Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
2.
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
3.
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.

4.
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
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.
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
7.
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
8.
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
9.
Invest Ophthalmol Vis Sci ; 60(13): 4249-4256, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31618761

RESUMEN

Purpose: To investigate the role of two deep-intronic ABCA4 variants, that showed a mild splice defect in vitro and can occur on the same allele as the low penetrant c.5603A>T, in Stargardt disease (STGD1). Methods: Ophthalmic data were assessed of 18 STGD1 patients who harbored c.769-784C>T or c.4253+43G>A in combination with a severe ABCA4 variant. Subjects carrying c.[769-784C>T; 5603A>T] were clinically compared with a STGD1 cohort previously published carrying c.5603A>T noncomplex. We calculated the penetrances of the intronic variants using ABCA4 allele frequency data of the general population and investigated the effect of c.769-784C>T on splicing in photoreceptor progenitor cells (PPCs). Results: Mostly, late-onset, foveal-sparing STGD1 was observed among subjects harboring c.769-784C>T or c.4253+43G>A (median age of onset, 54.5 and 52.0 years, respectively). However, ages of onset, phenotypes in fundo, and visual acuity courses varied widely. No significant clinical differences were observed between the c.[769-784C>T; 5603A>T] cohort and the c.4253+43G>A or the c.5603A>T cohort. The penetrances of c.769-784C>T (20.5%-39.6%) and c.4253+43G>A (35.8%-43.1%) were reduced, when not considering the effect of yet unidentified or known factors in cis, such as c.5603A>T (identified in 7/7 probands with c.769-784C>T; 1/8 probands with c.4253+43G>A). Variant c.769-784C>T resulted in a pseudo-exon insertion in 15% of the total mRNA (i.e., ∼30% of the c.769-784C>T allele alone). Conclusions: Two mild intronic ABCA4 variants could further explain missing heritability in late-onset STGD1, distinguishing it from AMD. The observed clinical variability and calculated reduced penetrance urge research into modifiers within and outside of the ABCA4 gene.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Enfermedad de Stargardt/genética , Anciano , Alelos , Femenino , Frecuencia de los Genes , Variación Genética , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Agudeza Visual
10.
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
11.
Invest Ophthalmol Vis Sci ; 59(8): 3220-3231, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29971439

RESUMEN

Purpose: To assess the occurrence and the disease expression of the common p.Asn1868Ile variant in patients with Stargardt disease (STGD1) harboring known, monoallelic causal ABCA4 variants. Methods: The coding and noncoding regions of ABCA4 were sequenced in 67 and 63 STGD1 probands respectively, harboring monoallelic ABCA4 variants. In case p.Asn1868Ile was detected, segregation analysis was performed whenever possible. Probands and affected siblings harboring p.Asn1868Ile without additional variants in cis were clinically evaluated retrospectively. Two asymptomatic siblings carrying the same ABCA4 variants as their probands were clinically examined. The penetrance of p.Asn1868Ile was calculated using allele frequency data of ABCA4 variants in non-Finnish European individuals. Results: The p.Asn1868Ile variant was found in cis with known variants in 14/67 probands. In 27/67 probands, we identified p.Asn1868Ile without additional variants in cis, in combination with known, mainly severe ABCA4 variants. In 23/27 probands, the trans configuration was established. Among 27 probands and 6/7 STGD1 siblings carrying p.Asn1868Ile, 42% manifested late-onset disease (>44 years). We additionally identified four asymptomatic relatives carrying a combination of a severe variant and p.Asn1868Ile; ophthalmologic examination in two persons did not reveal STGD1. Based on ABCA4 allele frequency data, we conservatively estimated the penetrance of p.Asn1868Ile, when present in trans with a severe variant, to be below 5%. Conclusions: A significant fraction of genetically unexplained STGD1 cases carries p.Asn1868Ile as a second variant. Our findings suggest exceptional differences in disease expression or even nonpenetrance of this ABCA4 variant, pointing toward an important role for genetic or environmental modifiers in STGD1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Degeneración Macular/congénito , Mutación , Polimorfismo de Nucleótido Simple , Adulto , Edad de Inicio , Anciano , Electrorretinografía , Femenino , Angiografía con Fluoresceína , Frecuencia de los Genes , Prueba de Complementación Genética , Humanos , Degeneración Macular/diagnóstico , Degeneración Macular/genética , Degeneración Macular/fisiopatología , Masculino , Persona de Mediana Edad , Linaje , Penetrancia , Estudios Retrospectivos , Análisis de Secuencia de ADN , Hermanos , Enfermedad de Stargardt , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología
12.
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
13.
Genome Biol ; 18(1): 48, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28274275

RESUMEN

BACKGROUND: Hirschsprung disease (HSCR), which is congenital obstruction of the bowel, results from a failure of enteric nervous system (ENS) progenitors to migrate, proliferate, differentiate, or survive within the distal intestine. Previous studies that have searched for genes underlying HSCR have focused on ENS-related pathways and genes not fitting the current knowledge have thus often been ignored. We identify and validate novel HSCR genes using whole exome sequencing (WES), burden tests, in silico prediction, unbiased in vivo analyses of the mutated genes in zebrafish, and expression analyses in zebrafish, mouse, and human. RESULTS: We performed de novo mutation (DNM) screening on 24 HSCR trios. We identify 28 DNMs in 21 different genes. Eight of the DNMs we identified occur in RET, the main HSCR gene, and the remaining 20 DNMs reside in genes not reported in the ENS. Knockdown of all 12 genes with missense or loss-of-function DNMs showed that the orthologs of four genes (DENND3, NCLN, NUP98, and TBATA) are indispensable for ENS development in zebrafish, and these results were confirmed by CRISPR knockout. These genes are also expressed in human and mouse gut and/or ENS progenitors. Importantly, the encoded proteins are linked to neuronal processes shared by the central nervous system and the ENS. CONCLUSIONS: Our data open new fields of investigation into HSCR pathology and provide novel insights into the development of the ENS. Moreover, the study demonstrates that functional analyses of genes carrying DNMs are warranted to delineate the full genetic architecture of rare complex diseases.


Asunto(s)
Exoma , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Secuenciación de Nucleótidos de Alto Rendimiento , Enfermedad de Hirschsprung/genética , Alelos , Animales , Estudios de Casos y Controles , Biología Computacional/métodos , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Genotipo , Humanos , Mutación , Fenotipo , Pez Cebra
14.
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
15.
Eur J Hum Genet ; 25(5): 591-599, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28224992

RESUMEN

Inherited eye disorders have a large clinical and genetic heterogeneity, which makes genetic diagnosis cumbersome. An exome-sequencing approach was developed in which data analysis was divided into two steps: the vision gene panel and exome analysis. In the vision gene panel analysis, variants in genes known to cause inherited eye disorders were assessed for pathogenicity. If no causative variants were detected and when the patient consented, the entire exome data was analyzed. A total of 266 Dutch patients with different types of inherited eye disorders, including inherited retinal dystrophies, cataract, developmental eye disorders and optic atrophy, were investigated. In the vision gene panel analysis (likely), causative variants were detected in 49% and in the exome analysis in an additional 2% of the patients. The highest detection rate of (likely) causative variants was in patients with inherited retinal dystrophies, for instance a yield of 63% in patients with retinitis pigmentosa. In patients with developmental eye defects, cataract and optic atrophy, the detection rate was 50, 33 and 17%, respectively. An exome-sequencing approach enables a genetic diagnosis in patients with different types of inherited eye disorders using one test. The exome approach has the same detection rate as targeted panel sequencing tests, but offers a number of advantages. For instance, the vision gene panel can be frequently and easily updated with additional (novel) eye disorder genes. Determination of the genetic diagnosis improved the clinical diagnosis, regarding the assessment of the inheritance pattern as well as future disease perspective.


Asunto(s)
Exoma , Enfermedades Hereditarias del Ojo/genética , Patrón de Herencia , Trastornos de la Visión/genética , Adolescente , Adulto , Estudios de Casos y Controles , Niño , Enfermedades Hereditarias del Ojo/patología , Humanos , Países Bajos , Trastornos de la Visión/patología
16.
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
17.
Sci Rep ; 6: 28253, 2016 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-27339364

RESUMEN

X-linked cone dysfunction disorders such as Blue Cone Monochromacy and X-linked Cone Dystrophy are characterized by complete loss (of) or reduced L- and M- cone function due to defects in the OPN1LW/OPN1MW gene cluster. Here we investigated 24 affected males from 16 families with either a structurally intact gene cluster or at least one intact single (hybrid) gene but harbouring rare combinations of common SNPs in exon 3 in single or multiple OPN1LW and OPN1MW gene copies. We assessed twelve different OPN1LW/MW exon 3 haplotypes by semi-quantitative minigene splicing assay. Nine haplotypes resulted in aberrant splicing of ≥20% of transcripts including the known pathogenic haplotypes (i.e. 'LIAVA', 'LVAVA') with absent or minute amounts of correctly spliced transcripts, respectively. De novo formation of the 'LIAVA' haplotype derived from an ancestral less deleterious 'LIAVS' haplotype was observed in one family with strikingly different phenotypes among affected family members. We could establish intrachromosomal gene conversion in the male germline as underlying mechanism. Gene conversion in the OPN1LW/OPN1MW genes has been postulated, however, we are first to demonstrate a de novo gene conversion within the lineage of a pedigree.


Asunto(s)
Defectos de la Visión Cromática/genética , Conversión Génica , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Mutación de Línea Germinal , Opsinas de Bastones/genética , Defectos de la Visión Cromática/diagnóstico por imagen , Defectos de la Visión Cromática/fisiopatología , Electrorretinografía , Exones , Femenino , Genes Ligados a X , Haplotipos , Humanos , Masculino , Familia de Multigenes , Linaje , Polimorfismo de Nucleótido Simple
18.
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
19.
Eur J Hum Genet ; 24(4): 542-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26173970

RESUMEN

Variants in CIB2 can underlie either Usher syndrome type I (USH1J) or nonsyndromic hearing impairment (NSHI) (DFNB48). Here, a novel homozygous missense variant c.196C>T and compound heterozygous variants, c.[97C>T];[196C>T], were found, respectively, in two unrelated families of Dutch origin. Besides, the previously reported c.272 T>C functional missense variant in CIB2 was identified in two families of Pakistani origin. The missense variants are demonstrated not to affect subcellular localization of CIB2 in vestibular hair cells in ex vivo expression experiments. Furthermore, these variants do not affect the ATP-induced calcium responses in COS-7 cells. However, based on the residues affected, the variants are suggested to alter αIIß integrin binding. HI was nonsyndromic in all four families. However, deafness segregating with the c.272T>C variant in one Pakistani family is remarkably less severe than that in all other families with this mutation. Our results contribute to the insight in genotype-phenotype correlations of CIB2 mutations.


Asunto(s)
Proteínas de Unión al Calcio/genética , Calcio/metabolismo , Sordera/genética , Células Ciliadas Auditivas/metabolismo , Adolescente , Adulto , Animales , Células COS , Proteínas de Unión al Calcio/metabolismo , Niño , Chlorocebus aethiops , Sordera/metabolismo , Femenino , Humanos , Integrina alfa2beta1/metabolismo , Masculino , Mutación Missense , Linaje , Unión Proteica
20.
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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...