Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
PLoS Genet ; 18(12): e1010504, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36480544

RESUMEN

Ollier disease (OD) and Maffucci Syndrome (MS) are rare disorders characterized by multiple enchondromas, commonly causing bone deformities, limb length discrepancies, and pathological fractures. MS is distinguished from OD by the development of vascular anomalies. Both disorders are cancer predisposition syndromes with malignancies developing in ~50% of the individuals with OD or MS. Somatic gain-of-function variants in IDH1 and IDH2 have been described in the enchondromas, vascular anomalies and chondrosarcomas of approximately 80% of the individuals with OD and MS. To date, however, no investigation of germline causative variants for these diseases has been comprehensively performed. To search for germline causative variants, we performed whole exome sequencing or whole genome sequencing of blood or saliva DNA in 94 unrelated probands (68 trios). We found that 7 had rare germline missense variants in HIF1A, 6 had rare germline missense variants in VHL, and 3 had IDH1 variants including 2 with mosaic IDH1-p.Arg132His variant. A burden analysis using 94 probands assigned as cases and 2,054 unrelated individuals presenting no OD- or MS-related features as controls, found that variants in HIF1A, VHL, and IDH1 were all significantly enriched in cases compared to controls. To further investigate the role of HIF-1 pathway in the pathogenesis of OD and MS, we performed RNA sequencing of fibroblasts from 4 probands with OD or MS at normoxia and at hypoxia. When cultured in hypoxic conditions, both proband and control cells showed altered expression of a subset of HIF-1 regulated genes. However, the set of differentially expressed genes in proband fibroblasts included a significantly reduced number of HIF-1 regulated genes compared to controls. Our findings suggest that germline or early post-zygotic variants identified in HIF1A, VHL, and IDH1 in probands with OD and MS underlie the development of the phenotypic abnormalities in a subset of individuals with OD and MS, but extensive functional studies are needed to further confirm it.


Asunto(s)
Neoplasias Óseas , Condrosarcoma , Encondromatosis , Enfermedades Vasculares , Humanos , Encondromatosis/complicaciones , Encondromatosis/genética , Encondromatosis/patología , Condrosarcoma/patología , Análisis de Secuencia de ADN , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética
3.
Mol Genet Genomic Med ; 9(4): e1655, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33719213

RESUMEN

BACKGROUND: Orofacial clefts (OFCs) are congenital malformations of the face and palate, with an incidence of 1 per 700 live births. Clubfoot or congenital talipes equinovarus (CTEV) is a three-dimensional abnormality of the leg, ankle, and feet that leads to the anomalous positioning of foot and ankle joints and has an incidence of 1 per 1000 live births. OFCs and CTEV may occur together or separately in certain genetic syndromes in addition to other congenital abnormalities. Here, we sought to decipher the genetic etiology of OFC and CTEV that occurred together in six probands. METHODS: At the time of recruitment, the most clinically obvious congenital anomalies in these individuals were the OFC and CTEV. We carried out whole-exome sequencing (WES) on DNA samples from probands and available parents employing the Agilent SureSelect XT kit and Illumina HiSeq2500 platform, followed by bioinformatics analyses. WES variants were validated by clinical Sanger Sequencing. RESULTS: Of the six probands, we observed probable pathogenic genetic variants in four. In three probands with probable pathogenic genetic variants, each individual had variants in three different genes, whereas one proband had probable pathogenic variant in just one gene. In one proband, we observed variants in DIS3L2, a gene associated with Perlman syndrome. A second proband had variants in EPG5 (associated with Vici Syndrome), BARX1 and MKI67, while another proband had potentially etiologic variants in FRAS1 (associated with Fraser Syndrome 1), TCOF1 (associated with Treacher Collins Syndrome 1) and MKI67. The last proband had variants in FRAS1, PRDM16 (associated with Cardiomyopathy, dilated, 1LL/Left ventricular noncompaction 8) and CHD7 (associated with CHARGE syndrome/Hypogonadotropic hypogonadism 5 with or without anosmia). CONCLUSION: Our results suggest that clubfoot and OFCs are two congenital abnormalities that can co-occur in certain individuals with varying genetic causes and expressivity, warranting the need for deep phenotyping.


Asunto(s)
Labio Leporino/genética , Fisura del Paladar/genética , Pie Equinovaro/genética , Heterogeneidad Genética , Adulto , África del Sur del Sahara , Proteínas Relacionadas con la Autofagia/genética , Preescolar , Labio Leporino/patología , Fisura del Paladar/patología , Pie Equinovaro/patología , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Proteínas de la Matriz Extracelular/genética , Femenino , Proteínas de Homeodominio/genética , Humanos , Lactante , Recién Nacido , Antígeno Ki-67/genética , Masculino , Síndrome , Factores de Transcripción/genética , Proteínas de Transporte Vesicular/genética , Secuenciación Completa del Genoma
4.
Hum Mol Genet ; 29(22): 3662-3678, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33276377

RESUMEN

The genetic causes of multiple congenital anomalies are incompletely understood. Here, we report novel heterozygous predicted loss-of-function (LoF) and predicted damaging missense variants in the WW domain binding protein 11 (WBP11) gene in seven unrelated families with a variety of overlapping congenital malformations, including cardiac, vertebral, tracheo-esophageal, renal and limb defects. WBP11 encodes a component of the spliceosome with the ability to activate pre-messenger RNA splicing. We generated a Wbp11 null allele in mouse using CRISPR-Cas9 targeting. Wbp11 homozygous null embryos die prior to E8.5, indicating that Wbp11 is essential for development. Fewer Wbp11 heterozygous null mice are found than expected due to embryonic and postnatal death. Importantly, Wbp11 heterozygous null mice are small and exhibit defects in axial skeleton, kidneys and esophagus, similar to the affected individuals, supporting the role of WBP11 haploinsufficiency in the development of congenital malformations in humans. LoF WBP11 variants should be considered as a possible cause of VACTERL association as well as isolated Klippel-Feil syndrome, renal agenesis or esophageal atresia.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Unión al ADN/genética , Haploinsuficiencia/genética , Riñón/metabolismo , Factores de Empalme de ARN/genética , Anomalías Múltiples/patología , Canal Anal/anomalías , Canal Anal/patología , Animales , Esófago/anomalías , Esófago/metabolismo , Esófago/patología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Heterocigoto , Humanos , Riñón/anomalías , Riñón/patología , Deformidades Congénitas de las Extremidades/genética , Deformidades Congénitas de las Extremidades/patología , Mutación con Pérdida de Función/genética , Ratones , Empalme del ARN/genética , Columna Vertebral/anomalías , Columna Vertebral/patología , Tráquea/anomalías , Tráquea/patología
5.
Am J Med Genet A ; 182(7): 1796-1800, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32420688

RESUMEN

Variants in MBTPS1 (membrane-bound transcription factor peptidase, site 1) encoding the protein convertase site-1 protease (S1P) were recently reported in a single individual with skeletal dysplasia and elevated plasma lysosomal enzymes. Here, we report the second individual with this newly described autosomal recessive spondyloepiphyseal dysplasia (OMIM #618392), presenting severe growth retardation, cataract and dysmorphic features, mainly retromicrognathia. Epilepsy and craniosynostosis were novel findings in our proband. She was found to be homozygous for a novel nonsense variant p.Trp983Ter in MBTPS1. In addition, she had normal levels of lysosomal enzyme activity in leukocytes but elevated levels in plasma. Our description confirms the existence of this new skeletal dysplasia and expands the phenotype and genotype of the disease.


Asunto(s)
Catarata/genética , Enfermedades por Almacenamiento Lisosomal/genética , Lisosomas/genética , Proproteína Convertasas/genética , Serina Endopeptidasas/genética , Catarata/diagnóstico por imagen , Catarata/patología , Preescolar , Femenino , Humanos , Enfermedades por Almacenamiento Lisosomal/sangre , Enfermedades por Almacenamiento Lisosomal/diagnóstico por imagen , Enfermedades por Almacenamiento Lisosomal/patología , Lisosomas/enzimología , Linaje , Fenotipo
6.
Am J Hum Genet ; 105(1): 151-165, 2019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31230722

RESUMEN

Genomic technologies such as next-generation sequencing (NGS) are revolutionizing molecular diagnostics and clinical medicine. However, these approaches have proven inefficient at identifying pathogenic repeat expansions. Here, we apply a collection of bioinformatics tools that can be utilized to identify either known or novel expanded repeat sequences in NGS data. We performed genetic studies of a cohort of 35 individuals from 22 families with a clinical diagnosis of cerebellar ataxia with neuropathy and bilateral vestibular areflexia syndrome (CANVAS). Analysis of whole-genome sequence (WGS) data with five independent algorithms identified a recessively inherited intronic repeat expansion [(AAGGG)exp] in the gene encoding Replication Factor C1 (RFC1). This motif, not reported in the reference sequence, localized to an Alu element and replaced the reference (AAAAG)11 short tandem repeat. Genetic analyses confirmed the pathogenic expansion in 18 of 22 CANVAS-affected families and identified a core ancestral haplotype, estimated to have arisen in Europe more than twenty-five thousand years ago. WGS of the four RFC1-negative CANVAS-affected families identified plausible variants in three, with genomic re-diagnosis of SCA3, spastic ataxia of the Charlevoix-Saguenay type, and SCA45. This study identified the genetic basis of CANVAS and demonstrated that these improved bioinformatics tools increase the diagnostic utility of WGS to determine the genetic basis of a heterogeneous group of clinically overlapping neurogenetic disorders.


Asunto(s)
Ataxia Cerebelosa/etiología , Biología Computacional/métodos , Intrones , Repeticiones de Microsatélite , Polineuropatías/etiología , Proteína de Replicación C/genética , Trastornos de la Sensación/etiología , Enfermedades Vestibulares/etiología , Algoritmos , Ataxia Cerebelosa/patología , Estudios de Cohortes , Familia , Femenino , Genómica , Humanos , Masculino , Persona de Mediana Edad , Polineuropatías/patología , Trastornos de la Sensación/patología , Síndrome , Enfermedades Vestibulares/patología , Secuenciación Completa del Genoma
7.
Eur J Med Genet ; 61(3): 134-138, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29169895

RESUMEN

Gillespie syndrome (GS) [MIM: 206700] is a very rare condition characterized by bilateral iris defect, congenital hypotonia, cerebellar ataxia and intellectual disability. The typical iris anomaly is considered necessary to the diagnosis of GS. Recently, variants in ITPR1 were described causing GS. Non-neurological features were reported in few patients. Here we describe two consanguineous siblings with GS and a novel homozygous ITPR1 pathogenic variant (p.N984fs). They also present a cardiac defect (pulmonary valve stenosis) and one sib had a genitourinary malformation (ureteropelvic junction obstruction). Our report reinforces ITPR1 as the cause of GS and suggests a possible role of ITPR1 in the development of other organs.


Asunto(s)
Aniridia/genética , Aniridia/patología , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/patología , Homocigoto , Receptores de Inositol 1,4,5-Trifosfato/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Mutación , Brasil , Preescolar , Consanguinidad , Femenino , Humanos , Lactante , Masculino , Linaje , Hermanos
8.
Curr Protoc Hum Genet ; 95: 9.31.1-9.31.15, 2017 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-29044468

RESUMEN

In well over half of the individuals with rare disease who undergo clinical or research next-generation sequencing, the responsible gene cannot be determined. Some reasons for this relatively low yield include unappreciated phenotypic heterogeneity; locus heterogeneity; somatic and germline mosaicism; variants of uncertain functional significance; technically inaccessible areas of the genome; incorrect mode of inheritance investigated; and inadequate communication between clinicians and basic scientists with knowledge of particular genes, proteins, or biological systems. To facilitate such communication and improve the search for patients or model organisms with similar phenotypes and variants in specific candidate genes, we have developed the Matchmaker Exchange (MME). MME was created to establish a federated network connecting databases of genomic and phenotypic data using a common application programming interface (API). To date, seven databases can exchange data using the API (GeneMatcher, PhenomeCentral, DECIPHER, MyGene2, matchbox, Australian Genomics Health Alliance Patient Archive, and Monarch Initiative; the latter included for model organism matching). This article guides usage of the MME for rare disease gene discovery. © 2017 by John Wiley & Sons, Inc.


Asunto(s)
Bases de Datos Genéticas , Estudios de Asociación Genética/métodos , Predisposición Genética a la Enfermedad , Enfermedades Raras/genética , Animales , Biología Computacional/métodos , Genómica/métodos , Humanos , Programas Informáticos , Navegador Web
9.
Am J Med Genet A ; 173(10): 2763-2771, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28777491

RESUMEN

Horstick et al. (2013) previously reported a homozygous p.Trp284Ser variant in STAC3 as the cause of Native American myopathy (NAM) in 5 Lumbee Native American families with congenital hypotonia and weakness, cleft palate, short stature, ptosis, kyphoscoliosis, talipes deformities, and susceptibility to malignant hyperthermia (MH). Here we present two non-Native American families, who were found to have STAC3 pathogenic variants. The first proband and her affected older sister are from a consanguineous Qatari family with a suspected clinical diagnosis of Carey-Fineman-Ziter syndrome (CFZS) based on features of hypotonia, myopathic facies with generalized weakness, ptosis, normal extraocular movements, cleft palate, growth delay, and kyphoscoliosis. We identified the homozygous c.851G>C;p.Trp284Ser variant in STAC3 in both sisters. The second proband and his affected sister are from a non-consanguineous, Puerto Rican family who was evaluated for a possible diagnosis of Moebius syndrome (MBS). His features included facial and generalized weakness, minimal limitation of horizontal gaze, cleft palate, and hypotonia, and he has a history of MH. The siblings were identified to be compound heterozygous for STAC3 variants c.851G>C;p.Trp284Ser and c.763_766delCTCT;p.Leu255IlefsX58. Given the phenotypic overlap of individuals with CFZS, MBS, and NAM, we screened STAC3 in 12 individuals diagnosed with CFZS and in 50 individuals diagnosed with MBS or a congenital facial weakness disorder. We did not identify any rare coding variants in STAC3. NAM should be considered in patients presenting with facial and generalized weakness, normal or mildly abnormal extraocular movement, hypotonia, cleft palate, and scoliosis, particularly if there is a history of MH.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Síndrome de Mobius/genética , Enfermedades Musculares/genética , Mutación , Síndrome de Pierre Robin/genética , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Síndrome de Mobius/complicaciones , Síndrome de Mobius/patología , Enfermedades Musculares/complicaciones , Enfermedades Musculares/patología , Linaje , Síndrome de Pierre Robin/complicaciones , Síndrome de Pierre Robin/patología , Pronóstico , Adulto Joven
10.
Am J Med Genet A ; 164A(5): 1170-4, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24478002

RESUMEN

Van den Ende-Gupta Syndrome (VDEGS) is an autosomal recessive disorder characterized by blepharophimosis, distinctive nose, hypoplastic maxilla, and skeletal abnormalities. Using homozygosity mapping in four VDEGS patients from three consanguineous families, Anastacio et al. [Anastacio et al. (2010); Am J Hum Genet 87:553-559] identified homozygous mutations in SCARF2, located at 22q11.2. Bedeschi et al. [2010] described a VDEGS patient with sclerocornea and cataracts with compound heterozygosity for the common 22q11.2 microdeletion and a hemizygous SCARF2 mutation. Because sclerocornea had been described in DiGeorge-velo-cardio-facial syndrome but not in VDEGS, they suggested that the ocular abnormalities were caused by the 22q11.2 microdeletion. We report on a 23-year-old male who presented with bilateral sclerocornea and the VDGEGS phenotype who was subsequently found to be homozygous for a 17 bp deletion in exon 4 of SCARF2. The occurrence of bilateral sclerocornea in our patient together with that of Bedeschi et al., suggests that the full VDEGS phenotype may include sclerocornea resulting from homozygosity or compound heterozygosity for loss of function variants in SCARF2.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Aracnodactilia/diagnóstico , Aracnodactilia/genética , Blefarofimosis/diagnóstico , Blefarofimosis/genética , Contractura/diagnóstico , Contractura/genética , Córnea/anomalías , Enfermedades de la Córnea/diagnóstico , Enfermedades de la Córnea/genética , Homocigoto , Receptores Depuradores de Clase F/genética , Eliminación de Secuencia , Adulto , Huesos/diagnóstico por imagen , Huesos/patología , Cromosomas Humanos Par 22 , Exones , Facies , Deformidades Congénitas de la Mano , Humanos , Masculino , Fenotipo , Radiografía , Análisis de Secuencia de ADN , Adulto Joven
11.
Genome Res ; 21(10): 1720-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21890680

RESUMEN

Translocations are a common class of chromosomal aberrations and can cause disease by physically disrupting genes or altering their regulatory environment. Some translocations, apparently balanced at the microscopic level, include deletions, duplications, insertions, or inversions at the molecular level. Traditionally, chromosomal rearrangements have been investigated with a conventional banded karyotype followed by arduous positional cloning projects. More recently, molecular cytogenetic approaches using fluorescence in situ hybridization (FISH), array comparative genomic hybridization (aCGH), or whole-genome SNP genotyping together with molecular methods such as inverse PCR and quantitative PCR have allowed more precise evaluation of the breakpoints. These methods suffer, however, from being experimentally intensive and time-consuming and of less than single base pair resolution. Here we describe targeted breakpoint capture followed by next-generation sequencing (TBCS) as a new approach to the general problem of determining the precise structural characterization of translocation breakpoints and related chromosomal aberrations. We tested this approach in three patients with complex chromosomal translocations: The first had craniofacial abnormalities and an apparently balanced t(2;3)(p15;q12) translocation; the second has cleidocranial dysplasia (OMIM 119600) associated with a t(2;6)(q22;p12.3) translocation and a breakpoint in RUNX2 on chromosome 6p; and the third has acampomelic campomelic dysplasia (OMIM 114290) associated with a t(5;17)(q23.2;q24) translocation, with a breakpoint upstream of SOX9 on chromosome 17q. Preliminary studies indicated complex rearrangements in patients 1 and 3 with a total of 10 predicted breakpoints in the three patients. By using TBCS, we quickly and precisely defined eight of the 10 breakpoints.


Asunto(s)
Cromosomas Humanos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Translocación Genética , Adulto , Secuencia de Bases , Displasia Campomélica/genética , Puntos de Rotura del Cromosoma , Mapeo Cromosómico , Displasia Cleidocraneal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Anomalías Craneofaciales/genética , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Factor de Transcripción SOX9/genética
12.
PLoS Genet ; 6(9): e1001111, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20838461

RESUMEN

We present the analysis of twenty human genomes to evaluate the prospects for identifying rare functional variants that contribute to a phenotype of interest. We sequenced at high coverage ten "case" genomes from individuals with severe hemophilia A and ten "control" genomes. We summarize the number of genetic variants emerging from a study of this magnitude, and provide a proof of concept for the identification of rare and highly-penetrant functional variants by confirming that the cause of hemophilia A is easily recognizable in this data set. We also show that the number of novel single nucleotide variants (SNVs) discovered per genome seems to stabilize at about 144,000 new variants per genome, after the first 15 individuals have been sequenced. Finally, we find that, on average, each genome carries 165 homozygous protein-truncating or stop loss variants in genes representing a diverse set of pathways.


Asunto(s)
Genoma Humano/genética , Análisis de Secuencia de ADN , Secuencia de Bases , Estudios de Casos y Controles , Variaciones en el Número de Copia de ADN/genética , Bases de Datos Genéticas , Exones/genética , Factor VIII/genética , Duplicación de Gen/genética , Técnicas de Inactivación de Genes , Genética de Población , Genotipo , Hemofilia A/genética , Humanos , Mutación INDEL/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Sistemas de Lectura Abierta/genética , Polimorfismo Genético , Polimorfismo de Nucleótido Simple/genética
13.
PLoS Genet ; 6(6): e1000991, 2010 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-20577567

RESUMEN

Although more than 2,400 genes have been shown to contain variants that cause Mendelian disease, there are still several thousand such diseases yet to be molecularly defined. The ability of new whole-genome sequencing technologies to rapidly indentify most of the genetic variants in any given genome opens an exciting opportunity to identify these disease genes. Here we sequenced the whole genome of a single patient with the dominant Mendelian disease, metachondromatosis (OMIM 156250), and used partial linkage data from her small family to focus our search for the responsible variant. In the proband, we identified an 11 bp deletion in exon four of PTPN11, which alters frame, results in premature translation termination, and co-segregates with the phenotype. In a second metachondromatosis family, we confirmed our result by identifying a nonsense mutation in exon 4 of PTPN11 that also co-segregates with the phenotype. Sequencing PTPN11 exon 4 in 469 controls showed no such protein truncating variants, supporting the pathogenicity of these two mutations. This combination of a new technology and a classical genetic approach provides a powerful strategy to discover the genes responsible for unexplained Mendelian disorders.


Asunto(s)
Ligamiento Genético , Predisposición Genética a la Enfermedad , Genoma Humano , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Exones , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Mutación , Linaje , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...