Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Genes Chromosomes Cancer ; 52(2): 202-13, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23077088

RESUMEN

ETV6-RUNX1 fusion [t(12;21)(p13;q22)] occurs in 25% of childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL) and is associated with a favorable outcome. Additional abnormalities involving der(21)t(12;21) and nonrearranged chromosome 12 are well characterized but aberrations involving the der(12)t(12;21) have rarely been described. Herein, we describe two novel abnormalities affecting the der(12)t(12;21): a deletion (20/247, 8%) and duplication (10/247, 4%). All 30 patients were under 10 years of age, had a median white blood count of 12.4 × 10(9)/L and 19.2 × 10(9)/L, respectively, with a good outcome. Deletions of der(12)t(12;21) on both sides of the breakpoint were confirmed and mapped: centromeric (12p11.21-12p13.2) and telomeric (21q22.12-21q22.3). The size of these deletions extended from 0.4-13.4 to 0.8-2.5 Mb, respectively. The centromeric deletion encompassed the following genes: LRP6, BCL2L14, DUSP16, CREBL2, and CDKN1B. We postulate that this deletion occurs at the same time as the translocation because it was present in all ETV6-RUNX1-positive cells. A second abnormality representing duplication of the reciprocal RUNX1-ETV6 fusion gene was a secondary event, which we hypothesize arose through mitotic recombination errors. This led to the formation of the following chromosome: der(12)(21qter→21q22.12::12p13.2-12p12.3::12p12.3→12qter). Both abnormalities affect the reciprocal RUNX1-ETV6 fusion product which could either eliminate or amplify its expression and thus contribute to leukemogenesis. However, other consequences such as haploinsufficiency of tumor suppressor genes and amplification of oncogenes could also be driving forces behind these aberrations. In conclusion, this study has defined novel abnormalities in ETV6-RUNX1 BCP-ALL, which implicate new genes involved in leukemogenesis.


Asunto(s)
Cromosomas Humanos Par 12/genética , Cromosomas Humanos Par 21/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Translocación Genética , Centrómero/genética , Niño , Preescolar , Puntos de Rotura del Cromosoma , Deleción Cromosómica , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Fosfatasas de Especificidad Dual/genética , Femenino , Duplicación de Gen , Humanos , Hibridación Fluorescente in Situ , Lactante , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Masculino , Análisis por Micromatrices/métodos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Reacción en Cadena de la Polimerasa Multiplex , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-bcl-2/genética , Telómero/genética
2.
Blood ; 116(15): e56-65, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20616218

RESUMEN

To obtain a comprehensive genomic profile of presenting multiple myeloma cases we performed high-resolution single nucleotide polymorphism mapping array analysis in 114 samples alongside 258 samples analyzed by U133 Plus 2.0 expression array (Affymetrix). We examined DNA copy number alterations and loss of heterozygosity (LOH) to define the spectrum of minimally deleted regions in which relevant genes of interest can be found. The most frequent deletions are located at 1p (30%), 6q (33%), 8p (25%), 12p (15%), 13q (59%), 14q (39%), 16q (35%), 17p (7%), 20 (12%), and 22 (18%). In addition, copy number-neutral LOH, or uniparental disomy, was also prevalent on 1q (8%), 16q (9%), and X (20%), and was associated with regions of gain and loss. Based on fluorescence in situ hybridization and expression quartile analysis, genes of prognostic importance were found to be located at 1p (FAF1, CDKN2C), 1q (ANP32E), and 17p (TP53). In addition, we identified common homozygously deleted genes that have functions relevant to myeloma biology. Taken together, these analyses indicate that the crucial pathways in myeloma pathogenesis include the nuclear factor-κB pathway, apoptosis, cell-cycle regulation, Wnt signaling, and histone modifications. This study was registered at http://isrctn.org as ISRCTN68454111.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos/genética , Variaciones en el Número de Copia de ADN/genética , Mieloma Múltiple/genética , Anciano , Deleción Cromosómica , Femenino , Reordenamiento Génico de Cadena Pesada de Linfocito B , Humanos , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Pronóstico , Translocación Genética , Disomía Uniparental
3.
Genes Chromosomes Cancer ; 50(10): 765-74, 2011 10.
Artículo en Inglés | MEDLINE | ID: mdl-21961181

RESUMEN

Hemizygous deletion of 17p (del(17p)) has been identified as a variable associated with poor prognosis in myeloma, although its impact in the context of thalidomide therapy is not well described. The clinical outcome of 85 myeloma patients with del(17p) treated in a clinical trial incorporating both conventional and thalidomide-based induction therapies was examined. The clinical impact of deletion, low expression, and mutation of TP53 was also determined. Patients with del(17p) did not have inferior response rates compared to patients without del(17p), but, despite this, del(17p) was associated with impaired overall survival (OS) (median OS 26.6 vs. 48.5 months, P < 0.001). Within the del(17p) group, thalidomide induction therapy was associated with improved response rates compared to conventional therapy, but there was no impact on OS. Thalidomide maintenance was associated with impaired OS, although our analysis suggests that this effect may have been due to confounding variables. A minimally deleted region on 17p13.1 involving 17 genes was identified, of which only TP53 and SAT2 were underexpressed. TP53 was mutated in <1% in patients without del(17p) and in 27% of patients with del(17p). The higher TP53 mutation rate in samples with del(17p) suggests a role for TP53 in these clinical outcomes. In conclusion, del(17p) defined a patient group associated with short survival in myeloma, and although thalidomide induction therapy was associated with improved response rates, it did not impact OS, suggesting that alternative therapeutic strategies are required for this group.


Asunto(s)
Acetiltransferasas/genética , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Cromosomas Humanos Par 17/química , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Talidomida/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores Farmacológicos/análisis , Deleción Cromosómica , Mapeo Cromosómico , Cromosomas Humanos Par 17/genética , Análisis Mutacional de ADN , Femenino , Estudios de Seguimiento , Expresión Génica , Hemicigoto , Humanos , Hibridación Fluorescente in Situ , Masculino , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Mutación , Tasa de Mutación , Tasa de Supervivencia , Talidomida/uso terapéutico , Resultado del Tratamiento , Reino Unido
4.
Blood ; 113(1): 100-7, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18838613

RESUMEN

Inactivation of the tumor suppressor gene, CDKN2A, can occur by deletion, methylation, or mutation. We assessed the principal mode of inactivation in childhood acute lymphoblastic leukemia (ALL) and frequency in biologically relevant subgroups. Mutation or methylation was rare, whereas genomic deletion occurred in 21% of B-cell precursor ALL and 50% of T-ALL patients. Single nucleotide polymorphism arrays revealed copy number neutral (CNN) loss of heterozygosity (LOH) in 8% of patients. Array-based comparative genomic hybridization demonstrated that the mean size of deletions was 14.8 Mb and biallelic deletions composed a large and small deletion (mean sizes, 23.3 Mb and 1.4 Mb). Among 86 patients, only 2 small deletions were below the resolution of detection by fluorescence in situ hybridization. Patients with high hyperdiploidy, ETV6-RUNX1, or 11q23/MLL rearrangements had low rates of deletion (11%, 15%, 13%), whereas patients with t(9;22), t(1;19), TLX3, or TLX1 rearrangements had higher frequencies (61%, 42%, 78%, and 89%). In conclusion, CDKN2A deletion is a significant secondary abnormality in childhood ALL strongly correlated with phenotype and genotype. The variation in the incidence of CDKN2A deletions by cytogenetic subgroup may explain its inconsistent association with outcome. CNN LOH without apparent CDKN2A inactivation suggests the presence of other relevant genes in this region.


Asunto(s)
Eliminación de Gen , Dosificación de Gen , Regulación Leucémica de la Expresión Génica , Genes p16 , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Niño , Metilación de ADN , Femenino , Genómica , Hormona de Crecimiento Humana , Humanos , Hibridación Fluorescente in Situ , Incidencia , Pérdida de Heterocigocidad , Masculino , Mutación , Fenotipo , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiología
5.
Proc Natl Acad Sci U S A ; 105(44): 17050-4, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18957548

RESUMEN

The search for target genes involved in unbalanced acquired chromosomal abnormalities has been largely unsuccessful, because the breakpoints of these rearrangements are too variable. Here, we use the example of dicentric chromosomes in B cell precursor acute lymphoblastic leukemia to show that, despite this heterogeneity, single genes are targeted through a variety of mechanisms. FISH showed that, although they were heterogeneous, breakpoints on 9p resulted in the partial or complete deletion of PAX5. Molecular copy number counting further delineated the breakpoints and facilitated cloning with long-distance inverse PCR. This approach identified 5 fusion gene partners with PAX5: LOC392027 (7p12.1), SLCO1B3 (12p12), ASXL1 (20q11.1), KIF3B (20q11.21), and C20orf112 (20q11.1). In each predicted fusion protein, the DNA-binding paired domain of PAX5 was present. Using quantitative PCR, we demonstrated that both the deletion and gene fusion events resulted in the same underexpression of PAX5, which extended to the differential expression of the PAX5 target genes, EBF1, ALDH1A1, ATP9A, and FLT3. Further molecular analysis showed deletion and mutation of the homologous PAX5 allele, providing further support for the key role of PAX5. Here, we show that specific gene loci may be the target of heterogeneous translocation breakpoints in human cancer, acting through a variety of mechanisms. This approach indicates an application for the identification of cancer genes in solid tumours, where unbalanced chromosomal rearrangements are particularly prevalent and few genes have been identified. It can be extrapolated that this strategy will reveal that the same mechanisms operate in cancer pathogenesis in general.


Asunto(s)
Rotura Cromosómica , Genes Relacionados con las Neoplasias , Factor de Transcripción PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Translocación Genética/genética , Alelos , Secuencia de Bases , Cromosomas Humanos Par 9/genética , Dosificación de Gen , Humanos , Hibridación Fluorescente in Situ , Modelos Genéticos , Datos de Secuencia Molecular , Mutación , Proteínas de Fusión Oncogénica/genética , Factor de Transcripción PAX5/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo
6.
Genes Chromosomes Cancer ; 49(3): 253-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19998443

RESUMEN

This study describes the cytogenetics of 33 children with ETV6-RUNX1 positive acute lymphoblastic leukemia (ALL) who had been in continuous complete remission for a minimum of 8.8 years [median event-free survival (EFS) 10.9 years]. The results were compared with a published series of 16 fusion positive patients treated on the same childhood ALL trial, who had relapsed (median EFS, 2.3 years). Interphase fluorescence in situ hybridization (FISH) at diagnosis showed deletion of the second ETV6 signal from all fusion positive cells in 45% of the long-term survivors but in none of the relapsed patients, whereas patients with mixed populations with retained or lost second signals were more frequent among those who had relapsed (69%) than the long-term survivors (21%). Interphase populations with two fusion signals in 18% of the long-term survivors and 31% of relapsed patients were smaller in the long-term survivors (median, 4% of total cells) than in the relapsed patients (median, 84%). The additional copy of chromosome 21 in 30% of long-term survivors and in 69% of relapsed patients was a derived chromosome 21 in 20% and 55% of patients, respectively. Metaphase FISH for 26 long-term survivors and 15 relapsed patients revealed complex karyotypes in both groups. Variant translocations involved different chromosome arms between the long-term survivors and relapsed patients. It appears that the two groups have some distinguishing cytogenetic features at the time of diagnosis, which may provide pointers to relapse that are worthy of more detailed study.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Fusión Génica , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Represoras/genética , Sobrevivientes , Adolescente , Niño , Preescolar , Estudios de Seguimiento , Humanos , Hibridación Fluorescente in Situ , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Recurrencia , Factores de Tiempo , Proteína ETS de Variante de Translocación 6
7.
Methods Mol Biol ; 538: 29-55, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19277578

RESUMEN

Cytogenetics is integral to the diagnosis of childhood leukaemia, particularly in relation to the risk stratification of patients for treatment. Fluorescence in situ hybridization (FISH) has become an important complementary technique, expanding chromosomal analysis into the molecular arena. It has greatly improved the accuracy and applicability of cytogenetics and led to the discovery of novel chromosomal changes of prognostic significance. Many probes are now commercially available, providing robust and reliable detection of chromosomal abnormalities. Since the cloning of the human genome, it is now possible to access detailed genomic information and develop FISH probes for virtually any known DNA sequence. The range of procedures necessary for the successful application of FISH in the accurate detection of significant chromosomal abnormalities in childhood acute leukaemia is described here.


Asunto(s)
Aberraciones Cromosómicas , Hibridación Fluorescente in Situ/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Médula Ósea/patología , Sondas de ADN/genética , Humanos , Hibridación de Ácido Nucleico
8.
Genes Chromosomes Cancer ; 47(12): 1118-25, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18767146

RESUMEN

The ETV6-RUNX1 fusion is the molecular consequence of the t(12;21)(p13;q22) seen in approximately 25% of children with acute lymphoblastic leukemia (ALL). Studies have shown that the fusion alone is insufficient for the initiation of leukemia; additional genetic changes are required. Genomic profiling identified copy number alterations at high frequencies in these patients. Focal deletions of TBL1XR1 were observed in 15% of cases; 3 patients exhibited deletions distal to the gene. Fluorescence in situ hybridization confirmed these deletions and quantitative RT-PCR showed that the TBL1XR1 gene was significantly under-expressed. TBL1XR1 is a key component of the SMRT and N-CoR compressor complexes, which control hormone-receptor mediated gene expression. Differential expression of the retinoic acid target genes, RARB, CRABP1, and CRABP2, indicated that deletion of TBL1XR1 compromised the function of SMRT/N-CoR in the appropriate control of gene expression. This study identifies deletions of TBL1XR1 as a recurrent abnormality in ETV6-RUNX1 positive ALL. We provide evidence that implicates this deletion in the inappropriate control of gene expression in these patients. The target of the interaction between TBL1XR1 and the signaling pathways described here may be exploited in cancer therapy.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Represoras/genética , Eliminación de Secuencia , Cromosomas Humanos Par 12/genética , Cromosomas Humanos Par 21/genética , Humanos , Modelos Genéticos , Translocación Genética
9.
Cancer Epidemiol Biomarkers Prev ; 20(8): 1703-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21680536

RESUMEN

BACKGROUND: Several cancer types have differences in incidence and clinical outcome dependent on gender, but these are not well described in myeloma. The aim of this study was to characterize gender disparities in myeloma. METHODS: We investigated the association of gender with the prevalence of tumor genetic lesions and the clinical outcome of 1,960 patients enrolled in the phase III clinical trial MRC Myeloma IX. Genetic lesions were characterized by FISH. RESULTS: Disparities were found in the prevalence of primary genetic lesions with immunoglobulin heavy chain gene (IGH) translocations being more common in women (50% of female patients vs. 38% of male patients, P < 0.001) and hyperdiploidy being more common in men (50% female vs. 62% male, P < 0.001). There were also differences in secondary genetic events with del(13q) (52% female vs. 41% male, P < 0.001) and +1q (43% female vs. 36% male, P = 0.042) being found more frequently in female myeloma patients. Female gender was associated with inferior overall survival (median: 44.8 months female vs. 49.9 months male, P = 0.020). CONCLUSIONS: We found gender-dependent differences in the prevalence of the primary genetic events of myeloma, with IGH translocations being more common in women and hyperdiploidy more common in men. This genetic background may impact subsequent genetic events such as +1q and del(13q), which were both more frequent in women. The higher prevalence of lesions associated with poor prognosis in the female myeloma population, such as t(4;14), t(14;16) and +1q, may adversely affect clinical outcome. IMPACT: These differences suggest that gender influences the primary genetic events of myeloma.


Asunto(s)
Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Adulto , Anciano , Anciano de 80 o más Años , Aberraciones Cromosómicas/estadística & datos numéricos , Femenino , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Mieloma Múltiple/epidemiología , Prevalencia , Factores Sexuales , Análisis de Supervivencia , Resultado del Tratamiento
10.
Clin Cancer Res ; 17(24): 7776-84, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21994415

RESUMEN

PURPOSE: Regions on 1p with recurrent deletions in presenting myeloma patients were examined with the purpose of defining the deletions and assessing their survival impact. EXPERIMENTAL DESIGN: Gene mapping, gene expression, FISH, and mutation analyses were conducted on patient samples from the MRC Myeloma IX trial and correlated with clinical outcome data. RESULTS: 1p32.3 was deleted in 11% of cases, and deletion was strongly associated with impaired overall survival (OS) in patients treated with autologous stem cell transplant (ASCT). In patients treated less intensively, del(1)(p32.3) was not associated with adverse progression-free survival (PFS) or OS. The target of homozygous deletions was CDKN2C, however its role in the adverse outcome of cases with hemizygous deletion was less certain. 1p22.1-21.2 was the most frequently deleted region and contained the candidate genes MTF2 and TMED5. No mutations were identified in these genes. 1p12 was deleted in 19% of cases, and deletion was associated with impaired OS in univariate analysis. The target of homozygous deletion was FAM46C, which was mutated in 3.4% of cases. When cases with FAM46C deletion or mutation were considered together, they were strongly associated with impaired OS in the intensive treatment setting. CONCLUSION: Deletion of 1p32.3 and 1p12 was associated with impaired OS in myeloma patients receiving ASCT. FAM46C was identified as a gene with potential pathogenic and prognostic significance based on the occurrence of recurrent homozygous deletions and mutations.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 1/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Mieloma Múltiple/genética , Anciano , Mapeo Cromosómico , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Humanos , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Análisis Multivariante , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Complejo Represivo Polycomb 2 , Modelos de Riesgos Proporcionales , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA