Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Prenat Diagn ; 42(6): 762-774, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34643287

RESUMEN

BACKGROUND: Adding rapid exome sequencing (rES) to conventional genetic tests improves the diagnostic yield of pregnancies showing ultrasound abnormalities but also carries a higher chance of unsolicited findings. We evaluated how rES, including pre- and post-test counseling, was experienced by parents investigating its impact on decision-making and experienced levels of anxiety. METHODS: A mixed-methods approach was adopted. Participating couples (n = 46) were asked to fill in two surveys (pre-test and post-test counseling) and 11 couples were approached for an additional interview. RESULTS: All couples accepted the rES test-offer with the most important reason for testing emphasizing their hope of finding an underlying diagnosis that would aid decision-making. The actual impact on decision-making was low, however, since most parents decided to terminate the pregnancy based on the major and multiple fetal ultrasound anomalies and did not wait for their rES results. Anxiety was elevated for most participants and decreased over time. CONCLUSION: Major congenital anomalies detected on ultrasound seem to have more impact on prenatal parental decision-making and anxiety then the offer and results of rES. However, the impact of rES on reproductive decision-making and experienced anxiety requires further investigation, especially in pregnancies where less (severe) fetal anomalies are detected on ultrasound.


Asunto(s)
Anomalías Múltiples , Diagnóstico Prenatal , Anomalías Múltiples/diagnóstico por imagen , Anomalías Múltiples/genética , Exoma , Femenino , Feto/diagnóstico por imagen , Humanos , Padres , Embarazo , Diagnóstico Prenatal/métodos , Ultrasonografía Prenatal/métodos , Secuenciación del Exoma/métodos
2.
Genet Med ; 22(3): 598-609, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31700164

RESUMEN

PURPOSE: Most classical aniridia is caused by PAX6 haploinsufficiency. PAX6 missense variants can be hypomorphic or mimic haploinsufficiency. We hypothesized that missense variants also cause previously undescribed disease by altering the affinity and/or specificity of PAX6 genomic interactions. METHODS: We screened PAX6 in 372 individuals with bilateral microphthalmia, anophthalmia, or coloboma (MAC) from the Medical Research Council Human Genetics Unit eye malformation cohort (HGUeye) and reviewed data from the Deciphering Developmental Disorders study. We performed cluster analysis on PAX6-associated ocular phenotypes by variant type and molecular modeling of the structural impact of 86 different PAX6 causative missense variants. RESULTS: Eight different PAX6 missense variants were identified in 17 individuals (15 families) with MAC, accounting for 4% (15/372) of our cohort. Seven altered the paired domain (p.[Arg26Gln]x1, p.[Gly36Val]x1, p.[Arg38Trp]x2, p.[Arg38Gln]x1, p.[Gly51Arg]x2, p.[Ser54Arg]x2, p.[Asn124Lys]x5) and one the homeodomain (p.[Asn260Tyr]x1). p.Ser54Arg and p.Asn124Lys were exclusively associated with severe bilateral microphthalmia. MAC-associated variants were predicted to alter but not ablate DNA interaction, consistent with the electrophoretic mobility shifts observed using mutant paired domains with well-characterized PAX6-binding sites. We found no strong evidence for novel PAX6-associated extraocular disease. CONCLUSION: Altering the affinity and specificity of PAX6-binding genome-wide provides a plausible mechanism for the worse-than-null effects of MAC-associated missense variants.


Asunto(s)
Anomalías del Ojo/genética , Predisposición Genética a la Enfermedad , Microftalmía/genética , Factor de Transcripción PAX6/genética , Adolescente , Adulto , Sitios de Unión/genética , Niño , Preescolar , Estudios de Cohortes , Proteínas de Unión al ADN/genética , Anomalías del Ojo/patología , Femenino , Heterocigoto , Humanos , Lactante , Masculino , Microftalmía/patología , Mutación Missense/genética , Linaje , Adulto Joven
3.
Genet Med ; 22(3): 547-556, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31649276

RESUMEN

PURPOSE: Treacher Collins syndrome (TCS) is a rare autosomal dominant mandibulofacial dysostosis, with a prevalence of 0.2-1/10,000. Features include bilateral and symmetrical malar and mandibular hypoplasia and facial abnormalities due to abnormal neural crest cell (NCC) migration and differentiation. To date, three genes have been identified: TCOF1, POLR1C, and POLR1D. Despite a large number of patients with a molecular diagnosis, some remain without a known genetic anomaly. METHODS: We performed exome sequencing for four individuals with TCS but who were negative for pathogenic variants in the known causative genes. The effect of the pathogenic variants was investigated in zebrafish. RESULTS: We identified three novel pathogenic variants in POLR1B. Knockdown of polr1b in zebrafish induced an abnormal craniofacial phenotype mimicking TCS that was associated with altered ribosomal gene expression, massive p53-associated cellular apoptosis in the neuroepithelium, and reduced number of NCC derivatives. CONCLUSION: Pathogenic variants in the RNA polymerase I subunit POLR1B might induce massive p53-dependent apoptosis in a restricted neuroepithelium area, altering NCC migration and causing cranioskeletal malformations. We identify POLR1B as a new causative gene responsible for a novel TCS syndrome (TCS4) and establish a novel experimental model in zebrafish to study POLR1B-related TCS.


Asunto(s)
Anomalías Craneofaciales/genética , ARN Polimerasas Dirigidas por ADN/genética , Disostosis Mandibulofacial/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Animales , Apoptosis/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Anomalías Craneofaciales/patología , Predisposición Genética a la Enfermedad , Humanos , Disostosis Mandibulofacial/patología , Mutación , Cresta Neural/anomalías , Cresta Neural/patología , Proteína p53 Supresora de Tumor/genética , Secuenciación del Exoma , Pez Cebra/genética
4.
Prenat Diagn ; 40(2): 197-205, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31697852

RESUMEN

OBJECTIVES: To give an overview of the genetic and structural abnormalities occurring in fetuses with nuchal translucency (NT) measurement exceeding the 95th percentile at first-trimester screening and to investigate which of these abnormalities would be missed if cell-free fetal DNA (cfDNA) were used as a first-tier screening test for chromosomal abnormalities. METHODS: This is a national study including 1901 pregnancies with NT≥95th percentile referred to seven university hospitals in the Netherlands between 1 January 2010 and 1 January 2016. All cases with unknown pregnancy outcome were excluded. Results of detailed ultrasound examinations, karyotyping, genotyping, pregnancy and neonatal outcomes, investigation by a clinical geneticist and post-mortem investigations were collected. RESULTS: In total, 821 (43%) pregnancies had at least one abnormality. The rate of abnormalities was 21% for fetuses with NT between 95th and 99th percentile and 62% for fetuses with NT≥99th percentile. Prevalence of single-gene disorders, submicroscopic, chromosomal and structural abnormalities was 2%, 2%, 30% and 9%, respectively. CONCLUSION: Although cfDNA is superior to the combined test, especially for the detection of trisomy 21, 34% of the congenital abnormalities occurring in fetuses with increased NT may remain undetected in the first trimester of pregnancy, unless cfDNA is used in combination with fetal sonographic assessment, including NT measurement.


Asunto(s)
Trastornos de los Cromosomas/diagnóstico por imagen , Trastornos de los Cromosomas/genética , Anomalías Congénitas/diagnóstico por imagen , Anomalías Congénitas/genética , Medida de Translucencia Nucal , Cariotipo Anormal , Adolescente , Adulto , Aneuploidia , Síndrome de DiGeorge/diagnóstico por imagen , Síndrome de DiGeorge/genética , Síndrome de Down/diagnóstico por imagen , Síndrome de Down/genética , Displasia Ectodérmica/diagnóstico por imagen , Displasia Ectodérmica/genética , Facies , Insuficiencia de Crecimiento/diagnóstico por imagen , Insuficiencia de Crecimiento/genética , Femenino , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/genética , Humanos , Síndrome LEOPARD/diagnóstico por imagen , Síndrome LEOPARD/genética , Persona de Mediana Edad , Países Bajos , Pruebas Prenatales no Invasivas , Síndrome de Noonan/diagnóstico por imagen , Síndrome de Noonan/genética , Embarazo , Primer Trimestre del Embarazo , Síndrome de la Trisomía 13/diagnóstico por imagen , Síndrome de la Trisomía 13/genética , Síndrome de la Trisomía 18/diagnóstico por imagen , Síndrome de la Trisomía 18/genética , Ultrasonografía Prenatal , Adulto Joven
5.
Prenat Diagn ; 40(10): 1300-1309, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32627857

RESUMEN

OBJECTIVE: Conventional genetic tests (quantitative fluorescent-PCR [QF-PCR] and single nucleotide polymorphism-array) only diagnose ~40% of fetuses showing ultrasound abnormalities. Rapid exome sequencing (rES) may improve this diagnostic yield, but includes challenges such as uncertainties in fetal phenotyping, variant interpretation, incidental unsolicited findings, and rapid turnaround times. In this study, we implemented rES in prenatal care to increase diagnostic yield. METHODS: We prospectively studied 55 fetuses. Inclusion criteria were: (a) two or more independent major fetal anomalies, (b) hydrops fetalis or bilateral renal cysts alone, or (c) one major fetal anomaly and a first-degree relative with the same anomaly. In addition to conventional genetic tests, we performed trio rES analysis using a custom virtual gene panel of ~3850 Online Mendelian Inheritance in Man (OMIM) genes. RESULTS: We established a genetic rES-based diagnosis in 8 out of 23 fetuses (35%) without QF-PCR or array abnormalities. Diagnoses included MIRAGE (SAMD9), Zellweger (PEX1), Walker-Warburg (POMGNT1), Noonan (PTNP11), Kabuki (KMT2D), and CHARGE (CHD7) syndrome and two cases of Osteogenesis Imperfecta type 2 (COL1A1). In six cases, rES diagnosis aided perinatal management. The median turnaround time was 14 (range 8-20) days. CONCLUSION: Implementing rES as a routine test in the prenatal setting is challenging but technically feasible, with a promising diagnostic yield and significant clinical relevance.


Asunto(s)
Anomalías Múltiples/diagnóstico , Secuenciación del Exoma , Diagnóstico Prenatal/métodos , Anomalías Múltiples/epidemiología , Anomalías Múltiples/genética , Adulto , Pruebas Diagnósticas de Rutina/estadística & datos numéricos , Estudios de Factibilidad , Femenino , Feto/diagnóstico por imagen , Pruebas Genéticas/métodos , Pruebas Genéticas/estadística & datos numéricos , Humanos , Recién Nacido , Masculino , Países Bajos/epidemiología , Reacción en Cadena de la Polimerasa/métodos , Polimorfismo de Nucleótido Simple , Valor Predictivo de las Pruebas , Embarazo , Resultado del Embarazo/epidemiología , Diagnóstico Prenatal/estadística & datos numéricos , Estudios Prospectivos , Ultrasonografía Prenatal
6.
Proc Natl Acad Sci U S A ; 114(13): E2739-E2747, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28292896

RESUMEN

Megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS) is a congenital visceral myopathy characterized by severe dilation of the urinary bladder and defective intestinal motility. The genetic basis of MMIHS has been ascribed to spontaneous and autosomal dominant mutations in actin gamma 2 (ACTG2), a smooth muscle contractile gene. However, evidence suggesting a recessive origin of the disease also exists. Using combined homozygosity mapping and whole exome sequencing, a genetically isolated family was found to carry a premature termination codon in Leiomodin1 (LMOD1), a gene preferentially expressed in vascular and visceral smooth muscle cells. Parents heterozygous for the mutation exhibited no abnormalities, but a child homozygous for the premature termination codon displayed symptoms consistent with MMIHS. We used CRISPR-Cas9 (CRISPR-associated protein) genome editing of Lmod1 to generate a similar premature termination codon. Mice homozygous for the mutation showed loss of LMOD1 protein and pathology consistent with MMIHS, including late gestation expansion of the bladder, hydronephrosis, and rapid demise after parturition. Loss of LMOD1 resulted in a reduction of filamentous actin, elongated cytoskeletal dense bodies, and impaired intestinal smooth muscle contractility. These results define LMOD1 as a disease gene for MMIHS and suggest its role in establishing normal smooth muscle cytoskeletal-contractile coupling.


Asunto(s)
Anomalías Múltiples/genética , Autoantígenos/fisiología , Colon/anomalías , Proteínas del Citoesqueleto/fisiología , Seudoobstrucción Intestinal/genética , Proteínas Musculares/fisiología , Vejiga Urinaria/anomalías , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Codón sin Sentido , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Femenino , Humanos , Recién Nacido , Ratones , Contracción Muscular/genética , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso/fisiología
7.
Gastroenterology ; 155(1): 130-143.e15, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29604290

RESUMEN

BACKGROUND & AIMS: Congenital diarrheal disorders are rare inherited intestinal disorders characterized by intractable, sometimes life-threatening, diarrhea and nutrient malabsorption; some have been associated with mutations in diacylglycerol-acyltransferase 1 (DGAT1), which catalyzes formation of triacylglycerol from diacylglycerol and acyl-CoA. We investigated the mechanisms by which DGAT1 deficiency contributes to intestinal failure using patient-derived organoids. METHODS: We collected blood samples from 10 patients, from 6 unrelated pedigrees, who presented with early-onset severe diarrhea and/or vomiting, hypoalbuminemia, and/or (fatal) protein-losing enteropathy with intestinal failure; we performed next-generation sequencing analysis of DNA from 8 patients. Organoids were generated from duodenal biopsies from 3 patients and 3 healthy individuals (controls). Caco-2 cells and patient-derived dermal fibroblasts were transfected or transduced with vectors that express full-length or mutant forms of DGAT1 or full-length DGAT2. We performed CRISPR/Cas9-guided disruption of DGAT1 in control intestinal organoids. Cells and organoids were analyzed by immunoblot, immunofluorescence, flow cytometry, chromatography, quantitative real-time polymerase chain reaction, and for the activity of caspases 3 and 7. RESULTS: In the 10 patients, we identified 5 bi-allelic loss-of-function mutations in DGAT1. In patient-derived fibroblasts and organoids, the mutations reduced expression of DGAT1 protein and altered triacylglycerol metabolism, resulting in decreased lipid droplet formation after oleic acid addition. Expression of full-length DGAT2 in patient-derived fibroblasts restored formation of lipid droplets. Organoids derived from patients with DGAT1 mutations were more susceptible to lipid-induced cell death than control organoids. CONCLUSIONS: We identified a large cohort of patients with congenital diarrheal disorders with mutations in DGAT1 that reduced expression of its product; dermal fibroblasts and intestinal organoids derived from these patients had altered lipid metabolism and were susceptible to lipid-induced cell death. Expression of full-length wildtype DGAT1 or DGAT2 restored normal lipid metabolism in these cells. These findings indicate the importance of DGAT1 in fat metabolism and lipotoxicity in the intestinal epithelium. A fat-free diet might serve as the first line of therapy for patients with reduced DGAT1 expression. It is important to identify genetic variants associated with congenital diarrheal disorders for proper diagnosis and selection of treatment strategies.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/genética , Duodeno/metabolismo , Fibroblastos/metabolismo , Hipoalbuminemia/genética , Trastornos del Metabolismo de los Lípidos/genética , Organoides/metabolismo , Enteropatías Perdedoras de Proteínas/genética , Células CACO-2 , Estudios de Casos y Controles , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Niño , Preescolar , Consanguinidad , Dermis/citología , Diacilglicerol O-Acetiltransferasa/deficiencia , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Países Bajos , Forboles , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Turquía
8.
Genet Med ; 21(8): 1751-1760, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30643219

RESUMEN

PURPOSE: Using exome sequencing, the underlying variants in many persons with autosomal recessive diseases remain undetected. We explored autosomal recessive Stargardt disease (STGD1) as a model to identify the missing heritability. METHODS: Sequencing of ABCA4 was performed in 8 STGD1 cases with one variant and p.Asn1868Ile in trans, 25 cases with one variant, and 3 cases with no ABCA4 variant. The effect of intronic variants was analyzed using in vitro splice assays in HEK293T cells and patient-derived fibroblasts. Antisense oligonucleotides were used to correct splice defects. RESULTS: In 24 of the probands (67%), one known and five novel deep-intronic variants were found. The five novel variants resulted in messenger RNA pseudoexon inclusions, due to strengthening of cryptic splice sites or by disrupting a splicing silencer motif. Variant c.769-784C>T showed partial insertion of a pseudoexon and was found in cis with c.5603A>T (p.Asn1868Ile), so its causal role could not be fully established. Variant c.4253+43G>A resulted in partial skipping of exon 28. Remarkably, antisense oligonucleotides targeting the aberrant splice processes resulted in (partial) correction of all splicing defects. CONCLUSION: Our data demonstrate the importance of assessing noncoding variants in genetic diseases, and show the great potential of splice modulation therapy for deep-intronic variants.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Oligonucleótidos Antisentido/genética , Isoformas de Proteínas/genética , Enfermedad de Stargardt/genética , Adolescente , Adulto , Anciano , Niño , Exones/genética , Células HEK293 , Humanos , Intrones/genética , Persona de Mediana Edad , Mutación/genética , Oligonucleótidos Antisentido/farmacología , Linaje , Polimorfismo de Nucleótido Simple/genética , Empalme del ARN/genética , Enfermedad de Stargardt/patología , Adulto Joven
9.
Hum Mol Genet ; 25(3): 571-83, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26647307

RESUMEN

Megacystis Microcolon Intestinal Hypoperistalsis Syndrome (MMIHS) is a rare congenital disorder, in which heterozygous missense variants in the Enteric Smooth Muscle actin γ-2 (ACTG2) gene have been recently identified. To investigate the mechanism by which ACTG2 variants lead to MMIHS, we screened a cohort of eleven MMIHS patients, eight sporadic and three familial cases, and performed immunohistochemistry, molecular modeling and molecular dynamics (MD) simulations, and in vitro assays. In all sporadic cases, a heterozygous missense variant in ACTG2 was identified. ACTG2 expression was detected in all intestinal layers where smooth muscle cells are present in different stages of human development. No histopathological abnormalities were found in the patients. Using molecular modeling and MD simulations, we predicted that ACTG2 variants lead to significant changes to the protein function. This was confirmed by in vitro studies, which showed that the identified variants not only impair ACTG2 polymerization, but also contribute to reduced cell contractility. Taken together, our results confirm the involvement of ACTG2 in sporadic MMIHS, and bring new insights to MMIHS pathogenesis.


Asunto(s)
Anomalías Múltiples/genética , Actinas/genética , Colon/anomalías , Mucosa Intestinal/metabolismo , Seudoobstrucción Intestinal/genética , Contracción Muscular/genética , Músculo Liso/metabolismo , Mutación Missense , Vejiga Urinaria/anomalías , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Actinas/química , Actinas/metabolismo , Colon/metabolismo , Colon/patología , Resultado Fatal , Femenino , Expresión Génica , Heterocigoto , Humanos , Recién Nacido , Seudoobstrucción Intestinal/metabolismo , Seudoobstrucción Intestinal/patología , Intestinos/patología , Masculino , Simulación de Dinámica Molecular , Músculo Liso/patología , Linaje , Multimerización de Proteína , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Adulto Joven
10.
Hum Mol Genet ; 25(23): 5265-5275, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27702942

RESUMEN

Hirschsprung disease (HSCR) is the most common cause of neonatal intestinal obstruction. It is characterized by the absence of ganglia in the nerve plexuses of the lower gastrointestinal tract. So far, three common disease-susceptibility variants at the RET, SEMA3 and NRG1 loci have been detected through genome-wide association studies (GWAS) in Europeans and Asians to understand its genetic etiologies. Here we present a trans-ethnic meta-analysis of 507 HSCR cases and 1191 controls, combining all published GWAS results on HSCR to fine-map these loci and narrow down the putatively causal variants to 99% credible sets. We also demonstrate that the effects of RET and NRG1 are universal across European and Asian ancestries. In contrast, we detected a European-specific association of a low-frequency variant, rs80227144, in SEMA3 [odds ratio (OR) = 5.2, P = 4.7 × 10-10]. Conditional analyses on the lead SNPs revealed a secondary association signal, corresponding to an Asian-specific, low-frequency missense variant encoding RET p.Asp489Asn (rs9282834, conditional OR = 20.3, conditional P = 4.1 × 10-14). When in trans with the RET intron 1 enhancer risk allele, rs9282834 increases the risk of HSCR from 1.1 to 26.7. Overall, our study provides further insights into the genetic architecture of HSCR and has profound implications for future study designs.


Asunto(s)
Predisposición Genética a la Enfermedad , Enfermedad de Hirschsprung/genética , Neurregulina-1/genética , Proteínas Proto-Oncogénicas c-ret/genética , Semaforina-3A/genética , Alelos , Pueblo Asiatico/genética , Etnicidad/genética , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Enfermedad de Hirschsprung/patología , Humanos , Intrones/genética , Masculino , Polimorfismo de Nucleótido Simple , Población Blanca/genética
11.
Am J Hum Genet ; 96(4): 581-96, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25839327

RESUMEN

Innervation of the gut is segmentally lost in Hirschsprung disease (HSCR), a consequence of cell-autonomous and non-autonomous defects in enteric neuronal cell differentiation, proliferation, migration, or survival. Rare, high-penetrance coding variants and common, low-penetrance non-coding variants in 13 genes are known to underlie HSCR risk, with the most frequent variants in the ret proto-oncogene (RET). We used a genome-wide association (220 trios) and replication (429 trios) study to reveal a second non-coding variant distal to RET and a non-coding allele on chromosome 7 within the class 3 Semaphorin gene cluster. Analysis in Ret wild-type and Ret-null mice demonstrates specific expression of Sema3a, Sema3c, and Sema3d in the enteric nervous system (ENS). In zebrafish embryos, sema3 knockdowns show reduction of migratory ENS precursors with complete ablation under conjoint ret loss of function. Seven candidate receptors of Sema3 proteins are also expressed within the mouse ENS and their expression is also lost in the ENS of Ret-null embryos. Sequencing of SEMA3A, SEMA3C, and SEMA3D in 254 HSCR-affected subjects followed by in silico protein structure modeling and functional analyses identified five disease-associated alleles with loss-of-function defects in semaphorin dimerization and binding to their cognate neuropilin and plexin receptors. Thus, semaphorin 3C/3D signaling is an evolutionarily conserved regulator of ENS development whose dys-regulation is a cause of enteric aganglionosis.


Asunto(s)
Epistasis Genética/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética , Enfermedad de Hirschsprung/genética , Proteínas Proto-Oncogénicas c-ret/genética , Semaforinas/genética , Animales , Secuencia de Bases , Estudio de Asociación del Genoma Completo , Ratones , Datos de Secuencia Molecular , Semaforinas/deficiencia , Semaforinas/metabolismo , Análisis de Secuencia de ADN
12.
Am J Hum Genet ; 97(3): 475-82, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26299364

RESUMEN

Adams-Oliver syndrome (AOS) is a rare developmental disorder characterized by the presence of aplasia cutis congenita (ACC) of the scalp vertex and terminal limb-reduction defects. Cardiovascular anomalies are also frequently observed. Mutations in five genes have been identified as a cause for AOS prior to this report. Mutations in EOGT and DOCK6 cause autosomal-recessive AOS, whereas mutations in ARHGAP31, RBPJ, and NOTCH1 lead to autosomal-dominant AOS. Because RBPJ, NOTCH1, and EOGT are involved in NOTCH signaling, we hypothesized that mutations in other genes involved in this pathway might also be implicated in AOS pathogenesis. Using a candidate-gene-based approach, we prioritized DLL4, a critical NOTCH ligand, due to its essential role in vascular development in the context of cardiovascular features in AOS-affected individuals. Targeted resequencing of the DLL4 gene with a custom enrichment panel in 89 independent families resulted in the identification of seven mutations. A defect in DLL4 was also detected in two families via whole-exome or genome sequencing. In total, nine heterozygous mutations in DLL4 were identified, including two nonsense and seven missense variants, the latter encompassing four mutations that replace or create cysteine residues, which are most likely critical for maintaining structural integrity of the protein. Affected individuals with DLL4 mutations present with variable clinical expression with no emerging genotype-phenotype correlations. Our findings demonstrate that DLL4 mutations are an additional cause of autosomal-dominant AOS or isolated ACC and provide further evidence for a key role of NOTCH signaling in the etiology of this disorder.


Asunto(s)
Displasia Ectodérmica/genética , Displasia Ectodérmica/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Deformidades Congénitas de las Extremidades/genética , Deformidades Congénitas de las Extremidades/patología , Mutación/genética , Dermatosis del Cuero Cabelludo/congénito , Transducción de Señal/genética , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas de Unión al Calcio , Heterocigoto , Humanos , Datos de Secuencia Molecular , Linaje , Receptores Notch/genética , Dermatosis del Cuero Cabelludo/genética , Dermatosis del Cuero Cabelludo/patología , Análisis de Secuencia de ADN
13.
Biochim Biophys Acta ; 1852(11): 2352-61, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26282049

RESUMEN

Congenital Short Bowel Syndrome (CSBS) is a rare gastrointestinal disorder in which the mean length of the small intestine is substantially reduced when compared to its normal counterpart. Families with several affected members have been described and CSBS has been suggested to have a genetic basis. Recently, our group found mutations in CLMP as the cause of the recessive form of CSBS, and mutations in FLNA as the cause of the X-linked form of the disease. These findings have improved the quality of genetic counselling for CSBS patients and made prenatal diagnostics possible. Moreover, they provided a reliable starting point to further investigate the pathogenesis of CSBS, and to better understand the development of the small intestine. In this review, we present our current knowledge on CSBS and discuss hypotheses on how the recent genetic findings can help understand the cause of CSBS.

14.
Am J Hum Genet ; 87(1): 60-74, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20598273

RESUMEN

The major gene for Hirschsprung disease (HSCR) encodes the receptor tyrosine kinase RET. In a study of 690 European- and 192 Chinese-descent probands and their parents or controls, we demonstrate the ubiquity of a >4-fold susceptibility from a C-->T allele (rs2435357: p = 3.9 x 10(-43) in European ancestry; p = 1.1 x 10(-21) in Chinese samples) that probably arose once within the intronic RET enhancer MCS+9.7. With in vitro assays, we now show that the T variant disrupts a SOX10 binding site within MCS+9.7 that compromises RET transactivation. The T allele, with a control frequency of 20%-30%/47% and case frequency of 54%-62%/88% in European/Chinese-ancestry individuals, is involved in all forms of HSCR. It is marginally associated with proband gender (p = 0.13) and significantly so with length of aganglionosis (p = 7.6 x 10(-5)) and familiality (p = 6.2 x 10(-4)). The enhancer variant is more frequent in the common forms of male, short-segment, and simplex families whereas multiple, rare, coding mutations are the norm in the less common and more severe forms of female, long-segment, and multiplex families. The T variant also increases penetrance in patients with rare RET coding mutations. Thus, both rare and common mutations, individually and together, make contributions to the risk of HSCR. The distribution of RET variants in diverse HSCR patients suggests a "cellular-recessive" genetic model where both RET alleles' function is compromised. The RET allelic series, and its genotype-phenotype correlations, shows that success in variant identification in complex disorders may strongly depend on which patients are studied.


Asunto(s)
Enfermedad de Hirschsprung/genética , Proteínas Proto-Oncogénicas c-ret/genética , Pueblo Asiatico , Secuencia de Bases , Estudios de Casos y Controles , Elementos de Facilitación Genéticos , Femenino , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Haplotipos , Enfermedad de Hirschsprung/etnología , Enfermedad de Hirschsprung/fisiopatología , Humanos , Masculino , Mutación , Penetrancia , Polimorfismo de Nucleótido Simple , Unión Proteica , Proteínas Proto-Oncogénicas c-ret/metabolismo , Factores de Transcripción SOXE/metabolismo , Factores Sexuales , Activación Transcripcional , Población Blanca
15.
Gastroenterology ; 142(3): 453-462.e3, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22155368

RESUMEN

BACKGROUND & AIMS: Short-bowel syndrome usually results from surgical resection of the small intestine for diseases such as intestinal atresias, volvulus, and necrotizing enterocolitis. Patients with congenital short-bowel syndrome (CSBS) are born with a substantial shortening of the small intestine, to a mean length of 50 cm, compared with a normal length at birth of 190-280 cm. They also are born with intestinal malrotation. Because CSBS occurs in many consanguineous families, it is considered to be an autosomal-recessive disorder. We aimed to identify and characterize the genetic factor causing CSBS. METHODS: We performed homozygosity mapping using 610,000 K single-nucleotide polymorphism arrays to analyze the genomes of 5 patients with CSBS. After identifying a gene causing the disease, we determined its expression pattern in human embryos. We also overexpressed forms of the gene product that were and were not associated with CSBS in Chinese Hamster Ovary and T84 cells and generated a zebrafish model of the disease. RESULTS: We identified loss-of-function mutations in Coxsackie- and adenovirus receptor-like membrane protein (CLMP) in CSBS patients. CLMP is a tight-junction-associated protein that is expressed in the intestine of human embryos throughout development. Mutations in CLMP prevented its normal localization to the cell membrane. Knock-down experiments in zebrafish resulted in general developmental defects, including shortening of the intestine and the absence of goblet cells. Because goblet cells are characteristic for the midintestine in zebrafish, which resembles the small intestine in human beings, the zebrafish model mimics CSBS. CONCLUSIONS: Loss-of-function mutations in CLMP cause CSBS in human beings, likely by interfering with tight-junction formation, which disrupts intestinal development. Furthermore, we developed a zebrafish model of CSBS.


Asunto(s)
Intestino Delgado/anomalías , Mutación Missense , Receptores Virales/genética , Síndrome del Intestino Corto/genética , Adolescente , Adulto , Animales , Células CHO , Niño , Preescolar , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Heterocigoto , Homocigoto , Humanos , Lactante , Recién Nacido , Intestino Delgado/metabolismo , Masculino , Morfogénesis , Fenotipo , Polimorfismo de Nucleótido Simple , Receptores Virales/metabolismo , Síndrome del Intestino Corto/embriología , Síndrome del Intestino Corto/metabolismo , Síndrome del Intestino Corto/patología , Transfección , Adulto Joven , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
Genet Med ; 15(4): 310-3, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23037936

RESUMEN

PURPOSE: Autosomal recessive congenital short bowel syndrome is caused by mutations in CLMP. No mutations were found in the affected males of a family with presumed X-linked congenital short bowel syndrome or in an isolated male patient. Our aim was to identify the disease-causing mutation in these patients. METHODS: We performed mutation analysis of the second exon of FLNA in the two surviving affected males of the presumed X-linked family and in the isolated patient. RESULTS: We identified a novel 2-base-pair deletion in the second exon of FLNA in all these male patients. The deletion is located between two nearby methionines at the N-terminus of filamin A. Previous studies showed that translation of FLNA occurs from both methionines, resulting in two isoforms of the protein. We hypothesized that the longer isoform is no longer translated due to the mutation and that this mutation is therefore not lethal for males in utero. CONCLUSION: Our findings emphasize that congenital short bowel syndrome can be the presenting symptom in male patients with mutations in FLNA.


Asunto(s)
Filaminas/genética , Mutación , Síndrome del Intestino Corto/diagnóstico , Síndrome del Intestino Corto/genética , Adolescente , Adulto , Secuencia de Bases , Exones , Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Humanos , Masculino , Linaje , Fenotipo , Eliminación de Secuencia , Adulto Joven
17.
Genet Med ; 15(3): 195-202, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22995989

RESUMEN

PURPOSE: Radial ray deficiencies are characterized by unilateral or bilateral absence of varying portions of the radius and thumb. Both isolated and syndromic forms have been described, and although for some of the syndromes the causal gene has been identified, many patients remain without a genetic diagnosis. METHODS: In this study, a cohort of 54 patients with radial ray deficiencies was screened for genomic aberrations by molecular karyotyping. RESULTS: In 8 of 54 cases, an aberration was detected. Two unrelated patients inherited a 1q21.1 microduplication from a healthy parent, whereas in a third patient, a 16p13.11 microduplication was identified. Two other interesting microdeletions were detected: a 10q24.3 deletion at the split hand-foot malformation (SHFM3) locus and a 7p22.1 deletion including the RAC1 gene. CONCLUSION: The finding of these microduplications may just be coincidental or, alternatively, they may illustrate the broad phenotypic spectrum of these microduplications. Duplications in the 10q24.3 region result in split hand-foot malformations, and our observation indicates that deletions may cause radial ray defects. Finally, a candidate gene for radial ray deficiencies was detected in the 7p22.1 deletion. RAC1 plays an important role in the canonical Wnt pathway and conditional RAC1 knockout mice exhibit truncated-limb defects.


Asunto(s)
Deformidades Congénitas de las Extremidades Superiores/genética , Duplicación Cromosómica , Cromosomas Humanos Par 10 , Cromosomas Humanos Par 7 , Hibridación Genómica Comparativa , Proteínas F-Box/genética , Femenino , Humanos , Masculino , Radiografía , Deformidades Congénitas de las Extremidades Superiores/diagnóstico , Deformidades Congénitas de las Extremidades Superiores/diagnóstico por imagen , Proteína de Unión al GTP rac1/genética
18.
HGG Adv ; 4(2): 100181, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-36785559

RESUMEN

A significant number of individuals with a rare disorder such as Usher syndrome (USH) and (non-)syndromic autosomal recessive retinitis pigmentosa (arRP) remain genetically unexplained. Therefore, we assessed subjects suspected of USH2A-associated disease and no or mono-allelic USH2A variants using whole genome sequencing (WGS) followed by an improved pipeline for variant interpretation to provide a conclusive diagnosis. One hundred subjects were screened using WGS to identify causative variants in USH2A or other USH/arRP-associated genes. In addition to the existing variant interpretation pipeline, a particular focus was put on assessing splice-affecting properties of variants, both in silico and in vitro. Also structural variants were extensively addressed. For variants resulting in pseudoexon inclusion, we designed and evaluated antisense oligonucleotides (AONs) using minigene splice assays and patient-derived photoreceptor precursor cells. Biallelic variants were identified in 49 of 100 subjects, including novel splice-affecting variants and structural variants, in USH2A or arRP/USH-associated genes. Thirteen variants were shown to affect USH2A pre-mRNA splicing, including four deep-intronic USH2A variants resulting in pseudoexon inclusion, which could be corrected upon AON treatment. We have shown that WGS, combined with a thorough variant interpretation pipeline focused on assessing pre-mRNA splicing defects and structural variants, is a powerful method to provide subjects with a rare genetic condition, a (likely) conclusive genetic diagnosis. This is essential for the development of future personalized treatments and for patients to be eligible for such treatments.


Asunto(s)
Retinitis Pigmentosa , Síndromes de Usher , Humanos , Síndromes de Usher/diagnóstico , Precursores del ARN , Mutación , Linaje , Retinitis Pigmentosa/diagnóstico , Secuenciación Completa del Genoma , Proteínas de la Matriz Extracelular/genética
19.
NPJ Genom Med ; 7(1): 65, 2022 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-36351915

RESUMEN

Pathogenic variants in the OPN1LW/OPN1MW gene cluster are causal for a range of mild to severe visual impairments with color deficiencies. The widely utilized short-read next-generation sequencing (NGS) is inappropriate for the analysis of the OPN1LW/OPN1MW gene cluster and many patients with pathogenic variants stay underdiagnosed. A diagnostic genetic assay was developed for the OPN1LW/OPN1MW gene cluster, consisting of copy number analysis via multiplex ligation-dependent probe amplification and sequence analysis via long-read circular consensus sequencing. Performance was determined on 50 clinical samples referred for genetic confirmation of the clinical diagnosis (n = 43) or carrier status analysis (n = 7). A broad range of pathogenic haplotypes were detected, including deletions, hybrid genes, single variants and combinations of variants. The developed genetic assay for the OPN1LW/OPN1MW gene cluster is a diagnostic test that can detect both structural and nucleotide variants with a straightforward analysis, improving diagnostic care of patients with visual impairment.

20.
Ophthalmology ; 118(1): 160-167.e1-3, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20801516

RESUMEN

PURPOSE: To develop a high-throughput, cost-effective diagnostic strategy for the identification of known and new mutations in 90 retinal disease genes. DESIGN: Evidence-based study. PARTICIPANTS: Sixty patients with a variety of retinal disorders, including Leber's congenital amaurosis, ocular albinism, pseudoxanthoma elasticum, retinitis pigmentosa, and Stargardt's disease. METHODS: We designed a custom 300-kb resequencing chip. Polymerase chain reaction (PCR) amplification, DNA fragmentation, and chip hybridization were performed according to Affymetrix recommendations. Hybridization signals were analyzed using Sequence pilot module seq-C mutation detection software (2009). This resequencing approach was validated by Sanger sequence technology. MAIN OUTCOME MEASURES: Disease-causing sequence changes. RESULTS: We developed a retinal resequencing chip that covers all exons of 90 retinal disease genes. We developed and tested multiplex primer sets for 1445 amplicons representing the genes included on the chip. We validated our approach by screening 87 exons from 25 retinal disease genes containing 87 known sequence changes previously identified in our patient group using Sanger sequencing. Call rates for successfully hybridized amplicons were 98% to 100%. Of the known single nucleotide changes, 99% could be detected on the chip. As expected, deletions could not be detected reliably. CONCLUSIONS: We designed a custom resequencing chip that can detect known and new sequence changes in 90 retinal disease genes using a new high-throughput strategy with a high sensitivity and specificity for one tenth of the cost of conventional direct sequencing. The developed amplification strategy allows for the pooling of multiple patients with non-overlapping phenotypes, enabling many patients to be analyzed simultaneously in a fast and cost-effective manner.


Asunto(s)
Análisis Mutacional de ADN/métodos , Proteínas del Ojo/genética , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Enfermedades de la Retina/genética , Análisis Costo-Beneficio , Análisis Mutacional de ADN/economía , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos/economía , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA