Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Haematol ; 192(5): 832-842, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33529373

RESUMEN

In acute myeloid leukaemia (AML) t(8;16)(p11;p13)/MYST3-CREBBP is a very rare abnormality. Previous small series suggested poor outcome. We report on 59 patients with t(8;16) within an international, collaborative study. Median age was 52 (range: 16-75) years. AML was de novo in 58%, therapy-related (t-AML) in 37% and secondary after myelodysplastic syndrome (s-AML) in 5%. Cytogenetics revealed a complex karyotype in 43%. Besides MYST3-CREBBP, whole-genome sequencing on a subset of 10 patients revealed recurrent mutations in ASXL1, BRD3, FLT3, MLH1, POLG, TP53, SAMD4B (n = 3, each), EYS, KRTAP9-1 SPTBN5 (n = 4, each), RUNX1 and TET2 (n = 2, each). Complete remission after intensive chemotherapy was achieved in 84%. Median follow-up was 5·48 years; five-year survival rate was 17%. Patients with s-/t-AML (P = 0·01) and those with complex karyotype (P = 0·04) had an inferior prognosis. Allogeneic haematopoietic cell transplantation (allo-HCT) was performed in 21 (36%) patients, including 15 in first complete remission (CR1). Allo-HCT in CR1 significantly improved survival (P = 0·04); multivariable analysis revealed that allo-HCT in CR1 was effective in de novo AML but not in patients with s-AML/t-AML and less in patients exhibiting a complex karyotype. In summary, outcomes of patients with t(8;16) are dismal with chemotherapy, and may be substantially improved with allo-HCT performed in CR1.


Asunto(s)
Cromosomas Humanos Par 16/ultraestructura , Cromosomas Humanos Par 8/ultraestructura , Leucemia Mieloide Aguda/genética , Translocación Genética , Cariotipo Anormal , Adolescente , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Terapia Combinada , Quimioterapia de Consolidación , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Trasplante de Células Madre Hematopoyéticas , Humanos , Cooperación Internacional , Leucemia Mieloide Aguda/epidemiología , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Mutación , Síndromes Mielodisplásicos/epidemiología , Neoplasias Primarias Secundarias/inducido químicamente , Neoplasias Primarias Secundarias/epidemiología , Proteínas de Fusión Oncogénica/genética , Inducción de Remisión , Análisis de Supervivencia , Secuenciación Completa del Genoma
2.
Cytogenet Genome Res ; 160(1): 11-17, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31982875

RESUMEN

Small supernumerary marker chromosomes (sSMCs) are characterized as additional centric chromosome fragments which are too small to be classified by cytogenetic banding alone and smaller than or equal to the size of chromosome 20 of the same metaphase spread. Here, we report a patient who presented with slight neutropenia and oral aphthous ulcers. A mosaic de novo sSMC, which originated from 5 discontinuous regions of chromosome 8, was detected in the patient. Formation of the sSMC(8) can probably be explained by a multi-step process beginning with maternal meiotic nondisjunction, followed by post-zygotic anaphase lag, and resulting in chromothripsis. Chromothripsis is a chromosomal rearrangement which occurs by breakage of one or more chromosomes leading to a fusion of surviving chromosome pieces. This case is a good example for emphasizing the importance of conventional karyotyping from PHA-induced peripheral blood lymphocytes and examining tissues other than bone marrow in patients with inconsistent genotype and phenotype.


Asunto(s)
Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/ultraestructura , Neutropenia/genética , Úlceras Bucales/genética , Estomatitis Aftosa/genética , Preescolar , Aberraciones Cromosómicas , Trastornos de los Cromosomas/genética , Citogenética , Femenino , Marcadores Genéticos , Genotipo , Humanos , Cariotipificación , Linfocitos/metabolismo , Metafase , Mosaicismo , Neutropenia/complicaciones , Neutropenia/diagnóstico , Análisis de Secuencia por Matrices de Oligonucleótidos , Úlceras Bucales/complicaciones , Úlceras Bucales/diagnóstico , Fenotipo , Estomatitis Aftosa/complicaciones , Estomatitis Aftosa/diagnóstico
3.
Am J Hematol ; 93(3): 375-382, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29194741

RESUMEN

Chronic lymphocytic leukemia (CLL) with 17p deletion (17p-) is associated with a lack of response to standard treatment and thus the worst possible clinical outcome. Various chromosomal abnormalities (including unbalanced translocations, deletions, ring chromosomes and isochromosomes) result in the loss of 17p and one copy of the TP53 gene. The objective of the present study was to determine whether the type of chromosomal abnormality leading to 17p- and the additional aberrations influenced the prognosis in a series of 195 patients with 17p-CLL. Loss of 17p resulted primarily from an unbalanced translocation (70%) with several chromosome partners (the most frequent being chromosome 18q), followed by deletion 17p (23%), monosomy 17 (8%), isochromosome 17q [i(17q)] (5%) and a ring chromosome 17 (2%). In a univariate analysis, monosomy 17, a highly complex karyotype (≥5 abnormalities), and 8q24 gain were associated with poor treatment-free survival, and i(17q) (P = .04), unbalanced translocations (P = .03) and 8q24 gain (P = .001) were significantly associated with poor overall survival. In a multivariate analysis, 8q24 gain remained a significant predictor of poor overall survival. We conclude that 17p deletion and 8q24 gain have a synergistic impact on outcome, and so patients with this "double-hit" CLL have a particularly poor prognosis. Systematic, targeting screening for 8q24 gain should therefore be considered in cases of 17p- CLL.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 17/ultraestructura , Cromosomas Humanos Par 8/ultraestructura , Leucemia Linfocítica Crónica de Células B/genética , Translocación Genética , Trisomía , Cariotipo Anormal , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Femenino , Genes p53 , Humanos , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Leucemia Linfocítica Crónica de Células B/mortalidad , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/genética , Pronóstico , Estudios Retrospectivos
4.
Ann Hematol ; 99(11): 2715-2717, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32671454
5.
Cytogenet Genome Res ; 142(1): 1-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24135068

RESUMEN

Branchio-oto-renal (BOR) syndrome is an autosomal dominantly inherited developmental disorder, which is characterized by anomalies of the ears, the branchial arches and the kidneys. It is caused by mutations in the genes EYA1,SIX1 and SIX5. Genomic rearrangements of chromosome 8 affecting the EYA1 gene have also been described. Owing to this fact, methods for the identification of abnormal copy numbers such as multiplex ligation-dependent probe amplification (MLPA) have been introduced as routine laboratory techniques for molecular diagnostics of BOR syndrome. The advantages of these techniques are clear compared to standard cytogenetic and array approaches as well as Southern blot. MLPA detects deletions or duplications of a part or the entire gene of interest, but not balanced structural aberrations such as inversions and translocations. Consequently, disruption of a gene by a genomic rearrangement may escape detection by a molecular genetic analysis, although this gene interruption results in haploinsufficiency and, therefore, causes the disease. In a patient with clinical features of BOR syndrome, such as hearing loss, preauricular fistulas and facial dysmorphisms, but no renal anomalies, neither sequencing of the 3 genes linked to BOR syndrome nor array comparative genomic hybridization and MLPA were able to uncover a causative mutation. By routine cytogenetic analysis, we finally identified a pericentric inversion of chromosome 8 in the affected female. High-resolution multicolor banding confirmed the chromosome 8 inversion and narrowed down the karyotype to 46,XX,inv(8)(p22q13). By applying fluorescence in situ hybridization, we narrowed down both breakpoints on chromosome 8 and found the EYA1 gene in q13.3 to be directly disrupted. We conclude that standard karyotyping should not be neglected in the genetic diagnostics of BOR syndrome or other Mendelian disorders, particularly when molecular testing failed to detect any causative alteration in patients with a convincing phenotype.


Asunto(s)
Síndrome Branquio Oto Renal/genética , Inversión Cromosómica , Cromosomas Humanos Par 8/ultraestructura , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Proteínas Tirosina Fosfatasas/genética , Adulto , Síndrome Branquio Oto Renal/patología , Puntos de Rotura del Cromosoma , Cromosomas Humanos Par 8/genética , Hibridación Genómica Comparativa , Femenino , Pérdida Auditiva Sensorineural/etiología , Proteínas de Homeodominio/genética , Humanos , Hibridación Fluorescente in Situ , Péptidos y Proteínas de Señalización Intracelular/química , Reacción en Cadena de la Polimerasa Multiplex , Proteínas Nucleares/química , Fenotipo , Proteínas Tirosina Fosfatasas/química
7.
Pediatr Blood Cancer ; 61(6): 1107-10, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24281971

RESUMEN

Chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL) is a disease of older adults. Pediatric CLL/SLL is vanishingly rare in the literature. We present a case of CLL/SLL diagnosed in a 17-year-old male. The pathologic findings of this case were those of classic CLL/SLL with an ATM deletion, a characteristic genetic abnormality in CLL/SLL. Management guidelines for CLL/SLL are tailored to older adults making determination of the optimal therapy for this patient a unique challenge.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/diagnóstico , Adolescente , Factores de Edad , Proteínas de la Ataxia Telangiectasia Mutada/genética , Biomarcadores de Tumor , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 11/ultraestructura , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/ultraestructura , Diagnóstico por Imagen , Dosificación de Gen , Genes myc , Humanos , Inmunofenotipificación , Hibridación Fluorescente in Situ , Leucemia Linfocítica Crónica de Células B/epidemiología , Leucemia Linfocítica Crónica de Células B/genética , Ganglios Linfáticos/patología , Masculino , Eliminación de Secuencia
8.
Morphologie ; 98(321): 86-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24656633

RESUMEN

The Wolf-Hirschhorn syndrome (WHS) encompasses deletions at the distal part of the short arm of one chromosome 4 (4p16 region). Clinical signs frequently include a typical facial appearance, mental retardation, intrauterine and postnatal growth retardation, hypotonia with decreased muscle bulk and seizures besides congenital heart malformations, midline defects, urinary tract malformations and brain, hearing and ophthalmologic malformations. Pathogenesis of WHS is multigenic and many factors are involved in prediction of prognosis such as extent of deletion, the occurrence of severe chromosome anomalies, the severe of seizures, the existence of serious internal, mainly cardiac, abnormalities and the degree of mental retardation. The phenotype of adult with WHS is in general similar to that of childhood being facial dysmorphism, growth retardation and mental retardation the rule in both adults and children. Avoid long-term complications and provide rehabilitation programs and genetic counseling may be essential in these patients.


Asunto(s)
Síndrome de Wolf-Hirschhorn/patología , Adolescente , Deleción Cromosómica , Cromosomas Humanos Par 4/genética , Cromosomas Humanos Par 4/ultraestructura , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/ultraestructura , Ventrículo Derecho con Doble Salida/genética , Epilepsia Generalizada/genética , Facies , Femenino , Hallux Valgus/genética , Humanos , Hibridación Fluorescente in Situ , Discapacidad Intelectual/genética , Cifosis/genética , Masculino , Fenotipo , Translocación Genética , Síndrome de Wolf-Hirschhorn/genética
10.
Ann Hematol ; 92(6): 759-69, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23400675

RESUMEN

The translocation t(8;9)(p22;p24) is a rare event that results in the fusion of JAK2 to PCM1 and thus leads to the activation of the Janus Kinase 2. In 2008, the WHO introduced a new entity called "Myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB or FGFR1", which are characterized by the formation of a fusion gene encoding an aberrant tyrosine kinase. These disorders share characteristics with myeloproliferative neoplasms and typically show an eosinophilia. We here now report on 6 new cases with PCM1-JAK2 fusion. These patients show characteristics with respect to epidemiology, clinical presentation, and genetic changes that are very similar to patients with rearrangements of PDGFRA, PDGFRB, or FGFR1. Our data suggests the integration of cases with JAK2-PCM1 fusion in the respective WHO category of myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB, or FGFR1.


Asunto(s)
Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 9/genética , Eosinofilia/genética , Leucemia/clasificación , Trastornos Mieloproliferativos/clasificación , Proteínas de Fusión Oncogénica/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Translocación Genética/genética , Adulto , Anciano , Médula Ósea/patología , Niño , Cromosomas Humanos Par 8/ultraestructura , Cromosomas Humanos Par 9/ultraestructura , Femenino , Humanos , Hibridación Fluorescente in Situ , Leucemia/diagnóstico , Leucemia/genética , Leucemia/patología , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Linfocitos/patología , Masculino , Persona de Mediana Edad , Mutación , Células Mieloides/patología , Trastornos Mieloproliferativos/diagnóstico , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Organización Mundial de la Salud
11.
Ann Hematol ; 97(3): 529-531, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29124313
12.
Ann Hematol ; 92(2): 163-71, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23053179

RESUMEN

Core binding factor (CBF)-positive acute myeloid leukemia (AML) presents a favorable prognosis, except for patients with KIT mutation, especially D816 mutation. The current retrospective study attempted to validate a prognostic role of KIT mutation in 121 Korean patients with CBF AML. The study patients consisted of 121 patients with CBF AML (82 patients with RUNX1/RUNX1T1 [67.8 %] and 39 patients with CBFB/MYH11 [32.2 %]) recruited from eight institutions in Korea. All patients received idarubicin plus cytarabine or behenoyl cytosine arabinoside 3 + 7 induction chemotherapy. The KIT gene mutation status was determined by direct sequencing analyses. A KIT mutation was detected in 32 cases (26.4 %) in our series of patients. The KIT mutation was most frequent in exon 17 (n = 18, 14.9 %; n = 16 with D816 mutation), followed by exon 8 (n = 10, 8.3 %). The presence of KIT D816 mutation was associated with adverse outcomes for the event-free survival (p = 0.03) and for the overall survival (p = 0.02). The unfavorable impact of D816 mutation was more prominent when the analysis was confined to the RUNX1/RUNX1T1 subtype. The KIT mutation was detected in 26.4 % of Korean patients with CBF AML. The KIT D816 mutation demonstrated an unfavorable prognostic implication, particularly in the RUNX1/RUNX1T1 subtype.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/genética , Mutación Puntual , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Adolescente , Adulto , Anciano , Inversión Cromosómica , Cromosomas Humanos Par 16/genética , Cromosomas Humanos Par 16/ultraestructura , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 21/ultraestructura , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/ultraestructura , Terapia Combinada , Factores de Unión al Sitio Principal/análisis , Factores de Unión al Sitio Principal/genética , Citarabina/administración & dosificación , Citarabina/análogos & derivados , Supervivencia sin Enfermedad , Exones/genética , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Idarrubicina/administración & dosificación , Estimación de Kaplan-Meier , Corea (Geográfico)/epidemiología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/cirugía , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/análisis , Pronóstico , Proteína 1 Compañera de Translocación de RUNX1 , Translocación Genética , Resultado del Tratamiento , Adulto Joven
13.
Nat Rev Cancer ; 1(3): 214-21, 2001 12.
Artículo en Inglés | MEDLINE | ID: mdl-11902576

RESUMEN

In 1979, the first chromosome alteration associated with familial cancer was reported. Five years later, a fragile site was observed in the same chromosome region. The product of the fragile histidine triad (FHIT) gene, which encompasses this fragile site, is partially or entirely lost in most human cancers, indicating that it has a tumour-suppressor function. Inactivation of only one FHIT allele compromises this suppressor function, indicating that a 'one-hit' mechanism of tumorigenesis is operative. Are genes disrupted at other fragile sites? And, are these genes also tumour suppressors?


Asunto(s)
Ácido Anhídrido Hidrolasas , Transformación Celular Neoplásica/genética , Fragilidad Cromosómica , Cromosomas Humanos Par 3/genética , Genes Supresores de Tumor , Proteínas de Neoplasias/fisiología , Adulto , Alelos , Secuencias de Aminoácidos , Animales , Apoptosis/genética , Rotura Cromosómica , Sitios Frágiles del Cromosoma , Fragilidad Cromosómica/genética , Cromosomas Humanos Par 3/ultraestructura , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/ultraestructura , Secuencia Conservada , Replicación del ADN , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/prevención & control , Predicción , Neoplasias Gastrointestinales/inducido químicamente , Neoplasias Gastrointestinales/genética , Eliminación de Gen , Predisposición Genética a la Enfermedad , Terapia Genética , Humanos , Neoplasias Renales/genética , Ratones , Ratones Noqueados , Modelos Genéticos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Recombinación Genética , Relación Estructura-Actividad , Translocación Genética
14.
Nat Genet ; 15(2): 170-4, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9020842

RESUMEN

Pleiomorphic adenoma of the salivary glands is a benign epithelial tumour occurring primarily in the major and minor salivary glands. It is by far the most common type of salivary gland tumour. Microscopically, pleiomorphic adenomas show a marked histological diversity with epithelial, myoepithelial and mesenchymal components in a variety of patterns. In addition to a cytogenetic subgroup with normal karyotypes, pleiomorphic adenomas are characterized by recurrent chromosome rearrangements, particularly reciprocal translocations, with breakpoints at 8q12, 3p21, and 12q13-15, in that order of frequency. The most common abnormality is a reciprocal t(3;8)(p21;q12). We here demonstrate that the t(3;8)(p21;q12) results in promoter swapping between PLAG1, a novel, developmentally regulated zinc finger gene at 8q12, and the constitutively expressed gene for beta-catenin (CTNNB1), a protein interface functioning in the WG/WNT signalling pathway and specification of cell fate during embryogenesis. Fusions occur in the 5'-non-coding regions of both genes, exchanging regulatory control elements while preserving the coding sequences. Due to the t(3;8)(p21;q12), PLAG1 is activated and expression levels of CTNNB1 are reduced. Activation of PLAG1 was also observed in an adenoma with a variant translocation t(8;15)(q12;q14). Our results indicate that PLAG1 activation due to promoter swapping is a crucial event in salivary gland tumourigenesis.


Asunto(s)
Adenoma Pleomórfico/genética , Cromosomas Humanos Par 3/genética , Cromosomas Humanos Par 8/genética , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Genes , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas , Neoplasias de las Glándulas Salivales/genética , Transactivadores , Translocación Genética/genética , Dedos de Zinc/genética , Adenoma Pleomórfico/patología , Secuencia de Aminoácidos , Secuencia de Bases , Paseo de Cromosoma , Cromosomas Artificiales de Levadura/genética , Cromosomas Humanos Par 3/ultraestructura , Cromosomas Humanos Par 8/ultraestructura , Humanos , Datos de Secuencia Molecular , Neoplasias de las Glándulas Salivales/patología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , beta Catenina
15.
Ann Hematol ; 96(12): 2127-2129, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28840298
17.
Med Oncol ; 26(2): 251-5, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18509766

RESUMEN

An 87-year-old woman was diagnosed with unclassified myeloproliferative disease having an acquired jumping translocation with the long arm of chromosome 3 translocating to the short arm telomeric region of chromosome 8 (major clone) and the long arm telomeric region of chromosome 10 (minor clone). Each abnormal clone was also associated with an extra copy of chromosome 8. Although there was no evidence of transformation to an acute leukemia, the patient deteriorated until her demise 7 months after disease presentation. There have been fewer than 70 cases of acquired jumping translocations reported in the literature. To our knowledge, this is the first acquired jumping translocation case to be reported in a patient with myeloproliferative disease.


Asunto(s)
Cromosomas Humanos Par 3 , Trastornos Mieloproliferativos/genética , Translocación Genética , Anciano de 80 o más Años , Bronconeumonía/etiología , Cromosomas Humanos Par 10/ultraestructura , Cromosomas Humanos Par 3/ultraestructura , Cromosomas Humanos Par 8/ultraestructura , Femenino , Humanos , Hidroxiurea/uso terapéutico , Secuencias Repetitivas Esparcidas , Trastornos Mieloproliferativos/complicaciones , Trastornos Mieloproliferativos/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA