Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
2.
Genes Chromosomes Cancer ; 39(2): 99-109, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14695989

ABSTRACT

Well-differentiated liposarcomas (WDLs) are common soft-tissue tumors in adults. They are characterized by large marker chromosomes and/or ring chromosomes containing 12q-derived sequences in which MDM2 is consistently amplified. WDLs are subdivided into two subtypes according to their karyotype. Type D cells exhibit a near-diploid karyotype, with very few or no chromosome changes. Type H cells exhibit a near-tetraploid karyotype and many structural changes. Expression of P14ARF, MDM2, and TP53 proteins was assayed in the two WDL subtypes to establish whether distinct expression profiles correlated with cell ploidy. Although a transcriptionally functional TP53 was present in most tumors independent of their karyotype, type H cells were characterized by high levels of P14ARF and MDM2 proteins. Although amplified within similar chromosome markers in type D tumors, MDM2 did not appear to be overexpressed. In addition, it was present as a C-terminal truncated protein, indicative of alternatively spliced variants of MDM2 mRNA. As the existence of karyotypically distinct tumors could result from alterations of the mitotic machinery, we investigated the centrosome behavior in the two WDL subtypes. Centrosome amplification occurred in WDL tumors types H and D independent of their ploidy status. Moreover, no functional centrosome difference was found between the two tumor subtypes.


Subject(s)
Centrosome/metabolism , Gene Amplification/genetics , Liposarcoma/genetics , Nuclear Proteins , Proto-Oncogene Proteins/genetics , Retroperitoneal Neoplasms/genetics , Soft Tissue Neoplasms/genetics , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p53/genetics , Adult , Aged , Aged, 80 and over , Centrosome/chemistry , Centrosome/pathology , Chromosome Aberrations , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , Karyotyping , Lipoma/genetics , Lipoma/pathology , Liposarcoma/classification , Liposarcoma/pathology , Middle Aged , Mitosis/genetics , Proto-Oncogene Proteins c-mdm2 , Retroperitoneal Neoplasms/pathology , Soft Tissue Neoplasms/classification , Soft Tissue Neoplasms/pathology , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Tumor Cells, Cultured
3.
Ann Pathol ; 21(4): 303-10, 2001 Aug.
Article in French | MEDLINE | ID: mdl-11685128

ABSTRACT

AIM: Spindle cell lipomas are rare adipose tissues tumors. Histologically, these lesions are composed of mature adipocytes and spindle cells associated with collagen bundles. Spindle cell lipomas are benign tumors that can be difficult to distinguish from malignant tumors such as spindle cell liposarcomas, myxoid liposarcomas or well-differentiated liposarcomas. RESULTS: We report herein the description of two new cases. The first case was a deeply situated and infiltrating tumor located in the retromastoidian area. The karyotype showed the presence of two chromosomal abnormalities, a partial deletion of the long arm of chromosome 13, del(13)(q12) and a balanced reciprocal translocation t(2;6)(p16~21;p21). The second case was a subcutaneous tumor of posterior cervical localization. The karyotype showed a 13q deletion associated with a complex rearrangement of chromosomes 5, 6 and 10. The presence of the 13q deletion allowed us to confirm the diagnosis of spindle cell lipoma in both cases. This deletion has been previously described in six out the eleven published karyotype reports. The 13q deletion is usually associated with partial monosomy 16. The present case confirms that it can occur independently. The 6p21 rearrangement may also play a role in the pathobiology of this tumor, as suggested by the positive HMGIY expression detected by immunohistochemistry. CONCLUSION: Our study further illustrates that spindle cell lipomas can infiltrate the surrounding muscle and emphasizes the usefulness of cytogenetic analysis in the differential diagnosis of soft tissue tumors.


Subject(s)
Chromosomes, Human, Pair 13 , Cytogenetic Analysis , Gene Deletion , Lipoma/genetics , Neoplasms, Adipose Tissue/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 2 , Chromosomes, Human, Pair 5 , Chromosomes, Human, Pair 6 , Diagnosis, Differential , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Karyotyping , Lipoma/diagnosis , Lipoma/pathology , Male , Middle Aged , Neoplasms, Adipose Tissue/diagnosis , Neoplasms, Adipose Tissue/pathology , Translocation, Genetic
4.
Mol Reprod Dev ; 60(2): 172-80, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11553915

ABSTRACT

The eukaryotic MutHLS-like system plays a crucial role in both mitosis and meiosis. Until now, a number of works have focused on the function of MutS and MutL homologs during mismatch DNA repair. Nevertheless, little is known about the role of these proteins during meiosis. MSH4 is a meiosis specific protein that is necessary for meiotic recombination in Saccharomyces cerevisiae. The human MSH4 protein is only found in testis and ovary. It is involved first in synapsis and second during recombination together with MLH1 (MutL Homolog 1). Here, we report the identification of the mouse Msh4 gene that is located on chromosome 3. We examined the expression of mMsh4 in testes of increasing developmental age and in elutriated germ cells. The pattern of expression during spermatogenesis is consistent with a role for MSH4 both during zygonema and pachynema. We demonstrated a promoter activity of the mMsh4 5'-flanking region by cell transfection experiments with a luciferase reporter gene. We found several SRY/Sox binding sites in this region and co-transfection experiments showed that SRY could down regulate mMsh4 promoter transcriptional activity. We propose that the regulation of mMsh4 expression could be one of the reasons for the persistence of SRY and/or SRY-related proteins in adult testis.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Nuclear Proteins , Proteins/genetics , Transcription Factors , 3T3 Cells , Animals , Base Sequence , Cell Cycle Proteins , Chromosome Painting , Cloning, Molecular , DNA-Binding Proteins/genetics , Genes, Reporter/genetics , Humans , In Situ Hybridization, Fluorescence , Male , Meiosis/genetics , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Proteins/chemistry , Proteins/metabolism , Sex-Determining Region Y Protein , Testis/cytology , Testis/metabolism , Tissue Distribution , Transcription, Genetic , Transfection
5.
Genes Chromosomes Cancer ; 31(1): 85-90, 2001 May.
Article in English | MEDLINE | ID: mdl-11284039

ABSTRACT

A prenatal tumor located in the lumbar paravertebral area was discovered during a routine ultrasound examination at 32 weeks of pregnancy and surgically removed at 4 months of life. The histopathological diagnosis was first suggested to be an infantile desmoid fibromatosis. The tumor karyotype showed a three-way translocation involving both chromosomes 2 and a chromosome 11, t(2;11;2)(p23;p15;q31). Fluorescence in situ hybridization with a probe flanking the ALK gene at 2p23 demonstrated a rearrangement, as previously described in inflammatory myofibroblastic tumors (IMTs). In light of the genetic analysis, the histopathological diagnosis was revised to IMT, although inflammatory cells were scarce. IMTs are pseudosarcomatous inflammatory lesions that primarily occur in the soft tissue and viscera of children and young adults. Our report describes for the first time the occurrence of IMT during prenatal life. The ALK rearrangement may represent the molecular definition of a subgroup of mesenchymal tumors, not always with complete morphological features of IMT, similar to the model of EWS rearrangement in the Ewing sarcoma family of tumors.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 2/genetics , DNA Probes , Fetal Diseases/genetics , Granuloma, Plasma Cell/classification , Granuloma, Plasma Cell/genetics , Protein-Tyrosine Kinases/genetics , Translocation, Genetic/genetics , Adult , Anaplastic Lymphoma Kinase , Chromosome Painting , Female , Fetal Diseases/classification , Fetal Diseases/diagnostic imaging , Fetal Diseases/pathology , Granuloma, Plasma Cell/diagnostic imaging , Granuloma, Plasma Cell/pathology , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Pregnancy , Receptor Protein-Tyrosine Kinases , Ultrasonography, Prenatal
6.
Nat Genet ; 26(2): 207-10, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017079

ABSTRACT

Optic atrophy type 1 (OPA1, MIM 165500) is a dominantly inherited optic neuropathy occurring in 1 in 50,000 individuals that features progressive loss in visual acuity leading, in many cases, to legal blindness. Phenotypic variations and loss of retinal ganglion cells, as found in Leber hereditary optic neuropathy (LHON), have suggested possible mitochondrial impairment. The OPA1 gene has been localized to 3q28-q29 (refs 13-19). We describe here a nuclear gene, OPA1, that maps within the candidate region and encodes a dynamin-related protein localized to mitochondria. We found four different OPA1 mutations, including frameshift and missense mutations, to segregate with the disease, demonstrating a role for mitochondria in retinal ganglion cell pathophysiology.


Subject(s)
Chromosomes, Human, Pair 3 , GTP Phosphohydrolases/genetics , Mutation , Optic Atrophy/genetics , Amino Acid Sequence , Cell Nucleus/genetics , Chromosome Mapping , Dynamins , Exons , Female , GTP Phosphohydrolases/chemistry , Genes, Dominant , Humans , In Situ Hybridization, Fluorescence , Male , Mitochondria/genetics , Molecular Sequence Data , Pedigree , Polymorphism, Genetic , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/genetics , Sequence Alignment , Sequence Homology, Amino Acid
7.
Genes Chromosomes Cancer ; 29(2): 117-29, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10959091

ABSTRACT

Supernumerary ring and large marker chromosomes are a characteristic of atypical lipomas and well-differentiated liposarcomas (ALP-WDLPS) and are composed of amplified 12q14-15 sequences in association with variable segments from other chromosomes. Although stably transmitted, these chromosomes contain centromeric alterations, showing no detectable alpha-satellite sequences. We performed C-banding, fluorescence in situ hybridization, and immunostaining with anti-centromere antibodies in 8 cases of liposarcomas with supernumerary rings and large markers, including 5 ALP-WDLPS and 3 dedifferentiated-LPS and high-grade LPS. Our results with alpha-satellite probes and anti-CENPB antibodies confirm the lack of detectable alpha-satellite sequences in the five ALP-WDLPS supernumerary chromosomes, whereas centromeric activity was proved by the detection of kinetochores by using anti-CENPC antibodies. In contrast, the high grade and dedifferentiated liposarcomas showed a different pattern. In 2 cases, amplified chromosome 12 sequences, including amplification of alpha-satellite 12 sequences in 1 case, were present on chromosomes with typical centromeres. In another case, the rings were similar to WDLPS-ALP rings, but a large marker contained a chromosome 5 centromere and amplified alpha-satellite sequences from chromosome 8. ALP-WDLPS is the first example of a tumor class for which the presence of stable analphoid chromosomes is a constant and specific abnormality. Formation of newly derived centromeres, so-called neocentromeres, could be an original and effective way to maintain a selective advantage in neoplastic cells by conferring stability to the supernumerary chromosomes of ALP-WDLPS. The activation of normally non-centromeric sequences might be obtained by an epigenetic mechanism due to the peculiar chromatin conformation of these highly complex chromosomes.


Subject(s)
Centromere/genetics , Liposarcoma/genetics , Blotting, Southern , Cell Differentiation/genetics , Female , Genetic Markers/genetics , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Karyotyping , Liposarcoma/classification , Liposarcoma/pathology , Male , Tumor Cells, Cultured
8.
Pathol Res Pract ; 196(12): 853-6, 2000.
Article in English | MEDLINE | ID: mdl-11156329

ABSTRACT

Nephrogenic adenofibroma is a benign renal tumor in children and young adults described by Hennigar and Beckwith in 1992. Seven cases have been described, and we report the first case in an 11-month-old child, in good health, revealed by a macroscopic hematuria. Nephrogenic adenofibroma is an unusual tumor, which was difficult to distinguish from nephroblastoma and mesoblastic nephroma. Beckwith makes a distinction between this principal differential diagnosis in child renal tumors based upon morphologic and immunohistochemical patterns. In our observation, the diagnosis remained difficult and needed several reviews of our case. Beckwith proposed the final diagnosis: nephrogenic adenofibroma with stromal predominance. The prognosis is excellent and no treatment is indicated. A FISH analysis of the tumor cells found a trisomy 11. Trisomy 11 has been reported in mesoblastic nephroma as the most frequent chromosomal abnormality. This finding in tumor cells provides an argument for excluding the diagnosis of nephroblastoma but can not clarify the difference between nephrogenic adenofibroma and mesoblastic nephroma.


Subject(s)
Adenofibroma/pathology , Kidney Neoplasms/pathology , Adenofibroma/diagnostic imaging , Adenofibroma/drug therapy , Adenofibroma/surgery , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Dactinomycin/administration & dosage , Diagnosis, Differential , Female , Humans , Kidney/pathology , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/drug therapy , Kidney Neoplasms/surgery , Magnetic Resonance Imaging , Radiography , Vincristine/administration & dosage , Wilms Tumor/diagnosis
9.
Cancer Genet Cytogenet ; 115(2): 89-95, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10598139

ABSTRACT

Comparative genomic hybridization (CGH) was used to detect chromosomal gains and losses in a series of 90 frozen soft tissue primary tumors (STTs), all untreated. The material consisted of 69 malignant sarcomas, including 20 malignant fibrous histiocytomas (MFH), 23 liposarcomas (LPS), 6 leiomyosarcomas (LMS), 4 synovial sarcomas, 4 primitive neuroectodermal tumors (PNETs), and various others subtypes, in addition to 21 benign tumors. Within the benign tumors, only 2 of the 3 schwannomas showed genetic changes. In malignant sarcomas, genetic changes were detected in 64 of the 69 samples analyzed (92%), with a mean of 4.5 per sample (range 0-10). Gains and losses on chromosome 13 were observed in 32% of the sarcomas with genomic imbalance. Recurring low-level copy number increases were found at new sites on chromosomes 7 (6 MFH samples, 30%) and 8 (10 LPS samples, 43%), the minimal common regions being 7p15-pter and 8q24. No new recurring high-level amplifications were found. Surprisingly, losses of DNA sequences were more frequent than gains; particularly, losses were the main feature in LMS, with highly recurrent common minimal losses at 11q14-qter and 13q21-q22 (4 samples, 66%, and 5 samples, 83%, respectively). Losses of chromosome 2 sequences (minimal common regions at 2p24-pter and 2q32-qter) were observed in 50% of the MFH analyzed. New recurrent losses of whole or part of chromosome 14 were found in 57% of the pleomorphic LPS (PLPS) analyzed. This study uncovers new clues for the diagnosis of malignant STTs and shows the importance of deletions as events in the early steps involved in the tumorigenesis of STTs.


Subject(s)
Chromosome Deletion , Histiocytoma, Benign Fibrous/genetics , In Situ Hybridization/methods , Liposarcoma/genetics , Soft Tissue Neoplasms/genetics , Chromosomes, Human , Chromosomes, Human, Pair 13 , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 2 , Humans , Neurilemmoma/genetics
10.
Genomics ; 60(3): 371-4, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10493837

ABSTRACT

In the Ewing family of tumors (EFT), the EWS gene is rearranged with members of the ets oncogene family. Variability in genomic breakpoint locations is the source of significant heterogeneity in fusion product structure. As a consequence of variably included exon sequences from the two partner genes, a variable amount of additional peptide sequence is inserted in between the minimal transforming domains. Some of this molecular diversity has recently been correlated with disparate clinical outcome. Here we report on cryptic exons found in the chimeric RNA of three EFT with different EWS-FLI1 fusions. In two tumors, the emergence of a cryptic exon from FLI1 intron 5 in the chimeric RNA was apparently unrelated to the genomic rearrangement that occurred in FLI1 introns 4 and 5, respectively. In one case, a novel exon was generated through the creation of an artificial splice acceptor site in FLI1 intron 6 by the genomic rearrangement that occurred in EWS intron 8. These results further extend the spectrum of molecular diversity in EFT.


Subject(s)
Oncogene Proteins, Fusion/genetics , Recombinant Fusion Proteins/genetics , Sarcoma, Ewing/genetics , Transcription Factors/genetics , Adolescent , Amino Acid Sequence , Child , Exons , Female , Gene Rearrangement , Genetic Variation , Humans , Male , Molecular Sequence Data , Proto-Oncogene Protein c-fli-1 , RNA/genetics , RNA-Binding Protein EWS , Sequence Analysis, DNA
11.
Clin Exp Pathol ; 47(2): 88-91, 1999.
Article in French | MEDLINE | ID: mdl-10398580

ABSTRACT

The authors report a child male case of secretory carcinoma of the breast, associated with a abnormal tumoral karyotype (monosomy 22). The diagnosis was made by histopathological examination. This breast carcinoma is characterized by an abundant intra and extracellulary secretory material, which present a reactivity with antimilk proteins antibodies and includes spherical dense bodies, identified by ultrastructural study. The prognosis is not bad, whatever the age of the patient, particularly for the children and young women, displaying an locoregional aggressivity. The signification of a monosomy 22 in secretory carcinoma is unknown.


Subject(s)
Breast Neoplasms, Male/pathology , Breast Neoplasms, Male/genetics , Breast Neoplasms, Male/metabolism , Child , Chromosomes, Human, Pair 22 , Humans , Karyotyping , Male , Monosomy
13.
Genes Chromosomes Cancer ; 24(1): 30-41, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892106

ABSTRACT

Supernumerary ring or giant rod marker chromosomes are a characteristic of well-differentiated liposarcomas (WDLPS) and atypical lipomas (ALP) and are often observed as the sole cytogenetic abnormality, but are rare in lipomas. Using a combination of different methods, we extensively investigated the structure and composition of rings and giant rods in a series of 17 WDLPS-ALP samples and three intra- or intermuscular lipomas (IMLP), revealing a unique combination of particular features strikingly related to these tumors. Although the rings and rods displayed in vitro and in vivo stability, the presence of alpha-satellites could not be detected on these supernumerary structures. Comparative genomic hybridization analysis, in combination with fluorescence in situ hybridization, identified the chromosomal regions contributing to the formation of these chromosomes: in WDLPS-ALP, all carried amplifications of 12q 14-15 and the MDM2 gene, with variable other noncontiguous regions. In the three IMLP, the rings consistently carried amplifications of 12q15-21 and 1q21, but increased copies of MDM2 were found in only one case. Other genes located more proximal in 12q14-15 were amplified in several WDLPS-ALP, but showed a normal copy number in IMLP. Furthermore, the immunohistochemical expression of the MDM2 protein was detected in most (12/14) WDLPS-ALP, in 1-30% of the cells, but never in IMLP. These supernumerary chromosomes represent a peculiar kind of amplification structure, midway between double minute chromosomes and homogeneously staining regions, but the mechanisms underlying the formation of these structures remain obscure.


Subject(s)
Chromosome Aberrations/pathology , Liposarcoma/genetics , Liposarcoma/pathology , Neoplasms, Adipose Tissue/genetics , Neoplasms, Adipose Tissue/pathology , Nuclear Proteins , Ring Chromosomes , Aged , Aged, 80 and over , Blotting, Southern , Centromere/chemistry , Centromere/genetics , Centromere/pathology , Chromosome Aberrations/genetics , Chromosome Disorders , Chromosomes, Human, Pair 12/genetics , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/genetics , DNA Probes , Female , Humans , In Situ Hybridization, Fluorescence , Liposarcoma/chemistry , Male , Middle Aged , Neoplasms, Adipose Tissue/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2
14.
Genes Chromosomes Cancer ; 23(3): 263-6, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9790508

ABSTRACT

Dermatofibrosarcoma protuberans (DP), an infiltrative skin tumor of intermediate malignancy, presents specific cytogenetic features such as reciprocal translocations t(17;22)(q22;q13.1) or, more often, supernumerary ring chromosomes derived from t(17;22). Different translocations, including t(2;17) and t(X;7), have also been described. We have shown previously that both r(17;22) and t(17;22) present the same molecular rearrangement fusing the COL1A1 gene on chromosome 17 and the PDGFB gene on chromosome 22. Out of our series of 16 DPs, we detected an extra ring chromosome in tumor T96-1175, which juxtaposed sequences from chromosomes 4 and 17. As shown by fluorescence in situ hybridization (FISH) using chromosome painting and alpha-satellite probes, T96-1175 apparently lacked chromosome 22 material in the ring. However, involvement of chromosome 22 through a rearrangement of PDGFB was shown by Southern blotting, reverse transcriptase-polymerase chain reaction (RT-PCR), and FISH. This study demonstrates that a cryptic molecular rearrangement between chromosomes 17 and 22 occurred in addition to the recombination of chromosomes 4 and 17 initially identified by FISH. Assessment for cryptic molecular events should be performed in other variant DP rearrangements.


Subject(s)
Collagen/genetics , Dermatofibrosarcoma/genetics , Platelet-Derived Growth Factor/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Ring Chromosomes , Adult , Chromosome Banding , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Proto-Oncogene Proteins c-sis , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic/genetics
16.
Genes Chromosomes Cancer ; 23(2): 187-93, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9739023

ABSTRACT

Dermatofibrosarcoma protuberans (DFSP) and its juvenile form, giant-cell fibroblastoma (GCF), are uncommon infiltrative tumors of the dermis, which present unique cytogenetic features, such as the reciprocal translocation t(17;22) or, more commonly, supernumerary ring chromosomes containing sequences from chromosomes 17 and 22. We have recently shown that these aberrations are cytogenetic manifestations of gene fusions between the platelet-derived growth factor B-chain gene (PDGFB), the cellular equivalent of the v-sis oncogene, and the collagen type 1 alpha 1 gene (COL1A1), the major protein constituent of the extracellular matrix in connective tissue of skin. We now report characterization of COL1A1/PDGFB chimeric genes at the RNA and DNA sequence levels in a series of DFSPs and GCFs. All 16 tumors studied contained the COL1A1/PDGFB gene. The location of breakpoints within COL1A1 varied greatly, but was always limited to the region encoding the alpha-helical domain. The PDGFB segment of the chimeric transcript always starts with exon 2, placing PDGFB under the control of the COL1A1 promoter and removing all known elements negatively controlling PDGFB transcription and translation. Production of these aberrant transcripts in fibroblasts, the suspected cell of origin of DFSP/GCF, likely causes autocrine stimulation and cell proliferation. No specific function has yet been assigned to exon 2 of PDGFB, and this exon does not encode for the mature growth factor. Its retention in all chimeric COL1A1/PDGFB genes suggests that it is important for the normal processing of the PDGFB polypeptide.


Subject(s)
Collagen/genetics , Dermatofibrosarcoma/genetics , Exons/genetics , Fibroma/genetics , Giant Cell Tumors/genetics , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins/genetics , Skin Neoplasms/genetics , Translocation, Genetic/genetics , Adult , Child, Preschool , Dermatofibrosarcoma/chemistry , Female , Fibroma/chemistry , Giant Cell Tumors/chemistry , Humans , Infant , Male , Middle Aged , Molecular Sequence Data , Platelet-Derived Growth Factor/chemistry , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-sis , Skin Neoplasms/chemistry
17.
Arch Pediatr ; 5(12): 1338-40, 1998 Dec.
Article in French | MEDLINE | ID: mdl-9885740

ABSTRACT

BACKGROUND: Clinical features of Jacobsen syndrome include facial dysmorphism, congenital heart defects, digit anomalies and mild to moderate psychomotor retardation. Thrombocytopenia or pancytopenia is observed in one half of patients. PATIENTS: Two unrelated children, a 6-month- and a 12-year-old, presented with a moderate thrombocytopenia associated with the clinical features of Jacobsen syndrome. Bone marrow aspirates showed, in both patients, normal cellularity with an increased number of micromegacaryocytes. Chromosome analysis showed a partial deletion of the long arm of chromosome 11. The 12-year-old patient had a history of upper respiratory airways infections with immune humoral deficiency (decreased level of IgA and IgM) which, to our knowledge, has never been reported. CONCLUSION: Jacobsen syndrome is a cause of inherited thrombocytopenia in children. Humoral immune functions must be explored in patients with a history of repeated infections.


Subject(s)
Dysgammaglobulinemia/complications , Face/abnormalities , Fingers/abnormalities , Heart Defects, Congenital/complications , IgA Deficiency/complications , Immunoglobulin M/deficiency , Psychomotor Disorders/complications , Thrombocytopenia/complications , Child , Chromosome Deletion , Chromosomes, Human, Pair 11 , Female , Humans , Infant , Male , Syndrome
18.
Genomics ; 46(1): 70-7, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9403060

ABSTRACT

Cytogenetic studies of banded chromosomes and fluorescence in situ hybridization (FISH) of several yeast artificial chromosomes (YACs) that are part of a 128-kb resolution physical map of a portion of 12q13 revealed that 4/14 (28%) lipomas have breakpoints in 12q13. These breakpoints are more than 10 Mb away from the HMGIC gene at 12q14-q15, which is known to be modified in some lipomas. FISH with individual YACs at 12q13 enabled us to identify four YACs that span three breakpoints. Our results suggest that genes other than HMGIC on human chromosome 12 may be involved in the etiology of lipoma development.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Lipoma/genetics , Translocation, Genetic/genetics , Chromosome Banding , Chromosome Mapping , Chromosomes, Artificial, Yeast , Genes, Neoplasm/genetics , Humans , In Situ Hybridization, Fluorescence
19.
Genomics ; 44(2): 188-94, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9299235

ABSTRACT

The Escherichia coli MutHLS system has been highly conserved throughout evolution. The eukaryotic pathway results in a specialization of MutS homologs that have evolved to play crucial roles in both DNA mismatch repair and meiotic recombination. So far, meiosis-specific genes belonging to this family have only been identified in yeast. In Saccharomyces cerevisiae, MSH4 (MutS homolog 4) is a meiosis-specific protein that is not involved in mismatch correction. This protein is required for reciprocal recombination and proper segregation of homologous chromosomes at meiosis I. We have identified the human MSH4 homolog gene. The predicted amino acid sequence shows 28.7% identity with the S. cerevisiae MSH4 protein. By Northern blot analysis, human MSH4 transcripts are only detectable in testis and in ovary with a lower level of expression. We have mapped MSH4 to human chromosome 1 at band p31 by fluorescence in situ hybridization. The identification of such a gene provides a powerful tool for clarifying the respective roles of MSH and MLH genes in mammalian meiosis.


Subject(s)
Adenosine Triphosphatases , Bacterial Proteins/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins , Escherichia coli Proteins , Meiosis/genetics , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 1/genetics , Cloning, Molecular , DNA Primers/genetics , DNA, Complementary/genetics , Escherichia coli/genetics , Evolution, Molecular , Female , Fungal Proteins/genetics , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Male , Molecular Sequence Data , MutS DNA Mismatch-Binding Protein , Saccharomyces cerevisiae/genetics
20.
Cancer Genet Cytogenet ; 97(1): 12-9, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9242212

ABSTRACT

The t(11;22)(q24;q12) translocation is found in about 85% of Ewing tumors and leads in all cases to an EWS/FLI1 fusion gene. In a few instances, complex translocations, involving chromosomes 11, 22 and a third chromosome or other variant translocations not involving chromosome 11 also have been reported. These rearrangements generate an active fusion gene between the EWS gene and either the human FLI1 gene or other members of the ETS gene family: the ERG gene localized in band 21q22.2, the ETV1 gene localized in band 7p22, or the E1AF gene localized in band 17q12. Using fluorescence in situ hybridization (FISH) on interphase nuclei or metaphases, we report here the characterization of particular karyotypes in Ewing tumors, namely a complex t(2;11;22) translocation, a variant t(12;22) translocation, and one Ewing tumor without specific rearrangements but with an EWS/ERG fusion gene demonstrated by molecular analysis. These molecular cytogenetic methods allowed us (1) to precisely localize the genomic breakpoints within-EWSR1 and EWSR2 and to identify the chromosome carrying the fusion gene; (2) to determine the nature of events generating the fusion genes; (3) to demonstrate that some variant translocations represent masked complex translocations; and (4) to show that the generation of an EWS/ERG fusion gene cannot be obtained through a simple balanced translocation.


Subject(s)
Chromosome Aberrations , DNA-Binding Proteins/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins , Ribonucleoproteins/genetics , Sarcoma, Ewing/genetics , Trans-Activators/genetics , Translocation, Genetic , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 2/genetics , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 22/genetics , Cosmids , Heterogeneous-Nuclear Ribonucleoproteins , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL