Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Blood Cancer ; 69(1): e29361, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34597466

RESUMEN

INTRODUCTION: One-quarter of the relapses in children with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) occur very early (within 18 months, before completion of treatment), and prognosis in these patients is worse compared to cases that relapse after treatment has ended. METHODS: In this study, we performed a genomic analysis of diagnosis-relapse pairs of 12 children who relapsed very early, followed by a deep-sequencing validation of all identified mutations. In addition, we included one case with a good initial treatment response and on-treatment relapse at the end of upfront therapy. RESULTS: We observed a dynamic clonal evolution in all cases, with relapse almost exclusively originating from a subclone at diagnosis. We identified several driver mutations that may have influenced the outgrowth of a minor clone at diagnosis to become the major clone at relapse. For example, a minimal residual disease (MRD)-based standard-risk patient with ETV6-RUNX1-positive leukemia developed a relapse from a TP53-mutated subclone after loss of the wildtype allele. Furthermore, two patients with TCF3-PBX1-positive leukemia that developed a very early relapse carried E1099K WHSC1 mutations at diagnosis, a hotspot mutation that was recurrently encountered in other very early TCF3-PBX1-positive leukemia relapses as well. In addition to alterations in known relapse drivers, we found two cases with truncating mutations in the cohesin gene RAD21. CONCLUSION: Comprehensive genomic characterization of diagnosis-relapse pairs shows that very early relapses in BCP-ALL frequently arise from minor subclones at diagnosis. A detailed understanding of the therapeutic pressure driving these events may aid the development of improved therapies.


Asunto(s)
Enfermedad Injerto contra Huésped , Leucemia-Linfoma Linfoblástico de Células Precursoras , Niño , Evolución Clonal/genética , Genómica , Humanos , Pronóstico , Recurrencia
2.
Haematologica ; 106(12): 3046-3055, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33147938

RESUMEN

Genomic studies of pediatric acute lymphoblastic leukemia (ALL) have shown remarkable heterogeneity in initial diagnosis, with multiple (sub)clones harboring lesions in relapse-associated genes. However, the clinical relevance of these subclonal alterations remains unclear. We assessed the clinical relevance and prognostic value of subclonal alterations in the relapse-associated genes IKZF1, CREBBP, KRAS, NRAS, PTPN11, TP53, NT5C2, and WHSC1 in 503 ALL cases. Using Molecular Inversion Probe sequencing and breakpoint-spanning PCR we reliably detected alterations below 1% allele frequency. We identified 660 genomic alterations in 285 diagnosis samples of which 495 (75%) were subclonal. RAS pathway mutations were common, particularly in minor subclones, and comparisons between RAS hotspot mutations revealed differences in their capacity to drive clonal expansion in ALL. We did not find an association of subclonal alterations with unfavorable outcome. Particularly for IKZF1, an established prognostic marker in ALL, all clonal but none of the subclonal alterations were preserved at relapse. We conclude that, for the genes tested, there is no basis to consider subclonal alterations detected at diagnosis for risk group stratification of ALL treatment.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Niño , Células Clonales , Genómica , Humanos , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Pronóstico
3.
J Pathol ; 244(2): 135-142, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29105096

RESUMEN

It is now well established that germline genomic aberrations can underlie high-penetrant familial polyposis and colorectal cancer syndromes, but a genetic cause has not yet been found for the major proportion of patients with polyposis. Since next-generation sequencing has become widely accessible, several novel, but rare, high-penetrant risk factors for adenomatous polyposis have been identified, all operating in pathways responsible for genomic maintenance and DNA repair. One of these is the base excision repair pathway. In addition to the well-established role of the DNA glycosylase gene MUTYH, biallelic mutations in which predispose to MUTYH-associated polyposis, a second DNA glycosylase gene, NTHL1, has recently been associated with adenomatous polyposis and a high colorectal cancer risk. Both recessive polyposis syndromes are associated with increased risks for several other cancer types as well, but the spectrum of benign and malignant tumours in individuals with biallelic NTHL1 mutations was shown to be broader; hence the name NTHL1-associated tumour syndrome. Colorectal tumours encountered in patients with these syndromes show unique, clearly distinct mutational signatures that may facilitate the identification of these syndromes. On the basis of the prevalence of pathogenic MUTYH and NTHL1 variants in the normal population, we estimate that the frequency of the novel NTHL1-associated tumour syndrome is five times lower than that of MUTYH-associated polyposis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Biomarcadores de Tumor/genética , ADN Glicosilasas/genética , Reparación del ADN/genética , Desoxirribonucleasa (Dímero de Pirimidina)/genética , Mutación , Poliposis Adenomatosa del Colon/epidemiología , Poliposis Adenomatosa del Colon/patología , Animales , Daño del ADN , Predisposición Genética a la Enfermedad , Humanos , Tasa de Mutación , Penetrancia , Fenotipo , Factores de Riesgo
4.
PLoS Genet ; 12(2): e1005880, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26901136

RESUMEN

Approximately 25-30% of colorectal cancer (CRC) cases are expected to result from a genetic predisposition, but in only 5-10% of these cases highly penetrant germline mutations are found. The remaining CRC heritability is still unexplained, and may be caused by a hitherto-undefined set of rare variants with a moderately penetrant risk. Here we aimed to identify novel risk factors for early-onset CRC using whole-exome sequencing, which was performed on a cohort of CRC individuals (n = 55) with a disease onset before 45 years of age. We searched for genes that were recurrently affected by rare variants (minor allele frequency ≤ 0.001) with potentially damaging effects and, subsequently, re-sequenced the candidate genes in a replication cohort of 174 early-onset or familial CRC individuals. Two functionally relevant genes with low frequency variants with potentially damaging effects, PTPN12 and LRP6, were found in at least three individuals. The protein tyrosine phosphatase PTP-PEST, encoded by PTPN12, is a regulator of cell motility and LRP6 is a component of the WNT-FZD-LRP5-LRP6 complex that triggers WNT signaling. All variants in LRP6 were identified in individuals with an extremely early-onset of the disease (≤30 years of age), and two of the three variants showed increased WNT signaling activity in vitro. In conclusion, we present PTPN12 and LRP6 as novel candidates contributing to the heterogeneous susceptibility to CRC.


Asunto(s)
Neoplasias Colorrectales/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Edad de Inicio , Secuencia de Aminoácidos , Segregación Cromosómica/genética , Estudios de Cohortes , Neoplasias Colorrectales/enzimología , Reparación de la Incompatibilidad de ADN/genética , Exoma/genética , Genes Relacionados con las Neoplasias , Humanos , Datos de Secuencia Molecular , Mutación Missense/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 12/química , Proteína Tirosina Fosfatasa no Receptora Tipo 12/genética , Análisis de Secuencia de ADN , Transducción de Señal/genética , Proteínas Wnt/metabolismo
5.
Genes Chromosomes Cancer ; 55(11): 855-63, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27239782

RESUMEN

Germline mutations in BUB1B, encoding BUBR1, one of the crucial components of the spindle assembly checkpoint (SAC), have been shown to cause variable phenotypes, including the recessive mosaic variegated aneuploidy (MVA) syndrome, which predisposes to cancer. Reduced levels of the wild-type BUBR1 protein have been linked to the development of gastrointestinal neoplasms. To determine whether mutations in BUB1B are enriched in individuals with colorectal cancer (CRC), we performed amplicon-based targeted next-generation sequencing of BUB1B on germline DNA of 192 individuals with early-onset CRC (≤50 years). None of the individuals was found to be homozygous or compound heterozygous for mutations in BUB1B. However, we did identify two rare heterozygous variants, p.Glu390del and p.Cys945Tyr, in patients who developed CRC at the ages of 41 and 43 years, respectively. Both variants were shown not to affect BUBR1 protein expression levels and protein localization. Since the p.Glu390del variant is located in the BUB3-binding domain, we also performed immunoprecipitation to examine whether this variant affects the binding of BUB1 or BUB3 to BUBR1 but, compared to wild-type BUBR1, no difference was observed. Our data suggest that mutations in BUB1B do not occur frequently in the germline of individuals with CRC and that BUB1B unlikely plays a major role in the predisposition to early-onset CRC. Whether carriers of pathogenic BUB1B mutations, such as the parents of MVA syndrome patients, have an increased risk for cancer remains of interest, as studies in mice have suggested that haploinsufficiency of BUB1B may cause an increase in carcinogen-induced tumors. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas de Ciclo Celular/genética , Neoplasias Colorrectales/genética , Mutación de Línea Germinal/genética , Proteínas Serina-Treonina Quinasas/genética , Adolescente , Adulto , Edad de Inicio , Alelos , Animales , Neoplasias Colorrectales/patología , Femenino , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Puntos de Control de la Fase M del Ciclo Celular/genética , Masculino , Ratones , Persona de Mediana Edad
6.
J Pathol ; 236(2): 155-64, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25712196

RESUMEN

Heritable genetic variants can significantly affect the lifetime risk of developing cancer, including polyposis and colorectal cancer (CRC). Variants in genes currently known to be associated with a high risk for polyposis or CRC, however, explain only a limited number of hereditary cases. The identification of additional genetic causes is, therefore, crucial to improve CRC prevention, detection and treatment. We have performed genome-wide and targeted DNA copy number profiling and resequencing in early-onset and familial polyposis/CRC patients, and show that deletions affecting the open reading frame of the tumour suppressor gene FOCAD are recurrent and significantly enriched in CRC patients compared with unaffected controls. All patients carrying FOCAD deletions exhibited a personal or family history of polyposis. RNA in situ hybridization revealed FOCAD expression in epithelial cells in the colonic crypt, the site of tumour initiation, as well as in colonic tumours and organoids. Our data suggest that monoallelic germline deletions in the tumour suppressor gene FOCAD underlie moderate genetic predisposition to the development of polyposis and CRC.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Neoplasias Colorrectales/genética , Eliminación de Gen , Mutación de Línea Germinal/genética , Proteínas Supresoras de Tumor/genética , Poliposis Adenomatosa del Colon/metabolismo , Adulto , Estudios de Casos y Controles , Cromosomas Humanos Par 9/genética , Neoplasias Colorrectales/metabolismo , Variaciones en el Número de Copia de ADN/genética , Células Epiteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Sistemas de Lectura Abierta/genética , Proteínas Supresoras de Tumor/metabolismo
7.
Am J Hum Genet ; 90(3): 426-33, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22341970

RESUMEN

Revertant mosaicism is an infrequently observed phenomenon caused by spontaneous correction of a pathogenic allele. We have observed such reversions caused by mitotic recombination of mutant TERC (telomerase RNA component) alleles in six patients from four families affected by dyskeratosis congenita (DC). DC is a multisystem disorder characterized by mucocutaneous abnormalities, dystrophic nails, bone-marrow failure, lung fibrosis, liver cirrhosis, and cancer. We identified a 4 nt deletion in TERC in a family with an autosomal-dominant form of DC. In two affected brothers without bone-marrow failure, sequence analysis revealed pronounced overrepresentation of the wild-type allele in blood cells, whereas no such skewing was observed in the other tissues tested. These observations suggest that this mosaic pattern might have resulted from somatic reversion of the mutated allele to the normal allele in blood-forming cells. SNP-microarray analysis on blood DNA from the two brothers indeed showed independent events of acquired segmental isodisomy of chromosome 3q, including TERC, indicating that the reversions must have resulted from mitotic recombination events. Subsequently, after developing a highly sensitive method of detecting mosaic homozygosity, we have found four additional cases with a mosaic-reversion pattern in blood cells; these four cases are part of a cohort of 17 individuals with germline TERC mutations. This shows that revertant mosaicism is a recurrent event in DC. This finding has important implications for improving diagnostic testing and understanding the variable phenotype of DC.


Asunto(s)
Disqueratosis Congénita/genética , Mitosis/genética , Mosaicismo , ARN/genética , Recombinación Genética , Telomerasa/genética , Adolescente , Adulto , Anciano , Alelos , Linaje de la Célula , Niño , Preescolar , Estudios de Cohortes , Femenino , Mutación de Línea Germinal , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Linaje , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN/métodos , Adulto Joven
8.
PLoS Genet ; 8(2): e1002533, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22359517

RESUMEN

Recurrent submicroscopic deletions in genes affecting key cellular pathways are a hallmark of pediatric acute lymphoblastic leukemia (ALL). To gain more insight into the mechanism underlying these deletions, we have studied the occurrence and nature of abnormalities in one of these genes, the B-cell translocation gene 1 (BTG1), in a large cohort of pediatric ALL cases. BTG1 was found to be exclusively affected by genomic deletions, which were detected in 65 out of 722 B-cell precursor ALL (BCP-ALL) patient samples (9%), but not in 109 T-ALL cases. Eight different deletion sizes were identified, which all clustered at the telomeric site in a hotspot region within the second (and last) exon of the BTG1 gene, resulting in the expression of truncated BTG1 read-through transcripts. The presence of V(D)J recombination signal sequences at both sites of virtually all deletions strongly suggests illegitimate RAG1/RAG2-mediated recombination as the responsible mechanism. Moreover, high levels of histone H3 lysine 4 trimethylation (H3K4me3), which is known to tether the RAG enzyme complex to DNA, were found within the BTG1 gene body in BCP-ALL cells, but not T-ALL cells. BTG1 deletions were rarely found in hyperdiploid BCP-ALLs, but were predominant in other cytogenetic subgroups, including the ETV6-RUNX1 and BCR-ABL1 positive BCP-ALL subgroups. Through sensitive PCR-based screening, we identified multiple additional BTG1 deletions at the subclonal level in BCP-ALL, with equal cytogenetic distribution which, in some cases, grew out into the major clone at relapse. Taken together, our results indicate that BTG1 deletions may act as "drivers" of leukemogenesis in specific BCP-ALL subgroups, in which they can arise independently in multiple subclones at sites that are prone to aberrant RAG1/RAG2-mediated recombination events. These findings provide further evidence for a complex and multiclonal evolution of ALL.


Asunto(s)
Evolución Clonal , Eliminación de Gen , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Enfermedad Aguda , Niño , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/patología
9.
Nat Genet ; 38(9): 999-1001, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16906164

RESUMEN

Submicroscopic genomic copy number changes have been identified only recently as an important cause of mental retardation. We describe the detection of three interstitial, overlapping 17q21.31 microdeletions in a cohort of 1,200 mentally retarded individuals associated with a clearly recognizable clinical phenotype of mental retardation, hypotonia and a characteristic face. The deletions encompass the MAPT and CRHR1 genes and are associated with a common inversion polymorphism.


Asunto(s)
Deleción Cromosómica , Inversión Cromosómica , Cromosomas Humanos Par 17 , Polimorfismo Genético , Adolescente , Adulto , Encéfalo/anomalías , Encéfalo/diagnóstico por imagen , Preescolar , Estudios de Cohortes , Cara/patología , Femenino , Dosificación de Gen , Frecuencia de los Genes , Haplotipos , Humanos , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Imagen por Resonancia Magnética , Masculino , Hipotonía Muscular/genética , Hipotonía Muscular/fisiopatología , Mapeo Físico de Cromosoma , Prevalencia , Radiografía , Receptores de Hormona Liberadora de Corticotropina/genética , Síndrome , Proteínas tau/genética
10.
Gastroenterology ; 145(3): 544-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23747338

RESUMEN

The spindle assembly checkpoint controls proper chromosome segregation during mitosis and prevents aneuploidy-an important feature of cancer cells. We performed genome-wide and targeted copy number and mutation analyses of germline DNA from 208 patients with familial or early-onset (40 years of age or younger) colorectal cancer; we identified haploinsufficiency or heterozygous mutations in the spindle assembly checkpoint genes BUB1 and BUB3 in 2.9% of them. Besides colorectal cancer, these patients had variegated aneuploidies in multiple tissues and variable dysmorphic features. These results indicate that mutations in BUB1 and BUB3 cause mosaic variegated aneuploidy and increase the risk of colorectal cancer at a young age.


Asunto(s)
Biomarcadores de Tumor/genética , Proteínas de Ciclo Celular/genética , Neoplasias Colorrectales/genética , Mutación de Línea Germinal , Proteínas Serina-Treonina Quinasas/genética , Adulto , Biomarcadores de Tumor/metabolismo , Estudios de Casos y Controles , Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorrectales/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Proteínas de Unión a Poli-ADP-Ribosa , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Riesgo
11.
Genes Chromosomes Cancer ; 52(1): 11-23, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22965931

RESUMEN

Uterine leiomyomas are benign solid tumors of mesenchymal origin which occur with an estimated incidence of up to 77% of all women of reproductive age. The majority of these tumors remains symptomless, but in about a quarter of cases they cause leiomyoma-associated symptoms including chronic pelvic pain, menorrhagia-induced anemia, and impaired fertility. As a consequence, they are the most common indication for pre-menopausal hysterectomy in the USA and Japan and annually translate into a multibillion dollar healthcare problem. Approximately 40% of these neoplasms present with recurring structural cytogenetic anomalies, including del(7)(q22), t(12;14)(q15;q24), t(1;2)(p36;p24), and anomalies affecting 6p21 and/or 10q22. Using positional cloning strategies, we and others previously identified HMGA1, HMGA2, RAD51L1, MORF, and, more recently, NCOA1 as primary target (fusion) genes associated with tumor initiation in four of these distinct cytogenetic subgroups. Despite the fact that the del(7)(q22) subgroup is the largest among leiomyomas, and was first described more than twenty years ago, the 7q22 leiomyoma target gene still awaits unequivocal identification. We here describe a positional cloning effort from two independent uterine leiomyomas, containing respectively a pericentric and a paracentric chromosomal inversion, both affecting band 7q22. We found that both chromosomal inversions target the cut-like homeobox 1 (CUX1) gene on chromosomal band 7q22.1 in a way which is functionally equivalent to the more frequently observed del(7q) cases, and which is compatible with a mono-allelic knock-out scenario, similar as was previously described for the cytogenetic subgroup showing chromosome 14q involvement.


Asunto(s)
Biomarcadores de Tumor/genética , Cromosomas Humanos Par 7 , Proteínas de Homeodominio/genética , Leiomioma/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Neoplasias Uterinas/genética , Secuencia de Aminoácidos , Secuencia de Bases , Femenino , Humanos , Hibridación Fluorescente in Situ , Persona de Mediana Edad , Datos de Secuencia Molecular , Factores de Transcripción
12.
Nat Genet ; 36(9): 955-7, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15300250

RESUMEN

CHARGE syndrome is a common cause of congenital anomalies affecting several tissues in a nonrandom fashion. We report a 2.3-Mb de novo overlapping microdeletion on chromosome 8q12 identified by array comparative genomic hybridization in two individuals with CHARGE syndrome. Sequence analysis of genes located in this region detected mutations in the gene CHD7 in 10 of 17 individuals with CHARGE syndrome without microdeletions, accounting for the disease in most affected individuals.


Asunto(s)
Anomalías Múltiples/genética , Atresia de las Coanas/genética , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Cardiopatías Congénitas/genética , Mutación , Coloboma/genética , Sordera/genética , Eliminación de Gen , Humanos , Análisis de Secuencia de ADN , Síndrome
13.
Blood ; 115(23): 4810-9, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20354172

RESUMEN

Resistance to glucocorticoids (GCs) is a major clinical problem in the treatment of acute lymphoblastic leukemia (ALL), but the underlying mechanisms are not well understood. Although mutations in the glucocorticoid receptor (GR) gene can give rise to therapy resistance in vitro, acquired somatic mutations in the GR are rarely encountered in patients. Here we report that the protein encoded by the BTG1 gene, which is frequently deleted in (pediatric) ALL, is a key determinant of GC responsiveness. Using RNA interference, we show that loss of BTG1 expression causes GC resistance both by decimating GR expression and by controlling GR-mediated transcription. Conversely, reexpression of BTG1 restores GC sensitivity by potentiating GC-induced GR expression, a phenomenon known as GR autoinduction. In addition, the arginine methyltransferase PRMT1, a BTG1-binding partner and transcriptional coactivator, is recruited to the GR gene promoter in a BTG1-dependent manner. These results implicate the BTG1/PRMT1 complex in GR-mediated gene expression and reveal that deregulation of a nuclear receptor coactivator complex can give rise to GC resistance. Further characterization of this complex as part of the GR regulatory circuitry could offer novel opportunities for improving the efficacy of GC-based therapies in ALL and other hematologic malignancies.


Asunto(s)
Resistencia a Antineoplásicos , Regulación Leucémica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Glucocorticoides/biosíntesis , Línea Celular Tumoral , Femenino , Eliminación de Gen , Glucocorticoides/efectos adversos , Glucocorticoides/farmacología , Glucocorticoides/uso terapéutico , Humanos , Masculino , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Regiones Promotoras Genéticas/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Interferencia de ARN , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
14.
Genes Chromosomes Cancer ; 50(12): 969-81, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21882283

RESUMEN

In acute lymphoblastic leukemia (ALL) specific genomic abnormalities provide important clinical information. In most routine clinical diagnostic laboratories conventional karyotyping, in conjunction with targeted screens using e.g., fluorescence in situ hybridization (FISH), is currently considered as the gold standard to detect such aberrations. Conventional karyotyping, however, is limited in its resolution and yield, thus hampering the genetic diagnosis of ALL. We explored whether microarray-based genomic profiling would be feasible as an alternative strategy in a routine clinical diagnostic setting. To this end, we compared conventional karyotypes with microarray-deduced copy number aberration (CNA) karyotypes in 60 ALL cases. Microarray-based genomic profiling resulted in a CNA detection rate of 90%, whereas for conventional karyotyping this was 61%. In addition, many small (< 5 Mb) genetic lesions were encountered, frequently harboring clinically relevant ALL-related genes such as CDKN2A/B, ETV6, PAX5, and IKZF1. From our data we conclude that microarray-based genomic profiling serves as a robust tool in the genetic diagnosis of ALL, outreaching conventional karyotyping in CNA detection both in terms of sensitivity and specificity. We also propose a practical workflow for a comprehensive and objective interpretation of CNAs obtained through microarray-based genomic profiling, thereby facilitating its application in a routine clinical diagnostic setting.


Asunto(s)
Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Niño , Aberraciones Cromosómicas , Dermatoglifia del ADN/métodos , Femenino , Dosificación de Gen , Humanos , Hibridación Fluorescente in Situ/métodos , Cariotipificación/métodos , Masculino , Polimorfismo de Nucleótido Simple , Sensibilidad y Especificidad
15.
Lancet Oncol ; 12(1): 49-55, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21145788

RESUMEN

BACKGROUND: Lynch syndrome is caused by germline mutations in MSH2, MLH1, MSH6, and PMS2 mismatch-repair genes and leads to a high risk of colorectal and endometrial cancer. We previously showed that constitutional 3' end deletions of EPCAM can cause Lynch syndrome through epigenetic silencing of MSH2 in EPCAM-expressing tissues, resulting in tissue-specific MSH2 deficiency. We aim to establish the risk of cancer associated with such EPCAM deletions. METHODS: We obtained clinical data for 194 carriers of a 3' end EPCAM deletion from 41 families known to us at the Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands and compared cancer risk with data from a previously described cohort of 473 carriers from 91 families with mutations in MLH1, MSH2, MSH6, or a combined EPCAM-MSH2 deletion. FINDINGS: 93 of the 194 EPCAM deletion carriers were diagnosed with colorectal cancer; three of the 92 women with EPCAM deletions were diagnosed with endometrial cancer. Carriers of an EPCAM deletion had a 75% (95% CI 65-85) cumulative risk of colorectal cancer before the age of 70 years (mean age at diagnosis 43 years [SD 12]), which did not differ significantly from that of carriers of combined EPCAM-MSH2 deletion (69% [95% CI 47-91], p=0·8609) or mutations in MSH2 (77% [64-90], p=0·5892) or MLH1 (79% [68-90], p=0·5492), but was higher than noted for carriers of MSH6 mutation (50% [38-62], p<0·0001). By contrast, women with EPCAM deletions had a 12% [0-27] cumulative risk of endometrial cancer, which was lower than was that noted for carriers of a combined EPCAM-MSH2 deletion (55% [20-90], p<0·0001) or of a mutation in MSH2 (51% [33-69], p=0·0006) or MSH6 (34% [20-48], p=0·0309), but did not differ significantly from that noted for MLH1 (33% [15-51], p=0·1193) mutation carriers. This risk seems to be restricted to deletions that extend close to the MSH2 gene promoter. Of 194 carriers of an EPCAM deletion, three had duodenal cancer and four had pancreatic cancer. INTERPRETATION: EPCAM deletion carriers have a high risk of colorectal cancer; only those with deletions extending close to the MSH2 promoter have an increased risk of endometrial cancer. These results underscore the effect of mosaic MSH2 deficiency, leading to variable cancer risks, and could form the basis of an optimised protocol for the recognition and targeted prevention of cancer in EPCAM deletion carriers.


Asunto(s)
Antígenos de Neoplasias/genética , Moléculas de Adhesión Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Endometriales/genética , Eliminación de Secuencia , Adolescente , Adulto , Anciano , Estudios de Cohortes , Neoplasias Colorrectales/etiología , Neoplasias Endometriales/etiología , Molécula de Adhesión Celular Epitelial , Femenino , Eliminación de Gen , Humanos , Masculino , Persona de Mediana Edad , Proteína 2 Homóloga a MutS/genética , Regiones Promotoras Genéticas , Riesgo
16.
Hum Mutat ; 32(4): 407-14, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21309036

RESUMEN

Recently, we identified 3' end deletions in the EPCAM gene as a novel cause of Lynch syndrome. These truncating EPCAM deletions cause allele-specific epigenetic silencing of the neighboring DNA mismatch repair gene MSH2 in tissues expressing EPCAM. Here we screened a cohort of unexplained Lynch-like families for the presence of EPCAM deletions. We identified 27 novel independent MSH2-deficient families from multiple geographical origins with varying deletions all encompassing the 3' end of EPCAM, but leaving the MSH2 gene intact. Within The Netherlands and Germany, EPCAM deletions appeared to represent at least 2.8% and 1.1% of the confirmed Lynch syndrome families, respectively. MSH2 promoter methylation was observed in epithelial tissues of all deletion carriers tested, thus confirming silencing of MSH2 as the causative defect. In a total of 45 families, 19 different deletions were found, all including the last two exons and the transcription termination signal of EPCAM. All deletions appeared to originate from Alu-repeat mediated recombination events. In 17 cases regions of microhomology around the breakpoints were found, suggesting nonallelic homologous recombination as the most likely mechanism. We conclude that 3' end EPCAM deletions are a recurrent cause of Lynch syndrome, which should be implemented in routine Lynch syndrome diagnostics.


Asunto(s)
Antígenos de Neoplasias/genética , Moléculas de Adhesión Celular/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Variación Genética , Mutación de Línea Germinal/genética , Eliminación de Secuencia/genética , Antígenos de Neoplasias/metabolismo , Secuencia de Bases , Moléculas de Adhesión Celular/metabolismo , Metilación de ADN , Molécula de Adhesión Celular Epitelial , Modelos Genéticos , Datos de Secuencia Molecular , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Países Bajos , Regiones Promotoras Genéticas , Recurrencia
17.
Int J Cancer ; 129(7): 1635-42, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21128281

RESUMEN

In the majority of colorectal cancers (CRCs) under clinical suspicion for a hereditary cause, the disease-causing genetic factors are still to be discovered. To identify such genetic factors we stringently selected a discovery cohort of 41 CRC index patients with microsatellite-stable tumors. All patients were below 40 years of age at diagnosis and/or exhibited an overt family history. We employed genome-wide copy number profiling using high-resolution SNP arrays on germline DNA, which resulted in the identification of novel copy number variants (CNVs) in six patients (15%) encompassing, among others, the cadherin gene CDH18, the bone morphogenetic protein antagonist family gene GREM1, and the breakpoint cluster region gene BCR. In addition, two genomic deletions were encountered encompassing two microRNA genes, hsa-mir-491/KIAA1797 and hsa-mir-646/AK309218. None of these CNVs has previously been reported in relation to CRC predisposition in humans, nor were they encountered in large control cohorts (>1,600 unaffected individuals). Since several of these newly identified candidate genes may be functionally linked to CRC development, our results illustrate the potential of this approach for the identification of novel candidate genes involved in CRC predisposition.


Asunto(s)
Variaciones en el Número de Copia de ADN , Predisposición Genética a la Enfermedad , Adulto , Cadherinas/genética , Estudios de Cohortes , Neoplasias Colorrectales/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética
18.
Hum Mol Genet ; 18(19): 3579-93, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19578123

RESUMEN

Genomic copy number variation (CNV) plays a major role in various human diseases as well as in normal phenotypic variability. For some recurrent disease-causing CNVs that convey genomic disorders, the causative mechanism is meiotic, non-allelic, homologous recombination between breakpoint regions exhibiting extensive sequence homology (e.g. low-copy repeats). For the majority of recently identified rare pathogenic CNVs, however, the mechanism is unknown. Recently, a model for CNV formation implicated mitotic replication-based mechanisms, such as (alternative) non-homologous end joining and fork stalling and template switching, in the etiology of human pathogenic CNVs. The extent to which such mitotic mechanisms contribute to rare pathogenic CNVs remains to be determined. In addition, it is unexplored whether genomic architectural features such as repetitive elements or sequence motifs associated with DNA breakage stimulate the formation of rare pathogenic CNVs. To this end, we have sequenced breakpoint junctions of 30 rare pathogenic microdeletions and eight tandem duplications, representing the largest series of such CNVs examined to date in this much detail. Our results demonstrate the presence of (micro)homology ranging from 2 to over 75 bp, in 79% of the breakpoint junctions. This indicates that microhomology-mediated repair mechanisms, including the recently reported fork stalling and template switching and/or microhomology-mediated break-induced replication, prevail in rare pathogenic CNVs. In addition, we found that the vast majority of all breakpoints (81%) were associated with at least one of the genomic architectural features evaluated. Moreover, 75% of tandem duplication breakpoints were associated with the presence of one of two novel sequence motifs. These data suggest that rare pathogenic microdeletions and tandem duplications do not occur at random genome sequences, but are stimulated and potentially catalyzed by various genomic architectural features.


Asunto(s)
Rotura Cromosómica , Enfermedad/genética , Duplicación de Gen , Genoma Humano , Eliminación de Secuencia , Elementos Alu , Secuencia de Bases , Femenino , Dosificación de Gen , Humanos , Masculino , Datos de Secuencia Molecular , Homología de Secuencia de Ácido Nucleico
19.
Am J Hum Genet ; 83(4): 504-10, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18940311

RESUMEN

Schizophrenia is a severe psychiatric disease with complex etiology, affecting approximately 1% of the general population. Most genetics studies so far have focused on disease association with common genetic variation, such as single-nucleotide polymorphisms (SNPs), but it has recently become apparent that large-scale genomic copy-number variants (CNVs) are involved in disease development as well. To assess the role of rare CNVs in schizophrenia, we screened 54 patients with deficit schizophrenia using Affymetrix's GeneChip 250K SNP arrays. We identified 90 CNVs in total, 77 of which have been reported previously in unaffected control cohorts. Among the genes disrupted by the remaining rare CNVs are MYT1L, CTNND2, NRXN1, and ASTN2, genes that play an important role in neuronal functioning but--except for NRXN1--have not been associated with schizophrenia before. We studied the occurrence of CNVs at these four loci in an additional cohort of 752 patients and 706 normal controls from The Netherlands. We identified eight additional CNVs, of which the four that affect coding sequences were found only in the patient cohort. Our study supports a role for rare CNVs in schizophrenia susceptibility and identifies at least three candidate genes for this complex disorder.


Asunto(s)
Variación Genética , Polimorfismo de Nucleótido Simple , Esquizofrenia/genética , Adolescente , Adulto , Estudios de Cohortes , Exones , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Modelos Genéticos , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Recurrencia
20.
Genet Med ; 13(7): 643-50, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21597380

RESUMEN

PURPOSE: Women who carry a fragile X mental retardation 1 premutation are at risk for fragile X-associated primary ovarian insufficiency and should be counseled for a potentially reduced fertility. Multiple factors can affect the age of onset and severity of fragile X-associated primary ovarian insufficiency. In this study, we assessed the predictive power of several factors with menopausal age, a surrogate measure of onset of fragile X-associated primary ovarian insufficiency. METHODS: Genetic, environmental, and reproductive factors were analyzed by Cox proportional hazard models in 1068 women, 385 of fragile X families ascertained through the Nijmegen study and 683 of fragile X families or general population families ascertained through the Atlanta study. RESULTS: The highest association with menopausal age among fragile X mental retardation 1 premutation carriers was found for risk index by CGG repeat size (hazard ratio, 1.43) and smoking (hazard ratio, 1.34). Women from the Nijmegen cohort showed an overall lower age at menopause onset, for which a correction was made. A prediction model based on these two parameters, mean menopausal age of first-degree relatives with the same mutation status and the correction for ascertainment site, estimated the probability of becoming postmenopausal for premutation carriers, with a margin of 7-10%. CONCLUSION: We conclude that this model serves as a first step toward clinical application of fragile X-associated primary ovarian insufficiency prediction.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Menopausia/genética , Insuficiencia Ovárica Primaria/genética , Adolescente , Adulto , Factores de Edad , Anciano , Secuencia de Bases , Estudios de Cohortes , Salud de la Familia , Femenino , Síndrome del Cromosoma X Frágil/complicaciones , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Persona de Mediana Edad , Mutación , Insuficiencia Ovárica Primaria/complicaciones , Insuficiencia Ovárica Primaria/diagnóstico , Pronóstico , Modelos de Riesgos Proporcionales , Medición de Riesgo/estadística & datos numéricos , Factores de Riesgo , Encuestas y Cuestionarios , Expansión de Repetición de Trinucleótido/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA