype, nonhyperdiploid karyotype (< 50 chromosomes), and poor event-free survival. These findings indicate that deletion of the p16INK4a/p15INK4b locus is one of the most common genetic abnormalities so far detected in pediatric ALL, and that loss of one or more of these cell cycle kinase inhibitors is important in leukemogenesis.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins , Chromosomes, Human, Pair 9/ultrastructure , DNA, Neoplasm/genetics , Gene Deletion , Genes, Tumor Suppressor , Leukemia-Lymphoma, Adult T-Cell/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tumor Suppressor Proteins , Adolescent , Alleles , Base Sequence , Blotting, Southern , Child , Child, Preschool , Chromosome Aberrations , Chromosome Mapping , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16 , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Leukemia-Lymphoma, Adult T-Cell/pathology , Male , Molecular Sequence Data , Polymerase Chain Reaction , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
14.
Cancer Res ; 55(4): 721-6, 1995 Feb 15.
Article in English | MEDLINE | ID: mdl-7850780

ABSTRACT

Multiple copies of N-myc proto-oncogene are only rarely detected in localized neuroblastomas (NBs), and the prognostic relevance of amplification in this subset of patients is not clear. We analyzed a series of 850 children with NB admitted to a Pediatric Oncology Group NB Biology Study and identified six patients with localized NBs harboring N-myc gene amplification. Three patients whose tumors showed favorable histology by Shimada classification and low-risk histological features according to the Joshi classification have remained disease-free, whereas two of three patients with unfavorable histology tumors have developed recurrent disease. Although earlier studies have indicated that N-myc amplification is associated with diploid DNA content, flow cytometric analysis revealed that only two of the localized tumors contained stem lines with diploid DNA content. Loss of chromosome 1p was not detected by fluorescence in situ hybridization in the two tumors examined. N-myc protein was detected by immunohistochemical studies in four of the five NBs analyzed. However, N-myc protein was not visualized in one of the tumors with stroma-rich histology, and Western blot analysis revealed only low levels of N-myc protein expression in another NB with favorable histology. These studies indicate that the presence of N-myc amplification in localized NBs does not necessarily portend an adverse outcome. Furthermore, the biological features of this subset of N-myc-amplified NBs appear to differ from those of more advanced N-myc-amplified tumors.


Subject(s)
Gene Amplification , Genes, myc , Neuroblastoma/genetics , Child , Child, Preschool , Chromosomes, Human, Pair 1 , DNA Probes , Humans , Immunohistochemistry , Infant , Neoplasm Staging , Neuroblastoma/pathology , Ploidies , Prognosis , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics
16.
Genomics ; 25(1): 130-8, 1995 Jan 01.
Article in English | MEDLINE | ID: mdl-7774910

ABSTRACT

The E2F transcription factor plays an important regulatory role in cell proliferation, mediating the expression of genes whose products are essential for inducing resting cells to enter the cell cycle and synthesize DNA. To investigate the possible involvement of E2F in hematopoietic malignancies, we isolated genomic clones encompassing the human E2F1 gene. We then used fluorescence in situ hybridization to localize E2F1 to human chromosome 20q11, telomeric to the p107 locus, a gene whose product is related to the retinoblastoma gene product (pRb). This finding contrasts with the 1p36 and 6q22 chromosomal locations previously assigned E2F2 and E2F3, two additional members of the E2F family. Although deletions or structural rearrangements of E2F1 were not detected in 14 primary acute leukemia or myelodysplasia samples with structural abnormalities of chromosome 20q11, the gene was amplified and overexpressed in HEL erythroleukemia cells and translocated to other chromosomes in several established human leukemia cell lines. This study provides the first evidence of gene amplification involving a member of the E2F family of transcription factors. We propose that E2F1 overexpression in erythroid progenitors may stimulate abnormal cell proliferation by overriding negative regulatory signals mediated by tumor suppressor proteins such as pRb.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Chromosomes, Human, Pair 20 , DNA-Binding Proteins , Leukemia, Erythroblastic, Acute/genetics , Myelodysplastic Syndromes/genetics , Transcription Factors/genetics , Animals , Base Sequence , Blotting, Northern , Blotting, Southern , Cell Division , Cell Line , Chromosome Mapping , Cloning, Molecular , Cricetinae , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , E2F3 Transcription Factor , Gene Amplification , Gene Expression , Genes, Tumor Suppressor , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Leukemia, Erythroblastic, Acute/metabolism , Molecular Sequence Data , Multigene Family , Myelodysplastic Syndromes/metabolism , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Retinoblastoma-Binding Protein 1 , Telomere , Transcription Factor DP1 , Transcription Factors/biosynthesis , Tumor Cells, Cultured
17.
Genomics ; 20(3): 468-73, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8034320

ABSTRACT

Interleukin-1 beta (IL-1 beta) mediates a wide range of immune and inflammatory responses. The active cytokine is generated by proteolytic cleavage of an inactive precursor by a protease called the IL-1 beta converting enzyme (ICE). A cDNA encoding this protease was recently isolated. A human genomic clone containing the ICE gene (IL1BC) was isolated using the cDNA as a probe. The gene consists of 10 exons spanning at least 10.6 kb. 5'-anchored polymerase chain reaction indicated a single transcription start site approximately 33 bp upstream of the initiator Met codon. The 5'-flanking region does not have an apparent TATA box but may contain an initiator (Inr) promoter element. However, transcriptional activity could not be detected with a fusion gene containing the 5'-flanking region linked to the bacterial chloramphenicol acetyltransferase gene (CAT) when transfected into the human acute monocytic leukemia cell line THP-1. Using the genomic IL1BC clone, we have confirmed the localization of the gene to chromosome 11 band q22.2-q22.3 by fluorescence in situ hybridization.


Subject(s)
Chromosomes, Human, Pair 11 , Hominidae/genetics , Metalloendopeptidases/genetics , Amino Acid Sequence , Animals , Base Sequence , Caspase 1 , Chromosome Banding , Chromosome Mapping , Cloning, Molecular , Codon , DNA Primers , DNA Probes , Exons , Genomic Library , Humans , In Situ Hybridization , Introns , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic , TATA Box , Transcription, Genetic
18.
Science ; 263(5151): 1281-4, 1994 Mar 04.
Article in English | MEDLINE | ID: mdl-8122112

ABSTRACT

The 2;5 chromosomal translocation occurs in most anaplastic large-cell non-Hodgkin's lymphomas arising from activated T lymphocytes. This rearrangement was shown to fuse the NPM nucleolar phosphoprotein gene on chromosome 5q35 to a previously unidentified protein tyrosine kinase gene, ALK, on chromosome 2p23. In the predicted hybrid protein, the amino terminus of nucleophosmin (NPM) is linked to the catalytic domain of anaplastic lymphoma kinase (ALK). Expressed in the small intestine, testis, and brain but not in normal lymphoid cells, ALK shows greatest sequence similarity to the insulin receptor subfamily of kinases. Unscheduled expression of the truncated ALK may contribute to malignant transformation in these lymphomas.


Subject(s)
Lymphoma, Large-Cell, Anaplastic/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Protein-Tyrosine Kinases/genetics , Translocation, Genetic , Amino Acid Sequence , Anaplastic Lymphoma Kinase , Base Sequence , Brain/enzymology , Cell Transformation, Neoplastic , Chromosome Walking , Chromosomes, Human, Pair 2 , Chromosomes, Human, Pair 5 , Cloning, Molecular , Gene Expression Regulation, Neoplastic , Humans , Intestine, Small/enzymology , Lymphoma, Large-Cell, Anaplastic/chemistry , Lymphoma, Large-Cell, Anaplastic/enzymology , Male , Molecular Sequence Data , Nuclear Proteins/chemistry , Nucleophosmin , Phosphoproteins/chemistry , Promoter Regions, Genetic , Protein-Tyrosine Kinases/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases , Sequence Alignment , Signal Transduction , Testis/enzymology , Tumor Cells, Cultured
19.
Genomics ; 17(3): 767-9, 1993 Sep.
Article in English | MEDLINE | ID: mdl-7902328

ABSTRACT

The murine Pax-7 gene and the cognate human gene, formerly designated HuP1, are members of the multigene paired-box-containing class of developmental regulatory genes first identified in Drosophila. By analysis of somatic cell hybrids segregating human chromosomes, the gene encoding PAX7 was localized to human chromosome 1. Fluorescence in situ hybridization confirmed this assignment and allowed mapping of the gene to the terminal region of the short arm (1p36) of the chromosome. Additionally, these results confirm the extensive homology between human chromosome 1p and the distal segment of mouse chromosome 4, extending from bands C5 through E2.


Subject(s)
Chromosomes, Human, Pair 1 , Genes, Homeobox , Animals , Chromosome Mapping , Drosophila/genetics , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Mice , Species Specificity
20.
Proc Natl Acad Sci U S A ; 90(12): 5539-43, 1993 Jun 15.
Article in English | MEDLINE | ID: mdl-8516298

ABSTRACT

The NF1 gene, which is altered in patients with type 1 neurofibromatosis, encodes neurofibromin, a protein whose GTPase-activating function can negatively regulate GTP-Ras by accelerating its conversion to inactive GDP-Ras. In schwannoma cell lines from patients with neurofibromatosis, loss of neurofibromin was previously shown to be associated with impaired regulation of GTP-Ras. Our analysis of other neural crest-derived tumor cell lines has shown that some melanoma and neuroblastoma cell lines established from tumors occurring in patients without neurofibromatosis contain reduced or undetectable levels of neurofibromin, with concomitant genetic abnormalities of the NF1 locus. In contrast to the schwannoma cell lines, GTP-Ras was appropriately regulated in the melanoma and neuroblastoma lines that were deficient in neurofibromin, even when c-H-ras was overexpressed in the lines. These results demonstrate that some neural crest tumors not associated with neurofibromatosis have acquired somatically inactivated NF1 genes and suggest a tumor-suppressor function for neurofibromin that is independent of Ras GTPase activation.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Neurofibromatosis 1 , Genes, ras , Protein Biosynthesis , Blotting, Northern , Blotting, Southern , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , GTPase-Activating Proteins , Gene Expression , Guanine Nucleotides/metabolism , Humans , In Situ Hybridization, Fluorescence , Kinetics , Melanoma , Neuroblastoma , Neurofibromin 1 , Phosphates/metabolism , Proteins/genetics , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Tumor Cells, Cultured , ras GTPase-Activating Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...