Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 110(5): 846-862, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37086723

RESUMEN

Craniosynostosis (CS) is the most common congenital cranial anomaly. Several Mendelian forms of syndromic CS are well described, but a genetic etiology remains elusive in a substantial fraction of probands. Analysis of exome sequence data from 526 proband-parent trios with syndromic CS identified a marked excess (observed 98, expected 33, p = 4.83 × 10-20) of damaging de novo variants (DNVs) in genes highly intolerant to loss-of-function variation (probability of LoF intolerance > 0.9). 30 probands harbored damaging DNVs in 21 genes that were not previously implicated in CS but are involved in chromatin modification and remodeling (4.7-fold enrichment, p = 1.1 × 10-11). 17 genes had multiple damaging DNVs, and 13 genes (CDK13, NFIX, ADNP, KMT5B, SON, ARID1B, CASK, CHD7, MED13L, PSMD12, POLR2A, CHD3, and SETBP1) surpassed thresholds for genome-wide significance. A recurrent gain-of-function DNV in the retinoic acid receptor alpha (RARA; c.865G>A [p.Gly289Arg]) was identified in two probands with similar CS phenotypes. CS risk genes overlap with those identified for autism and other neurodevelopmental disorders, are highly expressed in cranial neural crest cells, and converge in networks that regulate chromatin modification, gene transcription, and osteoblast differentiation. Our results identify several CS loci and have major implications for genetic testing and counseling.


Asunto(s)
Craneosinostosis , Tretinoina , Humanos , Mutación , Craneosinostosis/genética , Regulación de la Expresión Génica , Cromatina , Predisposición Genética a la Enfermedad
2.
Brain ; 147(4): 1553-1570, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38128548

RESUMEN

Hydrocephalus, characterized by cerebral ventriculomegaly, is the most common disorder requiring brain surgery in children. Recent studies have implicated SMARCC1, a component of the BRG1-associated factor (BAF) chromatin remodelling complex, as a candidate congenital hydrocephalus gene. However, SMARCC1 variants have not been systematically examined in a large patient cohort or conclusively linked with a human syndrome. Moreover, congenital hydrocephalus-associated SMARCC1 variants have not been functionally validated or mechanistically studied in vivo. Here, we aimed to assess the prevalence of SMARCC1 variants in an expanded patient cohort, describe associated clinical and radiographic phenotypes, and assess the impact of Smarcc1 depletion in a novel Xenopus tropicalis model of congenital hydrocephalus. To do this, we performed a genetic association study using whole-exome sequencing from a cohort consisting of 2697 total ventriculomegalic trios, including patients with neurosurgically-treated congenital hydrocephalus, that total 8091 exomes collected over 7 years (2016-23). A comparison control cohort consisted of 1798 exomes from unaffected siblings of patients with autism spectrum disorder and their unaffected parents were sourced from the Simons Simplex Collection. Enrichment and impact on protein structure were assessed in identified variants. Effects on the human fetal brain transcriptome were examined with RNA-sequencing and Smarcc1 knockdowns were generated in Xenopus and studied using optical coherence tomography imaging, in situ hybridization and immunofluorescence. SMARCC1 surpassed genome-wide significance thresholds, yielding six rare, protein-altering de novo variants localized to highly conserved residues in key functional domains. Patients exhibited hydrocephalus with aqueductal stenosis; corpus callosum abnormalities, developmental delay, and cardiac defects were also common. Xenopus knockdowns recapitulated both aqueductal stenosis and cardiac defects and were rescued by wild-type but not patient-specific variant SMARCC1. Hydrocephalic SMARCC1-variant human fetal brain and Smarcc1-variant Xenopus brain exhibited a similarly altered expression of key genes linked to midgestational neurogenesis, including the transcription factors NEUROD2 and MAB21L2. These results suggest de novo variants in SMARCC1 cause a novel human BAFopathy we term 'SMARCC1-associated developmental dysgenesis syndrome', characterized by variable presence of cerebral ventriculomegaly, aqueductal stenosis, developmental delay and a variety of structural brain or cardiac defects. These data underscore the importance of SMARCC1 and the BAF chromatin remodelling complex for human brain morphogenesis and provide evidence for a 'neural stem cell' paradigm of congenital hydrocephalus pathogenesis. These results highlight utility of trio-based whole-exome sequencing for identifying pathogenic variants in sporadic congenital structural brain disorders and suggest whole-exome sequencing may be a valuable adjunct in clinical management of congenital hydrocephalus patients.


Asunto(s)
Trastorno del Espectro Autista , Acueducto del Mesencéfalo/anomalías , Enfermedades Genéticas Ligadas al Cromosoma X , Hidrocefalia , Niño , Humanos , Trastorno del Espectro Autista/genética , Factores de Transcripción/genética , Hidrocefalia/diagnóstico por imagen , Hidrocefalia/genética , Epigénesis Genética , Proteínas del Ojo/genética , Péptidos y Proteínas de Señalización Intracelular/genética
3.
Am J Med Genet A ; 194(4): e63477, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37969032

RESUMEN

Germline pathogenic variants in the RAS/mitogen-activated protein kinase (MAPK) signaling pathway are the molecular cause of RASopathies, a group of clinically overlapping genetic syndromes. RASopathies constitute a wide clinical spectrum characterized by distinct facial features, short stature, predisposition to cancer, and variable anomalies in nearly all the major body systems. With increasing global recognition of these conditions, the 8th International RASopathies Symposium spotlighted global perspectives on clinical care and research, including strategies for building international collaborations and developing diverse patient cohorts in anticipation of interventional trials. This biannual meeting, organized by RASopathies Network, was held in a hybrid virtual/in-person format. The agenda featured emerging discoveries and case findings as well as progress in preclinical and therapeutic pipelines. Stakeholders including basic scientists, clinician-scientists, practitioners, industry representatives, patients, and family advocates gathered to discuss cutting edge science, recognize current gaps in knowledge, and hear from people with RASopathies about the experience of daily living. Presentations by RASopathy self-advocates and early-stage investigators were featured throughout the program to encourage a sustainable, diverse, long-term research and advocacy partnership focused on improving health and bringing treatments to people with RASopathies.


Asunto(s)
Síndrome de Costello , Displasia Ectodérmica , Cardiopatías Congénitas , Neoplasias , Síndrome de Noonan , Humanos , Proteínas ras/genética , Sistema de Señalización de MAP Quinasas/genética , Síndrome de Costello/genética , Neoplasias/genética , Displasia Ectodérmica/genética , Síndrome de Noonan/genética , Cardiopatías Congénitas/genética
4.
Am J Med Genet C Semin Med Genet ; 193(3): e32059, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37534870

RESUMEN

Facial analysis technology in rare diseases has the potential to shorten the diagnostic odyssey by providing physicians with a valuable diagnostic tool. Given that most clinical genetic resources focus on populations of European descent, we compare craniofacial features in genetic syndromes across different populations and review how machine learning algorithms perform on diagnosing genetic syndromes in geographically and ethnically diverse populations. We also discuss the value of populations from ancestrally diverse backgrounds in the training set of machine learning algorithms. Finally, this review demonstrates that across diverse population groups, machine learning models have outstanding accuracy as supported by the area under the curve values greater than 0.9. Artificial intelligence is only in its infancy in the diagnosis of rare disease in diverse populations and will become more accurate as larger and more diverse training sets, including a wider spectrum of ages, particularly infants, are studied.


Asunto(s)
Inteligencia Artificial , Grupos de Población , Humanos , Algoritmos , Aprendizaje Automático , Tecnología
5.
Am J Hum Genet ; 106(1): 121-128, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31883643

RESUMEN

In two independent ongoing next-generation sequencing projects for individuals with holoprosencephaly and individuals with disorders of sex development, and through international research collaboration, we identified twelve individuals with de novo loss-of-function (LoF) variants in protein phosphatase 1, regulatory subunit 12a (PPP1R12A), an important developmental gene involved in cell migration, adhesion, and morphogenesis. This gene has not been previously reported in association with human disease, and it has intolerance to LoF as illustrated by a very low observed-to-expected ratio of LoF variants in gnomAD. Of the twelve individuals, midline brain malformations were found in five, urogenital anomalies in nine, and a combination of both phenotypes in two. Other congenital anomalies identified included omphalocele, jejunal, and ileal atresia with aberrant mesenteric blood supply, and syndactyly. Six individuals had stop gain variants, five had a deletion or duplication resulting in a frameshift, and one had a canonical splice acceptor site loss. Murine and human in situ hybridization and immunostaining revealed PPP1R12A expression in the prosencephalic neural folds and protein localization in the lower urinary tract at critical periods for forebrain division and urogenital development. Based on these clinical and molecular findings, we propose the association of PPP1R12A pathogenic variants with a congenital malformations syndrome affecting the embryogenesis of the brain and genitourinary systems and including disorders of sex development.


Asunto(s)
Anomalías Múltiples/patología , Trastornos del Desarrollo Sexual/patología , Holoprosencefalia/patología , Mutación , Fosfatasa de Miosina de Cadena Ligera/genética , Anomalías Urogenitales/patología , Anomalías Múltiples/genética , Adolescente , Niño , Preescolar , Trastornos del Desarrollo Sexual/genética , Femenino , Edad Gestacional , Holoprosencefalia/genética , Humanos , Masculino , Fenotipo , Embarazo , Anomalías Urogenitales/genética
6.
Am J Hum Genet ; 107(1): 164-172, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32553196

RESUMEN

CNOT1 is a member of the CCR4-NOT complex, which is a master regulator, orchestrating gene expression, RNA deadenylation, and protein ubiquitination. We report on 39 individuals with heterozygous de novo CNOT1 variants, including missense, splice site, and nonsense variants, who present with a clinical spectrum of intellectual disability, motor delay, speech delay, seizures, hypotonia, and behavioral problems. To link CNOT1 dysfunction to the neurodevelopmental phenotype observed, we generated variant-specific Drosophila models, which showed learning and memory defects upon CNOT1 knockdown. Introduction of human wild-type CNOT1 was able to rescue this phenotype, whereas mutants could not or only partially, supporting our hypothesis that CNOT1 impairment results in neurodevelopmental delay. Furthermore, the genetic interaction with autism-spectrum genes, such as ASH1L, DYRK1A, MED13, and SHANK3, was impaired in our Drosophila models. Molecular characterization of CNOT1 variants revealed normal CNOT1 expression levels, with both mutant and wild-type alleles expressed at similar levels. Analysis of protein-protein interactions with other members indicated that the CCR4-NOT complex remained intact. An integrated omics approach of patient-derived genomics and transcriptomics data suggested only minimal effects on endonucleolytic nonsense-mediated mRNA decay components, suggesting that de novo CNOT1 variants are likely haploinsufficient hypomorph or neomorph, rather than dominant negative. In summary, we provide strong evidence that de novo CNOT1 variants cause neurodevelopmental delay with a wide range of additional co-morbidities. Whereas the underlying pathophysiological mechanism warrants further analysis, our data demonstrate an essential and central role of the CCR4-NOT complex in human brain development.


Asunto(s)
Discapacidades del Desarrollo/genética , Expresión Génica/genética , Trastornos del Neurodesarrollo/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , ARN/genética , Receptores CCR4/genética , Factores de Transcripción/genética , Alelos , Femenino , Variación Genética/genética , Haploinsuficiencia/genética , Heterocigoto , Humanos , Masculino , Malformaciones del Sistema Nervioso/genética , Fenotipo , Estabilidad Proteica
7.
Genet Med ; 25(12): 100947, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37534744

RESUMEN

PURPOSE: Variants of uncertain significance (VUS) are a common result of diagnostic genetic testing and can be difficult to manage with potential misinterpretation and downstream costs, including time investment by clinicians. We investigated the rate of VUS reported on diagnostic testing via multi-gene panels (MGPs) and exome and genome sequencing (ES/GS) to measure the magnitude of uncertain results and explore ways to reduce their potentially detrimental impact. METHODS: Rates of inconclusive results due to VUS were collected from over 1.5 million sequencing test results from 19 clinical laboratories in North America from 2020 to 2021. RESULTS: We found a lower rate of inconclusive test results due to VUSs from ES/GS (22.5%) compared with MGPs (32.6%; P < .0001). For MGPs, the rate of inconclusive results correlated with panel size. The use of trios reduced inconclusive rates (18.9% vs 27.6%; P < .0001), whereas the use of GS compared with ES had no impact (22.2% vs 22.6%; P = ns). CONCLUSION: The high rate of VUS observed in diagnostic MGP testing warrants examining current variant reporting practices. We propose several approaches to reduce reported VUS rates, while directing clinician resources toward important VUS follow-up.


Asunto(s)
Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Pruebas Genéticas/métodos , Genómica , Exoma/genética , América del Norte
8.
Am J Med Genet A ; 191(1): 271-274, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36301051

RESUMEN

Hajdu-Cheney syndrome is an ultra-rare autosomal dominant disorder caused by a heterozygous variant in NOTCH2 gene. Characteristic features include osteolysis, distinct facial appearance, skull deformity, joint laxity, osteoporosis, and short stature. Associated abnormalities are congenital heart disease, congenital defects of the kidney, and neurological problems. Here, we present the first reported case of an African child with a variant in NOTCH2 gene and features of Hajdu-Cheney syndrome in whom we detected a congenital heart defect that has not been previously reported in association with the syndrome. To appropriately characterize this disease and document correct proportion of cardiovascular malformation associations, echocardiography is recommended for all cases of Hajdu Cheney syndrome.


Asunto(s)
Anomalías Cardiovasculares , Síndrome de Hajdu-Cheney , Osteoporosis , Niño , Humanos , Síndrome de Hajdu-Cheney/diagnóstico , Síndrome de Hajdu-Cheney/genética , Receptor Notch2/genética , Osteoporosis/genética , Heterocigoto , Anomalías Cardiovasculares/complicaciones , Anomalías Cardiovasculares/diagnóstico , Anomalías Cardiovasculares/genética
9.
Am J Med Genet A ; 191(9): 2411-2415, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37313780

RESUMEN

Neurofibromatosis type 1 is an autosomal dominant multisystemic disease caused by mutation of the neurofibromin (NF1) gene located on chromosome 17q11. We report a case of Neurofibromatosis 1 with ambiguous genitalia, giant congenital melanocytic nevus, and associated subpulmonic outlet ventricular septal defect, hitherto unreported in sub-Saharan Africa. In addition, a literature review of congenital heart diseases associated with Neurofibromatosis 1 is presented.


Asunto(s)
Trastornos del Desarrollo Sexual , Defectos del Tabique Interventricular , Neurofibromatosis 1 , Nevo Pigmentado , Humanos , Niño , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/diagnóstico , Neurofibromatosis 1/genética , Nevo Pigmentado/diagnóstico , Nevo Pigmentado/genética , Nevo Pigmentado/congénito
10.
Proc Natl Acad Sci U S A ; 117(28): 16481-16491, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32601182

RESUMEN

Differences between female and male immunity may contribute to variations in response to infections and predisposition to autoimmunity. We previously reported that neutrophils from reproductive-age males are more immature and less activated than their female counterparts. To further characterize the mechanisms that drive differential neutrophil phenotypes, we performed RNA sequencing on circulating neutrophils from healthy adult females and males. Female neutrophils displayed significant up-regulation of type I IFN (IFN)-stimulated genes (ISGs). Single-cell RNA-sequencing analysis indicated that these differences are neutrophil specific, driven by a distinct neutrophil subset and related to maturation status. Neutrophil hyperresponsiveness to type I IFNs promoted enhanced responses to Toll-like receptor agonists. Neutrophils from young adult males had significantly increased mitochondrial metabolism compared to those from females and this was modulated by estradiol. Assessment of ISGs and neutrophil maturation genes in Klinefelter syndrome (47, XXY) males and in prepubescent children supported that differences in neutrophil phenotype between adult male and female neutrophils are hormonally driven and not explained by X chromosome gene dosage. Our results indicate that there are distinct sex differences in neutrophil biology related to responses to type I IFNs, immunometabolism, and maturation status that may have prominent functional and pathogenic implications.


Asunto(s)
Interferón Tipo I/inmunología , Neutrófilos/inmunología , Adulto , Femenino , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Síndrome de Klinefelter/genética , Síndrome de Klinefelter/inmunología , Síndrome de Klinefelter/metabolismo , Masculino , Factores Sexuales , Adulto Joven
11.
Am J Hum Genet ; 104(5): 990-993, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31006510

RESUMEN

Holoprosencephaly is the incomplete separation of the forebrain during embryogenesis. Both genetic and environmental etiologies have been determined for holoprosencephaly; however, a genetic etiology is not found in most cases. In this report, we present two unrelated individuals with semilobar holoprosencephaly who have the identical de novo missense variant in the gene CCR4-NOT transcription complex, subunit 1 (CNOT1). The variant (c.1603C>T [p.Arg535Cys]) is predicted to be deleterious and is not present in public databases. CNOT1 has not been previously associated with holoprosencephaly or other brain malformations. In situ hybridization analyses of mouse embryos show that Cnot1 is expressed in the prosencephalic neural folds at gestational day 8.25 during the critical period for subsequent forebrain division. Combining human and mouse data, we show that CNOT1 is associated with incomplete forebrain division.


Asunto(s)
Holoprosencefalia/genética , Holoprosencefalia/patología , Mutación Missense , Prosencéfalo/anomalías , Factores de Transcripción/genética , Animales , Niño , Femenino , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Prosencéfalo/metabolismo
12.
Am J Med Genet A ; 188(10): 3121-3125, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35860951

RESUMEN

POLE is a pleiotropic gene with phenotypic expression of pathogenic variants depending on the type of variant, impact on the protein, and mode of inheritance. Heterozygous missense variants located within the exonuclease domain have been shown to result in polymerase proofreading-associated polyposis (PPAP) which is characterized by an increased risk for colon polyps and colorectal cancer. Biallelic variants resulting in markedly reduced amounts of normal protein have been reported in two separate recessive pediatric syndromes: facial dysmorphism, immunodeficiency, livedo, and short stature as well as intrauterine growth restriction, metaphyseal dysplasia, adrenal hypoplasia congenital, and genital anomalies. Here we report two siblings identified to have POLE c.1686 + 32C > G in trans with POLE p.(Glu709*) via exome sequencing. A detailed review of the reported phenotypes in these two siblings and from available literature revealed that individuals with biallelic POLE pathogenic variants resulting in partial loss-of-function present with a similar phenotype: short stature and facial dysmorphism with or without immunodeficiency. These data suggest a phenotypic continuum between the previously reported POLE-related recessive disorders.


Asunto(s)
Enanismo , Anomalías Musculoesqueléticas , Osteocondrodisplasias , Enanismo/diagnóstico , Enanismo/genética , Humanos , Mutación Missense , Osteocondrodisplasias/genética , Fenotipo , Secuenciación del Exoma
13.
Curr Opin Pediatr ; 34(4): 447-460, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35796656

RESUMEN

PURPOSE OF REVIEW: Turner syndrome is the most common sex chromosome abnormality in female individuals, affecting 1/2000-1/2500 female newborns. Despite the high incidence of this condition, the mechanisms underlying the development of multiorgan dysfunction have not been elucidated. RECENT FINDINGS: Clinical features involve multiple organ systems and include short stature, dysmorphic facial features, delayed puberty and gonadal failure, cardiac and renal abnormalities, audiologic abnormalities, and a high prevalence of endocrine and autoimmune disorders. Paucity of available genotype/phenotype correlation limits the ability of clinicians to provide accurate guidance and management. Given the advent of robust genetic testing and analysis platforms, developments in the genetic basis of disease are materializing at a rapid pace. SUMMARY: The objective of this review is to highlight the recent advances in knowledge and to provide a framework with which to apply new data to the foundational understanding of the condition.


Asunto(s)
Enanismo , Síndrome de Turner , Femenino , Humanos , Síndrome de Turner/diagnóstico , Síndrome de Turner/epidemiología , Síndrome de Turner/genética
14.
Genet Med ; 23(10): 1952-1960, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34113005

RESUMEN

PURPOSE: Rare genetic variants in KDR, encoding the vascular endothelial growth factor receptor 2 (VEGFR2), have been reported in patients with tetralogy of Fallot (TOF). However, their role in disease causality and pathogenesis remains unclear. METHODS: We conducted exome sequencing in a familial case of TOF and large-scale genetic studies, including burden testing, in >1,500 patients with TOF. We studied gene-targeted mice and conducted cell-based assays to explore the role of KDR genetic variation in the etiology of TOF. RESULTS: Exome sequencing in a family with two siblings affected by TOF revealed biallelic missense variants in KDR. Studies in knock-in mice and in HEK 293T cells identified embryonic lethality for one variant when occurring in the homozygous state, and a significantly reduced VEGFR2 phosphorylation for both variants. Rare variant burden analysis conducted in a set of 1,569 patients of European descent with TOF identified a 46-fold enrichment of protein-truncating variants (PTVs) in TOF cases compared to controls (P = 7 × 10-11). CONCLUSION: Rare KDR variants, in particular PTVs, strongly associate with TOF, likely in the setting of different inheritance patterns. Supported by genetic and in vivo and in vitro functional analysis, we propose loss-of-function of VEGFR2 as one of the mechanisms involved in the pathogenesis of TOF.


Asunto(s)
Tetralogía de Fallot , Receptor 2 de Factores de Crecimiento Endotelial Vascular , Animales , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Ratones , Tetralogía de Fallot/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Secuenciación del Exoma
15.
Prenat Diagn ; 41(6): 778-790, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33522008

RESUMEN

OBJECTIVE: Report a single-center 12-year experience in the fetal diagnosis of diencephalic-mesencephalic junction dysplasia (DMJD) to expand the phenotype with Magnetic resonance imaging (MRI)-based classification, evaluate genetic etiologies, and ascertain outcomes. METHODS: Retrospective medical record and imaging review of all fetal MRI exams with DMJD were performed at our institution. RESULTS: Thirty-three pregnancies with fetal MRI findings of DMJD at 24 (18-37) weeks gestational age were studied; 70% were referred for fetal hydrocephalus. Three fetal MRI patterns were recognized. Type A (butterfly/hypothalamus-midbrain union) was seen in two cases (6%), Type B (partial thalamus-midbrain union) in 22 fetuses (70%), and Type C (complete/near complete midbrain-thalamic continuity) in nine fetuses (24%). L1CAM mutations were identified in four cases, and biallelic VRK1 variants in another. Among 14 live-born cases, 11 survived infancy, and 10 underwent postnatal brain MRI which confirmed the fetal MRI diagnosis in all but one case. Development was delayed in all surviving infants, most with additional neurological sequelae. CONCLUSIONS: DMJD may be identified by prenatal MRI as early as 18 weeks gestation. We propose three distinct phenotypic forms of DMJD, Types A-C. Next-generation sequencing provides an underlying molecular diagnosis in some patients, but further studies on associated genetic diagnoses and clinical outcomes are indicated.


Asunto(s)
Feto/anomalías , Enfermedades Genéticas Congénitas/diagnóstico , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Adulto , Femenino , Feto/diagnóstico por imagen , Enfermedades Genéticas Congénitas/epidemiología , Edad Gestacional , Humanos , Imagen por Resonancia Magnética/métodos , Evaluación de Resultado en la Atención de Salud/métodos , Fenotipo , Embarazo , Diagnóstico Prenatal/métodos , Diagnóstico Prenatal/estadística & datos numéricos , Estudios Retrospectivos
16.
Am J Med Genet C Semin Med Genet ; 184(1): 5-6, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32083375

RESUMEN

In this special issue of the American Journal of Medical Genetics Part C, we focus on the "State of Congenital Heart Disease." We anticipate that after viewing this journal, the reader will be up-to-date on the epidemiology of congenital heart disease (CHD), the genetic basis of CHD, ethical concerns, and the global impact of CHD. And most importantly, we are confident that this special issue conveys the message that CHD is complex and that much work is still needed in genetic and genomic research.


Asunto(s)
Genómica , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/epidemiología , Humanos
17.
Am J Med Genet C Semin Med Genet ; 184(1): 178-186, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31833661

RESUMEN

Congenital heart disease (CHD) is the most prevalent birth defect and is the result of multiple etiologies including genetic and environmental causes. This article reviews the genetic workup for structural CHD in the clinical setting, beginning with CHD epidemiology and etiology and then moving to genetic testing, clinical evaluation, and genetic counseling. An algorithm is presented as a guide to genetic test selection, and available tests are explained with their respective advantages and limitations. Finally, future advances are discussed. As this review focuses on structural heart disease, isolated cardiomyopathies, inherited primary arrhythmia syndromes and aortopathies are not discussed.


Asunto(s)
Cardiomiopatías/genética , Asesoramiento Genético , Cardiopatías Congénitas/genética , Algoritmos , Pruebas Genéticas , Cardiopatías Congénitas/patología , Humanos
18.
Am J Med Genet C Semin Med Genet ; 184(1): 154-158, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32022405

RESUMEN

Comorbidity of holoprosencephaly (HPE) and congenital heart disease (CHD) in individuals with genetic variants in known HPE-related genes has been recurrently observed. Morphogenesis of the brain and heart from very early stages are regulated by several biological pathways, some of them involved in both heart and brain development as evidenced by genetic studies on model organisms. For instance, downregulation of Hedgehog or Nodal signaling pathways, both known as major triggers of HPE, has been shown to play a role in the pathogenesis of CHD, including structural defects and left-right asymmetry defects. In this study, individuals with various types of HPE were investigated clinically and by genomic sequencing. Cardiac phenotypes were assessed in 434 individuals with HPE who underwent targeted sequencing. CHDs were identified in 8% (n = 33) of individuals, including 10 (30%) cases of complex heart disease. Only four individuals (4/33) had damaging variants in the known HPE genes STAG2, SIX3, and SHH. Interestingly, no CHD was identified in the 37 individuals of our cohort with pathogenic variants in ZIC2. These findings suggest that CHD occurs more frequently in HPE-affected individuals with or without identifiable genetic variants, and this co-occurrence may be genetically driven and gene-specific.


Asunto(s)
Cardiopatías Congénitas/genética , Holoprosencefalia/genética , Proteínas de Homeodominio/genética , Adolescente , Adulto , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Ciclo Celular/genética , Niño , Preescolar , Comorbilidad , Proteínas del Ojo/genética , Femenino , Cardiopatías Congénitas/patología , Holoprosencefalia/patología , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Fenotipo , Factores de Transcripción/genética , Adulto Joven , Proteína Homeobox SIX3
19.
Am J Med Genet C Semin Med Genet ; 184(1): 124-128, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32030882

RESUMEN

Sudden cardiac death (SCD) is one of the leading causes of mortality in the U.S. military and competitive athletes. In this study, we simulate how genetic screening may be implemented in the military to prevent an SCD endpoint resulting from hypertrophic cardiomyopathy (HCM). We created a logistic regression model to predict variant pathogenicity in the most common HCM associated genes MYH7 and MYBPC3. Model predictions were used in conjunction with the gnomAD database to identify frequencies of pathogenic variants. Extrapolating these variants to a military population, lives saved and cost benefit analyses were conducted for screening for HCM related to pathogenic variants in MYH7 and MYBPC3. Genetic screening for HCM followed by echocardiography in individuals with pathogenic variants is predicted to save an average of 2.9 lives per accession cohort, based on historical cohort sizes, and result in a break-even cost of ~$7 per test. The false positives, defined as disqualified individuals for military service who do not have HCM, are predicted to be 0 individuals per accession cohort. This study suggests that the main barriers for the implementation of genetic screening for the U.S. military are the low detection rate and variant interpretation.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatía Hipertrófica/genética , Proteínas Portadoras/genética , Muerte Súbita Cardíaca/prevención & control , Cadenas Pesadas de Miosina/genética , Adulto , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/epidemiología , Muerte Súbita Cardíaca/epidemiología , Ecocardiografía/métodos , Femenino , Pruebas Genéticas , Genética de Población , Humanos , Masculino , Medicina Militar , Personal Militar , Mutación/genética , Linaje , Fenotipo
20.
Am J Med Genet C Semin Med Genet ; 184(1): 47-52, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32052942

RESUMEN

Congenital heart disease (CHD) in low-and-middle income countries (LMIC) is often characterized by late presentation resulting from inadequate screening and healthcare access in these regions. Accurate estimates of the burden of CHD among school children are often lacking. The objective of this study was to determine the prevalence and distribution of CHD among school children in two communities (urban and semi-urban) in south western Nigeria. Using clinical assessment and portable echocardiography, 4107 school children aged 5 years to 16 years in Lagos, Nigeria, were selected using a multistage sampling procedure and screened for CHD. Diagnosis of CHD was made after echocardiography. Children identified with CHD were referred to a tertiary hospital for appropriate cardiac care. The 4,107 children screened had a mean age of 11.3 ± 2.7 years and 53.7% were females. Twenty seven children had echocardiography-confirmed CHD, representing a prevalence of CHD among school children in Lagos, Nigeria of 6.6 per 1000 children. Acyanotic CHD constituted 96.3% of detected cases. Two children diagnosed with CHD (Tetralogy of Fallot and severe pulmonary valve stenosis respectively) had successful intervention. The prevalence of previously undiagnosed CHD among school children in Lagos Nigeria is substantial and highlights gaps in the health care system and school health programs. Echocardiographic screening of school children provides an opportunity for missed early diagnosis and treatment of CHD and reduces the prevalence of first-diagnosed CHD in adulthood. Therefore, focused clinical examination of school children followed by echocardiography is a strategy that could bridge this diagnostic and treatment gap in CHD.


Asunto(s)
Cardiopatías Congénitas/epidemiología , Estenosis de la Válvula Pulmonar/diagnóstico , Tetralogía de Fallot/diagnóstico , Adolescente , Adulto , Niño , Preescolar , Ecocardiografía , Femenino , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/patología , Humanos , Masculino , Nigeria/epidemiología , Estenosis de la Válvula Pulmonar/epidemiología , Estenosis de la Válvula Pulmonar/patología , Instituciones Académicas , Tetralogía de Fallot/epidemiología , Tetralogía de Fallot/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA