Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Hum Genet ; 109(8): 1472-1483, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35931051

RESUMEN

Dyskeratosis congenita (DC) is an inherited bone-marrow-failure disorder characterized by a triad of mucocutaneous features that include abnormal skin pigmentation, nail dystrophy, and oral leucoplakia. Despite the identification of several genetic variants that cause DC, a significant proportion of probands remain without a molecular diagnosis. In a cohort of eight independent DC-affected families, we have identified a remarkable series of heterozygous germline variants in the gene encoding thymidylate synthase (TYMS). Although the inheritance appeared to be autosomal recessive, one parent in each family had a wild-type TYMS coding sequence. Targeted genomic sequencing identified a specific haplotype and rare variants in the naturally occurring TYMS antisense regulator ENOSF1 (enolase super family 1) inherited from the other parent. Lymphoblastoid cells from affected probands have severe TYMS deficiency, altered cellular deoxyribonucleotide triphosphate pools, and hypersensitivity to the TYMS-specific inhibitor 5-fluorouracil. These defects in the nucleotide metabolism pathway resulted in genotoxic stress, defective transcription, and abnormal telomere maintenance. Gene-rescue studies in cells from affected probands revealed that post-transcriptional epistatic silencing of TYMS is occurring via elevated ENOSF1. These cell and molecular abnormalities generated by the combination of germline digenic variants at the TYMS-ENOSF1 locus represent a unique pathogenetic pathway for DC causation in these affected individuals, whereas the parents who are carriers of either of these variants in a singular fashion remain unaffected.


Asunto(s)
Disqueratosis Congénita , Timidilato Sintasa , Disqueratosis Congénita/genética , Células Germinativas , Heterocigoto , Humanos , Nucleótidos , Timidilato Sintasa/deficiencia , Timidilato Sintasa/genética
2.
Proc Natl Acad Sci U S A ; 117(29): 17151-17155, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32636268

RESUMEN

Inherited bone marrow failure (BMF) syndromes are a heterogeneous group of diseases characterized by defective hematopoiesis and often predisposing to myelodysplastic syndrome (MDS) and acute myelogenous leukemia. We have studied a large family consisting of several affected individuals with hematologic abnormalities, including one family member who died of acute leukemia. By whole-exome sequencing, we identified a novel frameshift variant in the ubiquitously expressed transcription factor specificity protein 1 (SP1). This heterozygous variant (c.1995delA) truncates the canonical Sp1 molecule in the highly conserved C-terminal DNA-binding zinc finger domains. Transcriptomic analysis and gene promoter characterization in patients' blood revealed a hypermorphic effect of this Sp1 variant, triggering superactivation of Sp1-mediated transcription and driving significant up-regulation of Sp1 target genes. This familial genetic study indicates a central role for Sp1 in causing autosomal dominant transmission of BMF, thereby confirming its critical role in hematopoiesis in humans.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/genética , Mutación del Sistema de Lectura/genética , Factor de Transcripción Sp1/genética , Transcripción Genética/genética , Femenino , Humanos , Masculino , Linaje , Transcriptoma/genética , Regulación hacia Arriba/genética , Dedos de Zinc/genética
4.
Hum Genet ; 140(6): 945-955, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33709208

RESUMEN

Telomere biology disorders are complex clinical conditions that arise due to mutations in genes required for telomere maintenance. Telomere length has been utilised as part of the diagnostic work-up of patients with these diseases; here, we have tested the utility of high-throughput STELA (HT-STELA) for this purpose. HT-STELA was applied to a cohort of unaffected individuals (n = 171) and a retrospective cohort of mutation carriers (n = 172). HT-STELA displayed a low measurement error with inter- and intra-assay coefficient of variance of 2.3% and 1.8%, respectively. Whilst telomere length in unaffected individuals declined as a function of age, telomere length in mutation carriers appeared to increase due to a preponderance of shorter telomeres detected in younger individuals (< 20 years of age). These individuals were more severely affected, and age-adjusted telomere length differentials could be used to stratify the cohort for overall survival (Hazard Ratio = 5.6 (1.5-20.5); p < 0.0001). Telomere lengths of asymptomatic mutation carriers were shorter than controls (p < 0.0001), but longer than symptomatic mutation carriers (p < 0.0001) and telomere length heterogeneity was dependent on the diagnosis and mutational status. Our data show that the ability of HT-STELA to detect short telomere lengths, that are not readily detected with other methods, means it can provide powerful diagnostic discrimination and prognostic information. The rapid format, with a low measurement error, demonstrates that HT-STELA is a new high-quality laboratory test for the clinical diagnosis of an underlying telomeropathy.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/diagnóstico , Disqueratosis Congénita/diagnóstico , Retardo del Crecimiento Fetal/diagnóstico , Tamización de Portadores Genéticos/métodos , Discapacidad Intelectual/diagnóstico , Microcefalia/diagnóstico , Telómero/patología , Adolescente , Adulto , Factores de Edad , Anciano , Enfermedades Asintomáticas , Trastornos de Fallo de la Médula Ósea/genética , Trastornos de Fallo de la Médula Ósea/patología , Estudios de Casos y Controles , Niño , Preescolar , Disqueratosis Congénita/genética , Disqueratosis Congénita/patología , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/patología , Heterocigoto , Humanos , Lactante , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Masculino , Microcefalia/genética , Microcefalia/patología , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Telómero/metabolismo , Homeostasis del Telómero
5.
Proc Natl Acad Sci U S A ; 115(30): 7777-7782, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29987015

RESUMEN

Biallelic variants in the ERCC excision repair 6 like 2 gene (ERCC6L2) are known to cause bone marrow failure (BMF) due to defects in DNA repair and mitochondrial function. Here, we report on eight cases of BMF from five families harboring biallelic variants in ERCC6L2, two of whom present with myelodysplasia. We confirm that ERCC6L2 patients' lymphoblastoid cell lines (LCLs) are hypersensitive to DNA-damaging agents that specifically activate the transcription coupled nucleotide excision repair (TCNER) pathway. Interestingly, patients' LCLs are also hypersensitive to transcription inhibitors that interfere with RNA polymerase II (RNA Pol II) and display an abnormal delay in transcription recovery. Using affinity-based mass spectrometry we found that ERCC6L2 interacts with DNA-dependent protein kinase (DNA-PK), a regulatory component of the RNA Pol II transcription complex. Chromatin immunoprecipitation PCR studies revealed ERCC6L2 occupancy on gene bodies along with RNA Pol II and DNA-PK. Patients' LCLs fail to terminate transcript elongation accurately upon DNA damage and display a significant increase in nuclear DNA-RNA hybrids (R loops). Collectively, we conclude that ERCC6L2 is involved in regulating RNA Pol II-mediated transcription via its interaction with DNA-PK to resolve R loops and minimize transcription-associated genome instability. The inherited BMF syndrome caused by biallelic variants in ERCC6L2 can be considered as a primary transcription deficiency rather than a DNA repair defect.


Asunto(s)
Alelos , Enfermedades de la Médula Ósea/metabolismo , ADN Helicasas/metabolismo , Reparación del ADN , Enfermedades Genéticas Congénitas/metabolismo , Inestabilidad Genómica , Transcripción Genética , Células A549 , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/patología , ADN Helicasas/genética , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Femenino , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Células HeLa , Humanos , Masculino , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Síndrome
6.
Am J Hum Genet ; 99(1): 115-24, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27346687

RESUMEN

A substantial number of individuals with bone marrow failure (BMF) present with one or more extra-hematopoietic abnormality. This suggests a constitutional or inherited basis, and yet many of them do not fit the diagnostic criteria of the known BMF syndromes. Through exome sequencing, we have now identified a subgroup of these individuals, defined by germline biallelic mutations in DNAJC21 (DNAJ homolog subfamily C member 21). They present with global BMF, and one individual developed a hematological cancer (acute myeloid leukemia) in childhood. We show that the encoded protein associates with rRNA and plays a highly conserved role in the maturation of the 60S ribosomal subunit. Lymphoblastoid cells obtained from an affected individual exhibit increased sensitivity to the transcriptional inhibitor actinomycin D and reduced amounts of rRNA. Characterization of mutations revealed impairment in interactions with cofactors (PA2G4, HSPA8, and ZNF622) involved in 60S maturation. DNAJC21 deficiency resulted in cytoplasmic accumulation of the 60S nuclear export factor PA2G4, aberrant ribosome profiles, and increased cell death. Collectively, these findings demonstrate that mutations in DNAJC21 cause a cancer-prone BMF syndrome due to corruption of early nuclear rRNA biogenesis and late cytoplasmic maturation of the 60S subunit.


Asunto(s)
Anemia Aplásica/complicaciones , Anemia Aplásica/genética , Enfermedades de la Médula Ósea/complicaciones , Enfermedades de la Médula Ósea/genética , Proteínas del Choque Térmico HSP40/genética , Hemoglobinuria Paroxística/complicaciones , Hemoglobinuria Paroxística/genética , Mutación/genética , Neoplasias/complicaciones , Neoplasias/genética , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/patología , Secuencia de Aminoácidos , Trastornos de Fallo de la Médula Ósea , Proliferación Celular , Forma de la Célula , Niño , Preescolar , Femenino , Proteínas del Choque Térmico HSP40/química , Humanos , Leucemia Mieloide Aguda/complicaciones , Leucemia Mieloide Aguda/genética , Masculino , Unión Proteica , ARN Ribosómico/biosíntesis
7.
Am J Hum Genet ; 94(2): 246-56, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24507776

RESUMEN

Exome sequencing was performed in three index cases with bone marrow failure and neurological dysfunction and whose parents are first-degree cousins. Homozygous truncating mutations were identified in ERCC6L2 in two of the individuals. Both of these mutations affect the subcellular localization and stability of ERCC6L2. We show here that knockdown of ERCC6L2 in human A549 cells significantly reduced their viability upon exposure to the DNA-damaging agents mitomycin C and Irofulven, but not etoposide and camptothecin, suggesting a role in nucleotide excision repair. ERCC6L2-knockdown cells also displayed H2AX phosphorylation, which significantly increased upon genotoxic stress, suggesting an early DNA-damage response. Intriguingly, ERCC6L2 was seen to translocate to the mitochondria and the nucleus in response to DNA damage, and ERCC6L2 knockdown induced intracellular reactive oxygen species (ROS). Treatment with the ROS scavenger N-acetyl cysteine attenuated the Irofulven-induced cytotoxicity in ERCC6L2-knockdown cells and abolished ERCCGL2 traffic to the mitochondria and nucleus in response to this DNA-damaging agent. Collectively, these observations identify a distinct bone-marrow-failure syndrome due to mutations in ERCC6L2, a gene implicated in DNA repair and mitochondrial function.


Asunto(s)
ADN Helicasas/genética , Reparación del ADN/genética , Hemoglobinuria Paroxística/genética , Mitocondrias/genética , Acetilcisteína/metabolismo , Anemia Aplásica , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Mitocondrias/efectos de los fármacos , Mitomicina/toxicidad , Mutación , Linaje , Fosforilación , Especies Reactivas de Oxígeno , Sesquiterpenos/toxicidad
8.
Blood ; 126(2): 176-84, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26024875

RESUMEN

Dyskeratosis congenita (DC) and related diseases are a heterogeneous group of disorders characterized by impaired telomere maintenance, known collectively as the telomeropathies. Disease-causing variants have been identified in 10 telomere-related genes including the reverse transcriptase (TERT) and the RNA component (TERC) of the telomerase complex. Variants in TERC and TERT can impede telomere elongation causing stem cells to enter premature replicative senescence and/or apoptosis as telomeres become critically short. This explains the major impact of the disease on highly proliferative tissues such as the bone marrow and skin. However, telomerase variants are not always fully penetrant and in some families disease-causing variants are seen in asymptomatic family members. As a result, determining the pathogenic status of newly identified variants in TERC or TERT can be quite challenging. Over a 3-year period, we have identified 26 telomerase variants (16 of which are novel) in 23 families. Additional investigations (including family segregation and functional studies) enabled these to be categorized into 3 groups: (1) disease-causing (n = 15), (2) uncertain status (n = 6), and (3) bystanders (n = 5). Remarkably, this process has also enabled us to identify families with novel mechanisms of inheriting human telomeropathies. These include triallelic mutations, involving 2 different telomerase genes, and an epigenetic-like inheritance of short telomeres in the absence of a telomerase mutation. This study therefore highlights that telomerase variants have highly variable functional and clinical manifestations and require thorough investigation to assess their pathogenic contribution.


Asunto(s)
Disqueratosis Congénita/genética , Epigénesis Genética , Telomerasa/genética , Alelos , Secuencia de Bases , Células Cultivadas , Estudios de Cohortes , Familia , Humanos , Patrón de Herencia , Datos de Secuencia Molecular , Mutación , Linaje , ARN/genética
9.
Am J Hum Genet ; 92(3): 448-53, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23453664

RESUMEN

Dyskeratosis congenita (DC) and its phenotypically severe variant, Hoyeraal-Hreidarsson syndrome (HHS), are multisystem bone-marrow-failure syndromes in which the principal pathology is defective telomere maintenance. The genetic basis of many cases of DC and HHS remains unknown. Using whole-exome sequencing, we identified biallelic mutations in RTEL1, encoding a helicase essential for telomere maintenance and regulation of homologous recombination, in an individual with familial HHS. Additional screening of RTEL1 identified biallelic mutations in 6/23 index cases with HHS but none in 102 DC or DC-like cases. All 11 mutations in ten HHS individuals from seven families segregated in an autosomal-recessive manner, and telomere lengths were significantly shorter in cases than in controls (p = 0.0003). This group had significantly higher levels of telomeric circles, produced as a consequence of incorrect processing of telomere ends, than did controls (p = 0.0148). These biallelic RTEL1 mutations are responsible for a major subgroup (∼29%) of HHS. Our studies show that cells harboring these mutations have significant defects in telomere maintenance, but not in homologous recombination, and that incorrect resolution of T-loops is a mechanism for telomere shortening and disease causation in humans. They also demonstrate the severe multisystem consequences of its dysfunction.


Asunto(s)
ADN Helicasas/genética , Disqueratosis Congénita/genética , Retardo del Crecimiento Fetal/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Mutación , Adolescente , Secuencia de Aminoácidos , Niño , Preescolar , Exoma , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia de ADN/métodos , Análisis de Secuencia de Proteína/métodos , Telómero/genética , Acortamiento del Telómero/genética
10.
Haematologica ; 101(10): 1180-1189, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27612988

RESUMEN

Dyskeratosis congenita is a highly pleotropic genetic disorder. This heterogeneity can lead to difficulties in making an accurate diagnosis and delays in appropriate management. The aim of this study was to determine the underlying genetic basis in patients presenting with features of dyskeratosis congenita and who were negative for mutations in the classical dyskeratosis congenita genes. By whole exome and targeted sequencing, we identified biallelic variants in genes that are not associated with dyskeratosis congenita in 17 individuals from 12 families. Specifically, these were homozygous variants in USB1 (8 families), homozygous missense variants in GRHL2 (2 families) and identical compound heterozygous variants in LIG4 (2 families). All patients had multiple somatic features of dyskeratosis congenita but not the characteristic short telomeres. Our case series shows that biallelic variants in USB1, LIG4 and GRHL2, the genes mutated in poikiloderma with neutropenia, LIG4/Dubowitz syndrome and the recently recognized ectodermal dysplasia/short stature syndrome, respectively, cause features that overlap with dyskeratosis congenita. Strikingly, these genes also overlap in their biological function with the known dyskeratosis congenita genes that are implicated in telomere maintenance and DNA repair pathways. Collectively, these observations demonstrate the marked overlap of dyskeratosis congenita with four other genetic syndromes, confounding accurate diagnosis and subsequent management. This has important implications for establishing a genetic diagnosis when a new patient presents in the clinic. Patients with clinical features of dyskeratosis congenita need to have genetic analysis of USB1, LIG4 and GRHL2 in addition to the classical dyskeratosis congenita genes and telomere length measurements.


Asunto(s)
Disqueratosis Congénita/diagnóstico , Disqueratosis Congénita/genética , Exoma/genética , Variación Genética/genética , ADN Ligasa (ATP)/genética , Proteínas de Unión al ADN/genética , Humanos , Linaje , Hidrolasas Diéster Fosfóricas/genética , Análisis de Secuencia de ADN , Síndrome , Factores de Transcripción/genética
11.
Am J Hum Genet ; 90(5): 888-92, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22541560

RESUMEN

Aplastic anemia (AA) and myelodysplasia (MDS) are forms of bone marrow failure that are often part of the same progressive underlying disorder. While most cases are simplex and idiopathic, some show a clear pattern of inheritance; therefore, elucidating the underlying genetic cause could lead to a greater understanding of this spectrum of disorders. We used a combination of exome sequencing and SNP haplotype analysis to identify causative mutations in a family with a history of autosomal-dominant AA/MDS. We identified a heterozygous mutation in SRP72, a component of the signal recognition particle (SRP) that is responsible for the translocation of nascent membrane-bound and excreted proteins to the endoplasmic reticulum. A subsequent screen revealed another autosomal-dominant family with an inherited heterozygous SRP72 mutation. Transfection of these sequences into mammalian cells suggested that these proteins localize incorrectly within the cell. Furthermore, coimmunoprecipitation of epitope-tagged SRP72 indicated that the essential RNA component of the SRP did not fully associate with one of the SRP72 variants. These results suggest that inherited mutations in a component of the SRP have a role in the pathophysiology of AA/MDS, identifying a third pathway for developing these disorders alongside transcription factor and telomerase mutations.


Asunto(s)
Anemia Aplásica/genética , Exoma , Mutación , Síndromes Mielodisplásicos/genética , Partícula de Reconocimiento de Señal/genética , Anemia Aplásica/fisiopatología , Análisis Mutacional de ADN , Bases de Datos Genéticas , Femenino , Biblioteca de Genes , Heterocigoto , Humanos , Masculino , Síndromes Mielodisplásicos/fisiopatología , Linaje , ARN/genética , Partícula de Reconocimiento de Señal/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
N Engl J Med ; 365(16): 1502-8, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-22010916

RESUMEN

We performed genetic and immunohistochemical studies in a sister and brother with autosomal recessive neonatal inflammatory skin and bowel lesions. The girl died suddenly at 12 years of age from parvovirus B19-associated myocarditis; her brother had mild cardiomyopathy. We identified a loss-of-function mutation in ADAM17, which encodes a disintegrin and metalloproteinase 17 (also called tumor necrosis factor α [TNF-α]-converting enzyme, or TACE), as the probable cause of this syndrome. Peripheral-blood mononuclear cells (PBMCs) obtained from the brother at 17 years of age showed high levels of lipopolysaccharide-induced production of interleukin-1ß and interleukin-6 but impaired release of TNF-α. Despite repeated skin infections, this young man has led a relatively normal life. (Funded by Barts and the London Charity and the European Commission Seventh Framework Programme.).


Asunto(s)
Proteínas ADAM/genética , Enfermedades Inflamatorias del Intestino/genética , Eliminación de Secuencia , Enfermedades de la Piel/genética , Proteína ADAM17 , Adolescente , Niño , Resultado Fatal , Femenino , Humanos , Masculino , Miocarditis/genética , Miocarditis/virología , Linaje
15.
Haematologica ; 98(3): 334-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22899577

RESUMEN

Dyskeratosis congenita and its variants have overlapping phenotypes with many disorders including Coats plus, and their underlying pathology is thought to be one of defective telomere maintenance. Recently, biallelic CTC1 mutations have been described in patients with syndromes overlapping Coats plus. CTC1, STN1 and TEN1 are part of the telomere-capping complex involved in maintaining telomeric structural integrity. Based on phenotypic overlap we screened 73 genetically uncharacterized patients with dyskeratosis congenita and related bone marrow failure syndromes for mutations in this complex. Biallelic CTC1 mutations were identified in 6 patients but none in either STN1 or TEN1. We have expanded the phenotypic spectrum associated with CTC1 mutations and report that intracranial and retinal abnormalities are not a defining feature, as well as showing that the effect of these mutations on telomere length is variable. The study also demonstrates the lack of disease-causing mutations in other components of the telomere-capping complex.


Asunto(s)
Disqueratosis Congénita/genética , Hemoglobinuria Paroxística/genética , Mutación , Proteínas de Unión a Telómeros/genética , Anemia Aplásica , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Disqueratosis Congénita/diagnóstico , Femenino , Hemoglobinuria Paroxística/diagnóstico , Heterocigoto , Humanos , Masculino , Linaje , Sistema de Registros , Telómero/metabolismo , Acortamiento del Telómero
16.
Nat Genet ; 36(5): 447-9, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15098033

RESUMEN

Telomerase is a ribonucleoprotein complex that is required to synthesize DNA repeats at the ends of each chromosome. The RNA component of this reverse transcriptase is mutated in the bone marrow failure syndrome autosomal dominant dyskeratosis congenita. Here we show that disease anticipation is observed in families with this disease and that this is associated with progressive telomere shortening.


Asunto(s)
Disqueratosis Congénita/genética , Mutación/genética , ARN/genética , Telomerasa/genética , Telómero/genética , Adolescente , Adulto , Niño , Preescolar , Disqueratosis Congénita/diagnóstico , Familia , Femenino , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Linaje , Eliminación de Secuencia
17.
Hum Mol Genet ; 19(22): 4453-61, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20817924

RESUMEN

Dyskeratosis congenita (DC) is an inherited poikiloderma which in addition to the skin abnormalities is typically associated with nail dystrophy, leucoplakia, bone marrow failure, cancer predisposition and other features. Approximately 50% of DC patients remain genetically uncharacterized. All the DC genes identified to date are important in telomere maintenance. To determine the genetic basis of the remaining cases of DC, we undertook linkage analysis in 20 families and identified a common candidate gene region on chromosome 16 in a subset of these. This region included the C16orf57 gene recently identified to be mutated in poikiloderma with neutropenia (PN), an inherited poikiloderma displaying significant clinical overlap with DC. Analysis of the C16orf57 gene in our uncharacterized DC patients revealed homozygous mutations in 6 of 132 families. In addition, three of six families previously classified as Rothmund-Thomson syndrome (RTS-a poikiloderma that is sometimes confused with PN) were also found to have homozygous C16orf57 mutations. Given the role of the previous DC genes in telomere maintenance, telomere length was analysed in these patients and found to be comparable to age-matched controls. These findings suggest that mutations in C16orf57 unify a distinct set of families which clinically can be categorized as DC, PN or RTS. This study also highlights the multi-system nature (wider than just poikiloderma and neutropenia) of the clinical features of affected individuals (and therefore house-keeping function of C16orf57), a possible role for C16orf57 in apoptosis, as well as a distinct difference from previously characterized DC patients because telomere length was normal.


Asunto(s)
Disqueratosis Congénita/genética , Neutropenia/genética , Proteínas Nucleares/genética , Trastornos de la Pigmentación/genética , Síndrome Rothmund-Thomson/genética , Telómero/genética , Estudios de Casos y Controles , Consanguinidad , Análisis Mutacional de ADN , Femenino , Proteínas Activadoras de GTPasa , Ligamiento Genético , Homocigoto , Humanos , Masculino , Mutación , Polimorfismo de Nucleótido Simple , Telómero/metabolismo
18.
Br J Haematol ; 158(2): 242-248, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22533337

RESUMEN

The myelodysplastic syndromes (MDS) are heterogeneous and can evolve into acute myeloid leukaemia (AML). Rare familial cases are reported in which five disease genes have been identified to date (RUNX1, CEBPA, TERC, TERT and GATA2). Here we report the genetic categorization of 27 families with familial MDS/AML. All of these families were screened for RUNX1, CEBPA, TERC, TERT and GATA2 as well as TET2 and NPM1. Five of the 27 families had telomerase mutations; one had a RUNX1 mutation, while none were found to have TET2, CEBPA or NPM1 mutations. We identified four families with heterozygous GATA2 mutations, each associated with a different phenotype. While one of these mutations is novel, three have been previously reported: one has been described in dendritic cell, monocyte, B and NK lymphoid (DCML) deficiency and one is in a family that has been reported in a series with primary lymphoedema with a predisposition to AML (Emberger syndrome). In summary, genetic characterization was shown in 10 (four GATA2, three TERT, two TERC, one RUNX1) of these families; however 17 remain uncharacterized, highlighting marked genetic heterogeneity in familial MDS/AML and the scope for further functional pathways that could give rise to this group of disorders.


Asunto(s)
Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Adolescente , Adulto , Niño , Preescolar , Cromatografía Líquida de Alta Presión/métodos , ADN de Neoplasias/genética , Factor de Transcripción GATA2/genética , Genes Relacionados con las Neoplasias , Predisposición Genética a la Enfermedad , Humanos , Persona de Mediana Edad , Mutación , Nucleofosmina , Linaje , Fenotipo , Adulto Joven
19.
Haematologica ; 97(4): 524-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22180433

RESUMEN

The primary cause of aplastic anemia remains unknown in many patients. The aim of this study was to clarify the genetic cause of familial aplastic anemia. Genomic DNA of an affected individual from a multiplex consanguineous family was hybridized to a Nimblegen exome library before being sequenced on a GAIIx genome analyzer. Once the disease causing homozygous mutation had been confirmed in the consanguineous family, this gene was then analyzed for mutation in 33 uncharacterized index cases of aplastic anemia (<13 years) using denaturing HPLC. Abnormal traces were confirmed by direct sequencing. Exome sequencing identified a novel homozygous nonsense mutation in the thrombopoietin receptor gene MPL. An additional novel homozygous MPL mutation was identified in the screen of 33 aplastic anemia patients. This study shows for the first time a link between homozygous MPL mutations and familial aplastic anemia. It also highlights the important role of MPL in trilineage hematopoiesis.


Asunto(s)
Anemia Aplásica/genética , Receptores de Trombopoyetina/genética , Preescolar , Consanguinidad , Exoma , Familia , Femenino , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Linaje , Análisis de Secuencia de ADN , Telómero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA