Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Am J Med Genet A ; 194(5): e63516, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38168088

RESUMEN

The NFIX gene encodes a DNA-binding protein belonging to the nuclear factor one (NFI) family of transcription factors. Pathogenic variants of NFIX are associated with two autosomal dominant Mendelian disorders, Malan syndrome (MIM 614753) and Marshall-Smith syndrome (MIM 602535), which are clinically distinct due to different disease-causing mechanisms. NFIX variants associated with Malan syndrome are missense variants mostly located in exon 2 encoding the N-terminal DNA binding and dimerization domain or are protein-truncating variants that trigger nonsense-mediated mRNA decay (NMD) resulting in NFIX haploinsufficiency. NFIX variants associated with Marshall-Smith syndrome are protein-truncating and are clustered between exons 6 and 10, including a recurrent Alu-mediated deletion of exons 6 and 7, which can escape NMD. The more severe phenotype of Marshall-Smith syndrome is likely due to a dominant-negative effect of these protein-truncating variants that escape NMD. Here, we report a child with clinical features of Malan syndrome who has a de novo NFIX intragenic duplication. Using genome sequencing, exon-level microarray analysis, and RNA sequencing, we show that this duplication encompasses exons 6 and 7 and leads to NFIX haploinsufficiency. To our knowledge, this is the first reported case of Malan Syndrome caused by an intragenic NFIX duplication.


Asunto(s)
Anomalías Múltiples , Enfermedades del Desarrollo Óseo , Anomalías Craneofaciales , Discapacidad Intelectual , Megalencefalia , Displasia Septo-Óptica , Síndrome de Sotos , Niño , Humanos , Factores de Transcripción NFI/genética , Síndrome de Sotos/genética , Exones/genética , Megalencefalia/genética , Discapacidad Intelectual/genética , Análisis de Secuencia de ARN
2.
Pediatr Res ; 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38969815

RESUMEN

BACKGROUND: Despite advances in treatment and survival, individuals with congenital heart defects (CHD) have a higher risk of heart failure (HF) compared to the general population. OBJECTIVE: To evaluate comorbidities associated with HF in patients with CHD with a goal of identifying potentially modifiable risk factors that may reduce HF-associated morbidity and mortality. METHODS: Five surveillance sites in the United States linked population-based healthcare data and vital records. Individuals with an ICD-9-CM code for CHD aged 11-64 years were included and were stratified by presence of HF diagnosis code. Prevalence of death and cardiovascular risk factors based on diagnosis codes were compared by HF status using log-linear regression. RESULTS: A total of 25,343 individuals met inclusion/exclusion criteria. HF was documented for 2.2% of adolescents and 12.9% of adults with CHD. Adolescents and adults with HF had a higher mortality than those without HF. In both age groups, HF was positively associated with coronary artery disease, hypertension, obesity, diabetes, and increased healthcare utilization compared to those without HF. CONCLUSIONS: Within this population-based cohort, over 1 in 50 adolescents and 1 in 8 adults with CHD had HF, which was associated with increased mortality. Modifiable cardiovascular comorbidities were associated with HF. IMPACT: Five sites in the United States linked population-based healthcare data and vital records to establish surveillance network for identifying the factors which influence congenital heart disease (CHD) outcomes. Survivors of CHD frequently develop heart failure across the lifespan. Over 1 in 50 adolescent and 1 in 8 adult survivors of CHD have heart failure which is associated with increased mortality compared to CHD survivors without heart failure. Heart failure development is associated with potentially modifiable cardiovascular risk factors such as hypertension, coronary artery disease, and diabetes. Controlling modifiable cardiovascular risk factors may serve to lower the risk of heart failure and mortality in survivors of congenital heart disease of all ages.

3.
Brain ; 146(4): 1357-1372, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-36074901

RESUMEN

The vacuolar H+-ATPase is an enzymatic complex that functions in an ATP-dependent manner to pump protons across membranes and acidify organelles, thereby creating the proton/pH gradient required for membrane trafficking by several different types of transporters. We describe heterozygous point variants in ATP6V0C, encoding the c-subunit in the membrane bound integral domain of the vacuolar H+-ATPase, in 27 patients with neurodevelopmental abnormalities with or without epilepsy. Corpus callosum hypoplasia and cardiac abnormalities were also present in some patients. In silico modelling suggested that the patient variants interfere with the interactions between the ATP6V0C and ATP6V0A subunits during ATP hydrolysis. Consistent with decreased vacuolar H+-ATPase activity, functional analyses conducted in Saccharomyces cerevisiae revealed reduced LysoSensor fluorescence and reduced growth in media containing varying concentrations of CaCl2. Knockdown of ATP6V0C in Drosophila resulted in increased duration of seizure-like behaviour, and the expression of selected patient variants in Caenorhabditis elegans led to reduced growth, motor dysfunction and reduced lifespan. In summary, this study establishes ATP6V0C as an important disease gene, describes the clinical features of the associated neurodevelopmental disorder and provides insight into disease mechanisms.


Asunto(s)
Epilepsia , ATPasas de Translocación de Protón Vacuolares , Humanos , ATPasas de Translocación de Protón Vacuolares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Epilepsia/genética , Adenosina Trifosfato
4.
PLoS Genet ; 17(3): e1009413, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33684136

RESUMEN

Previous research on risk factors for obstructive heart defects (OHDs) focused on maternal and infant genetic variants, prenatal environmental exposures, and their potential interaction effects. Less is known about the role of paternal genetic variants or environmental exposures and risk of OHDs. We examined parent-of-origin effects in transmission of alleles in the folate, homocysteine, or transsulfuration pathway genes on OHD occurrence in offspring. We used data on 569 families of liveborn infants with OHDs born between October 1997 and August 2008 from the National Birth Defects Prevention Study to conduct a family-based case-only study. Maternal, paternal, and infant DNA were genotyped using an Illumina Golden Gate custom single nucleotide polymorphism (SNP) panel. Relative risks (RR), 95% confidence interval (CI), and likelihood ratio tests from log-linear models were used to estimate the parent-of-origin effect of 877 SNPs in 60 candidate genes in the folate, homocysteine, and transsulfuration pathways on the risk of OHDs. Bonferroni correction was applied for multiple testing. We identified 3 SNPs in the transsulfuration pathway and 1 SNP in the folate pathway that were statistically significant after Bonferroni correction. Among infants who inherited paternally-derived copies of the G allele for rs6812588 in the RFC1 gene, the G allele for rs1762430 in the MGMT gene, and the A allele for rs9296695 and rs4712023 in the GSTA3 gene, RRs for OHD were 0.11 (95% CI: 0.04, 0.29, P = 9.16x10-7), 0.30 (95% CI: 0.17, 0.53, P = 9.80x10-6), 0.34 (95% CI: 0.20, 0.57, P = 2.28x10-5), and 0.34 (95% CI: 0.20, 0.58, P = 3.77x10-5), respectively, compared to infants who inherited maternally-derived copies of the same alleles. We observed statistically significant decreased risk of OHDs among infants who inherited paternal gene variants involved in folate and transsulfuration pathways.


Asunto(s)
Predisposición Genética a la Enfermedad , Variación Genética , Cardiopatías Congénitas/genética , Patrón de Herencia , Adulto , Alelos , Cardiomiopatía Hipertrófica Familiar/genética , Mapeo Cromosómico , Femenino , Genotipo , Cardiopatías Congénitas/epidemiología , Cardiopatías Congénitas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Fenotipo , Polimorfismo de Nucleótido Simple , Medición de Riesgo , Adulto Joven
5.
Ann Neurol ; 92(5): 895-901, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35947102

RESUMEN

NOTCH1 belongs to the NOTCH family of proteins that regulate cell fate and inflammatory responses. Somatic and germline NOTCH1 variants have been implicated in cancer, Adams-Oliver syndrome, and cardiovascular defects. We describe 7 unrelated patients grouped by the presence of leukoencephalopathy with calcifications and heterozygous de novo gain-of-function variants in NOTCH1. Immunologic profiling showed upregulated CSF IP-10, a cytokine secreted downstream of NOTCH1 signaling. Autopsy revealed extensive leukoencephalopathy and microangiopathy with vascular calcifications. This evidence implicates that heterozygous gain-of-function variants in NOTCH1 lead to a chronic central nervous system (CNS) inflammatory response resulting in a calcifying microangiopathy with leukoencephalopathy. ANN NEUROL 2022;92:895-901.


Asunto(s)
Displasia Ectodérmica , Leucoencefalopatías , Humanos , Receptor Notch1/genética , Receptor Notch1/metabolismo , Quimiocina CXCL10 , Sistema Nervioso Central/metabolismo
6.
J Pediatr ; 240: 79-86.e1, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34508749

RESUMEN

OBJECTIVES: To assess associations between maternal smoking and congenital heart defects (CHDs) in offspring. STUDY DESIGN: We performed a retrospective case-control study using data for cases of CHD (n = 8339) and nonmalformed controls (n = 11 020) from all years (1997-2011) of the National Birth Defects Prevention Study. Maternal self-reported smoking 1 month before through 3 months after conception was evaluated as a binary (none, any) and categorical (light, medium, heavy) exposure. Multivariable logistic regression was used to estimate aOR and 95% CIs. Stratified analyses were performed for septal defects according to maternal age, prepregnancy body mass index, and maternal race/ethnicity. RESULTS: Multiple CHDs displayed modest associations with any level of maternal periconceptional smoking independent of potential confounders; the strongest associations were for aggregated septal defects (OR, 1.5; 95% CI, 1.3-1.7), tricuspid atresia (OR, 1.7; 95% CI, 1.0-2.7), and double outlet right ventricle (DORV) (OR, 1.5; 95% CI, 1.1-2.1). Tricuspid atresia and DORV also displayed dose-response relationships. Among heavy smokers, the highest odds were again observed for tricuspid atresia (aOR 3.0; 95% CI, 1.5-6.1) and DORV (aOR 1.5; 95% CI, 1.1-2.2). Heavy smokers ≥35 years old more frequently had a child with a septal defect when compared with similarly aged nonsmokers (aOR 2.3; 95% CI, 1.4-3.9). CONCLUSIONS: Maternal periconceptional smoking is most strongly associated with septal defects, tricuspid atresia, and DORV; the risk for septal defects is modified by maternal age.


Asunto(s)
Cannabis , Cardiopatías Congénitas , Efectos Tardíos de la Exposición Prenatal , Adulto , Anciano , Estudios de Casos y Controles , Niño , Femenino , Cardiopatías Congénitas/epidemiología , Cardiopatías Congénitas/etiología , Humanos , Lactante , Embarazo , Efectos Tardíos de la Exposición Prenatal/epidemiología , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Fumar/efectos adversos
7.
Am J Med Genet C Semin Med Genet ; 187(2): 199-212, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33982868

RESUMEN

People with skeletal dysplasias have left traces in art and antiquities through ages and cultures worldwide, in Ancient Egypt, Classical Greece, Sub-Saharan Africa, Asia, and Europe. Such traces record the impact of people with skeletal dysplasia on society and culture-in daily life, religion, and mythology. However, identifying ("diagnosing") skeletal dysplasia in artifacts and interpreting what such depictions meant within the culture in which they were created is extremely challenging and at times impossible. The objectives of this short and necessarily selective survey are to present a few examples of art through different ages and cultures as a springboard for discussion not only on potential medical diagnoses but also on the lives of people with chondrodysplasia and how they were valued in the society in which they lived. The artifacts were selected from Ancient Egypt, Classical Greece, Mesoamerica (Maya), Sub-Saharan Africa (Kingdom of Benin), Tang China, and 17th Century Europe. In some cases, surviving artifacts with likely depictions of skeletal dysplasia are few and their cultural context incompletely understood. Nevertheless, certain themes and attitudes seem to repeat across different times and regions, though some cultures, such as those in Ancient Egypt, appeared to have had a comparatively positive view of people with restricted growth and chondrodysplasia.


Asunto(s)
Anomalías Musculoesqueléticas , Osteocondrodisplasias , Asia , China , Europa (Continente) , Humanos
8.
Am J Med Genet A ; 185(2): 517-527, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33398909

RESUMEN

Bone dysplasias (osteochondrodysplasias) are a large group of conditions associated with short stature, skeletal disproportion, and radiographic abnormalities of skeletal elements. Nearly all are genetic in origin. We report a series of seven children with similar findings of chondrodysplasia and growth failure following early hematopoietic stem cell transplantation (HSCT) for pediatric non-oncologic disease: hemophagocytic lymphohistiocytosis or HLH (five children, three with biallelic HLH-associated variants [in PRF1 and UNC13D] and one with HLH secondary to visceral Leishmaniasis), one child with severe combined immunodeficiency and one with Omenn syndrome (both children had biallelic RAG1 pathogenic variants). All children had normal growth and no sign of chondrodysplasia at birth and prior to their primary disease. After HSCT, all children developed growth failure, with standard deviation scores for height at or below -3. Radiographically, all children had changes in the spine, metaphyses and epiphyses, compatible with a spondyloepimetaphyseal dysplasia. Genomic sequencing failed to detect pathogenic variants in genes associated with osteochondrodysplasias. We propose that such chondrodysplasia with growth failure is a novel, rare, but clinically important complication following early HSCT for non-oncologic pediatric diseases. The pathogenesis is unknown but could possibly involve loss or perturbation of the cartilage-bone stem cell population.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/efectos adversos , Linfohistiocitosis Hemofagocítica/genética , Osteocondrodisplasias/genética , Niño , Preescolar , Femenino , Humanos , Linfohistiocitosis Hemofagocítica/complicaciones , Linfohistiocitosis Hemofagocítica/diagnóstico , Linfohistiocitosis Hemofagocítica/terapia , Masculino , Proteínas de la Membrana/genética , Osteocondrodisplasias/complicaciones , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/terapia , Perforina/genética , Resultado del Tratamiento
9.
Am J Med Genet C Semin Med Genet ; 184(1): 23-35, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32083404

RESUMEN

Congenital heart disease (CHD) is common, costly, and critical. Approximately half of all infant deaths due to congenital anomalies are associated with CHD or neural tube defects. As infant mortality improves due to better infection control and peripartum care, congenital anomalies are becoming a key driver of pediatric survival and health. Improving CHD prevention and care globally will play a significant role toward key goals such as United Nation's sustainable development goals (SDGs) of good health and well-being (SDG 3) and reduced inequalities (SDG 10). This review addresses two questions: how can we reinterpret and reframe available data on CHD to spur action in prevention and care? How can we re-engineer how we currently track CHD in populations to efficiently generate new data to assess successes and detect gaps in prevention and care? Answering these questions requires understanding the causal chain of disease, from cause to CHD occurrence to health outcomes. This perspective provides a logical basis for two innovations. First, develop a data-driven message that reframes epidemiologic and clinical data in terms of incentives for action, evidence for change, and strategies for population-wide impact. Second, through partnerships between clinical and public health systems, implement an integrated "triple surveillance," which, in the same population, concurrently tracks the three elements of the causal chain-causes, disease occurrence, health outcomes. By streamlining activities and minimizing operational waste, such systems can have a vital role in improving prevention and care on a population level, including in many low and middle-income countries.


Asunto(s)
Cardiopatías Congénitas/epidemiología , Cardiopatías Congénitas/genética , Salud Pública , Cardiopatías Congénitas/patología , Humanos , Lactante , Mortalidad Infantil , Recién Nacido
10.
Hum Genet ; 139(8): 1077-1090, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32266521

RESUMEN

Our previous genome-wide association study (GWAS) for sagittal nonsyndromic craniosynostosis (sNCS) provided important insights into the genetics of midline CS. In this study, we performed a GWAS for a second midline NCS, metopic NCS (mNCS), using 215 non-Hispanic white case-parent triads. We identified six variants with genome-wide significance (P ≤ 5 × 10-8): rs781716 (P = 4.71 × 10-9; odds ratio [OR] = 2.44) intronic to SPRY3; rs6127972 (P = 4.41 × 10-8; OR = 2.17) intronic to BMP7; rs62590971 (P = 6.22 × 10-9; OR = 0.34), located ~ 155 kb upstream from TGIF2LX; and rs2522623, rs2573826, and rs2754857, all intronic to PCDH11X (P = 1.76 × 10-8, OR = 0.45; P = 3.31 × 10-8, OR = 0.45; P = 1.09 × 10-8, OR = 0.44, respectively). We performed a replication study of these variants using an independent non-Hispanic white sample of 194 unrelated mNCS cases and 333 unaffected controls; only the association for rs6127972 (P = 0.004, OR = 1.45; meta-analysis P = 1.27 × 10-8, OR = 1.74) was replicated. Our meta-analysis examining single nucleotide polymorphisms common to both our mNCS and sNCS studies showed the strongest association for rs6127972 (P = 1.16 × 10-6). Our imputation analysis identified a linkage disequilibrium block encompassing rs6127972, which contained an enhancer overlapping a CTCF transcription factor binding site (chr20:55,798,821-55,798,917) that was significantly hypomethylated in mesenchymal stem cells derived from fused metopic compared to open sutures from the same probands. This study provides additional insights into genetic factors in midline CS.


Asunto(s)
Proteína Morfogenética Ósea 7/genética , Craneosinostosis/genética , Variación Genética , Polimorfismo de Nucleótido Simple/genética , Alelos , Metilación de ADN , Genes Reporteros , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Intrones/genética , Desequilibrio de Ligamiento , Regiones Promotoras Genéticas/genética , Factores de Riesgo
11.
Mol Genet Metab ; 129(1): 13-19, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31836396

RESUMEN

BACKGROUND: Medium-Chain Acyl-CoA Dehydrogenase (MCAD) deficiency is a fatty acid oxidation disorder that can have variable clinical severity. There is still limited information on its clinical presentation and longitudinal history by genotype, and effectiveness of newborn screening (NBS). METHODS: Retrospective data were collected from 90 patients (44 female, 46 male) to compare biochemical data with clinical outcomes. The frequency of adverse events (number of hypoglycemia-related ER visits and admissions) was assessed by genotype (homozygosity or not for the common pathogenic variant, p.Lys329Glu, in the ACADM gene), and method of diagnosis (NBS vs. clinical). RESULTS: MCAD deficiency in Utah was more frequent compared to the United States average (1: 9266 versus 1:17,759 newborns). With age, C8-carnitine did not change significantly whereas C2-carnitine decreased (p < .001), possibly reflecting reduced carnitine supplementation typically seen with age. Children with MCAD deficiency had normal growth. p.Lys329Glu homozygotes had higher NBS C8-carnitine (23.4 ± 19.6 vs. 6.6 ± 3.0 µmol/L) and lifetime plasma C8-carnitine levels (6.2 ± 5 vs. 3.6 ± 1.9 µmol/L) compared to patients with at least one other pathogenic variant (p < .001 for both) and higher transaminases compared to compound heterozygotes (ALT 41.9 ± 6.2 vs. 31.5 ± 3.7 U/L, AST 63.9 ± 5.8 vs. 45.7 ± 1.8 U/L, p < .05 for both). On average, p.Lys329Glu homozygotes had more hypoglycemic events than compound heterozygotes (1.44 versus 0.49 events/patient) as did patients diagnosed clinically compared to those diagnosed by NBS (2.15 versus 0.62 events/patient), though these differences were not statistically significant. Neonatal death was observed before results of newborn screening were available in one patient homozygous for the common p.Lys329Glu pathogenic variant, but severe neonatal complications (hypoglycemia, cardiac arrhythmia) were also seen in patients with other mutations. No irreversible complications were observed after diagnosis in any patient with MCAD deficiency. DISCUSSION: Homozygosity for the common ACADM p.Lys329Glu pathogenic variant was associated with increased levels of C8-carnitine and transaminases. Newborn screening provides the opportunity to reduce morbidity and post-neonatal mortality in all patients with MCAD deficiency, regardless of genotype.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Genotipo , Homocigoto , Errores Innatos del Metabolismo Lipídico/diagnóstico , Tamizaje Neonatal , Acil-CoA Deshidrogenasa/genética , Adolescente , Adulto , Carnitina/sangre , Niño , Preescolar , Femenino , Variación Genética , Humanos , Recién Nacido , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/mortalidad , Masculino , Mutación , Fenotipo , Estudios Retrospectivos , Transaminasas/sangre , Estados Unidos , Utah , Adulto Joven
12.
Am J Obstet Gynecol ; 222(2): 176.e1-176.e11, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31454511

RESUMEN

BACKGROUND: Diabetes is associated with an increased risk for many birth defects and is likely to have an increasing impact on birth defect prevalence because of the rise in diabetes in the United States in recent decades. One of the first analyses in which specific birth defects were assessed for their relationship with both pregestational and gestational diabetes used data from the initial 6 years of the National Birth Defects Prevention Study. That analysis reported strong associations for pregestational diabetes with several birth defects, but few exposures among some of the less common birth defects led to unstable estimates with wide confidence intervals. Since that analysis, the study continued to collect data for another 8 years, including information on approximately 19,000 additional cases and 6900 additional controls. OBJECTIVE: Our objective was to use data from the National Birth Defects Prevention Study, the largest population-based birth defects case-control study in the United States, to provide updated and more precise estimates of the association between diabetes and birth defects, including some defects not previously assessed. STUDY DESIGN: We analyzed data on deliveries from October 1997 through December 2011. Mothers of case and control infants were interviewed about their health conditions and exposures during pregnancy, including diagnosis of pregestational (type 1 or type 2) diabetes before the index pregnancy or gestational diabetes during the index pregnancy. Using logistic regression, we separately assessed the association between pregestational and gestational diabetes with specific categories of structural birth defects for which there were at least 3 exposed case infants. For birth defect categories for which there were at least 5 exposed case infants, we calculated odds ratios adjusted for maternal body mass index, age, education, race/ethnicity, and study site; for defect categories with 3 or 4 exposed cases, we calculated crude odds ratios. RESULTS: Pregestational diabetes was reported by 0.6% of mothers of control infants (71 of 11,447) and 2.5% of mothers of case infants (775 of 31,007). Gestational diabetes during the index pregnancy was reported by 4.7% of mothers of control infants (536 of 11,447) and 5.3% of mothers of case infants (1,653 of 31,007). Pregestational diabetes was associated with strong, statistically significant odds ratios (range, 2.5-80.2) for 46 of 50 birth defects considered. The largest odds ratio was observed for sacral agenesis (adjusted odds ratio, 80.2; 95% confidence interval, 46.1-139.3). A greater than 10-fold increased risk was also observed for holoprosencephaly (adjusted odds ratio, 13.1; 95% confidence interval, 7.0-24.5), longitudinal limb deficiency (adjusted odds ratio, 10.1; 95% confidence interval, 6.2-16.5), heterotaxy (adjusted odds ratio, 12.3; 95% confidence interval, 7.3-20.5), truncus arteriosus (adjusted odds ratio, 14.9; 95% confidence interval, 7.6-29.3), atrioventricular septal defect (adjusted odds ratio, 10.5; 95% confidence interval, 6.2-17.9), and single ventricle complex (adjusted odds ratio, 14.7; 95% confidence interval, 8.9-24.3). For gestational diabetes, statistically significant odds ratios were fewer (12 of 56) and of smaller magnitude (range, 1.3- 2.1; 0.5 for gastroschisis). CONCLUSION: Pregestational diabetes is associated with a markedly increased risk for many specific births defects. Because glycemic control before pregnancy is associated with a reduced risk for birth defects, ongoing quality care for persons with diabetes is an important opportunity for prevention.


Asunto(s)
Anomalías Congénitas/epidemiología , Diabetes Gestacional/epidemiología , Embarazo en Diabéticas/epidemiología , Anomalías Múltiples/epidemiología , Adulto , Estudios de Casos y Controles , Femenino , Gastrosquisis/epidemiología , Cardiopatías Congénitas/epidemiología , Holoprosencefalia/epidemiología , Humanos , Deformidades Congénitas de las Extremidades/epidemiología , Meningocele/epidemiología , Malformaciones del Sistema Nervioso/epidemiología , Embarazo , Región Sacrococcígea/anomalías , Estados Unidos/epidemiología , Adulto Joven
13.
Hum Mutat ; 40(7): 908-925, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30817854

RESUMEN

Pathogenic de novo variants in the X-linked gene SLC35A2 encoding the major Golgi-localized UDP-galactose transporter required for proper protein and lipid glycosylation cause a rare type of congenital disorder of glycosylation known as SLC35A2-congenital disorders of glycosylation (CDG; formerly CDG-IIm). To date, 29 unique de novo variants from 32 unrelated individuals have been described in the literature. The majority of affected individuals are primarily characterized by varying degrees of neurological impairments with or without skeletal abnormalities. Surprisingly, most affected individuals do not show abnormalities in serum transferrin N-glycosylation, a common biomarker for most types of CDG. Here we present data characterizing 30 individuals and add 26 new variants, the single largest study involving SLC35A2-CDG. The great majority of these individuals had normal transferrin glycosylation. In addition, expanding the molecular and clinical spectrum of this rare disorder, we developed a robust and reliable biochemical assay to assess SLC35A2-dependent UDP-galactose transport activity in primary fibroblasts. Finally, we show that transport activity is directly correlated to the ratio of wild-type to mutant alleles in fibroblasts from affected individuals.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Uridina Difosfato Galactosa/metabolismo , Animales , Biopsia , Células CHO , Células Cultivadas , Trastornos Congénitos de Glicosilación/metabolismo , Trastornos Congénitos de Glicosilación/patología , Cricetulus , Femenino , Humanos , Masculino , Mutación
14.
Am J Hum Genet ; 99(2): 299-317, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27476657

RESUMEN

Vascular malformations are non-neoplastic expansions of blood vessels that arise due to errors during angiogenesis. They are a heterogeneous group of sporadic or inherited vascular disorders characterized by localized lesions of arteriovenous, capillary, or lymphatic origin. Vascular malformations that occur inside bone tissue are rare. Herein, we report loss-of-function mutations in ELMO2 (which translates extracellular signals into cellular movements) that are causative for autosomal-recessive intraosseous vascular malformation (VMOS) in five different families. Individuals with VMOS suffer from life-threatening progressive expansion of the jaw, craniofacial, and other intramembranous bones caused by malformed blood vessels that lack a mature vascular smooth muscle layer. Analysis of primary fibroblasts from an affected individual showed that absence of ELMO2 correlated with a significant downregulation of binding partner DOCK1, resulting in deficient RAC1-dependent cell migration. Unexpectedly, elmo2-knockout zebrafish appeared phenotypically normal, suggesting that there might be human-specific ELMO2 requirements in bone vasculature homeostasis or genetic compensation by related genes. Comparative phylogenetic analysis indicated that elmo2 originated upon the appearance of intramembranous bones and the jaw in ancestral vertebrates, implying that elmo2 might have been involved in the evolution of these novel traits. The present findings highlight the necessity of ELMO2 for maintaining vascular integrity, specifically in intramembranous bones.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Huesos/irrigación sanguínea , Proteínas del Citoesqueleto/genética , Mutación/genética , Transducción de Señal/genética , Malformaciones Vasculares/genética , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Alelos , Animales , Movimiento Celular , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/metabolismo , Evolución Molecular , Femenino , Homocigoto , Humanos , Masculino , Fenotipo , Filogenia , Especificidad de la Especie , Malformaciones Vasculares/metabolismo , Malformaciones Vasculares/patología , Pez Cebra/genética , Pez Cebra/fisiología , Proteínas de Unión al GTP rac/genética
15.
Am J Hum Genet ; 99(3): 695-703, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27545681

RESUMEN

Via whole-exome sequencing, we identified rare autosomal-recessive variants in UBA5 in five children from four unrelated families affected with a similar pattern of severe intellectual deficiency, microcephaly, movement disorders, and/or early-onset intractable epilepsy. UBA5 encodes the E1-activating enzyme of ubiquitin-fold modifier 1 (UFM1), a recently identified ubiquitin-like protein. Biochemical studies of mutant UBA5 proteins and studies in fibroblasts from affected individuals revealed that UBA5 mutations impair the process of ufmylation, resulting in an abnormal endoplasmic reticulum structure. In Caenorhabditis elegans, knockout of uba-5 and of human orthologous genes in the UFM1 cascade alter cholinergic, but not glutamatergic, neurotransmission. In addition, uba5 silencing in zebrafish decreased motility while inducing abnormal movements suggestive of seizures. These clinical, biochemical, and experimental findings support our finding of UBA5 mutations as a pathophysiological cause for early-onset encephalopathies due to abnormal protein ufmylation.


Asunto(s)
Alelos , Encefalopatías/genética , Mutación/genética , Proteínas/metabolismo , Enzimas Activadoras de Ubiquitina/genética , Edad de Inicio , Animales , Mapeo Encefálico , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Niño , Preescolar , Neuronas Colinérgicas/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Epilepsia/genética , Exoma/genética , Femenino , Fibroblastos , Genes Recesivos/genética , Humanos , Discapacidad Intelectual/genética , Imagen por Resonancia Magnética , Masculino , Microcefalia/genética , Trastornos del Movimiento , Proteínas/genética , Transmisión Sináptica/genética , Ubiquitina/genética , Ubiquitina/metabolismo , Enzimas Activadoras de Ubiquitina/deficiencia , Enzimas Activadoras de Ubiquitina/metabolismo , Ubiquitinas/genética , Ubiquitinas/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
Am J Med Genet A ; 179(5): 792-796, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30773818

RESUMEN

The NONO gene encodes a nuclear protein involved in RNA metabolism. Hemizygous loss-of-function NONO variants have been associated with syndromic intellectual disability and with left ventricular noncompaction (LVNC). A two-year-old boy presented to the University of Utah's Penelope Undiagnosed Disease Program with developmental delay, nonfamilial features, relative macrocephaly, and dilated cardiomyopathy with LVNC and Ebstein anomaly. Brain MRI showed a thick corpus callosum, mild Chiari I malformation, and a flattened pituitary. Exome sequencing identified a novel intronic deletion (c.154+5_154+6delGT) in the NONO gene. Splicing studies demonstrated intron 4 read-through and the use of an alternative donor causing the frameshift p.Asn52Serfs*6. Family segregation analysis showed that the variant occurred de novo in the boy's unaffected mother. MRI and endocrine findings suggest that hypopituitarism may contribute to growth failure, abnormal thyroid hormone levels, cryptorchidism, or delayed puberty in patients with NONO-associated disease. Also, including this case LVNC has been observed in five out of eight patients, and this report also confirms an association between loss of NONO and Ebstein anomaly. In some cases, unrelated individuals share the same pathogenic NONO variants but do not all have clinically significant LVNC, suggesting that additional modifiers may contribute to cardiac phenotypes.


Asunto(s)
Proteínas de Unión al ADN/genética , Genes Ligados a X , Predisposición Genética a la Enfermedad , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Mutación , Fenotipo , Proteínas de Unión al ARN/genética , Preescolar , Análisis Mutacional de ADN , Exoma , Facies , Femenino , Estudios de Asociación Genética , Sitios Genéticos , Humanos , Imagen por Resonancia Magnética , Masculino , Polimorfismo de Nucleótido Simple , Síndrome
17.
J Pediatr ; 203: 371-379.e7, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30268400

RESUMEN

OBJECTIVE: To assess longitudinal estimates of inpatient costs through early childhood in patients with critical congenital heart defects (CCHDs), for whom reliable estimates are scarce, using a population-based cohort of clinically validated CCHD cases. STUDY DESIGN: Longitudinal retrospective cohort of infants with CCHDs live born from 1997 to 2012 in Utah. Cases identified from birth defect registry data were linked to inpatient discharge abstracts and vital records to track inpatient days and costs through age 10 years. Costs were adjusted for inflation and discounted by 3% per year to generate present value estimates. Multivariable models identified infant and maternal factors potentially associated with higher resource utilization and were used to calculate adjusted costs by defect type. RESULTS: The final statewide cohort included 1439 CCHD cases among 803 509 livebirths (1.8/1000). The average cost per affected child through age 10 years was $136 682 with a median of $74 924 because of a small number of extremely high cost children; costs were highest for pulmonary atresia with ventricular septal defect and hypoplastic left heart syndrome. Inpatient costs increased by 1.6% per year during the study period. A single birth year cohort (~50 000 births/year) had estimated expenditures of $11 902 899 through age 10 years. Extrapolating to the US population, inpatient costs for a single birth year cohort through age 10 years were ~$1 billion. CONCLUSIONS: Inpatient costs for CCHDs throughout childhood are high and rising. These revised estimates will contribute to comparative effectiveness research aimed at improving the value of care on a patient and population level.


Asunto(s)
Costos de la Atención en Salud , Cardiopatías Congénitas/economía , Cardiopatías Congénitas/epidemiología , Tamizaje Neonatal/economía , Tamizaje Neonatal/métodos , Anomalías Congénitas , Bases de Datos Factuales , Femenino , Defectos del Tabique Interventricular/economía , Defectos del Tabique Interventricular/epidemiología , Hospitalización/economía , Humanos , Síndrome del Corazón Izquierdo Hipoplásico/economía , Síndrome del Corazón Izquierdo Hipoplásico/epidemiología , Lactante , Recién Nacido , Pacientes Internos , Estudios Longitudinales , Masculino , Análisis Multivariante , Atresia Pulmonar/economía , Atresia Pulmonar/epidemiología , Sistema de Registros , Estudios Retrospectivos , Utah/epidemiología
18.
Am J Med Genet A ; 176(12): 2901-2906, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30346094

RESUMEN

Interstitial deletions of chromosome 9q31 are very rare. The deletions in most reported patients have been detected by conventional cytogenetics, with reported breakpoints ranging between 9q21 and 9q34. Therefore, an accurate description of a "9q31 deletion syndrome" could not be established. However, based on microarray studies, a small region of overlap has recently been proposed. We report clinical features of two unrelated individuals with overlapping 9q deletions identified by SNP microarray analysis. Patient 1 has a 9 Mb deletion, while Patient 2's deletion was 21.6 Mb. The clinical features common to our patients and those in the literature include developmental delay and short stature. Patient 2 shows additional features not reported in other 9q31 deletions, such as hearing loss, ventriculomegaly, cleft lip and palate, and small kidneys, which could be due to the larger size of the deletion, hence the influence of the genes in the region beyond the smallest region of overlap. Based on the comparison of these patients with the previously reported patients, we redefine the smallest region of overlap and characterize the clinical features of the 9q31 deletion syndrome.


Asunto(s)
Deleción Cromosómica , Trastornos de los Cromosomas/diagnóstico , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 9 , Adolescente , Alelos , Hibridación Genómica Comparativa , Facies , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Lactante , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple , Síndrome
19.
Matern Child Health J ; 22(10): 1418-1429, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29574536

RESUMEN

Introduction While associations between active smoking and various adverse birth outcomes (ABOs) have been reported in the literature, less is known about the impact of secondhand smoke (SHS) on many pregnancy outcomes. Methods We examined the relationship between maternal exposure to SHS during pregnancy and preterm (< 37 weeks gestation) and small-for-gestational age (SGA; assessed using sex-, race/ethnic-, and parity-specific growth curves) singleton births using non-smoking controls from the National Birth Defects Prevention Study (1997-2011). Multivariable logistic regression models for household, workplace/school, and combined SHS exposure-controlled for maternal education, race/ethnicity, pre-pregnancy body mass index, and high blood pressure-were used to estimate adjusted odds ratios (aORs) and 95% confidence intervals (CIs). Interaction was assessed for maternal folic acid supplementation, alcohol use, age at delivery, and infant sex. Results Infants of 8855 mothers were examined in the preterm birth analysis with 666 (7.5%) categorized as preterm, 574 moderately preterm (32-36 weeks), and 92 very preterm (< 32 weeks). For the SGA analysis, infants of 8684 mothers were examined with 670 (7.7%) categorized as SGA. The aORs for mothers reporting both household and workplace/school SHS were elevated for preterm (aOR 1.99; 95% CI 1.13-3.50) and moderately preterm birth (32-36 weeks) (aOR 2.17; 95% CI 1.22-3.88). No results for the SGA analysis achieved significance, nor was evidence of interaction evident. Conclusion The findings suggest an association between SHS from multiple exposure sources and preterm birth, but no evidence for association with SGA births. Continued study of SHS and ABOs is needed to best inform public health prevention programs.


Asunto(s)
Recién Nacido Pequeño para la Edad Gestacional , Exposición Materna/efectos adversos , Nacimiento Prematuro/inducido químicamente , Contaminación por Humo de Tabaco/efectos adversos , Adulto , Escolaridad , Femenino , Edad Gestacional , Humanos , Lactante , Recién Nacido , Masculino , Embarazo , Resultado del Embarazo , Nacimiento Prematuro/epidemiología , Nicotiana , Contaminación por Humo de Tabaco/estadística & datos numéricos
20.
Am J Epidemiol ; 186(6): 719-729, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28520847

RESUMEN

Nutrients that regulate methylation processes may modify susceptibility to the effects of air pollutants. Data from the National Birth Defects Prevention Study (United States, 1997-2006) were used to estimate associations between maternal exposure to nitrogen dioxide (NO2), dietary intake of methyl nutrients, and the odds of congenital heart defects in offspring. NO2 concentrations, a marker of traffic-related air pollution, averaged across postconception weeks 2-8, were assigned to 6,160 nondiabetic mothers of cases and controls using inverse distance-squared weighting of air monitors within 50 km of maternal residences. Intakes of choline, folate, methionine, and vitamins B6 and B12 were assessed using a food frequency questionnaire. Hierarchical regression models, which accounted for similarities across defects, were constructed, and relative excess risks due to interaction were calculated. Relative to women with the lowest NO2 exposure and high methionine intake, women with the highest NO2 exposure and lowest methionine intake had the greatest odds of offspring with a perimembranous ventricular septal defect (odds ratio = 3.23, 95% confidence interval: 1.74, 6.01; relative excess risk due to interaction = 2.15, 95% confidence interval: 0.39, 3.92). Considerable departure from additivity was not observed for other defects. These results provide modest evidence of interaction between nutrition and NO2 exposure during pregnancy.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Ingestión de Alimentos , Cardiopatías Congénitas/inducido químicamente , Exposición Materna/efectos adversos , Dióxido de Nitrógeno/toxicidad , Adulto , Contaminantes Atmosféricos/análisis , Contaminación del Aire/efectos adversos , Contaminación del Aire/análisis , Estudios de Casos y Controles , Colina/análisis , Registros de Dieta , Femenino , Ácido Fólico/análisis , Análisis de los Alimentos , Humanos , Recién Nacido , Metionina/análisis , Dióxido de Nitrógeno/análisis , Oportunidad Relativa , Embarazo , Fenómenos Fisiologicos de la Nutrición Prenatal , Factores de Riesgo , Estados Unidos , Vitamina B 12/análisis , Vitamina B 6/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA