Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dev Biol ; 506: 72-84, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38110169

RESUMEN

The DGCR8 gene, encoding a critical miRNA processing protein, maps within the hemizygous region in patients with 22q11.2 deletion syndrome. Most patients have malformations of the cardiac outflow tract that is derived in part from the anterior second heart field (aSHF) mesoderm. To understand the function of Dgcr8 in the aSHF, we inactivated it in mice using Mef2c-AHF-Cre. Inactivation resulted in a fully penetrant persistent truncus arteriosus and a hypoplastic right ventricle leading to lethality by E14.5. To understand the molecular mechanism for this phenotype, we performed gene expression profiling of the aSHF and the cardiac outflow tract with right ventricle in conditional null versus normal mouse littermates at stage E9.5 prior to morphology changes. We identified dysregulation of mRNA gene expression, of which some are relevant to cardiogenesis. Many pri-miRNA genes were strongly increased in expression in mutant embryos along with reduced expression of mature miRNA genes. We further examined the individual, mature miRNAs that were decreased in expression along with pri-miRNAs that were accumulated that could be direct effects due to loss of Dgcr8. Among these genes, were miR-1a, miR-133a, miR-134, miR143 and miR145a, which have known functions in heart development. These early mRNA and miRNA changes may in part, explain the first steps that lead to the resulting phenotype in Dgcr8 aSHF conditional mutant embryos.


Asunto(s)
Ventrículos Cardíacos , MicroARNs , Humanos , Ratones , Animales , Ventrículos Cardíacos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Mamíferos/metabolismo , ARN Mensajero
2.
Dev Biol ; 494: 71-84, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36521641

RESUMEN

The morphogenesis of the otic vesicle (OV) to form inner ear organs serves as an excellent model system to understand cell fate acquisition on a single cell level. Tbx2 and Tbx3 (Tbx2/3) encode closely related T-box transcription factors that are expressed widely in the mammalian OV. Inactivation of both genes in the OV (Tbx2/3cKO) results in failed morphogenesis into inner ear organs. To understand the basis of these defects, single cell RNA-sequencing (scRNA-seq) was performed on the OV lineage, in controls versus Tbx2/3cKO embryos. We identified a multipotent population termed otic progenitors in controls that are marked by expression of the known otic placode markers Eya1, Sox2, and Sox3 as well as new markers Fgf18, Cxcl12, and Pou3f3. The otic progenitor population was increased three-fold in Tbx2/3cKO embryos, concomitant with dysregulation of genes in these cells as well as reduced progression to more differentiated states of prosensory and nonsensory cells. An ectopic neural population of cells was detected in the posterior OV of Tbx2/3cKO embryos but had reduced maturation to delaminated neural cells. As all three cell fates were affected in Tbx2/3cKO embryos, we suggest that Tbx2/3 promotes progression of multipotent otic progenitors to more differentiated cell types in the OV.


Asunto(s)
Oído Interno , Animales , Diferenciación Celular/genética , Oído Interno/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Mamíferos/metabolismo , Morfogénesis , Sistema Nervioso/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Dominio T Box
3.
Hum Mol Genet ; 31(8): 1197-1215, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686881

RESUMEN

CRK and CRKL encode cytoplasmic adaptors that contribute to the etiology of congenital heart disease. Neural crest cells (NCCs) are required for cardiac outflow tract (OFT) septation and aortic arch formation. The roles of Crk/Crkl in NCCs during mouse cardiovascular development remain unknown. To test this, we inactivated Crk and/or Crkl in NCCs. We found that the loss of Crk, rather than Crkl, in NCCs resulted in double outlet right ventricle, while loss of both Crk/Crkl in NCCs resulted in severe defects with earlier lethality due to failed OFT septation and severe dilation of the pharyngeal arch arteries (PAAs). We found that these defects are due to altered cell morphology resulting in reduced localization of NCCs to the OFT and failed integrity of the PAAs, along with reduced expression of Integrin signaling genes. Further, molecular studies identified reduced differentiation of vascular smooth muscle cells that may in part be due to altered Notch signaling. Additionally, there is increased cellular stress that leads to modest increase in apoptosis. Overall, this explains the mechanism for the Crk/Crkl phenotype.


Asunto(s)
Cardiopatías Congénitas , Cresta Neural , Animales , Diferenciación Celular/genética , Cardiopatías Congénitas/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Cresta Neural/metabolismo , Proteínas Proto-Oncogénicas c-crk/metabolismo , Transducción de Señal/genética
4.
Mol Psychiatry ; 28(5): 2071-2080, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36869225

RESUMEN

22q11.2 deletion is one of the strongest known genetic risk factors for schizophrenia. Recent whole-genome sequencing of schizophrenia cases and controls with this deletion provided an unprecedented opportunity to identify risk modifying genetic variants and investigate their contribution to the pathogenesis of schizophrenia in 22q11.2 deletion syndrome. Here, we apply a novel analytic framework that integrates gene network and phenotype data to investigate the aggregate effects of rare coding variants and identified modifier genes in this etiologically homogenous cohort (223 schizophrenia cases and 233 controls of European descent). Our analyses revealed significant additive genetic components of rare nonsynonymous variants in 110 modifier genes (adjusted P = 9.4E-04) that overall accounted for 4.6% of the variance in schizophrenia status in this cohort, of which 4.0% was independent of the common polygenic risk for schizophrenia. The modifier genes affected by rare coding variants were enriched with genes involved in synaptic function and developmental disorders. Spatiotemporal transcriptomic analyses identified an enrichment of coexpression between modifier and 22q11.2 genes in cortical brain regions from late infancy to young adulthood. Corresponding gene coexpression modules are enriched with brain-specific protein-protein interactions of SLC25A1, COMT, and PI4KA in the 22q11.2 deletion region. Overall, our study highlights the contribution of rare coding variants to the SCZ risk. They not only complement common variants in disease genetics but also pinpoint brain regions and developmental stages critical to the etiology of syndromic schizophrenia.


Asunto(s)
Síndrome de DiGeorge , Esquizofrenia , Humanos , Adulto Joven , Adulto , Esquizofrenia/genética , Síndrome de DiGeorge/genética , Encéfalo , Perfilación de la Expresión Génica , Secuenciación Completa del Genoma
5.
Mol Psychiatry ; 26(8): 4496-4510, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32015465

RESUMEN

Schizophrenia occurs in about one in four individuals with 22q11.2 deletion syndrome (22q11.2DS). The aim of this International Brain and Behavior 22q11.2DS Consortium (IBBC) study was to identify genetic factors that contribute to schizophrenia, in addition to the ~20-fold increased risk conveyed by the 22q11.2 deletion. Using whole-genome sequencing data from 519 unrelated individuals with 22q11.2DS, we conducted genome-wide comparisons of common and rare variants between those with schizophrenia and those with no psychotic disorder at age ≥25 years. Available microarray data enabled direct comparison of polygenic risk for schizophrenia between 22q11.2DS and independent population samples with no 22q11.2 deletion, with and without schizophrenia (total n = 35,182). Polygenic risk for schizophrenia within 22q11.2DS was significantly greater for those with schizophrenia (padj = 6.73 × 10-6). Novel reciprocal case-control comparisons between the 22q11.2DS and population-based cohorts showed that polygenic risk score was significantly greater in individuals with psychotic illness, regardless of the presence of the 22q11.2 deletion. Within the 22q11.2DS cohort, results of gene-set analyses showed some support for rare variants affecting synaptic genes. No common or rare variants within the 22q11.2 deletion region were significantly associated with schizophrenia. These findings suggest that in addition to the deletion conferring a greatly increased risk to schizophrenia, the risk is higher when the 22q11.2 deletion and common polygenic risk factors that contribute to schizophrenia in the general population are both present.


Asunto(s)
Síndrome de DiGeorge , Trastornos Psicóticos , Esquizofrenia , Adulto , Estudios de Casos y Controles , Estudios de Cohortes , Síndrome de DiGeorge/genética , Humanos , Esquizofrenia/genética
6.
PLoS Genet ; 15(8): e1008301, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31412026

RESUMEN

We investigated whether Tbx1, the gene for 22q11.2 deletion syndrome (22q11.2DS) and Foxi3, both required for segmentation of the pharyngeal apparatus (PA) to individual arches, genetically interact. We found that all Tbx1+/-;Foxi3+/- double heterozygous mouse embryos had thymus and parathyroid gland defects, similar to those in 22q11.2DS patients. We then examined Tbx1 and Foxi3 heterozygous, null as well as conditional Tbx1Cre and Sox172A-iCre/+ null mutant embryos. While Tbx1Cre/+;Foxi3f/f embryos had absent thymus and parathyroid glands, Foxi3-/- and Sox172A-iCre/+;Foxi3f/f endoderm conditional mutant embryos had in addition, interrupted aortic arch type B and retroesophageal origin of the right subclavian artery, which are all features of 22q11.2DS. Tbx1Cre/+;Foxi3f/f embryos had failed invagination of the third pharyngeal pouch with greatly reduced Gcm2 and Foxn1 expression, thereby explaining the absence of thymus and parathyroid glands. Immunofluorescence on tissue sections with E-cadherin and ZO-1 antibodies in wildtype mouse embryos at E8.5-E10.5, revealed that multilayers of epithelial cells form where cells are invaginating as a normal process. We noted that excessive multilayers formed in Foxi3-/-, Sox172A-iCre/+;Foxi3f/f as well as Tbx1 null mutant embryos where invagination should have occurred. Several genes expressed in the PA epithelia were downregulated in both Tbx1 and Foxi3 null mutant embryos including Notch pathway genes Jag1, Hes1, and Hey1, suggesting that they may, along with other genes, act downstream to explain the observed genetic interaction. We found Alcam and Fibronectin extracellular matrix proteins were reduced in expression in Foxi3 null but not Tbx1 null embryos, suggesting that some, but not all of the downstream mechanisms are shared.


Asunto(s)
Síndrome de DiGeorge/patología , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Dominio T Box/metabolismo , Animales , Región Branquial/embriología , Síndrome de DiGeorge/genética , Modelos Animales de Enfermedad , Embrión de Mamíferos , Endodermo/embriología , Femenino , Factores de Transcripción Forkhead/genética , Corazón/embriología , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación , Miocardio/patología , Proteínas de Dominio T Box/genética
7.
Hum Mol Genet ; 28(22): 3724-3733, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31884517

RESUMEN

The majority (99%) of individuals with 22q11.2 deletion syndrome (22q11.2DS) have a deletion that is caused by non-allelic homologous recombination between two of four low copy repeat clusters on chromosome 22q11.2 (LCR22s). However, in a small subset of patients, atypical deletions are observed with at least one deletion breakpoint within unique sequence between the LCR22s. The position of the chromosome breakpoints and the mechanisms driving those atypical deletions remain poorly studied. Our large-scale, whole genome sequencing study of >1500 subjects with 22q11.2DS identified six unrelated individuals with atypical deletions of different types. Using a combination of whole genome sequencing data and fiber-fluorescence in situ hybridization, we mapped the rearranged alleles in these subjects. In four of them, the distal breakpoints mapped within one of the LCR22s and we found that the deletions likely occurred by replication-based mechanisms. Interestingly, in two of them, an inversion probably preceded inter-chromosomal 'allelic' homologous recombination between differently oriented LCR22-D alleles. Inversion associated allelic homologous recombination (AHR) may well be a common mechanism driving (atypical) deletions on 22q11.2.


Asunto(s)
Síndrome de DiGeorge/genética , Síndrome de DiGeorge/metabolismo , Recombinación Homóloga/genética , Adulto , Alelos , Puntos de Rotura del Cromosoma , Deleción Cromosómica , Inversión Cromosómica/genética , Mapeo Cromosómico/métodos , Cromosomas/genética , Cromosomas Humanos Par 22/genética , Femenino , Humanos , Hibridación Fluorescente in Situ/métodos , Masculino , Duplicaciones Segmentarias en el Genoma/genética , Secuenciación Completa del Genoma/métodos
8.
Hum Mol Genet ; 27(11): 1847-1857, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29509905

RESUMEN

Non-allelic homologous recombination events on chromosome 22q11.2 during meiosis can result in either the deletion (22q11.2DS) or duplication (22q11.2DupS) syndrome. Although the spectrum and frequency of congenital heart disease (CHD) are known for 22q11.2DS, there is less known for 22q11.2DupS. We now evaluated cardiac phenotypes in 235 subjects with 22q11.2DupS including 102 subjects we collected and 133 subjects that were previously reported as a confirmation and found 25% have CHD, mostly affecting the cardiac outflow tract (OFT). Previous studies have shown that global loss or gain of function (LOF; GOF) of mouse Tbx1, encoding a T-box transcription factor mapping to the region of synteny to 22q11.2, results in similar OFT defects. To further evaluate Tbx1 function in the progenitor cells forming the cardiac OFT, termed the anterior heart field, Tbx1 was overexpressed using the Mef2c-AHF-Cre driver (Tbx1 GOF). Here we found that all resulting conditional GOF embryos had a persistent truncus arteriosus (PTA), similar to what was previously reported for conditional Tbx1 LOF mutant embryos. To understand the basis for the PTA in the conditional GOF embryos, we found that proliferation in the Mef2c-AHF-Cre lineage cells before migrating to the heart, was reduced and critical genes were oppositely changed in this tissue in Tbx1 GOF embryos versus conditional LOF embryos. These results suggest that a major function of TBX1 in the AHF is to maintain the normal balance of expression of key cardiac developmental genes required to form the aorta and pulmonary trunk, which is disrupted in 22q11.2DS and 22q11.2DupS.


Asunto(s)
Desarrollo Embrionario/genética , Cardiopatías Congénitas/genética , Corazón/crecimiento & desarrollo , Proteínas de Dominio T Box/genética , Anomalías Múltiples/genética , Anomalías Múltiples/fisiopatología , Animales , Aorta/fisiopatología , Duplicación Cromosómica/genética , Cromosomas Humanos Par 22/genética , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/fisiopatología , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica/genética , Corazón/fisiopatología , Cardiopatías Congénitas/patología , Recombinación Homóloga/genética , Humanos , Meiosis/genética , Ratones , Mutación , Tronco Arterial Persistente/genética , Tronco Arterial Persistente/fisiopatología
9.
Hum Mol Genet ; 27(7): 1150-1163, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29361080

RESUMEN

Recurrent, de novo, meiotic non-allelic homologous recombination events between low copy repeats, termed LCR22s, leads to the 22q11.2 deletion syndrome (22q11.2DS; velo-cardio-facial syndrome/DiGeorge syndrome). Although most 22q11.2DS patients have a similar sized 3 million base pair (Mb), LCR22A-D deletion, some have nested LCR22A-B or LCR22A-C deletions. Our goal is to identify additional recurrent 22q11.2 deletions associated with 22q11.2DS, serving as recombination hotspots for meiotic chromosomal rearrangements. Here, using data from Affymetrix 6.0 microarrays on 1680 22q11.2DS subjects, we identified what appeared to be a nested proximal 22q11.2 deletion in 38 (2.3%) of them. Using molecular and haplotype analyses from 14 subjects and their parent(s) with available DNA, we found essentially three types of scenarios to explain this observation. In eight subjects, the proximal breakpoints occurred in a small sized 12 kb LCR distal to LCR22A, referred to LCR22A+, resulting in LCR22A+-B or LCR22A+-D deletions. Six of these eight subjects had a nested 22q11.2 deletion that occurred during meiosis in a parent carrying a benign 0.2 Mb duplication of the LCR22A-LCR22A+ region with a breakpoint in LCR22A+. Another six had a typical de novo LCR22A-D deletion on one allele and inherited the LCR22A-A+ duplication from the other parent thus appearing on microarrays to have a nested deletion. LCR22A+ maps to an evolutionary breakpoint between mice and humans and appears to serve as a local hotspot for chromosome rearrangements on 22q11.2.


Asunto(s)
Alelos , Mapeo Cromosómico , Síndrome de DiGeorge/genética , Meiosis , Deleción Cromosómica , Cromosomas Humanos Par 22/genética , Femenino , Humanos , Masculino
10.
Am J Hum Genet ; 101(4): 616-622, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28965848

RESUMEN

Inversion polymorphisms between low-copy repeats (LCRs) might predispose chromosomes to meiotic non-allelic homologous recombination (NAHR) events and thus lead to genomic disorders. However, for the 22q11.2 deletion syndrome (22q11.2DS), the most common genomic disorder, no such inversions have been uncovered as of yet. Using fiber-FISH, we demonstrate that parents transmitting the de novo 3 Mb LCR22A-D 22q11.2 deletion, the reciprocal duplication, and the smaller 1.5 Mb LCR22A-B 22q11.2 deletion carry inversions of LCR22B-D or LCR22C-D. Hence, the inversions predispose chromosome 22q11.2 to meiotic rearrangements and increase the individual risk for transmitting rearrangements. Interestingly, the inversions are nested or flanking rather than coinciding with the deletion or duplication sizes. This finding raises the possibility that inversions are a prerequisite not only for 22q11.2 rearrangements but also for all NAHR-mediated genomic disorders.


Asunto(s)
Inversión Cromosómica , Síndrome de DiGeorge/genética , Predisposición Genética a la Enfermedad , Meiosis , Polimorfismo de Nucleótido Simple , Deleción Cromosómica , Variaciones en el Número de Copia de ADN , Síndrome de DiGeorge/patología , Recombinación Homóloga , Humanos , Hibridación Fluorescente in Situ/métodos
11.
PLoS Genet ; 13(12): e1007142, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29281626

RESUMEN

Rare variants of major effect play an important role in human complex diseases and can be discovered by sequencing-based genome-wide association studies. Here, we introduce an integrated approach that combines the rare variant association test with gene network and phenotype information to identify risk genes implicated by rare variants for human complex diseases. Our data integration method follows a 'discovery-driven' strategy without relying on prior knowledge about the disease and thus maintains the unbiased character of genome-wide association studies. Simulations reveal that our method can outperform a widely-used rare variant association test method by 2 to 3 times. In a case study of a small disease cohort, we uncovered putative risk genes and the corresponding rare variants that may act as genetic modifiers of congenital heart disease in 22q11.2 deletion syndrome patients. These variants were missed by a conventional approach that relied on the rare variant association test alone.


Asunto(s)
Predisposición Genética a la Enfermedad , Variación Genética , Estudio de Asociación del Genoma Completo/métodos , Análisis de Secuencia de ADN/métodos , Estudios de Casos y Controles , Simulación por Computador , Interpretación Estadística de Datos , Síndrome de DiGeorge/genética , Humanos , Fenotipo , Factores de Riesgo , Análisis de Secuencia de ADN/estadística & datos numéricos
12.
PLoS Genet ; 13(3): e1006687, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28346476

RESUMEN

The 22q11.2 deletion syndrome (22q11.2DS; velo-cardio-facial syndrome; DiGeorge syndrome) is a congenital anomaly disorder in which haploinsufficiency of TBX1, encoding a T-box transcription factor, is the major candidate for cardiac outflow tract (OFT) malformations. Inactivation of Tbx1 in the anterior heart field (AHF) mesoderm in the mouse results in premature expression of pro-differentiation genes and a persistent truncus arteriosus (PTA) in which septation does not form between the aorta and pulmonary trunk. Canonical Wnt/ß-catenin has major roles in cardiac OFT development that may act upstream of Tbx1. Consistent with an antagonistic relationship, we found the opposite gene expression changes occurred in the AHF in ß-catenin loss of function embryos compared to Tbx1 loss of function embryos, providing an opportunity to test for genetic rescue. When both alleles of Tbx1 and one allele of ß-catenin were inactivated in the Mef2c-AHF-Cre domain, 61% of them (n = 34) showed partial or complete rescue of the PTA defect. Upregulated genes that were oppositely changed in expression in individual mutant embryos were normalized in significantly rescued embryos. Further, ß-catenin was increased in expression when Tbx1 was inactivated, suggesting that there may be a negative feedback loop between canonical Wnt and Tbx1 in the AHF to allow the formation of the OFT. We suggest that alteration of this balance may contribute to variable expressivity in 22q11.2DS.


Asunto(s)
Anomalías Cardiovasculares/genética , Síndrome de DiGeorge/genética , Modelos Animales de Enfermedad , Proteínas de Dominio T Box/genética , beta Catenina/genética , Animales , Apoptosis/genética , Anomalías Cardiovasculares/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/genética , Síndrome de DiGeorge/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Miocitos Cardíacos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/metabolismo , Tronco Arterial/citología , Tronco Arterial/embriología , Tronco Arterial/metabolismo , beta Catenina/metabolismo
13.
J Mol Cell Cardiol ; 125: 98-105, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30347193

RESUMEN

NOTCH and WNT signaling pathways play critical roles in cardiac chamber formation. Here we explored the potential interactions between the two pathways in this developmental process by using genetically modified mouse models and whole embryo culture systems. By deletion of Notch1 to inactivate NOTCH1 signaling in the endocardium in vivo and ex vivo rescue experiments, we showed that myocardial WNT5A mediated endocardial NOTCH1 signaling to maintain the gene regulatory network essential for cardiac chamber formation. Furthermore, genetic deletion of ß-catenin in the myocardium and inhibition of the WNT/Ca2+ signaling by FK506 resulted in a similar disruption of the gene regulatory network as inactivation of endocardial NOTCH1 signaling. Together, these findings identify WNT5A as a key myocardial factor that mediates the endocardial NOTCH signaling to maintain the gene regulatory network essential for cardiac chamber formation through WNT/ß-catenin and WNT/Ca2+ signaling pathways.


Asunto(s)
Receptor Notch1/metabolismo , Receptores Notch/metabolismo , Proteína Wnt-5a/metabolismo , beta Catenina/metabolismo , Animales , Endocardio/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Hibridación in Situ , Masculino , Ratones , Miocardio/metabolismo , Receptor Notch1/genética , Receptores Notch/genética , Proteína Wnt-5a/genética , beta Catenina/genética
14.
Hum Mol Genet ; 25(17): 3754-3767, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27436579

RESUMEN

Velo-cardio-facial syndrome/DiGeorge syndrome/22q11.2 deletion syndrome (22q11.2DS) is caused by meiotic non-allelic homologous recombination events between flanking low copy repeats termed LCR22A and LCR22D, resulting in a 3 million base pair (Mb) deletion. Due to their complex structure, large size and high sequence identity, genetic variation within LCR22s among different individuals has not been well characterized. In this study, we sequenced 13 BAC clones derived from LCR22A/D and aligned them with 15 previously available BAC sequences to create a new genetic variation map. The thousands of variants identified by this analysis were not uniformly distributed in the two LCR22s. Moreover, shared single nucleotide variants between LCR22A and LCR22D were enriched in the Breakpoint Cluster Region pseudogene (BCRP) block, suggesting the existence of a possible recombination hotspot there. Interestingly, breakpoints for atypical 22q11.2 rearrangements have previously been located to BCRPs To further explore this finding, we carried out in-depth analyses of whole genome sequence (WGS) data from two unrelated probands harbouring a de novo 3Mb 22q11.2 deletion and their normal parents. By focusing primarily on WGS reads uniquely mapped to LCR22A, using the variation map from our BAC analysis to help resolve allele ambiguity, and by performing PCR analysis, we infer that the deletion breakpoints were most likely located near or within the BCRP module. In summary, we found a high degree of sequence variation in LCR22A and LCR22D and a potential recombination breakpoint near or within the BCRP block, providing a starting point for future breakpoint mapping using additional trios.


Asunto(s)
Puntos de Rotura del Cromosoma , Síndrome de DiGeorge/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Cromosomas Artificiales Bacterianos/genética , Cromosomas Humanos Par 22/genética , Estudio de Asociación del Genoma Completo , Humanos , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple , Eliminación de Secuencia
15.
Am J Hum Genet ; 97(6): 869-77, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26608785

RESUMEN

We performed whole exome sequence (WES) to identify genetic modifiers on 184 individuals with 22q11.2 deletion syndrome (22q11DS), of whom 89 case subjects had severe congenital heart disease (CHD) and 95 control subjects had normal hearts. Three genes including JMJD1C (jumonji domain containing 1C), RREB1 (Ras responsive element binding protein 1), and SEC24C (SEC24 family member C) had rare (MAF < 0.001) predicted deleterious single-nucleotide variations (rdSNVs) in seven case subjects and no control subjects (p = 0.005; Fisher exact and permutation tests). Because JMJD1C and RREB1 are involved in chromatin modification, we investigated other histone modification genes. Eighteen case subjects (20%) had rdSNVs in four genes (JMJD1C, RREB1, MINA, KDM7A) all involved in demethylation of histones (H3K9, H3K27). Overall, rdSNVs were enriched in histone modifier genes that activate transcription (Fisher exact p = 0.0004, permutations, p = 0.0003, OR = 5.16); however, rdSNVs in control subjects were not enriched. This implicates histone modification genes as influencing risk for CHD in presence of the deletion.


Asunto(s)
Proteínas de Unión al ADN/genética , Síndrome de DiGeorge/genética , Cardiopatías Congénitas/genética , Histonas/genética , Histona Demetilasas con Dominio de Jumonji/genética , Proteínas Nucleares/genética , Oxidorreductasas N-Desmetilantes/genética , Factores de Transcripción/genética , Estudios de Casos y Controles , Síndrome de DiGeorge/complicaciones , Síndrome de DiGeorge/patología , Dioxigenasas , Exoma , Regulación de la Expresión Génica , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Histona Demetilasas , Histonas/metabolismo , Humanos , Anotación de Secuencia Molecular , Fenotipo , Polimorfismo de Nucleótido Simple , Riesgo , Transcripción Genética , Proteínas de Transporte Vesicular/genética
16.
Am J Hum Genet ; 96(2): 235-44, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25658046

RESUMEN

The human chromosome 22q11.2 region is susceptible to rearrangements during meiosis leading to velo-cardio-facial/DiGeorge/22q11.2 deletion syndrome (22q11DS) characterized by conotruncal heart defects (CTDs) and other congenital anomalies. The majority of individuals have a 3 Mb deletion whose proximal region contains the presumed disease-associated gene TBX1 (T-box 1). Although a small subset have proximal nested deletions including TBX1, individuals with distal deletions that exclude TBX1 have also been identified. The deletions are flanked by low-copy repeats (LCR22A, B, C, D). We describe cardiac phenotypes in 25 individuals with atypical distal nested deletions within the 3 Mb region that do not include TBX1 including 20 with LCR22B to LCR22D deletions and 5 with nested LCR22C to LCR22D deletions. Together with previous reports, 12 of 37 (32%) with LCR22B-D deletions and 5 of 34 (15%) individuals with LCR22C-D deletions had CTDs including tetralogy of Fallot. In the absence of TBX1, we hypothesized that CRKL (Crk-like), mapping to the LCR22C-D region, might contribute to the cardiac phenotype in these individuals. We created an allelic series in mice of Crkl, including a hypomorphic allele, to test for gene expression effects on phenotype. We found that the spectrum of heart defects depends on Crkl expression, occurring with analogous malformations to that in human individuals, suggesting that haploinsufficiency of CRKL could be responsible for the etiology of CTDs in individuals with nested distal deletions and might act as a genetic modifier of individuals with the typical 3 Mb deletion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Cromosomas Humanos Par 22/genética , Cardiopatías Congénitas/genética , Proteínas Nucleares/genética , Fenotipo , Duplicaciones Segmentarias en el Genoma/genética , Eliminación de Secuencia/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Ecocardiografía , Cardiopatías Congénitas/patología , Humanos , Hibridación Fluorescente in Situ , Ratones , Proteínas Nucleares/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
Am J Hum Genet ; 96(5): 753-64, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25892112

RESUMEN

The 22q11.2 deletion syndrome (22q11DS; velocardiofacial/DiGeorge syndrome; VCFS/DGS) is the most common microdeletion syndrome and the phenotypic presentation is highly variable. Approximately 65% of individuals with 22q11DS have a congenital heart defect (CHD), mostly of the conotruncal type, and/or an aortic arch defect. The etiology of this phenotypic variability is not currently known. We hypothesized that copy-number variants (CNVs) outside the 22q11.2 deleted region might increase the risk of being born with a CHD in this sensitized population. Genotyping with Affymetrix SNP Array 6.0 was performed on two groups of subjects with 22q11DS separated by time of ascertainment and processing. CNV analysis was completed on a total of 949 subjects (cohort 1, n = 562; cohort 2, n = 387), 603 with CHDs (cohort 1, n = 363; cohort 2, n = 240) and 346 with normal cardiac anatomy (cohort 1, n = 199; cohort 2, n = 147). Our analysis revealed that a duplication of SLC2A3 was the most frequent CNV identified in the first cohort. It was present in 18 subjects with CHDs and 1 subject without (p = 3.12 × 10(-3), two-tailed Fisher's exact test). In the second cohort, the SLC2A3 duplication was also significantly enriched in subjects with CHDs (p = 3.30 × 10(-2), two-tailed Fisher's exact test). The SLC2A3 duplication was the most frequent CNV detected and the only significant finding in our combined analysis (p = 2.68 × 10(-4), two-tailed Fisher's exact test), indicating that the SLC2A3 duplication might serve as a genetic modifier of CHDs and/or aortic arch anomalies in individuals with 22q11DS.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Síndrome de DiGeorge/genética , Transportador de Glucosa de Tipo 3/genética , Cardiopatías Congénitas/genética , Adulto , Aorta Torácica/fisiopatología , Síndrome de DiGeorge/fisiopatología , Femenino , Genotipo , Cardiopatías Congénitas/fisiopatología , Humanos , Masculino , Polimorfismo de Nucleótido Simple
18.
Am J Med Genet A ; 176(10): 2070-2081, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30380194

RESUMEN

The 22q11.2 deletion syndrome (22q11.2DS) is a congenital malformation and neuropsychiatric disorder caused by meiotic chromosome rearrangements. One of the goals of this review is to summarize the current state of basic research studies of 22q11.2DS. It highlights efforts to understand the mechanisms responsible for the 22q11.2 deletion that occurs in meiosis. This mechanism involves the four sets of low copy repeats (LCR22) that are dispersed in the 22q11.2 region and the deletion is mediated by nonallelic homologous recombination events. This review also highlights selected genes mapping to the 22q11.2 region that may contribute to the typical clinical findings associated with the disorder and explain that mutations in genes on the remaining allele can uncover rare recessive conditions. Another important aspect of 22q11.2DS is the existence of phenotypic heterogeneity. While some patients are mildly affected, others have severe medical, cognitive, and/or psychiatric challenges. Variability may be due in part to the presence of genetic modifiers. This review discusses current genome-wide efforts to identify such modifiers that could shed light on molecular pathways required for normal human development, cognition or behavior.


Asunto(s)
Anomalías Múltiples/genética , Cromosomas Humanos Par 22 , Síndrome de DiGeorge/genética , Mutación , Deleción Cromosómica , Síndrome de DiGeorge/etiología , Genes Recesivos , Pruebas Genéticas , Humanos , Meiosis
19.
Am J Med Genet A ; 176(10): 2172-2181, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30289625

RESUMEN

The 22q11.2 deletion syndrome is caused by non-allelic homologous recombination events during meiosis between low copy repeats (LCR22) termed A, B, C, and D. Most patients have a typical LCR22A-D (AD) deletion of 3 million base pairs (Mb). In this report, we evaluated IQ scores in 1,478 subjects with 22q11.2DS. The mean of full scale IQ, verbal IQ, and performance IQ scores in our cohort were 72.41 (standard deviation-SD of 13.72), 75.91(SD of 14.46), and 73.01(SD of 13.71), respectively. To investigate whether IQ scores are associated with deletion size, we examined individuals with the 3 Mb, AD (n = 1,353) and nested 1.5 Mb, AB (n = 74) deletions, since they comprised the largest subgroups. We found that full scale IQ was decreased by 6.25 points (p = .002), verbal IQ was decreased by 8.17 points (p = .0002) and performance IQ was decreased by 4.03 points (p = .028) in subjects with the AD versus AB deletion. Thus, individuals with the smaller, 1.5 Mb AB deletion have modestly higher IQ scores than those with the larger, 3 Mb AD deletion. Overall, the deletion of genes in the AB region largely explains the observed low IQ in the 22q11.2DS population. However, our results also indicate that haploinsufficiency of genes in the LCR22B-D region (BD) exert an additional negative impact on IQ. Furthermore, we did not find evidence of a confounding effect of severe congenital heart disease on IQ scores in our cohort.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 22 , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/psicología , Adolescente , Adulto , Niño , Femenino , Humanos , Discapacidad Intelectual/genética , Pruebas de Inteligencia , Masculino
20.
Hum Mol Genet ; 23(16): 4215-31, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24705356

RESUMEN

Velo-cardio-facial/DiGeorge syndrome, also known as 22q11.2 deletion syndrome, is a congenital anomaly disorder characterized by craniofacial anomalies including velo-pharyngeal insufficiency, facial muscle hypotonia and feeding difficulties, in part due to hypoplasia of the branchiomeric muscles. Inactivation of both alleles of mouse Tbx1, encoding a T-box transcription factor, deleted on chromosome 22q11.2, results in reduction or loss of branchiomeric muscles. To identify downstream pathways, we performed gene profiling of microdissected pharyngeal arch one (PA1) from Tbx1(+/+) and Tbx1(-/-) embryos at stages E9.5 (somites 20-25) and E10.5 (somites 30-35). Basic helix-loop-helix (bHLH) transcription factors were reduced, while secondary heart field genes were increased in expression early and were replaced by an increase in expression of cellular stress response genes later, suggesting a change in gene expression patterns or cell populations. Lineage tracing studies using Mesp1(Cre) and T-Cre drivers showed that core mesoderm cells within PA1 were present at E9.5 but were greatly reduced by E10.5 in Tbx1(-/-) embryos. Using Tbx1(Cre) knock-in mice, we found that cells are lost due to apoptosis, consistent with increase in expression of cellular stress response genes at E10.5. To determine whether Tbx1 is required autonomously in the core mesoderm, we used Mesp1(Cre) and T-Cre mesodermal drivers in combination with inactivate Tbx1 and found reduction or loss of branchiomeric muscles from PA1. These mechanistic studies inform us that Tbx1 is required upstream of key myogenic genes needed for core mesoderm cell survival and fate, between E9.5 and E10.5, resulting in formation of the branchiomeric muscles.


Asunto(s)
Diferenciación Celular/genética , Masticación/genética , Músculos/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Animales , Apoptosis/genética , Supervivencia Celular/genética , Desarrollo Embrionario/genética , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Faringe/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA