Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
PLoS Genet ; 18(6): e1010236, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35737725

RESUMEN

Congenital heart disease (CHD) is a common group of birth defects with a strong genetic contribution to their etiology, but historically the diagnostic yield from exome studies of isolated CHD has been low. Pleiotropy, variable expressivity, and the difficulty of accurately phenotyping newborns contribute to this problem. We hypothesized that performing exome sequencing on selected individuals in families with multiple members affected by left-sided CHD, then filtering variants by population frequency, in silico predictive algorithms, and phenotypic annotations from publicly available databases would increase this yield and generate a list of candidate disease-causing variants that would show a high validation rate. In eight of the nineteen families in our study (42%), we established a well-known gene/phenotype link for a candidate variant or performed confirmation of a candidate variant's effect on protein function, including variants in genes not previously described or firmly established as disease genes in the body of CHD literature: BMP10, CASZ1, ROCK1 and SMYD1. Two plausible variants in different genes were found to segregate in the same family in two instances suggesting oligogenic inheritance. These results highlight the need for functional validation and demonstrate that in the era of next-generation sequencing, multiplex families with isolated CHD can still bring high yield to the discovery of novel disease genes.


Asunto(s)
Exoma , Cardiopatías Congénitas , Proteínas Morfogenéticas Óseas/genética , Proteínas de Unión al ADN/genética , Exoma/genética , Frecuencia de los Genes , Estudios de Asociación Genética , Cardiopatías Congénitas/genética , Humanos , Recién Nacido , Linaje , Factores de Transcripción/genética , Secuenciación del Exoma , Quinasas Asociadas a rho/genética
2.
J Neonatal Perinatal Med ; 15(1): 113-121, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34151866

RESUMEN

BACKGROUND: Bronchopulmonary dysplasia (BPD) is chronic lung disease of prematurity and pulmonary hypertension (PH) is a major contributor to morbidity and mortality in BPD patients. Nitric oxide (NO) is a vasodilator and apoptotic mediator made by NO synthase (NOS). NOS is inhibited by asymmetric dimethylarginine (ADMA), and dimethylarginine dimethylaminohydrolase (DDAH) hydrolyzes ADMA. Previously, in a BPD patient cohort, we identified single nucleotide polymorphism (SNP) DDAH1 rs480414 (G > A) that was protective against developing PH. This study aims to determine functional consequences of the DDAH1 SNP in lymphoblastoid cell lines (LCLs) derived from neonatal cord blood. We tested the hypothesis that DDAH1 SNP (AA) results in DDAH1 gain of function, leading to greater NO-mediated apoptosis compared to DDAH1 wild-type (GG) in LCLs. METHODS: LCLs were analyzed by Western blot (DDAH1, cleaved and total caspase-3 and -8, and ß-actin), and RT-PCR (DDAH1, iNOS). Cell media assayed for nitrites with chemiluminescence NO analyzer, and conversion of ADMA to L-citrulline was measured by spectrophotometry. RESULTS: LCLs with DDAH1 SNP had similar levels of DDAH1 protein and mRNA expression, as well as DDAH activity, compared to DDAH1 WT LCLs. There were also no changes in cleaved caspase-3 and -8 protein levels. LCLs with DDAH1 SNP had similar iNOS mRNA expression. Nitrite levels in media were lower for DDAH1 SNP LCLs compared to DDAH1 WT LCLs (p < 0.05). CONCLUSION: Contrary to our hypothesis, we found that NO production was lower in DDAH1 SNP LCLs, indicative of a loss of function phenotype.


Asunto(s)
Displasia Broncopulmonar , Hipertensión Pulmonar , Displasia Broncopulmonar/genética , Línea Celular , Sangre Fetal , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/prevención & control , Recién Nacido , Óxido Nítrico , Polimorfismo de Nucleótido Simple
3.
PLoS Genet ; 16(5): e1008639, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32453731

RESUMEN

Hypertrophic cardiomyopathy (HCM) is characterized by thickening of the ventricular muscle without dilation and is often associated with dominant pathogenic variants in cardiac sarcomeric protein genes. Here, we report a family with two infants diagnosed with infantile-onset HCM and mitral valve dysplasia that led to death before one year of age. Using exome sequencing, we discovered that one of the affected children had a homozygous frameshift variant in Myosin light chain 2 (MYL2:NM_000432.3:c.431_432delCT: p.Pro144Argfs*57;MYL2-fs), which alters the last 20 amino acids of the protein and is predicted to impact the most C-terminal of the three EF-hand domains in MYL2. The parents are unaffected heterozygous carriers of the variant and the variant is absent in control cohorts from gnomAD. The absence of the phenotype in carriers and the infantile presentation of severe HCM is in contrast to HCM associated with dominant MYL2 variants. Immunohistochemical analysis of the ventricular muscle of the deceased patient with the MYL2-fs variant showed a marked reduction of MYL2 expression compared to an unaffected control. In vitro overexpression studies further indicate that the MYL2-fs variant is actively degraded. In contrast, an HCM-associated missense variant (MYL2:p.Gly162Arg) and three other MYL2 stop-gain variants (p.E22*, p.K62*, p.E97*) that result in loss of the EF domains are stably expressed but show impaired localization. The degradation of the MYL2-fs can be rescued by inhibiting the cell's proteasome function supporting a post-translational effect of the variant. In vivo rescue experiments with a Drosophila MYL2-homolog (Mlc2) knockdown model indicate that neither the MYL2-fs nor the MYL2:p.Gly162Arg variant supports normal cardiac function. The tools that we have generated provide a rapid screening platform for functional assessment of variants of unknown significance in MYL2. Our study supports an autosomal recessive model of inheritance for MYL2 loss-of-function variants in infantile HCM and highlights the variant-specific molecular differences found in MYL2-associated cardiomyopathy.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica/genética , Familia , Mutación del Sistema de Lectura , Cadenas Ligeras de Miosina/genética , Adulto , Animales , Animales Modificados Genéticamente , Cardiomiopatía Hipertrófica/clasificación , Cardiomiopatía Hipertrófica/congénito , Cardiomiopatía Hipertrófica/patología , Células Cultivadas , Consanguinidad , Drosophila , Resultado Fatal , Femenino , Genes Dominantes , Genes Recesivos , Heterocigoto , Humanos , Lactante , Muerte del Lactante , Recién Nacido , Masculino , Linaje , Fenotipo , Hermanos
4.
Am J Med Genet A ; 173(8): 2176-2188, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28653806

RESUMEN

Congenital left-sided cardiac lesions (LSLs) are a significant contributor to the mortality and morbidity of congenital heart disease (CHD). Structural copy number variants (CNVs) have been implicated in LSL without extra-cardiac features; however, non-penetrance and variable expressivity have created uncertainty over the use of CNV analyses in such patients. High-density SNP microarray genotyping data were used to infer large, likely-pathogenic, autosomal CNVs in a cohort of 1,139 probands with LSL and their families. CNVs were molecularly confirmed and the medical records of individual carriers reviewed. The gene content of novel CNVs was then compared with public CNV data from CHD patients. Large CNVs (>1 MB) were observed in 33 probands (∼3%). Six of these were de novo and 14 were not observed in the only available parent sample. Associated cardiac phenotypes spanned a broad spectrum without clear predilection. Candidate CNVs were largely non-recurrent, associated with heterozygous loss of copy number, and overlapped known CHD genomic regions. Novel CNV regions were enriched for cardiac development genes, including seven that have not been previously associated with human CHD. CNV analysis can be a clinically useful and molecularly informative tool in LSLs without obvious extra-cardiac defects, and may identify a clinically relevant genomic disorder in a small but important proportion of these individuals.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Cardiopatías Congénitas/genética , Corazón/fisiopatología , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Genómica , Genotipo , Cardiopatías Congénitas/fisiopatología , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , Adulto Joven
5.
Circ Cardiovasc Genet ; 9(4): 320-9, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27418595

RESUMEN

BACKGROUND: Congenital heart disease (CHD) is the most common type of birth defect with family- and population-based studies supporting a strong genetic cause for CHD. The goal of this study was to determine whether a whole exome sequencing (WES) approach could identify pathogenic-segregating variants in multiplex CHD families. METHODS AND RESULTS: WES was performed on 9 kindreds with familial CHD, 4 with atrial septal defects, 2 with patent ductus arteriosus, 2 with tetralogy of Fallot, and 1 with pulmonary valve dysplasia. Rare variants (<1% minor allele frequency) that segregated with disease were identified by WES, and variants in 69 CHD candidate genes were further analyzed. These selected variants were subjected to in silico analysis to predict pathogenicity and resulted in the discovery of likely pathogenic mutations in 3 of 9 (33%) families. A GATA4 mutation in the transactivation domain, p.G115W, was identified in familial atrial septal defects and demonstrated decreased transactivation ability in vitro. A p.I263V mutation in TLL1 was identified in an atrial septal defects kindred and is predicted to affect the enzymatic functionality of TLL1. A disease-segregating splice donor site mutation in MYH11 (c.4599+1delG) was identified in familial patent ductus arteriosus and found to disrupt normal splicing of MYH11 mRNA in the affected individual. CONCLUSIONS: Our findings demonstrate the clinical utility of WES to identify causative mutations in familial CHD and demonstrate the successful use of a CHD candidate gene list to allow for a more streamlined approach enabling rapid prioritization and identification of likely pathogenic variants from large WES data sets. CLINICAL TRIAL REGISTRATION: URL: https://clinicaltrials.gov; Unique Identifier: NCT0112048.


Asunto(s)
Exoma , Cardiopatías Congénitas/genética , Mutación , Adolescente , Células Cultivadas , Niño , Preescolar , Simulación por Computador , Análisis Mutacional de ADN/métodos , Bases de Datos Genéticas , Conducto Arterioso Permeable/diagnóstico , Conducto Arterioso Permeable/genética , Femenino , Factor de Transcripción GATA4/genética , Frecuencia de los Genes , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/terapia , Defectos del Tabique Interatrial/diagnóstico , Defectos del Tabique Interatrial/genética , Herencia , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Modelos Genéticos , Tasa de Mutación , Cadenas Pesadas de Miosina/genética , Linaje , Fenotipo , Factores de Riesgo , Tetralogía de Fallot/diagnóstico , Tetralogía de Fallot/genética , Metaloproteinasas Similares a Tolloid/genética
6.
Hum Mol Genet ; 25(11): 2331-2341, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26965164

RESUMEN

Congenital heart defects involving left-sided lesions (LSLs) are relatively common birth defects with substantial morbidity and mortality. Previous studies have suggested a high heritability with a complex genetic architecture, such that only a few LSL loci have been identified. We performed a genome-wide case-control association study to address the role of common variants using a discovery cohort of 778 cases and 2756 controls. We identified a genome-wide significant association mapping to a 200 kb region on chromosome 20q11 [P= 1.72 × 10-8 for rs3746446; imputed Single Nucleotide Polymorphism (SNP) rs6088703 P= 3.01 × 10-9, odds ratio (OR)= 1.6 for both]. This result was supported by transmission disequilibrium analyses using a subset of 541 case families (lowest P in region= 4.51 × 10-5, OR= 1.5). Replication in a cohort of 367 LSL cases and 5159 controls showed nominal association (P= 0.03 for rs3746446) resulting in P= 9.49 × 10-9 for rs3746446 upon meta-analysis of the combined cohorts. In addition, a group of seven SNPs on chromosome 1q21.3 met threshold for suggestive association (lowest P= 9.35 × 10-7 for rs12045807). Both regions include genes involved in cardiac development-MYH7B/miR499A on chromosome 20 and CTSK, CTSS and ARNT on chromosome 1. Genome-wide heritability analysis using case-control genotyped SNPs suggested that the mean heritability of LSLs attributable to common variants is moderately high ([Formula: see text] range= 0.26-0.34) and consistent with previous assertions. These results provide evidence for the role of common variation in LSLs, proffer new genes as potential biological candidates, and give further insight to the complex genetic architecture of congenital heart disease.


Asunto(s)
Cromosomas Humanos Par 20/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Cardiopatías Congénitas/genética , Mapeo Cromosómico , Estudios de Cohortes , Femenino , Genotipo , Cardiopatías Congénitas/fisiopatología , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Polimorfismo de Nucleótido Simple
7.
BMC Med Genomics ; 7: 56, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25260786

RESUMEN

BACKGROUND: Bicuspid aortic valve (BAV) is the most common type of congenital heart disease with a population prevalence of 1-2%. While BAV is known to be highly heritable, mutations in single genes (such as GATA5 and NOTCH1) have been reported in few human BAV cases. Traditional gene sequencing methods are time and labor intensive, while next-generation high throughput sequencing remains costly for large patient cohorts and requires extensive bioinformatics processing. Here we describe an approach to targeted multi-gene sequencing with combinatorial pooling of samples from BAV patients. METHODS: We studied a previously described cohort of 78 unrelated subjects with echocardiogram-identified BAV. Subjects were identified as having isolated BAV or BAV associated with coarctation of aorta (BAV-CoA). BAV cusp fusion morphology was defined as right-left cusp fusion, right non-coronary cusp fusion, or left non-coronary cusp fusion. Samples were combined into 19 pools using a uniquely overlapping combinatorial design; a given mutation could be attributed to a single individual on the basis of which pools contained the mutation. A custom gene capture of 97 candidate genes was sequenced on the Illumina HiSeq 2000. Multistep bioinformatics processing was performed for base calling, variant identification, and in-silico analysis of putative disease-causing variants. RESULTS: Targeted capture identified 42 rare, non-synonymous, exonic variants involving 35 of the 97 candidate genes. Among these variants, in-silico analysis classified 33 of these variants as putative disease-causing changes. Sanger sequencing confirmed thirty-one of these variants, found among 16 individuals. There were no significant differences in variant burden among BAV fusion phenotypes or isolated BAV versus BAV-CoA. Pathway analysis suggests a role for the WNT signaling pathway in human BAV. CONCLUSION: We successfully developed a pooling and targeted capture strategy that enabled rapid and cost effective next generation sequencing of target genes in a large patient cohort. This approach identified a large number of putative disease-causing variants in a cohort of patients with BAV, including variants in 26 genes not previously associated with human BAV. The data suggest that BAV heritability is complex and polygenic. Our pooling approach saved over $39,350 compared to an unpooled, targeted capture sequencing strategy.


Asunto(s)
Válvula Aórtica/anomalías , Biología Computacional , Enfermedades de las Válvulas Cardíacas/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Adolescente , Aorta/metabolismo , Enfermedad de la Válvula Aórtica Bicúspide , Femenino , Variación Genética , Humanos , Masculino , Fenotipo
8.
Pediatr Res ; 76(2): 211-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24796370

RESUMEN

BACKGROUND: Bicuspid aortic valve (BAV) is the most common type of congenital heart disease (CHD) and has a proposed genetic etiology. BAV is categorized by cusp fusion, with right-left (R-L) cusp fusion being associated with additional CHD, and right-noncoronary cusp (R-NC) fusion being associated with aortic valve dysfunction. Loss of murine Gata5, which encodes a cardiac transcription factor, results in a partially penetrant R-NC BAV, and we hypothesize that mutations in GATA5 are associated with R-NC BAV in humans. METHODS: A cohort of 78 BAV patients (50 with isolated BAV and 28 with associated aortic coarctation) was analyzed using Sanger sequencing to identify GATA5 sequence variants. Biochemical assays were performed to identify functional deficits of identified sequence variants. RESULTS: We identified two rare heterozygous nonsynonymous variants, p.Gln3Arg and p.Leu233Pro, for a frequency of 2.6% (2/78). Both individuals with nonsynonymous variants had BAV and aortic coarctation, one R-L and one R-NC subtype. Of the nonsynonymous variants, only p.Gln3Arg demonstrated decreased transcriptional activity in vitro. CONCLUSION: Rare sequence variants in GATA5 are associated with human BAV. Our findings suggest a genotype-phenotype correlation in regards to associated CHD but not cusp fusion.


Asunto(s)
Válvula Aórtica/anomalías , Factor de Transcripción GATA5/genética , Variación Genética , Enfermedades de las Válvulas Cardíacas/genética , Fenotipo , Válvula Aórtica/patología , Secuencia de Bases , Enfermedad de la Válvula Aórtica Bicúspide , Estudios de Cohortes , Cartilla de ADN/genética , Femenino , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Immunoblotting , Masculino , Datos de Secuencia Molecular , Mutación Missense/genética , Ohio , Análisis de Secuencia de ADN
9.
Birth Defects Res A Clin Mol Teratol ; 91(3): 162-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21290564

RESUMEN

BACKGROUND: The left ventricular outflow tract (LVOT) defects aortic valve stenosis (AVS), coarctation of the aorta (COA), and hypoplastic left heart syndrome (HLHS) represent an embryologically related group of congenital cardiovascular malformations. They are common and cause substantial morbidity and mortality. Prior evidence suggests a strong genetic component in their causation. METHODS: We selected NRG1, ERBB3, and ERBB4 of the epidermal growth factor receptor (EGFR) signaling pathway as candidate genes for investigation of association with LVOT defects based on the importance of this pathway in cardiac development and the phenotypes in knockout mouse models. Single nucleotide polymorphism (SNP) genotyping was performed on 343 affected case-parent trios of European ancestry. RESULTS: We identified a specific haplotype in intron 3 of ERBB4 that was positively associated with the combined LVOT defects phenotype (p=0.0005) and in each anatomic defect AVS, COA, and HLHS separately. Mutation screening of individuals with an LVOT defect failed to identify a coding sequence or splice site change in ERBB4. RT-PCR on lymphoblastoid cells from LVOT subjects did not show altered splice variant ratios among those homozygous for the associated haplotype. CONCLUSION: These results suggest ERBB4 is associated with LVOT defects. Further replication will be required in separate cohorts to confirm the consistency of the observed association.


Asunto(s)
Receptores ErbB/genética , Polimorfismo de Nucleótido Simple , Obstrucción del Flujo Ventricular Externo/congénito , Obstrucción del Flujo Ventricular Externo/genética , Niño , Estudios de Cohortes , Femenino , Frecuencia de los Genes , Genes erbB/genética , Ligamiento Genético , Predisposición Genética a la Enfermedad , Genotipo , Cardiopatías Congénitas/genética , Humanos , Masculino , Neurregulina-1/genética , Polimorfismo de Nucleótido Simple/fisiología , Isoformas de Proteínas/genética , Receptor ErbB-4
10.
Kidney Int ; 79(1): 120-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20881942

RESUMEN

A novel COL4A5 mutation causes rapid progression to end-stage renal disease in males, despite the absence of clinical and biopsy findings associated with Alport syndrome. Affected males have proteinuria, variable hematuria, and an early progression to end-stage renal disease. Renal biopsy findings include global and segmental glomerulosclerosis, mesangial hypercellularity and basement membrane immune complex deposition. Exon sequencing of the COL4A5 locus identified a thymine to guanine transversion at nucleotide 665, resulting in a phenylalanine to cysteine missense mutation at codon 222. The phenylalanine at position 222 is absolutely conserved among vertebrates. This mutation was confirmed in 4 affected males and 4 female obligate carriers, but was absent in 6 asymptomatic male family members and 198 unrelated individuals. Immunostaining for α5(IV) collagen in renal biopsies from affected males was normal. This mutation, in a non-collagenous interruption associated with severe renal disease, provides evidence for the importance of this structural motif and suggests the range of phenotypes associated with COL4A5 mutations is more diverse than previously realized. Hence, COL4A5 mutation analysis should be considered when glomerulonephritis presents in an X-linked inheritance pattern, even with a presentation distinct from Alport syndrome.


Asunto(s)
Cromosomas Humanos X/genética , Colágeno Tipo IV/genética , Membrana Basal Glomerular/patología , Glomerulonefritis/genética , Fallo Renal Crónico/genética , Mutación Missense , Adolescente , Niño , Colágeno Tipo IV/metabolismo , Femenino , Heterocigoto , Humanos , Fallo Renal Crónico/patología , Masculino , Linaje
11.
Eur J Hum Genet ; 17(6): 811-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19142209

RESUMEN

The left ventricular outflow tract (LVOT) malformations aortic valve stenosis (AVS), coarctation of the aorta (CoA), and hypoplastic left heart syndrome (HLHS) are significant causes of infant mortality. These three malformations are thought to share developmental pathogenetic mechanisms. A strong genetic component has been demonstrated earlier, but the underlying genetic etiologies are unknown. Our objective was to identify genetic susceptibility loci for the broad phenotype of LVOT malformations. We genotyped 411 microsatellites spaced at an average of 10 cM in 43 families constituting 289 individuals, with an additional 5 cM spaced markers for fine mapping. A non-parametric linkage (NPL) analysis of the combined LVOT malformations gave three suggestive linkage peaks on chromosomes 16p12 (NPL score (NPLS)=2.52), 2p23 (NPLS=2.41), and 10q21 (NPLS=2.14). Individually, suggestive peaks for AVS families occurred on chromosomes 16p12 (NPLS=2.64), 7q36 (NPLS=2.31), and 2p25 (NPLS=2.14); and for CoA families on chromosome 1q24 (NPLS=2.61), 6p23 (NPLS=2.29), 7p14 (NPLS=2.27), 10q11 (NPLS=1.98), and 2p15 (NPLS=2.02). Significant NPLS in HLHS families were noted for chromosome 2p15 (NPLS=3.23), with additional suggestive peaks on 19q13 (NPLS=2.16) and 10q21 (NPLS=2.07). Overlapping linkage signals on 10q11 (AVS and CoA) and 16p12 (AVS, CoA, and HLHS) led to higher NPL scores when all malformations were analyzed together. In conclusion, we report suggestive evidence for linkage to chromosomes 2p23, 10q21, and 16p12 for the LVOT malformations of AVS, CoA, and HLHS individually and in a combined analysis, with a significant peak on 2p15 for HLHS. Overlapping linkage peaks provide evidence for a common genetic etiology.


Asunto(s)
Coartación Aórtica/genética , Estenosis de la Válvula Aórtica/genética , Ligamiento Genético , Síndrome del Corazón Izquierdo Hipoplásico/genética , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Obstrucción del Flujo Ventricular Externo/genética
12.
Hum Mol Genet ; 17(18): 2886-93, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18593716

RESUMEN

Congenital aortic valve stenosis (AVS), coarctation of the aorta (COA) and hypoplastic left heart syndrome (HLHS) are congenital cardiovascular malformations that all involve the left ventricular outflow tract (LVOT). They are presumably caused by a similar developmental mechanism involving the developing endothelium. The exact etiology for most LVOT malformations is unknown, but a strong genetic component has been established. We demonstrate here that mutations in the gene NOTCH1, coding for a receptor in a developmentally important signaling pathway, are found across the spectrum of LVOT defects. We identify two specific mutations that reduce ligand (JAGGED1) induced NOTCH1 signaling. One of these mutations perturbs the S1 cleavage of the receptor in the Golgi. These findings suggest that the levels of NOTCH1 signaling are tightly regulated during cardiovascular development, and that relatively minor alterations may promote LVOT defects. These results also establish for the first time that AVS, COA and HLHS can share a common pathogenetic mechanism at the molecular level, explaining observations of these defects co-occurring within families.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Ventrículos Cardíacos/anomalías , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Mutación Missense , Receptor Notch1/genética , Transducción de Señal , Obstrucción del Flujo Ventricular Externo/genética , Obstrucción del Flujo Ventricular Externo/fisiopatología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Ventrículos Cardíacos/fisiopatología , Humanos , Proteína Jagged-1 , Ligandos , Masculino , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Receptor Notch1/química , Receptor Notch1/metabolismo , Alineación de Secuencia , Proteínas Serrate-Jagged , Obstrucción del Flujo Ventricular Externo/congénito , Población Blanca/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...