Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 105(2): 283-301, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31353023

RESUMEN

The RNA polymerase II complex (pol II) is responsible for transcription of all ∼21,000 human protein-encoding genes. Here, we describe sixteen individuals harboring de novo heterozygous variants in POLR2A, encoding RPB1, the largest subunit of pol II. An iterative approach combining structural evaluation and mass spectrometry analyses, the use of S. cerevisiae as a model system, and the assessment of cell viability in HeLa cells allowed us to classify eleven variants as probably disease-causing and four variants as possibly disease-causing. The significance of one variant remains unresolved. By quantification of phenotypic severity, we could distinguish mild and severe phenotypic consequences of the disease-causing variants. Missense variants expected to exert only mild structural effects led to a malfunctioning pol II enzyme, thereby inducing a dominant-negative effect on gene transcription. Intriguingly, individuals carrying these variants presented with a severe phenotype dominated by profound infantile-onset hypotonia and developmental delay. Conversely, individuals carrying variants expected to result in complete loss of function, thus reduced levels of functional pol II from the normal allele, exhibited the mildest phenotypes. We conclude that subtle variants that are central in functionally important domains of POLR2A cause a neurodevelopmental syndrome characterized by profound infantile-onset hypotonia and developmental delay through a dominant-negative effect on pol-II-mediated transcription of DNA.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/genética , Hipotonía Muscular/patología , Mutación , Trastornos del Neurodesarrollo/patología , Saccharomyces cerevisiae/crecimiento & desarrollo , Adolescente , Edad de Inicio , Niño , Preescolar , Femenino , Células HeLa , Heterocigoto , Humanos , Masculino , Hipotonía Muscular/enzimología , Hipotonía Muscular/genética , Trastornos del Neurodesarrollo/enzimología , Trastornos del Neurodesarrollo/genética , Fenotipo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
2.
Clin Genet ; 100(3): 292-297, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33993487

RESUMEN

The congenital imprinting disorder, Beckwith-Wiedemann syndrome (BWS) is associated with variable clinical features including hemihypertrophy/lateralised overgrowth (LO) and embryonal tumour predisposition. BWS-associated (epi)genetic alterations occur in a subset of patients with isolated LO (ILO), leading to the concept of BWS spectrum disorder (BWSp). We investigated the relationship between clinical features and molecular diagnostic results in a cohort with LO using the BWSp international consensus group (BWSICG) clinical scoring system. Clinical/molecular findings in 94 previously-unreported patients with LO referred for BWSp molecular studies were reviewed retrospectively. The BWSICG score was assigned and diagnostic rate calculated. BWSp-associated (epi)genetic alteration was identified in 15/94 (16%). The molecular diagnostic rate by MS-MLPA (blood DNA) for BWS-related molecular findings in patients with LO was positively correlated with the BWSICG score. 3/48 with ILO had a molecular alteration. No individuals with ILO had developed an embryonal tumour at last follow up. Among a cohort of individuals with LO referred for BWSp molecular testing, the BWSICG score correlated with diagnostic yield. The embryonal tumour risk in children with ILO and negative molecular testing appeared very low, however longer- and more complete follow up is required to better define tumour risks in this group.


Asunto(s)
Síndrome de Beckwith-Wiedemann/fisiopatología , Hipertrofia/fisiopatología , Adolescente , Adulto , Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Niño , Preescolar , Estudios de Cohortes , Femenino , Pruebas Genéticas , Humanos , Hipertrofia/diagnóstico , Hipertrofia/genética , Lactante , Recién Nacido , Masculino , Repeticiones de Microsatélite , Técnicas de Diagnóstico Molecular , Neoplasias de Células Germinales y Embrionarias/complicaciones , Neoplasias de Células Germinales y Embrionarias/diagnóstico , Neoplasias de Células Germinales y Embrionarias/genética , Estudios Retrospectivos , Adulto Joven
3.
Clin Genet ; 97(3): 396-406, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31794058

RESUMEN

The 2017 classification of Ehlers-Danlos syndromes (EDS) identifies three types associated with causative variants in COL1A1/COL1A2 and distinct from osteogenesis imperfecta (OI). Previously, patients have been described with variable features of both disorders, and causative variants in COL1A1/COL1A2; but this phenotype has not been included in the current classification. Here, we expand and re-define this OI/EDS overlap as a missing EDS type. Twenty-one individuals from 13 families were reported, in whom COL1A1/COL1A2 variants were found after a suspicion of EDS. None of them could be classified as affected by OI or by any of the three recognized EDS variants associated with COL1A1/COL1A2. This phenotype is dominated by EDS-related features. OI-related features were limited to mildly reduced bone mass, occasional fractures and short stature. Eight COL1A1/COL1A2 variants were novel and five recurrent with a predominance of glycine substitutions affecting residues within the procollagen N-proteinase cleavage site of α1(I) and α2(I) procollagens. Selected variants were investigated by biochemical, ultrastructural and immunofluorescence studies. The pattern of observed changes in the dermis and in vitro for selected variants was more typical of EDS rather than OI. Our findings indicate the existence of a wider recognizable spectrum associated with COL1A1/COL1A2.


Asunto(s)
Colágeno Tipo I/genética , Enfermedades del Tejido Conjuntivo/clasificación , Síndrome de Ehlers-Danlos/clasificación , Variación Genética , Osteogénesis Imperfecta/clasificación , Adolescente , Adulto , Niño , Preescolar , Colágeno Tipo I/ultraestructura , Cadena alfa 1 del Colágeno Tipo I , Tejido Conectivo/ultraestructura , Enfermedades del Tejido Conjuntivo/genética , Demografía , Síndrome de Ehlers-Danlos/genética , Femenino , Heterocigoto , Humanos , Lactante , Masculino , Persona de Mediana Edad , Osteogénesis Imperfecta/genética , Fenotipo , Adulto Joven
4.
Hum Mutat ; 39(9): 1246-1261, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29924900

RESUMEN

Adams-Oliver syndrome (AOS) is a rare developmental disorder, characterized by scalp aplasia cutis congenita (ACC) and transverse terminal limb defects (TTLD). Autosomal dominant forms of AOS are linked to mutations in ARHGAP31, DLL4, NOTCH1 or RBPJ, while DOCK6 and EOGT underlie autosomal recessive inheritance. Data on the frequency and distribution of mutations in large cohorts are currently limited. The purpose of this study was therefore to comprehensively examine the genetic architecture of AOS in an extensive cohort. Molecular diagnostic screening of 194 AOS/ACC/TTLD probands/families was conducted using next-generation and/or capillary sequencing analyses. In total, we identified 63 (likely) pathogenic mutations, comprising 56 distinct and 22 novel mutations, providing a molecular diagnosis in 30% of patients. Taken together with previous reports, these findings bring the total number of reported disease variants to 63, with a diagnostic yield of 36% in familial cases. NOTCH1 is the major contributor, underlying 10% of AOS/ACC/TTLD cases, with DLL4 (6%), DOCK6 (6%), ARHGAP31 (3%), EOGT (3%), and RBPJ (2%) representing additional causality in this cohort. We confirm the relevance of genetic screening across the AOS/ACC/TTLD spectrum, highlighting preliminary but important genotype-phenotype correlations. This cohort offers potential for further gene identification to address missing heritability.


Asunto(s)
Displasia Ectodérmica/genética , Deformidades Congénitas de las Extremidades/genética , Dermatosis del Cuero Cabelludo/congénito , Proteínas de Unión al GTP rho/genética , Displasia Ectodérmica/fisiopatología , Extremidades/fisiopatología , Femenino , Estudios de Asociación Genética , Humanos , Deformidades Congénitas de las Extremidades/fisiopatología , Masculino , Mutación , Linaje , Receptores Notch/genética , Cuero Cabelludo/fisiopatología , Dermatosis del Cuero Cabelludo/genética , Dermatosis del Cuero Cabelludo/fisiopatología
5.
Am J Med Genet A ; 170(12): 3069-3082, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27648933

RESUMEN

Rubinstein-Taybi syndrome (RSTS) is a developmental disorder characterized by a typical face and distal limbs abnormalities, intellectual disability, and a vast number of other features. Two genes are known to cause RSTS, CREBBP in 60% and EP300 in 8-10% of clinically diagnosed cases. Both paralogs act in chromatin remodeling and encode for transcriptional co-activators interacting with >400 proteins. Up to now 26 individuals with an EP300 mutation have been published. Here, we describe the phenotype and genotype of 42 unpublished RSTS patients carrying EP300 mutations and intragenic deletions and offer an update on another 10 patients. We compare the data to 308 individuals with CREBBP mutations. We demonstrate that EP300 mutations cause a phenotype that typically resembles the classical RSTS phenotype due to CREBBP mutations to a great extent, although most facial signs are less marked with the exception of a low-hanging columella. The limb anomalies are more similar to those in CREBBP mutated individuals except for angulation of thumbs and halluces which is very uncommon in EP300 mutated individuals. The intellectual disability is variable but typically less marked whereas the microcephaly is more common. All types of mutations occur but truncating mutations and small rearrangements are most common (86%). Missense mutations in the HAT domain are associated with a classical RSTS phenotype but otherwise no genotype-phenotype correlation is detected. Pre-eclampsia occurs in 12/52 mothers of EP300 mutated individuals versus in 2/59 mothers of CREBBP mutated individuals, making pregnancy with an EP300 mutated fetus the strongest known predictor for pre-eclampsia. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteína de Unión a CREB/genética , Proteína p300 Asociada a E1A/genética , Preeclampsia/genética , Síndrome de Rubinstein-Taybi/genética , Adulto , Ensamble y Desensamble de Cromatina/genética , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense/genética , Preeclampsia/fisiopatología , Embarazo , Síndrome de Rubinstein-Taybi/patología , Eliminación de Secuencia
6.
Am J Hum Genet ; 90(2): 290-4, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22265017

RESUMEN

Genitopatellar syndrome (GPS) is a rare disorder in which patellar aplasia or hypoplasia is associated with external genital anomalies and severe intellectual disability. Using an exome-sequencing approach, we identified de novo mutations of KAT6B in five individuals with GPS; a single nonsense variant and three frameshift indels, including a 4 bp deletion observed in two cases. All identified mutations are located within the terminal exon of the gene and are predicted to generate a truncated protein product lacking evolutionarily conserved domains. KAT6B encodes a member of the MYST family of histone acetyltranferases. We demonstrate a reduced level of both histone H3 and H4 acetylation in patient-derived cells suggesting that dysregulation of histone acetylation is a direct functional consequence of GPS alleles. These findings define the genetic basis of GPS and illustrate the complex role of the regulation of histone acetylation during development.


Asunto(s)
Histona Acetiltransferasas/genética , Anomalías Musculoesqueléticas/genética , Mutación , Anomalías Urogenitales/genética , Acetilación , Alelos , Animales , Exoma , Exones , Femenino , Histonas/metabolismo , Humanos , Discapacidad Intelectual/enzimología , Discapacidad Intelectual/genética , Masculino , Ratones , Anomalías Musculoesqueléticas/enzimología , Análisis de Secuencia de ADN/métodos , Anomalías Urogenitales/enzimología
7.
Am J Med Genet A ; 167A(4): 831-6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25736269

RESUMEN

Congenital diaphragmatic hernia (CDH) is a relatively common, life--threatening birth defect. We present a family with recurrent CDH--paraesophageal and central--for whom exome sequencing (ES) revealed a frameshift mutation (c.4969_4970insA, p.Ile1657Asnfs*30) in the fibrillin 1 gene (FBN1) that causes Marfan syndrome. A diagnosis of Marfan syndrome had not been considered previously in this family. However, a review of the literature demonstrated that FBN1 mutations have an unusual pattern of CDH in which paraesophageal hernias are particularly common. Subsequent clinical evaluations revealed evidence for ectopia lentis in affected family members supporting a clinical diagnosis of Marfan syndrome. Since only two other cases of familial CDH have been described in association with FBN1 mutations, we investigated an oligogenic hypothesis by examining ES data for deleterious sequence changes in other CDH-related genes. This search revealed putatively deleterious sequence changes in four other genes that have been shown to cause diaphragm defects in humans and/or mice--FREM1, DES, PAX3 and MET. It is unclear whether these changes, alone or in aggregate, are contributing to the development of CDH in this family. However, their individual contribution is likely to be small compared to that of the frameshift mutation in FBN1. We conclude that ES can be used to identify both major and minor genetic factors that may contribute to CDH. These results also suggest that ES should be considered in the diagnostic evaluation of individuals and families with CDH, particularly when other diagnostic modalities have failed to reveal a molecular etiology.


Asunto(s)
Hernias Diafragmáticas Congénitas/diagnóstico , Síndrome de Marfan/diagnóstico , Proteínas de Microfilamentos/genética , Adulto , Preescolar , Análisis Mutacional de ADN , Exoma , Femenino , Fibrilina-1 , Fibrilinas , Mutación del Sistema de Lectura , Estudios de Asociación Genética , Hernias Diafragmáticas Congénitas/genética , Humanos , Masculino , Síndrome de Marfan/genética , Linaje
8.
Am J Hum Genet ; 88(5): 574-85, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21565291

RESUMEN

Regulation of cell proliferation and motility is essential for normal development. The Rho family of GTPases plays a critical role in the control of cell polarity and migration by effecting the cytoskeleton, membrane trafficking, and cell adhesion. We investigated a recognized developmental disorder, Adams-Oliver syndrome (AOS), characterized by the combination of aplasia cutis congenita (ACC) and terminal transverse limb defects (TTLD). Through a genome-wide linkage analysis, we detected a locus for autosomal-dominant ACC-TTLD on 3q generating a maximum LOD score of 4.93 at marker rs1464311. Candidate-gene- and exome-based sequencing led to the identification of independent premature truncating mutations in the terminal exon of the Rho GTPase-activating protein 31 gene, ARHGAP31, which encodes a Cdc42/Rac1 regulatory protein. Mutant transcripts are stable and increase ARHGAP31 activity in vitro through a gain-of-function mechanism. Constitutively active ARHGAP31 mutations result in a loss of available active Cdc42 and consequently disrupt actin cytoskeletal structures. Arhgap31 expression in the mouse is substantially restricted to the terminal limb buds and craniofacial processes during early development; these locations closely mirror the sites of impaired organogenesis that characterize this syndrome. These data identify the requirement for regulated Cdc42 and/or Rac1 signaling processes during early human development.


Asunto(s)
Displasia Ectodérmica/genética , Proteínas Activadoras de GTPasa/genética , Mutación , Actinas/metabolismo , Adhesión Celular , Movimiento Celular , Polaridad Celular , Proliferación Celular , Mapeo Cromosómico , Citoesqueleto/metabolismo , Análisis Mutacional de ADN , Displasia Ectodérmica/embriología , Femenino , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Deformidades Congénitas de las Extremidades/embriología , Deformidades Congénitas de las Extremidades/genética , Masculino , Dermatosis del Cuero Cabelludo/congénito , Dermatosis del Cuero Cabelludo/embriología , Dermatosis del Cuero Cabelludo/genética , Transducción de Señal , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo
9.
Am J Med Genet A ; 164A(11): 2707-23, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25123976

RESUMEN

22q11.2 deletion syndrome is one of the most common microdeletion syndromes. Most patients have a deletion resulting from a recombination of low copy repeat blocks LCR22-A and LCR22-D. Loss of the TBX1 gene is considered the most important cause of the phenotype. A limited number of patients with smaller, overlapping deletions distal to the TBX1 locus have been described in the literature. In these patients, the CRKL gene is deleted. Haploinsufficiency of this gene has also been implicated in the pathogenesis of 22q11.2 deletion syndrome. To distinguish these deletions (comprising the LCR22-B to LCR22-D region) from the more distal 22q11.2 deletions (located beyond LCR22-D), we propose the term "central 22q11.2 deletions". In the present study we report on 27 new patients with such a deletion. Together with information on previously published cases, we review the clinical findings of 52 patients. The prevalence of congenital heart anomalies and the frequency of de novo deletions in patients with a central deletion are substantially lower than in patients with a common or distal 22q11.2 deletion. Renal and urinary tract malformations, developmental delays, cognitive impairments and behavioral problems seem to be equally frequent as in patients with a common deletion. None of the patients had a cleft palate. Patients with a deletion that also encompassed the MAPK1 gene, located just distal to LCR22-D, have a different and more severe phenotype, characterized by a higher prevalence of congenital heart anomalies, growth restriction and microcephaly. Our results further elucidate genotype-phenotype correlations in 22q11.2 deletion syndrome spectrum.


Asunto(s)
Síndrome de DiGeorge/diagnóstico , Síndrome de DiGeorge/genética , Adolescente , Adulto , Niño , Preescolar , Facies , Familia , Femenino , Orden Génico , Sitios Genéticos , Humanos , Masculino , Fenotipo , Diagnóstico Prenatal , Adulto Joven
10.
Am J Hum Genet ; 86(6): 963-9, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20560210

RESUMEN

Urinary voiding dysfunction in childhood, manifesting as incontinence, dysuria, and urinary frequency, is a common condition. Urofacial syndrome (UFS) is a rare autosomal recessive disease characterized by facial grimacing when attempting to smile and failure of the urinary bladder to void completely despite a lack of anatomical bladder outflow obstruction or overt neurological damage. UFS individuals often have reflux of infected urine from the bladder to the upper renal tract, with a risk of kidney damage and renal failure. Whole-genome SNP mapping in one affected individual defined an autozygous region of 16 Mb on chromosome 10q23-q24, within which a 10 kb deletion encompassing exons 8 and 9 of HPSE2 was identified. Homozygous exonic deletions, nonsense mutations, and frameshift mutations in five further unrelated families confirmed HPSE2 as the causative gene for UFS. Mutations were not identified in four additional UFS patients, indicating genetic heterogeneity. We show that HPSE2 is expressed in the fetal and adult central nervous system, where it might be implicated in controlling facial expression and urinary voiding, and also in bladder smooth muscle, consistent with a role in renal tract morphology and function. Our findings have broader implications for understanding the genetic basis of lower renal tract malformations and voiding dysfunction.


Asunto(s)
Facies , Glucuronidasa/genética , Enfermedades Urológicas/genética , Encéfalo/metabolismo , Niño , Preescolar , Mapeo Cromosómico , Cromosomas Humanos Par 10 , Femenino , Genes Recesivos , Glucuronidasa/química , Glucuronidasa/metabolismo , Humanos , Masculino , Modelos Moleculares , Músculos/metabolismo , Mutación , Linaje , Síndrome , Vejiga Urinaria/metabolismo
11.
Am J Med Genet A ; 158A(8): 1977-81, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22711382

RESUMEN

Hearing impairment is common in individuals with branchio-oculo-facial (BOF) syndrome. The majority of described individuals have conductive hearing impairment due to malformed ossicles and/or external canal stenosis or atresia, although a sensorineural component to the hearing impairment in BOF syndrome is increasingly being reported. Sophisticated computed tomography (CT) of the temporal bone has revealed middle and inner ear malformations in three previous reports. We present middle and inner ear abnormalities in three additional individuals with mutation-proven BOF syndrome. We suggest that temporal bone CT imaging be included in the medical workup of a child with BOF syndrome, in order to guide management.


Asunto(s)
Síndrome Branquio Oto Renal/patología , Oído Interno/patología , Oído Medio/patología , Mutación , Síndrome Branquio Oto Renal/genética , Niño , Femenino , Humanos , Masculino , Síndrome , Tomografía Computarizada por Rayos X
12.
Am J Med Genet A ; 158A(11): 2733-42, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23023959

RESUMEN

Meier-Gorlin syndrome (MGS) is a rare autosomal recessive disorder characterized by primordial dwarfism, microtia, and patellar aplasia/hypoplasia. Recently, mutations in the ORC1, ORC4, ORC6, CDT1, and CDC6 genes, encoding components of the pre-replication complex, have been identified. This complex is essential for DNA replication and therefore mutations are expected to impair cell proliferation and consequently could globally reduce growth. However, detailed growth characteristics of MGS patients have not been reported, and so this is addressed here through study of 45 MGS patients, the largest cohort worldwide. Here, we report that growth velocity (length) is impaired in MGS during pregnancy and first year of life, but, thereafter, height increases in paralleled normal reference centiles, resulting in a mean adult height of -4.5 standard deviations (SD). Height is dependent on ethnic background and underlying molecular cause, with ORC1 and ORC4 mutations causing more severe short stature and microcephaly. Growth hormone therapy (n = 9) was generally ineffective, though in two patients with significantly reduced IGF1 levels, growth was substantially improved by GH treatment, with 2SD and 3.8 SD improvement in height. Growth parameters for monitoring growth in future MGS patients are provided and as well we highlight that growth is disproportionately affected in certain structures, with growth related minor genital abnormalities (42%) and mammary hypoplasia (100%) frequently present, in addition to established effects on ears and patellar growth.


Asunto(s)
Gráficos de Crecimiento , Trastornos del Crecimiento/diagnóstico , Micrognatismo/diagnóstico , Desarrollo Sexual , Proteínas de Ciclo Celular/genética , Preescolar , Estudios de Cohortes , Microtia Congénita , Oído/anomalías , Femenino , Trastornos del Crecimiento/tratamiento farmacológico , Trastornos del Crecimiento/genética , Hormona de Crecimiento Humana/sangre , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Lactante , Masculino , Micrognatismo/tratamiento farmacológico , Micrognatismo/genética , Mutación , Complejo de Reconocimiento del Origen/genética , Rótula/anomalías , Desarrollo Sexual/genética , Anomalías Urogenitales
13.
Am J Med Genet A ; 155A(9): 2203-11, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21834056

RESUMEN

Pierpont syndrome is a multiple congenital anomaly syndrome with learning disability first described in 1998. There are only three patients with Pierpont syndrome who have previously been published in the literature. Details of a series of patients with features of this condition were therefore obtained retrospectively to better characterize its key features. These patients were noted to have distinctive shared facial characteristics, in addition to plantar fat pads and other limb abnormalities. Further individuals with equally striking hand and foot findings were identified whose facies were less characteristic, and hence we considered them unlikely to be affected with the same condition. Despite several patients with possible Pierpont syndrome having had high-resolution array CGH or SNP array, the etiology of this phenotype remains unknown. Whilst it is as yet unclear whether it is a single entity, there appears to be a group of patients in whom Pierpont syndrome may be a recognizable condition, with typical facies, particularly when smiling, and characteristic hand and foot findings.


Asunto(s)
Anomalías Múltiples/genética , Discapacidades para el Aprendizaje/genética , Adulto , Niño , Preescolar , Anomalías Craneofaciales/genética , Discapacidades del Desarrollo , Cara/anomalías , Facies , Femenino , Deformidades Congénitas del Pie/genética , Deformidades Congénitas de la Mano/genética , Humanos , Cariotipo , Masculino , Persona de Mediana Edad , Fenotipo , Estudios Retrospectivos , Síndrome
14.
Am J Med Genet A ; 155A(1): 22-32, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21204207

RESUMEN

Branchio-oculo-facial syndrome (BOFS; OMIM#113620) is a rare autosomal dominant craniofacial disorder with variable expression. Major features include cutaneous and ocular abnormalities, characteristic facies, renal, ectodermal, and temporal bone anomalies. Having determined that mutations involving TFAP2A result in BOFS, we studied a total of 30 families (41 affected individuals); 26/30 (87%) fulfilled our cardinal diagnostic criteria. The original family with the 3.2 Mb deletion including the TFAP2A gene remains the only BOFS family without the typical CL/P and the only family with a deletion. We have identified a hotspot region in the highly conserved exons 4 and 5 of TFAP2A that harbors missense mutations in 27/30 (90%) families. Several of these mutations are recurrent. Mosaicism was detected in one family. To date, genetic heterogeneity has not been observed. Although the cardinal criteria for BOFS have been based on the presence of each of the core defects, an affected family member or thymic remnant, we documented TFAP2A mutations in three (10%) probands in our series without a classic cervical cutaneous defect or ectopic thymus. Temporal bone anomalies were identified in 3/5 patients investigated. The occurrence of CL/P, premature graying, coloboma, heterochromia irides, and ectopic thymus, are evidence for BOFS as a neurocristopathy. Intrafamilial clinical variability can be marked. Although there does not appear to be mutation-specific genotype-phenotype correlations at this time, more patients need to be studied. Clinical testing for TFAP2A mutations is now available and will assist geneticists in confirming the typical cases or excluding the diagnosis in atypical cases.


Asunto(s)
Síndrome Branquio Oto Renal/genética , Síndrome Branquio Oto Renal/patología , Cromosomas Humanos Par 6/genética , Fenotipo , Factor de Transcripción AP-2/genética , Secuencia de Aminoácidos , Secuencia de Bases , Deleción Cromosómica , Genotipo , Humanos , Datos de Secuencia Molecular , Mutación Missense/genética , Análisis de Secuencia de ADN
15.
Eur J Hum Genet ; 29(1): 141-153, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32860008

RESUMEN

Despite clear technical superiority of genome sequencing (GS) over other diagnostic methods such as exome sequencing (ES), few studies are available regarding the advantages of its clinical application. We analyzed 1007 consecutive index cases for whom GS was performed in a diagnostic setting over a 2-year period. We reported pathogenic and likely pathogenic (P/LP) variants that explain the patients' phenotype in 212 of the 1007 cases (21.1%). In 245 additional cases (24.3%), a variant of unknown significance (VUS) related to the phenotype was reported. We especially investigated patients which had had ES with no genetic diagnosis (n = 358). For this group, GS diagnostic yield was 14.5% (52 patients with P/LP out of 358). GS should be especially indicated for ES-negative cases since up to 29.6% of them  could benefit from GS testing (14.5% with P/LP, n = 52 and 15.1% with VUS, n = 54). Genetic diagnoses in most of the ES-negative/GS-positive cases were determined by technical superiority of GS, i.e., access to noncoding regions and more uniform coverage. Importantly, we reported 79 noncoding variants, of which, 41 variants were classified as P/LP. Interpretation of noncoding variants remains challenging, and in many cases, complementary methods based on direct enzyme assessment, biomarker testing and RNA analysis are needed for variant classification and diagnosis. We present the largest cohort of patients with GS performed in a clinical setting to date. The results of this study should direct the decision for GS as standard second-line, or even first-line stand-alone test.


Asunto(s)
Secuenciación del Exoma/normas , Enfermedades Genéticas Congénitas/diagnóstico , Pruebas Genéticas/normas , Adolescente , Niño , Preescolar , Femenino , Frecuencia de los Genes , Enfermedades Genéticas Congénitas/epidemiología , Enfermedades Genéticas Congénitas/genética , Pruebas Genéticas/estadística & datos numéricos , Humanos , Lactante , Recién Nacido , Masculino , Diagnóstico Prenatal/normas , Diagnóstico Prenatal/estadística & datos numéricos , Sensibilidad y Especificidad , Secuenciación del Exoma/estadística & datos numéricos
16.
Am J Med Genet A ; 152A(2): 333-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20101697

RESUMEN

Juvenile polyposis (JP) and hereditary hemorrhagic telangiectasia (HHT) are clinically distinct diseases caused by mutations in SMAD4 and BMPR1A (for JP) and endoglin and ALK1 (for HHT). Recently, a combined syndrome of JP-HHT was described that is also caused by mutations in SMAD4. Although both JP and JP-HHT are caused by SMAD4 mutations, a possible genotype:phenotype correlation was noted as all of the SMAD4 mutations in the JP-HHT patients were clustered in the COOH-terminal MH2 domain of the protein. If valid, this correlation would provide a molecular explanation for the phenotypic differences, as well as a pre-symptomatic diagnostic test to distinguish patients at risk for the overlapping but different clinical features of the disorders. In this study, we collected 19 new JP-HHT patients from which we identified 15 additional SMAD4 mutations. We also reviewed the literature for other reports of JP patients with HHT symptoms with confirmed SMAD4 mutations. Our combined results show that although the SMAD4 mutations in JP-HHT patients do show a tendency to cluster in the MH2 domain, mutations in other parts of the gene also cause the combined syndrome. Thus, any mutation in SMAD4 can cause JP-HHT. Any JP patient with a SMAD4 mutation is, therefore, at risk for the visceral manifestations of HHT and any HHT patient with SMAD4 mutation is at risk for early onset gastrointestinal cancer. In conclusion, a patient who tests positive for any SMAD4 mutation must be considered at risk for the combined syndrome of JP-HHT and monitored accordingly.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Mutación , Proteína Smad4/genética , Telangiectasia Hemorrágica Hereditaria/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Neoplasias Gastrointestinales/diagnóstico , Neoplasias Gastrointestinales/genética , Humanos , Lactante , Persona de Mediana Edad , Estructura Terciaria de Proteína , Síndrome
17.
Eur J Hum Genet ; 28(3): 367-372, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31822863

RESUMEN

Intellectual disability (ID) is one of most frequent reasons for genetic consultation. The complex molecular anatomy of ID ranges from complete chromosomal imbalances to single nucleotide variant changes occurring de novo, with thousands of genes identified. This extreme genetic heterogeneity challenges the molecular diagnosis, which mostly requires a genomic approach. CXorf56 is largely uncharacterized and was recently proposed as a candidate ID gene based on findings in a single Dutch family. Here, we describe nine cases (six males and three females) from three unrelated families. Exome sequencing and combined database analyses, identified family-specific CXorf56 variants (NM_022101.3:c.498_503del, p.(Glu167_Glu168del) and c.303_304delCTinsACCC, p.(Phe101Leufs*20)) that segregated with the ID phenotype. These variants are presumably leading to loss-of-function, which is the proposed disease mechanism. Clinically, CXorf56-related disease is a slowly progressive neurological disorder. The phenotype is more severe in hemizygote males, but might also manifests in heterozygote females, which showed skewed X-inactivation patterns in blood. Male patients might present previously unreported neurological features such as epilepsy, abnormal gait, tremor, and clonus, which extends the clinical spectrum of the disorder. In conclusion, we confirm the causative role of variants in CXorf56 for an X-linked form of intellectual disability with additional neurological features. The gene should be considered for molecular diagnostics of patients with ID, specifically when family history is suggestive of X-linked inheritance. Further work is needed to understand the role of this gene in neurodevelopment and intellectual disability.


Asunto(s)
Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Adulto , Discapacidades del Desarrollo/patología , Femenino , Hemicigoto , Humanos , Discapacidad Intelectual/patología , Mutación con Pérdida de Función , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Inactivación del Cromosoma X
18.
Clin Dysmorphol ; 16(3): 203-206, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17551339

RESUMEN

CFC syndrome is a genetically heterogenous condition. Missense mutations have been identified in BRAF, KRAS, MEK1 and MEK2. We have reported here a KRAS mutation in a baby girl with an early clinical diagnosis of CFC syndrome associated with a large ulcerating hemangioma. Although ectodermal abnormalities have been described in all individuals with this condition, features such as ichthyosis and hemangioma have been previously found only in those patients carrying a mutation in BRAF, and not in KRAS. The findings we have described contrast with these observations. The relatively high frequency of hemangiomas in CFC syndrome suggests that defects in the expression of the MAPK pathway may alter endothelial cell proliferation. Increased understanding of how the molecular pathways with which defects in CFC syndrome predispose affected individuals to hemangiomas might offer insights into the pathogenesis of this common childhood tumour in the general population.


Asunto(s)
Anomalías Múltiples/genética , Hemangioma/congénito , Hemangioma/complicaciones , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Secuencia de Bases , Análisis Mutacional de ADN , Femenino , Humanos , Recién Nacido , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas p21(ras) , Síndrome
19.
Eur J Hum Genet ; 23(9): 1165-70, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25424711

RESUMEN

KAT6B sequence variants have been identified previously in both patients with the Say-Barber-Biesecker type of blepharophimosis mental retardation syndromes (SBBS) and in the more severe genitopatellar syndrome (GPS). We report on the findings in a previously unreported group of 57 individuals with suggestive features of SBBS or GPS. Likely causative variants have been identified in 34/57 patients and were commonly located in the terminal exons of KAT6B. Of those where parental samples could be tested, all occurred de novo. Thirty out of thirty-four had truncating variants, one had a missense variant and the remaining three had the same synonymous change predicted to affect splicing. Variants in GPS tended to occur more proximally to those in SBBS patients, and genotype/phenotype analysis demonstrated significant clinical overlap between SBBS and GPS. The de novo synonymous change seen in three patients with features of SBBS occurred more proximally in exon 16. Statistical analysis of clinical features demonstrated that KAT6B variant-positive patients were more likely to display hypotonia, feeding difficulties, long thumbs/great toes and dental, thyroid and patella abnormalities than KAT6B variant-negative patients. The few reported patients with KAT6B haploinsufficiency had a much milder phenotype, though with some features overlapping those of SBBS. We report the findings in a previously unreported patient with a deletion of the KAT6B gene to further delineate the haploinsufficiency phenotype. The molecular mechanisms giving rise to the SBBS and GPS phenotypes are discussed.


Asunto(s)
Blefarofimosis/genética , Hipotiroidismo Congénito/genética , Anomalías Craneofaciales/genética , Exones , Cardiopatías Congénitas/genética , Histona Acetiltransferasas/genética , Discapacidad Intelectual/genética , Inestabilidad de la Articulación/genética , Riñón/anomalías , Mutación , Rótula/anomalías , Trastornos Psicomotores/genética , Escroto/anomalías , Anomalías Urogenitales/genética , Blefarofimosis/diagnóstico , Blefarofimosis/patología , Preescolar , Hipotiroidismo Congénito/diagnóstico , Hipotiroidismo Congénito/patología , Anomalías Craneofaciales/diagnóstico , Anomalías Craneofaciales/patología , Análisis Mutacional de ADN , Diagnóstico Diferencial , Exoma , Facies , Femenino , Expresión Génica , Estudios de Asociación Genética , Genotipo , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/patología , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Inestabilidad de la Articulación/diagnóstico , Inestabilidad de la Articulación/patología , Riñón/patología , Masculino , Rótula/patología , Fenotipo , Trastornos Psicomotores/diagnóstico , Trastornos Psicomotores/patología , Escroto/patología , Índice de Severidad de la Enfermedad , Anomalías Urogenitales/diagnóstico , Anomalías Urogenitales/patología
20.
Orphanet J Rare Dis ; 8: 110, 2013 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-23879989

RESUMEN

BACKGROUND: Mutations in EFTUD2 were proven to cause a very distinct mandibulofacial dysostosis type Guion-Almeida (MFDGA, OMIM #610536). Recently, gross deletions and mutations in EFTUD2 were determined to cause syndromic esophageal atresia (EA), as well. We set forth to find further conditions caused by mutations in the EFTUD2 gene (OMIM *603892). METHODS AND RESULTS: We performed exome sequencing in two familial cases with clinical features overlapping with MFDGA and EA, but which were previously assumed to represent distinct entities, a syndrome with esophageal atresia, hypoplasia of zygomatic complex, microcephaly, cup-shaped ears, congenital heart defect, and intellectual disability in a mother and her two children [AJMG 143A(11):1135-1142, 2007] and a supposedly autosomal recessive oto-facial syndrome with midline malformations in two sisters [AJMG 132(4):398-401, 2005]. While the analysis of our exome data was in progress, a recent publication made EFTUD2 mutations highly likely in these families. This hypothesis could be confirmed with exome as well as with Sanger sequencing. Also, in three further sporadic patients, clinically overlapping to these two families, de novo mutations within EFTUD2 were identified by Sanger sequencing. Our clinical and molecular workup of the patients discloses a broad phenotypic spectrum, and describes for the first time an instance of germline mosaicism for an EFTUD2 mutation. CONCLUSIONS: The clinical features of the eight patients described here further broaden the phenotypic spectrum caused by EFTUD2 mutations or deletions. We here show, that it not only includes mandibulofacial dysostosis type Guion-Almeida, which should be reclassified as an acrofacial dysostosis because of thumb anomalies (present in 12/35 or 34% of patients) and syndromic esophageal atresia [JMG 49(12). 737-746, 2012], but also the two new syndromes, namely oto-facial syndrome with midline malformations published by Mégarbané et al. [AJMG 132(4): 398-401, 2005] and the syndrome published by Wieczorek et al. [AJMG 143A(11): 1135-1142, 2007] The finding of mild phenotypic features in the mother of one family that could have been overlooked and the possibility of germline mosaicism in apparently healthy parents in the other family should be taken into account when counseling such families.


Asunto(s)
Atresia Esofágica/patología , Estudios de Asociación Genética , Discapacidad Intelectual/patología , Disostosis Mandibulofacial/patología , Mutación , Factores de Elongación de Péptidos/genética , Adolescente , Adulto , Niño , Preescolar , Atresia Esofágica/genética , Femenino , Humanos , Discapacidad Intelectual/genética , Masculino , Disostosis Mandibulofacial/genética , Fenotipo , Ribonucleoproteína Nuclear Pequeña U5 , Análisis de Secuencia de ADN , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA