Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Leukemia ; 31(4): 821-828, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27733777

RESUMO

The contribution of genetic predisposing factors to the development of pediatric acute lymphoblastic leukemia (ALL), the most frequently diagnosed cancer in childhood, has not been fully elucidated. Children presenting with multiple de novo leukemias are more likely to suffer from genetic predisposition. Here, we selected five of these patients and analyzed the mutational spectrum of normal and malignant tissues. In two patients, we identified germline mutations in TYK2, a member of the JAK tyrosine kinase family. These mutations were located in two adjacent codons of the pseudokinase domain (p.Pro760Leu and p.Gly761Val). In silico modeling revealed that both mutations affect the conformation of this autoregulatory domain. Consistent with this notion, both germline mutations promote TYK2 autophosphorylation and activate downstream STAT family members, which could be blocked with the JAK kinase inhibitor I. These data indicate that germline activating TYK2 mutations predispose to the development of ALL.


Assuntos
Mutação em Linhagem Germinativa , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , TYK2 Quinase/genética , Alelos , Substituição de Aminoácidos , Exoma , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Modelos Moleculares , Fosforilação , Polimorfismo de Nucleotídeo Único , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Fatores de Transcrição STAT/metabolismo , TYK2 Quinase/química , TYK2 Quinase/metabolismo
2.
Cytogenet Genome Res ; 135(3-4): 212-21, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21934286

RESUMO

Array-based comparative genomic hybridization analysis of genomic DNA was first applied in postnatal diagnosis for patients with intellectual disability (ID) and/or congenital anomalies (CA). Genome-wide single-nucleotide polymorphism (SNP) array analysis was subsequently implemented as the first line diagnostic test for ID/CA patients in our laboratory in 2009, because its diagnostic yield is significantly higher than that of routine cytogenetic analysis. In addition to the detection of copy number variations, the genotype information obtained with SNP array analysis enables the detection of stretches of homozygosity and thereby the possible identification of recessive disease genes, mosaic aneuploidy, or uniparental disomy. Patient-parent (trio) information analysis is used to screen for the presence of any form of uniparental disomy in the patient and can determine the parental origin of a de novo copy number variation. Moreover, the outcome of a genotype analysis is used as a final quality control by ruling out potential sample mismatches due to non-paternity or sample mix-up. SNP array analysis is now also used in our laboratory for patients with disorders for which locus heterogeneity is known (homozygosity pre-screening), in prenatal diagnosis in case of structural ultrasound anomalies, and for patients with leukemia. In this report, we summarize our array findings and experiences in the various diagnostic applications and demonstrate the power of a SNP-based array platform for molecular karyotyping, because it not only significantly improves the diagnostic yield in both constitutional and cancer genome diagnostics, but it also enhances the quality of the diagnostic laboratory workflow.


Assuntos
Hibridização Genômica Comparativa/métodos , Variações do Número de Cópias de DNA , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Polimorfismo de Nucleotídeo Único , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Hibridização Genômica Comparativa/normas , Anormalidades Congênitas/diagnóstico , Anormalidades Congênitas/genética , Interpretação Estatística de Dados , Feminino , Genótipo , Homozigoto , Humanos , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/genética , Masculino , Análise de Sequência com Séries de Oligonucleotídeos/normas , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Gravidez , Diagnóstico Pré-Natal/métodos , Valores de Referência
3.
Cytogenet Genome Res ; 123(1-4): 188-94, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19287155

RESUMO

Although cancer is mostly regarded as an acquired disease, familial predisposition plays a significant role in many cancer types. Thus far, several high penetrant cancer predisposing genes have been identified. As yet, however, these genes explain only a fraction of the familial and/or hereditary cases of cancer. This has led to the exploration of the human genome for novel cancer predisposing genes. The identification of such genes will not only increase our understanding of cancer predisposition and development, but will also have direct implications for genetic counseling and personalized management of the patients and their family members. Here we provide an inventory of currently known molecular mechanisms related to familial colorectal cancer development and an outline of copy number analysis-based strategies to identify new predisposing genes. Finally, we discuss a novel copy number-associated epigenetic mechanism underlying the predisposition to colorectal cancer.


Assuntos
Neoplasias Colorretais/genética , Predisposição Genética para Doença/genética , Alelos , Perfilação da Expressão Gênica , Humanos , Linhagem
4.
Cytogenet Genome Res ; 118(2-4): 157-65, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18000366

RESUMO

Renal cell carcinomas (RCCs) represent a heterogeneous group of neoplasms, which differ in histological, pathologic and clinical characteristics. The tumors originate from different locations within the nephron and are accompanied by different recurrent (cyto)genetic anomalies. Recently, a novel subgroup of RCCs has been defined, i.e., the MiT translocation subgroup of RCCs. These tumors originate from the proximal tubule of the nephron, exhibit pleomorphic histological features including clear cell morphologies and papillary structures, and are found predominantly in children and young adults. In addition, these tumors are characterized by the occurrence of recurrent chromosomal translocations, which result in disruption and fusion of either the TFE3 or TFEB genes, both members of the MiT family of basic helix-loop-helix/leucine-zipper transcription factor genes. Hence the name MiT translocation subgroup of RCCs. In this review several features of this RCC subgroup will be discussed, including the molecular mechanisms that may underlie their development.


Assuntos
Carcinoma de Células Renais/genética , Neoplasias Renais/genética , Translocação Genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Carcinoma de Células Renais/patologia , Fusão Gênica , Humanos , Neoplasias Renais/patologia , Proteínas de Neoplasias/genética
5.
Cytogenet Genome Res ; 115(3-4): 289-97, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17124412

RESUMO

Insulinomas represent the predominant syndromic subtype of endocrine pancreatic tumors (EPTs). Their metastatic potential cannot be predicted reliably using histopathological criteria. In the past few years, several attempts have been made to identify prognostic markers, among them TP53 mutations and immunostaining of p53 and recently cytokeratin 19 (CK19). In a previous study using conventional comparative genomic hybridization (CGH) we have shown that chromosomal instability (CIN) is associated with metastatic disease in insulinomas. It was our aim to evaluate these potential parameters in a single study. For the determination of CIN, we applied CGH to microarrays because it allows a high-resolution detection of DNA copy number changes in comparison with conventional CGH as well as the analysis of chromosomal regions close to the centromeres and telomeres, and at 1pter-->p32, 16p, 19 and 22. These regions are usually excluded from conventional CGH analysis, because they may show DNA gains in negative control hybridizations. Array CGH analysis of 30 insulinomas (15 tumors of benign, eight tumors of uncertain and seven tumors of malignant behavior) revealed that >or=20 chromosomal alterations and >or=6 telomeric losses were the best predictors of malignant progression. A subset of 22 insulinomas was further investigated for TP53 exon 5-8 gene mutations, and p53 and CK19 expression. Only one malignant tumor was shown to harbor an arginine 273 serine mutation and immunopositivity for p53. CK19 immunopositivity was detected in three malignant tumors and one tumor with uncertain behavior. In conclusion, our results indicate that CIN as well as telomeric loss are very powerful indicators for malignant progression in sporadic insulinomas. Our data do not support a critical role for p53 and CK19 as molecular parameters for this purpose.


Assuntos
Biologia Computacional/métodos , Insulinoma/diagnóstico , Insulinoma/patologia , Queratina-19/genética , Neoplasias Pancreáticas/genética , Proteína Supressora de Tumor p53/genética , Instabilidade Cromossômica , Análise Mutacional de DNA , Progressão da Doença , Éxons , Feminino , Humanos , Masculino , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Pancreáticas/patologia , Prognóstico
6.
Hum Mol Genet ; 15(19): 2936-44, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16926188

RESUMO

The synovial sarcoma-associated protein SS18 (also known as SYT or SSXT) is thought to act as a transcriptional co-activator. This activity appears to be mediated through the SWI/SNF proteins BRG1 and INI1 and the histone acetyl transferase p300. Here, we report that disruption of the mouse Ss18 gene results in a recessive embryonic lethal phenotype, due to placental failure caused by impairment of placental vascularization and/or chorio-allantoic fusion. This phenotype resembles the p300 knockout phenotype, but is distinct from the Brg1 and Ini1 knockout phenotypes. Through expression profiling of knockout embryos, we observed altered expression of genes known to affect placental development, including the peroxisome proliferator-activated receptor-binding protein (Pparbp). Since Pparbp null mutant embryos display a similar, lethal phenotype with placental failure, we suggest that the functional and phenotypic co-linearities between Ss18 and p300 may also include the transcriptional co-activator Pparbp. Additional interbreeding of Ss18 and Ss18l1 (Crest) mutant mice indicates that these two functionally and structurally related genes may act synergistically during critical stages of embryonic development.


Assuntos
Perda do Embrião/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Sarcoma Sinovial/genética , Fatores de Transcrição/genética , Animais , Feminino , Expressão Gênica , Marcação de Genes , Genes Letais , Genes Recessivos , Humanos , Subunidade 1 do Complexo Mediador , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Gravidez
7.
Cytogenet Genome Res ; 112(3-4): 222-6, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16484776

RESUMO

The highly conserved synovial sarcoma associated protein SS18 is thought to act as a transcriptional co-activator through interactions with various proteins involved in (epigenetic) gene regulation. The SS18 SNH domain appears to act as a major interface for these protein-protein interactions. Previously, we used this SNH domain to identify SS18 paralogs (SS18L1 and SS18L2) and orthologs in various species. The functional significance of these SS18-like proteins is embodied by the observations that SS18L1 and SS18L2 can replace SS18 in its various protein-protein interactions, and that SS18L1 may act as a fusion partner of SSX in synovial sarcoma. In the current study, we performed a comprehensive comparison of SNH-containing loci in several distinct species. By doing so, we found that the vertebrate SS18 and SS18L1 genes map within co-linear DNA segments that may have evolved through a relatively recent genomic duplication event. An additional phylogenetic study indicated that the more divergent SS18L2 gene is most likely derived from an earlier gene duplication event, again in the vertebrate lineage.


Assuntos
Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Sarcoma Sinovial/genética , Transativadores/genética , Algoritmos , Animais , Caenorhabditis elegans/genética , Galinhas/genética , Mapeamento Cromossômico , Evolução Molecular , Duplicação Gênica , Humanos , Camundongos/genética , Família Multigênica , Filogenia , Vertebrados/genética
8.
Cytogenet Genome Res ; 106(1): 68-73, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15218244

RESUMO

Recently we found that the human papillary renal cell carcinoma-associated protein PRCC interacts with the cell cycle control protein Mad2B, and translocates this protein to the nucleus where it exerts its mitotic checkpoint function. Here we have successfully isolated Xenopus laevis Mad2B and PRCC cDNAs. The full-length xMad2B cDNA encodes a 211 amino acid protein that is highly homologous to human Mad2B, thus pointing to an important function for this protein in higher eukaryotes. The full-length xPRCC cDNA encodes a 544 amino acid protein. Remarkably, this protein contains an amino-terminal region distinct from that in mouse and human, whereas the C-terminal region is highly conserved. Northern blot and RT-PCR analyses revealed a relatively low expression of both xMad2B and xPRCC in most tissues examined. However, an abundant expression was observed in testis and oocyte, indicating a role in meiotic division processes. Coimmunoprecipitation and immunofluorescence analyses revealed that, despite its distinct amino terminus, the xPRCC-protein is still capable of interacting with xMad2B and of shuttling this protein to the nucleus. Therefore, the well-established animal model Xenopus laevis can be used as a powerful system to study in detail the role of xPRCC and xMad2B in the intricate processes of cell cycle control.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Ciclo Celular/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Chlorocebus aethiops , Embrião não Mamífero/metabolismo , Feminino , Proteínas Mad2 , Masculino , Dados de Sequência Molecular , Oócitos/metabolismo , Transporte Proteico , RNA Mensageiro/análise , Homologia de Sequência de Aminoácidos , Testículo/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
9.
Leukemia ; 18(6): 1108-14, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15085164

RESUMO

Chromosomal rearrangements involving 3q26 either due to inversion or translocation with various partner chromosomes are a recurrent finding in malignant myeloid disorders. Typically, these chromosome aberrations contribute to ectopic expression of or to the formation of fusion genes involving the EVI1 proto-oncogene. Chromosomal translocations involving the short arm of chromosome 2 (p15-p23) and the distal part of the long arm of chromosome 3 (q26-q27) are a rare but recurrent finding in patients with myeloid malignancies, and are assumed to be part of this spectrum of disorders. Thus far, however, these translocations have been poorly studied. Here, we present 21 new cases with myelodysplasia, acute myeloid leukemia or CML in blast crisis, which upon karyotyping showed the presence of a t(2;3). Furthermore, an extensive literature review disclosed 29 additional cases. Morphological, clinical and cytogenetic assessment revealed the typical hallmarks of 3q26/EVI1 rearrangements, that is, trilineage dysplasia and dysmegakaryopoiesis, poor prognosis and additional monosomy 7. Molecular cytogenetic analysis and PCR in selected samples indicated that in most cases the translocation indeed targets the EVI1 locus. Mapping of the chromosome 2 breakpoints confirmed the initially suspected cytogenetic breakpoint heterogeneity at the 2p arm.


Assuntos
Cromossomos Humanos Par 2 , Cromossomos Humanos Par 3 , Leucemia Mieloide/genética , Leucemia Mieloide/patologia , Translocação Genética , Doença Aguda , Adulto , Idoso , Criança , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade , Proto-Oncogene Mas
10.
Genet Couns ; 14(2): 149-54, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12872808

RESUMO

Renal cell carcinomas (RCC) occur in both sporadic and familial forms. The best known example of a familial RCC syndrome is the Von Hippel Lindau cancer syndrome. In addition, RCC families segregating constitutional chromosome 3 translocations have been reported. The list of these latter families is rapidly expanding. We have initiated a survey of all Dutch families known to segregate chromosome 3 translocations for (i) the ocurrence of RCCs and (ii) the establishment of refined risk estimates. This information will be critical for genetic counseling and clinical patient management. Within the families 'at risk' that we have identified so far, this approach has already led to early RCC detection and surgical intervention.


Assuntos
Carcinoma de Células Renais/genética , Cromossomos Humanos Par 3/genética , Neoplasias Renais/genética , Translocação Genética/genética , Proteínas Supressoras de Tumor , Ubiquitina-Proteína Ligases , Feminino , Humanos , Ligases/genética , Masculino , Pessoa de Meia-Idade , Mutação Puntual/genética , Fatores de Risco , Proteína Supressora de Tumor Von Hippel-Lindau
12.
Leukemia ; 16(1): 13-21, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11840258

RESUMO

Using red cell phenotyping (RCP) and/or cytogenetics (CYT) we identified 19 patients with persisting mixed chimerism (MC) among 231 patients transplanted with partially T cell-depleted stem cell grafts from HLA-identical siblings. Persisting MC is defined as MC for more than 2 years in patients without any evidence of relapse. Median leukemia-free survival in these patients was 150 (range, 50-218) months. Diagnoses were ALL (n= 10); AML (n = 2); CML (n = 2); NHL (n = 2); MDS (n= 1); MM (n = 1) and SAA (n = 1). Purpose of this study was the long-term follow-up of MC and definition of patterns of chimerism in the various subsets of PBMCs and granulocytes. Using a PCR-STR technique CD3(+)/CD4(+) (T4 lymphocytes), CD3(+)/CD8(+) (T8 lymphocytes), CD45(+)/CD19(+) (B lymphocytes), CD45(+)/CD14(+) (monocytes), CD45(+)/CD15(+) (granulocytes) and CD3(-)/CD56(+) (NK-cells) were analyzed. The majority of patients with persisting MC were conditioned with a less intensive conditioning regimen and had little GVHD. Sequential monitoring of the chimerism resulted in a group of patients (n = 7) with very slow transient mixed chimerism that resulted in complete DC after median 7 years. Another nine patients had a relatively high percentage of persisting autologous cells for a median of 12 years and in three patients we observed a stable low percentage of autologous cells. Only two out of 19 patients (AML-CR1, CML-CP1) relapsed during follow-up. Both patients had a relatively high percentage of autologous cells. Chimerism in granulocytes and PBMC subsets was analyzed at a median of 8 years after SCT in nine patients. In five patients mixed chimerism simultaneously detected by RCP and CYT was associated with MC in all subsets. Within each individual patient the percentages of donor and recipient cells were very different between the different subsets. Two CML-CP1 patients were mixed chimera in only two subsets and in one patient these subsets represented pending relapse. In another two patients mixed chimerism with a very low number of autologous red cells was not found in the PBMCs because of the different sensitivity level of the RCP and the PCR-STR technique. We conclude that in patients with persisting mixed chimerism after partially T cell-depleted SCT a remarkable number of patients had lymphoid malignancies, the majority of the patients were conditioned with less intensive conditioning regimens and the mixed chimerism was not correlated with relapse. Chimerism in granulocytes and PBMC subsets did show great intra-individual differences in the subsets and these data correlated well with RCP and CYT data with the exception of the NK cells.


Assuntos
Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Adolescente , Adulto , Sobrevivência Celular , Intervalo Livre de Doença , Feminino , Seguimentos , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro/epidemiologia , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/prevenção & controle , Neoplasias Hematológicas/mortalidade , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Imunofenotipagem , Contagem de Linfócitos , Depleção Linfocítica , Subpopulações de Linfócitos , Masculino , Pessoa de Meia-Idade , Células Mieloides , Reação em Cadeia da Polimerase , Sensibilidade e Especificidade , Linfócitos T , Condicionamento Pré-Transplante , Transplante Homólogo
13.
Lab Invest ; 81(7): 919-28, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11454979

RESUMO

It is generally accepted that testicular seminomas and spermatocytic seminomas have separate pathogeneses, although the origin of these two types of germ cell tumors of the adult testis remains a matter of debate. Although an embryonic germ cell origin seems to be most likely for seminomas, a spermatogonia-spermatocyte origin has been suggested for spermatocytic seminoma. To shed more light on the etiology of spermatocytic seminomas, we undertook an immunohistochemical and molecular approach using SCP1 (synaptonemal complex protein 1), SSX (synovial sarcoma on X chromosome), and XPA (xeroderma pigmentosum type A) as targets. Although a stage-specific expression pattern has been reported for SCP1 and SSX in normal spermatogenesis, we demonstrate here that it also exists for XPA. In fact, immunohistochemistry shows that the proteins of SCP1 and XPA are specifically present in the stage of primary and pachytene spermatocytes. In contrast, SSX was found in spermatogonia and primary spermatocytes, as well as in germ cells, from at least the 17th week of intrauterine development onward. Although no protein encoded by any of these genes was detected in tumor cells of a series of testicular seminomas, all tested spermatocytic seminomas were positive, in agreement with expression analysis. These data support the model that seminomas originate from an embryonic germ cell, and they imply that the cell of origin of spermatocytic seminomas is at least capable of maturing to the stage of spermatogonia-pachytene spermatocyte.


Assuntos
Células Germinativas/citologia , Seminoma/patologia , Neoplasias Testiculares/patologia , Especificidade de Anticorpos , Sequência de Bases , Primers do DNA , Proteínas de Ligação a DNA , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteolipídeos/genética , Proteínas Associadas a Surfactantes Pulmonares , Surfactantes Pulmonares/genética , RNA Mensageiro/genética , Proteínas Repressoras/genética , Seminoma/diagnóstico , Seminoma/etiologia , Coloração e Rotulagem , Neoplasias Testiculares/diagnóstico , Neoplasias Testiculares/etiologia
14.
Cytogenet Cell Genet ; 92(3-4): 310-9, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11435705

RESUMO

We have previously isolated and characterized a mouse cDNA orthologous to the human synovial sarcoma associated SS18 (formerly named SSXT and SYT) cDNA. Here, we report the characterization of the genomic structure of the mouse Ss18 gene. Through in silico methods with sequence information contained in the public databases, we did the same for the human SS18 gene and two human SS18 homologous genes, SS18L1 and SS18L2. In addition, we identified a mouse Ss18 processed pseudogene and mapped it to chromosome 1, band A2-3. The mouse Ss18 gene, which is subject to extensive alternative splicing, is made up of 11 exons, spread out over approximately 45 kb of genomic sequence. The human SS18 gene is also composed of 11 exons with similar intron-exon boundaries, spreading out over about 70 kb of genomic sequence. One alternatively spliced exon, which is not included in the published SS18 cDNA, corresponds to a stretch of sequence which we previously identified in the mouse Ss18 cDNA. The human SS18L1 gene, which is also made up of 11 exons with similar intron-exon boundaries, was mapped to chromosome 20 band q13.3. The smaller SS18L2 gene, which is composed of three exons with similar boundaries as the first three exons of the other three genes, was mapped to chromosome 3 band p21. Through sequence and mutation analyses this gene could be excluded as a candidate gene for 3p21-associated renal cell cancer. In addition, we created a detailed BAC map around the human SS18 gene, placing it unequivocally between the CA-repeat marker AFMc014wf9 and the dihydrofolate reductase pseudogene DHFRP1. The next gene in this map, located distal to SS18, was found to be the TBP associated factor TAFII-105 (TAF2C2). Further analogies between the mouse Ss18 gene, the human SS18 gene and its two homologous genes were found in the putative promoter fragments. All four promoters resemble the promoters of housekeeping genes in that they are TATA-less and embedded in canonical CpG islands, thus explaining the high and widespread expression of the SS18 genes.


Assuntos
Cromossomos Humanos Par 20/genética , Cromossomos Humanos Par 3/genética , Proteínas/genética , Pseudogenes/genética , Sarcoma Sinovial/genética , Processamento Alternativo/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Mapeamento de Sequências Contíguas , Ilhas de CpG/genética , Éxons/genética , Humanos , Hibridização in Situ Fluorescente , Íntrons/genética , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Proteínas/química , Proteínas Proto-Oncogênicas , Sítios de Splice de RNA/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Proteínas Repressoras , Elementos de Resposta/genética , Homologia de Sequência , Fatores de Transcrição/metabolismo
15.
Cytogenet Cell Genet ; 92(3-4): 326-32, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11435707

RESUMO

In a subset of papillary renal cell carcinomas a t(X;1)(p11;q21) chromosome translocation has repeatedly been reported. Positional cloning has demonstrated that, as a result of this translocation, the transcription factor TFE3 gene on the X-chromosome becomes fused to a novel gene, PRCC, on chromosome 1. Since as yet little is known about the function of PRCC, we sought to identify the mouse counterpart of the PRCC gene. Isolation and sequence analysis of a mouse Prcc cDNA revealed a high level of conservation between man and mouse, both at the nucleotide and protein level. As the human PRCC gene, the mouse Prcc gene is ubiquitously expressed. It shows low expression in all mouse fetal tissues examined. In addition, we identified a genomic cosmid clone containing the complete Prcc gene. The mouse Prcc gene consists of seven exons, all of which contain coding sequences. The small second exon, which was found to be located adjacent to the t(X;1) breakpoint in the human gene on chromosome 1, is also conserved between man and mouse. In mouse, Prcc is located on chromosome 3. These cDNA and genomic clones will be instrumental in the creation of mouse models for a further elucidation of the function of PRCC.


Assuntos
Carcinoma Papilar/genética , Carcinoma de Células Renais/genética , Proteínas de Ciclo Celular , Éxons/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Neoplasias , Mapeamento Físico do Cromossomo , Proteínas/genética , Sequência de Aminoácidos , Animais , Cromossomos/genética , Clonagem Molecular , Hibridização in Situ Fluorescente , Íntrons/genética , Camundongos , Dados de Sequência Molecular , Proteínas/química , RNA Mensageiro/análise , RNA Mensageiro/genética , Alinhamento de Sequência
16.
Oncogene ; 20(25): 3281-9, 2001 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-11423977

RESUMO

As a result of the synovial sarcoma associated t(X;18) translocation, the human SYT gene on chromosome 18 is fused to either the SSX1 or the SSX2 gene on the X chromosome. Although preliminary evidence indicates that the (fusion) proteins encoded by these genes may play a role in transcriptional regulation, little is known about their exact function. We set out to isolate interacting proteins through yeast two hybrid screening of a human cDNA library using SYT as a bait. Of the positive clones isolated, two were found to correspond to the acute leukemia t(10;11) associated AF10 gene, a fusion partner of MLL. Confirmation of these results was obtained via co-immunoprecipitation of endogenous and exogenous, epitope-tagged, SYT and AF10 proteins from cell line extracts and colocalization of epitope-tagged SYT and AF10 proteins in transfected cells. Subsequent sequential mutation analysis revealed a highly specific interaction of N-terminal SYT fragments with C-terminal AF10 fragments. The N-terminal interaction domain of the SYT protein was also found to be present in several SYT orthologs and homologs. The C-terminal interaction domain of AF10 is located outside known functional domains. Based on these results, a model is proposed in which the SYT and AF10 proteins act in concert as bipartite transcription factors. This model has implications for the molecular mechanisms underlying the development of both human synovial sarcomas and acute leukemias.


Assuntos
Proteínas de Neoplasias/metabolismo , Proteínas/metabolismo , Sarcoma Sinovial/metabolismo , Fatores de Transcrição/metabolismo , Doença Aguda , Sequência de Aminoácidos , Animais , Sítios de Ligação , Humanos , Leucemia/genética , Camundongos , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/genética , Proteínas Proto-Oncogênicas , Proteínas Repressoras , Sarcoma Sinovial/genética , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/genética , Técnicas do Sistema de Duplo-Híbrido
17.
Genes Chromosomes Cancer ; 31(1): 23-32, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11284032

RESUMO

We identified a novel familial case of clear-cell renal cancer and a t(3;6)(q12;q15). Subsequent cytogenetic and molecular analyses showed the presence of several abnormalities within tumour samples obtained from different patients. Loss of the der(3) chromosome was noted in some, but not all, of the samples. A concomitant VHL gene mutation was found in one of the samples. In addition, cytogenetic and molecular evidence for heterogeneity was obtained through analysis of several biopsy samples from one of the tumours. Based on these results and those reported in the literature, we conclude that loss of der(3) and subsequent VHL gene mutation may represent critical steps in the development of renal cell cancers in persons carrying the chromosome 3 translocation. Moreover, preliminary data suggest that other (epi)genetic changes may be related to tumour initiation.


Assuntos
Carcinoma de Células Renais/genética , Cromossomos Humanos Par 3/genética , Cromossomos Humanos Par 6/genética , Neoplasias Renais/genética , Translocação Genética/genética , Adulto , Feminino , Humanos , Hibridização in Situ Fluorescente , Cariotipagem/métodos , Perda de Heterozigosidade/genética , Masculino , Pessoa de Meia-Idade , Hibridização de Ácido Nucleico/métodos , Linhagem , Células Tumorais Cultivadas
18.
Oncogene ; 20(12): 1414-24, 2001 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-11313885

RESUMO

A recurrent chromosomal abnormality associated with a subset of papillary renal cell carcinomas is t(X;1)(p11;q21). This translocation leads to the formation of two fusion genes, TFE3PRCC and the reciprocal product PRCCTFE3. Both fusion genes are expressed in t(X;1)-positive renal cell carcinomas and contain major parts of the coding regions of the parental transcription factor PRCC and TFE3 genes, respectively. To find out whether these fusion genes possess transforming capacity, we transfected NIH3T3 and rat-1 cells with the fusion products, either separately or combined. When using soft agar assays, we observed colony formation in all cases. NIH3T3 cells transfected with PRCCTFE3 or PRCCTFE3 together with TFE3PRCC yielded the highest colony forming capacities. Examination of other characteristics associated with malignant transformation, i.e., growth under low-serum conditions and formation of tumors in athymic nude mice, revealed that cells transfected with PRCCTFE3 exhibited all these transformation-associated characteristics. Upon transfection of the fusion products into conditionally immortalized kidney cells, derived from the proximal tubules of an H-2Kb-tsA58 transgenic mouse, and consecutive incubation under non-permissive conditions, growth arrest was observed, followed by differentiation except for those cells transfected with PRCCTFE3. Therefore, we conclude that PRCCTFE3 may be the t(X;1)-associated fusion product that is most critical for the development of papillary renal cell carcinomas.


Assuntos
Carcinoma de Células Renais/genética , Proteínas de Ciclo Celular , Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/genética , Neoplasias Renais/genética , Proteínas de Neoplasias , Proteínas de Fusão Oncogênica/genética , Fatores de Transcrição/genética , Células 3T3 , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Carcinoma Papilar/genética , Adesão Celular , Aberrações Cromossômicas , Transtornos Cromossômicos , Humanos , Rim/citologia , Camundongos , Proteínas/genética , Ratos , Transfecção , Translocação Genética
19.
Genomics ; 66(1): 35-42, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10843802

RESUMO

Previously, we located three novel human tumor-associated translocation breakpoints in the chromosome 11q13 region between the markers D11S4933 and D11S546. To facilitate the molecular analysis of these breakpoints, we have constructed a continuous sequence-ready cosmid and PAC contig of approximately 350 kb, including the markers D11S4933 and D11S546. In addition, a detailed transcript map was generated. This resulted in the precise positioning of 11 genes and ESTs within the contig, including 4 genes already known to map in the 11q13 region. Three other genes that we positioned within the contig showed homologies to unmapped genes from human and/or other species. Three ESTs were novel. Partial cosmid sequencing resulted in the establishment of the direction of transcription of several of the reported genes. This contig will be instrumental for the detailed characterization of the tumor-associated chromosomal breakpoints and the identification of other 11q13-associated disease genes.


Assuntos
Cromossomos Humanos Par 11 , Mapeamento de Sequências Contíguas , Cosmídeos/síntese química , Animais , Bacteriófagos , Quebra Cromossômica , Clonagem Molecular , DNA de Neoplasias , Etiquetas de Sequências Expressas , Perfilação da Expressão Gênica , Biblioteca Gênica , Marcadores Genéticos , Humanos , Hibridização in Situ Fluorescente , Metáfase , Dados de Sequência Molecular , Mapeamento por Restrição , Análise de Sequência de DNA , Sitios de Sequências Rotuladas , Translocação Genética , Células Tumorais Cultivadas
20.
Br J Haematol ; 108(1): 116-25, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10651734

RESUMO

Fifteen consecutive patients with Philadelphia chromosome (Ph)-positive chronic myeloid leukaemia (CML) who relapsed from T-cell-depleted bone marrow transplantation (BMT) were successfully treated with donor leucocyte infusions (DLIs). Chimaerism was analysed using red blood cell phenotyping (RCP), and the results were compared with cytogenetic analysis and outcome of qualitative and quantitative polymerase chain reaction (PCR) for breakpoint molecules. In all patients, an increase in autologous erythrocytes and/or a decrease in donor red cells indicated relapse. Donor erythrocytes started to increase from 4 to 20 (median 12) weeks after DLI. At 6 and 12 months after DLI, complete donor chimaerism was found in 11 and 15 patients, respectively, and all patients were in cytogenic remission. A high percentage of autologous red cells at the time of DLI predicted pancytopenia. During relapse and after DLI, the percentage of autologous red cells was strongly correlated with the reappearance and disappearance of Ph-positive metaphases (r = 0.90; P < 0.001 and r = 0.96; P < 0.001 respectively). The same was true for the correlation between the percentage of autologous red cells and positivity/negativity in PCR for Bcr-Abl breakpoint molecules (r = 0.94; P < 0.001). A normalized Bcr-Abl dose of greater than 10-3 in real-time quantitative PCR correlated well with relapse and the presence of autologous red blood cells (r = 0.77; P < 0.001). We conclude that RCP is a sensitive, easy to perform and fast technique for the prediction of pending relapse after BMT for CML. RCP also predicts the response to DLI and the occurrence of bone marrow aplasia after DLI.


Assuntos
Transplante de Medula Óssea/métodos , Eritrócitos/fisiologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Adulto , Idoso , Feminino , Proteínas de Fusão bcr-abl/genética , Doença Enxerto-Hospedeiro/etiologia , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/sangue , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Transfusão de Leucócitos/métodos , Masculino , Pancitopenia/etiologia , Fenótipo , Reação em Cadeia da Polimerase/métodos , Recidiva , Quimeras de Transplante/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA