Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
1.
Blood ; 116(23): 4958-67, 2010 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-20729466

RESUMEN

Genomic aberrations are of predominant importance to the biology and clinical outcome of patients with acute myelogenous leukemia (AML), and conventional karyotype-based risk classifications are routinely used in clinical decision making in AML. One of the known limitations of cytogenetic analysis is the inability to detect genomic abnormalities less than 5 Mb in size, and it is currently unclear whether overcoming this limitation with high-resolution genomic single-nucleotide polymorphism (SNP) array analysis would be clinically relevant. Furthermore, given the heterogeneity of molecular mechanisms/aberrations that underlie the conventional karyotype-based risk classifications, it is likely that further refinements in genomic risk prognostication can be achieved. In this study, we analyzed flow cytometer-sorted, AML blast-derived, and paired, buccal DNA from 114 previously untreated prospectively enrolled AML patients for acquired genomic copy number changes and loss of heterozygosity using Affymetrix SNP 6.0 arrays, and we correlated genomic lesion load and specific chromosomal abnormalities with patient survival. Using multivariate analyses, we found that having ≥ 2 genomic lesions detected through SNP 6.0 array profiling approximately doubles the risk of death when controlling for age- and karyotype-based risk. Finally, we identified an independent negative prognostic impact of p53 mutations, or p53 mutations and 17p-loss of heterozygosity combined on survival in AML.


Asunto(s)
Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Adulto , Anciano , Anciano de 80 o más Años , Separación Celular , Aberraciones Cromosómicas , Cromosomas Humanos Par 17/genética , Citometría de Flujo , Dosificación de Gen , Historia del Siglo XVI , Historia del Siglo XVII , Humanos , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Cariotipificación , Pérdida de Heterocigocidad , Pronóstico , Proteína p53 Supresora de Tumor/genética , Adulto Joven
2.
Am J Med Genet A ; 158A(10): 2591-601, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22903861

RESUMEN

We identified a novel 6.33 Mb deletion of 1q21.3q23.3 (hg18; chr1: 153035245-159367106) in two siblings presenting with blepharophimosis, ptosis, microbrachycephaly, severe psychomotor, and intellectual disability. Additional common features include small corpus callosum, normal birth length and head circumference, postnatal growth restriction, low anterior hairline, upturned nose, bilateral preauricular pits, widely spaced teeth, gingival hypertrophy, left ventricular dilatation with decreased biventricular systolic function, delayed bone age, 5th finger clinodactyly, short 3rd digit, hyperconvex nails, obstructive and central sleep apnea, and bilateral heel contractures. Fluorescence in situ hybridization (FISH) performed in the mother of both children showed an apparently balanced, intrachromosomal insertional translocation of 1q21.3q23.3 to 1q42.12. The sibling recurrence likely arose by a maternal meiotic crossing over on the rearranged chromosome 1 between the deleted region and the insertion. We hypothesize that the decreased cardiac function and contractures may be related to LMNA haploinsufficiency. This case illustrates the importance of FISH when attempting to determine inheritance of a copy-number variation and emphasize the value of evaluating known haploinsufficiency phenotypes for genes in deleted regions.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 1/genética , Hermanos , Translocación Genética , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Adolescente , Blefarofimosis/genética , Blefarofimosis/patología , Blefaroptosis/genética , Blefaroptosis/patología , Niño , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Femenino , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Humanos , Hibridación Fluorescente in Situ , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Masculino , Mutagénesis Insercional
3.
Clin Cancer Res ; 16(16): 4135-47, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20505189

RESUMEN

PURPOSE: This study was conducted to identify novel genes with importance to the biology of adult acute myelogenous leukemia (AML). EXPERIMENTAL DESIGN: We analyzed DNA from highly purified AML blasts and paired buccal cells from 95 patients for recurrent genomic microdeletions using ultra-high density Affymetrix single nucleotide polymorphism 6.0 array-based genomic profiling. RESULTS: Through fine mapping of microdeletions on 17q, we derived a minimal deleted region of approximately 0.9-Mb length that harbors 11 known genes; this region includes Neurofibromin 1 (NF1). Sequence analysis of all NF1 coding exons in the 11 AML cases with NF1 copy number changes identified acquired truncating frameshift mutations in two patients. These NF1 mutations were already present in the hematopoetic stem cell compartment. Subsequent expression analysis of NF1 mRNA in the entire AML cohort using fluorescence-activated cell sorting sorted blasts as a source of RNA identified six patients (one with a NF1 mutation) with absent NF1 expression. The NF1 null states were associated with increased Ras-bound GTP, and short hairpin RNA-mediated NF1 suppression in primary AML blasts with wild-type NF1 facilitated colony formation in methylcellulose. Primary AML blasts without functional NF1, unlike blasts with functional NF1, displayed sensitivity to rapamycin-induced apoptosis, thus identifying a dependence on mammalian target of rapamycin (mTOR) signaling for survival. Finally, colony formation in methylcellulose ex vivo of NF1 null CD34+/CD38- cells sorted from AML bone marrow samples was inhibited by low-dose rapamycin. CONCLUSIONS: NF1 null states are present in 7 of 95 (7%) of adult AML and delineate a disease subset that could be preferentially targeted by Ras or mammalian target of rapamycin-directed therapeutics.


Asunto(s)
Silenciador del Gen , Genes de Neurofibromatosis 1 , Leucemia Mieloide Aguda/genética , Transducción de Señal/genética , Adulto , Anciano , Anciano de 80 o más Años , Western Blotting , Separación Celular , Femenino , Citometría de Flujo , Mutación del Sistema de Lectura , Dosificación de Gen , Perfilación de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA