Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 587(7834): 460-465, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33149301

RESUMEN

Atrial fibrillation, the most common cardiac arrhythmia, is an important contributor to mortality and morbidity, and particularly to the risk of stroke in humans1. Atrial-tissue fibrosis is a central pathophysiological feature of atrial fibrillation that also hampers its treatment; the underlying molecular mechanisms are poorly understood and warrant investigation given the inadequacy of present therapies2. Here we show that calcitonin, a hormone product of the thyroid gland involved in bone metabolism3, is also produced by atrial cardiomyocytes in substantial quantities and acts as a paracrine signal that affects neighbouring collagen-producing fibroblasts to control their proliferation and secretion of extracellular matrix proteins. Global disruption of calcitonin receptor signalling in mice causes atrial fibrosis and increases susceptibility to atrial fibrillation. In mice in which liver kinase B1 is knocked down specifically in the atria, atrial-specific knockdown of calcitonin promotes atrial fibrosis and increases and prolongs spontaneous episodes of atrial fibrillation, whereas atrial-specific overexpression of calcitonin prevents both atrial fibrosis and fibrillation. Human patients with persistent atrial fibrillation show sixfold lower levels of myocardial calcitonin compared to control individuals with normal heart rhythm, with loss of calcitonin receptors in the fibroblast membrane. Although transcriptome analysis of human atrial fibroblasts reveals little change after exposure to calcitonin, proteomic analysis shows extensive alterations in extracellular matrix proteins and pathways related to fibrogenesis, infection and immune responses, and transcriptional regulation. Strategies to restore disrupted myocardial calcitonin signalling thus may offer therapeutic avenues for patients with atrial fibrillation.


Asunto(s)
Arritmias Cardíacas/metabolismo , Calcitonina/metabolismo , Fibrinógeno/biosíntesis , Atrios Cardíacos/metabolismo , Miocardio/metabolismo , Comunicación Paracrina , Animales , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Fibrilación Atrial , Colágeno Tipo I/metabolismo , Femenino , Fibroblastos/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Atrios Cardíacos/citología , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Humanos , Masculino , Ratones , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Receptores de Calcitonina/metabolismo
2.
Trends Biochem Sci ; 45(2): 108-122, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31679840

RESUMEN

Ticks are hematophagous arachnids that parasitize mammals and other hosts, feeding on their blood. Ticks secrete numerous salivary factors that enhance host blood flow or suppress the host inflammatory response. The recruitment of leukocytes, a hallmark of inflammation, is regulated by chemokines, which activate chemokine receptors on the leukocytes. Ticks target this process by secreting glycoproteins called Evasins, which bind to chemokines and prevent leukocyte recruitment. This review describes the recent discovery of numerous Evasins produced by ticks, their classification into two structural and functional classes, and the efficacy of Evasins in animal models of inflammatory diseases. The review also proposes a standard nomenclature system for Evasins and discusses the potential of repurposing or engineering Evasins as therapeutic anti-inflammatory agents.


Asunto(s)
Quimiocinas/antagonistas & inhibidores , Proteínas de Insectos/metabolismo , Proteínas y Péptidos Salivales/metabolismo , Garrapatas/metabolismo , Animales , Leucocitos/metabolismo , Receptores de Quimiocina/metabolismo , Terminología como Asunto
3.
J Biol Chem ; 295(32): 10926-10939, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32471866

RESUMEN

Chemokines mediate leukocyte migration and homeostasis and are key targets in inflammatory diseases including atherosclerosis, cytokine storm, and chronic autoimmune disease. Chemokine redundancy and ensuing network robustness has frustrated therapeutic development. Salivary evasins from ticks bind multiple chemokines to overcome redundancy and are effective in several preclinical disease models. Their clinical development has not progressed because of concerns regarding potential immunogenicity, parenteral delivery, and cost. Peptides mimicking protein activity can overcome the perceived limitations of therapeutic proteins. Here we show that peptides possessing multiple chemokine-binding and anti-inflammatory activities can be developed from the chemokine-binding site of an evasin. We used hydrogen-deuterium exchange MS to map the binding interface of the evasin P672 that physically interacts with C-C motif chemokine ligand (CCL) 8 and synthesized a 16-mer peptide (BK1.1) based on this interface region in evasin P672. Fluorescent polarization and native MS approaches showed that BK1.1 binds CCL8, CCL7, and CCL18 and disrupts CCL8 homodimerization. We show that a BK1.1 derivative, BK1.3, has substantially improved ability to disrupt P672 binding to CCL8, CCL2, and CCL3 in an AlphaScreen assay. Using isothermal titration calorimetry, we show that BK1.3 directly binds CCL8. BK1.3 also has substantially improved ability to inhibit CCL8, CCL7, CCL2, and CCL3 chemotactic function in vitro We show that local as well as systemic administration of BK1.3 potently blocks inflammation in vivo Identification and characterization of the chemokine-binding interface of evasins could thus inspire the development of novel anti-inflammatory peptides that therapeutically target the chemokine network in inflammatory diseases.


Asunto(s)
Antiinflamatorios/química , Quimiocina CCL8/metabolismo , Péptidos/química , Ingeniería de Proteínas , Receptores de Quimiocina/metabolismo , Secuencia de Aminoácidos , Animales , Antiinflamatorios/farmacología , Dimerización , Humanos , Espectrometría de Masas/métodos , Péptidos/farmacología , Unión Proteica , Homología de Secuencia de Aminoácido , Garrapatas/metabolismo
4.
Circ Res ; 124(4): 553-563, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30582441

RESUMEN

RATIONALE: Familial recurrence studies provide strong evidence for a genetic component to the predisposition to sporadic, nonsyndromic Tetralogy of Fallot (TOF), the most common cyanotic congenital heart disease phenotype. Rare genetic variants have been identified as important contributors to the risk of congenital heart disease, but relatively small numbers of TOF cases have been studied to date. OBJECTIVE: We used whole exome sequencing to assess the prevalence of unique, deleterious variants in the largest cohort of nonsyndromic TOF patients reported to date. METHODS AND RESULTS: Eight hundred twenty-nine TOF patients underwent whole exome sequencing. The presence of unique, deleterious variants was determined; defined by their absence in the Genome Aggregation Database and a scaled combined annotation-dependent depletion score of ≥20. The clustering of variants in 2 genes, NOTCH1 and FLT4, surpassed thresholds for genome-wide significance (assigned as P<5×10-8) after correction for multiple comparisons. NOTCH1 was most frequently found to harbor unique, deleterious variants. Thirty-one changes were observed in 37 probands (4.5%; 95% CI, 3.2%-6.1%) and included 7 loss-of-function variants 22 missense variants and 2 in-frame indels. Sanger sequencing of the unaffected parents of 7 cases identified 5 de novo variants. Three NOTCH1 variants (p.G200R, p.C607Y, and p.N1875S) were subjected to functional evaluation, and 2 showed a reduction in Jagged1-induced NOTCH signaling. FLT4 variants were found in 2.4% (95% CI, 1.6%-3.8%) of TOF patients, with 21 patients harboring 22 unique, deleterious variants. The variants identified were distinct to those that cause the congenital lymphoedema syndrome Milroy disease. In addition to NOTCH1, FLT4 and the well-established TOF gene, TBX1, we identified potential association with variants in several other candidates, including RYR1, ZFPM1, CAMTA2, DLX6, and PCM1. CONCLUSIONS: The NOTCH1 locus is the most frequent site of genetic variants predisposing to nonsyndromic TOF, followed by FLT4. Together, variants in these genes are found in almost 7% of TOF patients.


Asunto(s)
Exoma , Tasa de Mutación , Tetralogía de Fallot/genética , Autoantígenos/genética , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Proteínas de Homeodominio/genética , Humanos , Mutación con Pérdida de Función , Mutación Missense , Proteínas Nucleares/genética , Receptor Notch1/genética , Transactivadores/genética , Factores de Transcripción/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
5.
J Biol Chem ; 294(29): 11199-11212, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31167786

RESUMEN

Tick evasins (EVAs) bind either CC- or CXC-chemokines by a poorly understood promiscuous or "one-to-many" mechanism to neutralize inflammation. Because EVAs potently inhibit inflammation in many preclinical models, highlighting their potential as biological therapeutics for inflammatory diseases, we sought to further unravel the CXC-chemokine-EVA interactions. Using yeast surface display, we identified and characterized 27 novel CXC-chemokine-binding evasins homologous to EVA3 and defined two functional classes. The first, which included EVA3, exclusively bound ELR+ CXC-chemokines, whereas the second class bound both ELR+ and ELR- CXC-chemokines, in several cases including CXC-motif chemokine ligand 10 (CXCL10) but, surprisingly, not CXCL8. The X-ray crystal structure of EVA3 at a resolution of 1.79 Å revealed a single antiparallel ß-sheet with six conserved cysteine residues forming a disulfide-bonded knottin scaffold that creates a contiguous solvent-accessible surface. Swapping analyses identified distinct knottin scaffold segments necessary for different CXC-chemokine-binding activities, implying that differential ligand positioning, at least in part, plays a role in promiscuous binding. Swapping segments also transferred chemokine-binding activity, resulting in a hybrid EVA with dual CXCL10- and CXCL8-binding activities. The solvent-accessible surfaces of the knottin scaffold segments have distinctive shape and charge, which we suggest drives chemokine-binding specificity. These studies provide structural and mechanistic insight into how CXC-chemokine-binding tick EVAs achieve class specificity but also engage in promiscuous binding.


Asunto(s)
Quimiocinas CXC/metabolismo , Miniproteínas Nodales de Cistina/metabolismo , Receptores de Quimiocina/metabolismo , Garrapatas/metabolismo , Animales , Cristalografía por Rayos X , Unión Proteica , Conformación Proteica , Receptores de Quimiocina/genética , Receptores de Quimiocina/aislamiento & purificación , Especificidad de la Especie , Garrapatas/clasificación , Levaduras/genética
6.
J Biol Chem ; 293(16): 6134-6146, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29487134

RESUMEN

Tick chemokine-binding proteins (evasins) are an emerging class of biologicals that target multiple chemokines and show anti-inflammatory activities in preclinical disease models. Using yeast surface display, we identified a CCL8-binding evasin, P672, from the tick Rhipicephalus pulchellus We found that P672 binds CCL8 and eight other CC-class chemokines with a Kd < 10 nm and four other CC chemokines with a Kd between 10 and 100 nm and neutralizes CCL3, CCL3L1, and CCL8 with an IC50 < 10 nm The CC chemokine-binding profile was distinct from that of evasin 1 (EVA1), which does not bind CCL8. We also show that P672's binding activity can be markedly modulated by the location of a StrepII-His purification tag. Combining native MS and bottom-up proteomics, we further demonstrated that P672 is glycosylated and forms a 1:1 complex with CCL8, disrupting CCL8 homodimerization. Homology modeling of P672 using the crystal structure of the EVA1 and CCL3 complex as template suggested that 44 N-terminal residues of P672 form most of the contacts with CCL8. Replacing the 29 N-terminal residues of EVA1 with the 44 N-terminal residues of P672 enabled this hybrid evasin to bind and neutralize CCL8, indicating that the CCL8-binding properties of P672 reside, in part, in its N-terminal residues. This study shows that the function of certain tick evasins can be manipulated simply by adding a tag. We conclude that homology modeling helps identify regions with transportable chemokine-binding functions within evasins, which can be used to construct hybrid evasins with altered properties.


Asunto(s)
Proteínas de Artrópodos/metabolismo , Quimiocinas/metabolismo , Receptores de Quimiocina/metabolismo , Garrapatas/metabolismo , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Glicosilación , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Receptores de Quimiocina/química , Receptores de Quimiocina/genética , Saccharomyces cerevisiae/genética , Espectrometría de Masas en Tándem
7.
Development ; 143(15): 2716-23, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27317809

RESUMEN

The Polycomb repressive complexes PRC1 and PRC2 are key mediators of heritable gene silencing in multicellular organisms. Here, we characterise AEBP2, a known PRC2 co-factor which, in vitro, has been shown to stimulate PRC2 activity. We show that AEBP2 localises specifically to PRC2 target loci, including the inactive X chromosome. Proteomic analysis confirms that AEBP2 associates exclusively with PRC2 complexes. However, analysis of embryos homozygous for a targeted mutation of Aebp2 unexpectedly revealed a Trithorax phenotype, normally linked to antagonism of Polycomb function. Consistent with this, we observe elevated levels of PRC2-mediated histone H3K27 methylation at target loci in Aebp2 mutant embryonic stem cells (ESCs). We further demonstrate that mutant ESCs assemble atypical hybrid PRC2 subcomplexes, potentially accounting for enhancement of Polycomb activity, and suggesting that AEBP2 normally plays a role in defining the mutually exclusive composition of PRC2 subcomplexes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Proteómica/métodos , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Femenino , Histonas/metabolismo , Ratones , Mutación/genética , Proteínas Nucleares/genética , Complejo Represivo Polycomb 2/genética , Proteínas Represoras
8.
Hum Mol Genet ; 25(11): 2331-2341, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26965164

RESUMEN

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


Asunto(s)
Cromosomas Humanos Par 20/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Cardiopatías Congénitas/genética , Mapeo Cromosómico , Estudios de Cohortes , Femenino , Genotipo , Cardiopatías Congénitas/fisiopatología , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Polimorfismo de Nucleótido Simple
9.
BMC Dev Biol ; 17(1): 6, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28446132

RESUMEN

BACKGROUND: Loss of proprotein convertase subtilisin/kexin type 5 (Pcsk5) results in multiple developmental anomalies including cardiac malformations, caudal regression, pre-sacral mass, renal agenesis, anteroposterior patterning defects, and tracheo-oesophageal and anorectal malformations, and is a model for VACTERL/caudal regression/Currarino syndromes (VACTERL association - Vertebral anomalies, Anal atresia, Cardiac defects, Tracheoesophageal fistula and/or Esophageal atresia, Renal & Radial anomalies and Limb defects). RESULTS: Using magnetic resonance imaging (MRI), we examined heart development in mouse embryos with zygotic and cardiac specific deletion of Pcsk5. We show that conditional deletion of Pcsk5 in all epiblastic lineages recapitulates all developmental malformations except for tracheo-esophageal malformations. Using a conditional deletion strategy, we find that there is an essential and specific requirement for Pcsk5 in the cranio-cardiac mesoderm for cardiogenesis, but not for conotruncal septation or any other aspect of embryonic development. Surprisingly, deletion of Pcsk5 in cardiogenic or pharyngeal mesodermal progenitors that form later from the cranio-cardiac mesoderm does not affect heart development. Neither is Pcsk5 essential in the neural crest, which drives conotruncal septation. CONCLUSIONS: Our results suggest that Pcsk5 may have an essential and early role in the cranio-cardiac mesoderm for heart development. Alternatively, it is possible that Pcsk5 may still play a critical role in Nkx2.5-expressing cardiac progenitors, with persistence of mRNA or protein accounting for the lack of effect of deletion on heart development.


Asunto(s)
Corazón/embriología , Mesodermo/embriología , Organogénesis/genética , Proproteína Convertasa 5/genética , Proproteína Convertasa 5/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Ratones , Ratones Noqueados , Mutación
10.
Am J Hum Genet ; 94(4): 574-85, 2014 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-24702954

RESUMEN

Congenital heart defects (CHDs) are the most common birth defect worldwide and are a leading cause of neonatal mortality. Nonsyndromic atrioventricular septal defects (AVSDs) are an important subtype of CHDs for which the genetic architecture is poorly understood. We performed exome sequencing in 13 parent-offspring trios and 112 unrelated individuals with nonsyndromic AVSDs and identified five rare missense variants (two of which arose de novo) in the highly conserved gene NR2F2, a very significant enrichment (p = 7.7 × 10(-7)) compared to 5,194 control subjects. We identified three additional CHD-affected families with other variants in NR2F2 including a de novo balanced chromosomal translocation, a de novo substitution disrupting a splice donor site, and a 3 bp duplication that cosegregated in a multiplex family. NR2F2 encodes a pleiotropic developmental transcription factor, and decreased dosage of NR2F2 in mice has been shown to result in abnormal development of atrioventricular septa. Via luciferase assays, we showed that all six coding sequence variants observed in individuals significantly alter the activity of NR2F2 on target promoters.


Asunto(s)
Factor de Transcripción COUP II/genética , Cardiopatías Congénitas/genética , Animales , Sitios de Unión , Factor de Transcripción COUP II/metabolismo , Línea Celular , Exoma , Femenino , Humanos , Masculino , Ratones , Mutación Missense , Linaje , Estudios Prospectivos , Transcripción Genética
11.
Mol Ther ; 24(10): 1745-1759, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27480772

RESUMEN

Therapeutic angiogenesis is a major goal of regenerative medicine, but no clinically approved small molecule exists that enhances new blood vessel formation. Here we show, using a phenotype-driven high-content imaging screen of an annotated chemical library of 1,280 bioactive small molecules, that the retinoid agonist Tazarotene, enhances in vitro angiogenesis, promoting branching morphogenesis, and tubule remodeling. The proangiogenic phenotype is mediated by retinoic acid receptor but not retinoic X receptor activation, and is characterized by secretion of the proangiogenic factors hepatocyte growth factor, vascular endothelial growth factor, plasminogen activator, urokinase and placental growth factor, and reduced secretion of the antiangiogenic factor pentraxin-3 from adjacent fibroblasts. In vivo, Tazarotene enhanced the growth of mature and functional microvessels in Matrigel implants and wound healing models, and increased blood flow. Notably, in ear punch wound healing model, Tazarotene promoted tissue repair characterized by rapid ear punch closure with normal-appearing skin containing new hair follicles, and maturing collagen fibers. Our study suggests that Tazarotene, an FDA-approved small molecule, could be potentially exploited for therapeutic applications in neovascularization and wound healing.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Fibroblastos/citología , Ácidos Nicotínicos/administración & dosificación , Receptores de Ácido Retinoico/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Inductores de la Angiogénesis/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ácidos Nicotínicos/farmacología , Transducción de Señal
12.
Genet Med ; 18(2): 189-98, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25996639

RESUMEN

PURPOSE: The genetic etiology of atrioventricular septal defect (AVSD) is unknown in 40% cases. Conventional sequencing and arrays have identified the etiology in only a minority of nonsyndromic individuals with AVSD. METHODS: Whole-exome sequencing was performed in 81 unrelated probands with AVSD to identify potentially causal variants in a comprehensive set of 112 genes with strong biological relevance to AVSD. RESULTS: A significant enrichment of rare and rare damaging variants was identified in the gene set, compared with controls (odds ratio (OR): 1.52; 95% confidence interval (CI): 1.35-1.71; P = 4.8 × 10(-11)). The enrichment was specific to AVSD probands, compared with a cohort without AVSD with tetralogy of Fallot (OR: 2.25; 95% CI: 1.84-2.76; P = 2.2 × 10(-16)). Six genes (NIPBL, CHD7, CEP152, BMPR1a, ZFPM2, and MDM4) were enriched for rare variants in AVSD compared with controls, including three syndrome-associated genes (NIPBL, CHD7, and CEP152). The findings were confirmed in a replication cohort of 81 AVSD probands. CONCLUSION: Mutations in genes with strong biological relevance to AVSD, including syndrome-associated genes, can contribute to AVSD, even in those with isolated heart disease. The identification of a gene set associated with AVSD will facilitate targeted genetic screening in this cohort.


Asunto(s)
Exoma , Variación Genética , Defectos de los Tabiques Cardíacos/genética , Adolescente , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Masculino , Mutación , Fenotipo , Análisis de Secuencia de ADN
13.
Circ Res ; 115(1): 23-31, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24748541

RESUMEN

RATIONALE: 22q11 deletion syndrome arises from recombination between low-copy repeats on chromosome 22. Typical deletions result in hemizygosity for TBX1 associated with congenital cardiovascular disease. Deletions distal to the typically deleted region result in a similar cardiac phenotype but lack in extracardiac features of the syndrome, suggesting that a second haploinsufficient gene maps to this interval. OBJECTIVE: The transcription factor HIC2 is lost in most distal deletions, as well as in a minority of typical deletions. We used mouse models to test the hypothesis that HIC2 hemizygosity causes congenital heart disease. METHODS AND RESULTS: We created a genetrap mouse allele of Hic2. The genetrap reporter was expressed in the heart throughout the key stages of cardiac morphogenesis. Homozygosity for the genetrap allele was embryonic lethal before embryonic day E10.5, whereas the heterozygous condition exhibited a partially penetrant late lethality. One third of heterozygous embryos had a cardiac phenotype. MRI demonstrated a ventricular septal defect with over-riding aorta. Conditional targeting indicated a requirement for Hic2 within the Nkx2.5+ and Mesp1+ cardiovascular progenitor lineages. Microarray analysis revealed increased expression of Bmp10. CONCLUSIONS: Our results demonstrate a novel role for Hic2 in cardiac development. Hic2 is the first gene within the distal 22q11 interval to have a demonstrated haploinsufficient cardiac phenotype in mice. Together our data suggest that HIC2 haploinsufficiency likely contributes to the cardiac defects seen in distal 22q11 deletion syndrome.


Asunto(s)
Síndrome de Deleción 22q11/etiología , Corazón/embriología , Factores de Transcripción de Tipo Kruppel/fisiología , Proteínas Supresoras de Tumor/fisiología , Síndrome de Deleción 22q11/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Proteínas Morfogenéticas Óseas/fisiología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Cardiopatías Congénitas/etiología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Morfogénesis , Mutagénesis , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología , Proteínas Supresoras de Tumor/genética
14.
Hum Mutat ; 36(12): 1197-204, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26350204

RESUMEN

To identify genetic causes of intellectual disability (ID), we screened a cohort of 986 individuals with moderate to severe ID for variants in 565 known or candidate ID-associated genes using targeted next-generation sequencing. Likely pathogenic rare variants were found in ∼11% of the cases (113 variants in 107/986 individuals: ∼8% of the individuals had a likely pathogenic loss-of-function [LoF] variant, whereas ∼3% had a known pathogenic missense variant). Variants in SETD5, ATRX, CUL4B, MECP2, and ARID1B were the most common causes of ID. This study assessed the value of sequencing a cohort of probands to provide a molecular diagnosis of ID, without the availability of DNA from both parents for de novo sequence analysis. This modeling is clinically relevant as 28% of all UK families with dependent children are single parent households. In conclusion, to diagnose patients with ID in the absence of parental DNA, we recommend investigation of all LoF variants in known genes that cause ID and assessment of a limited list of proven pathogenic missense variants in these genes. This will provide 11% additional diagnostic yield beyond the 10%-15% yield from array CGH alone.


Asunto(s)
Estudios de Asociación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Discapacidad Intelectual/genética , Alelos , Estudios de Cohortes , Biología Computacional/métodos , Femenino , Humanos , Patrón de Herencia , Masculino , Mutación , Polimorfismo de Nucleótido Simple
15.
Hum Mol Genet ; 22(16): 3269-82, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23595884

RESUMEN

Glucocorticoids are vital for the structural and functional maturation of foetal organs, yet excessive foetal exposure is detrimental to adult cardiovascular health. To elucidate the role of glucocorticoid signalling in late-gestation cardiovascular maturation, we have generated mice with conditional disruption of glucocorticoid receptor (GR) in cardiomyocytes and vascular smooth muscle cells using smooth muscle protein 22-driven Cre recombinase (SMGRKO mice) and compared them with mice with global deficiency in GR (GR(-/-)). Echocardiography shows impaired heart function in both SMGRKO and GR(-/-) mice at embryonic day (E)17.5, associated with generalized oedema. Cardiac ultrastructure is markedly disrupted in both SMGRKO and GR(-/-) mice at E17.5, with short, disorganized myofibrils and cardiomyocytes that fail to align in the compact myocardium. Failure to induce critical genes involved in contractile function, calcium handling and energy metabolism underpins this common phenotype. However, although hearts of GR(-/-) mice are smaller, with 22% reduced ventricular volume at E17.5, SMGRKO hearts are normally sized. Moreover, while levels of mRNA encoding atrial natriuretic peptide are reduced in E17.5 GR(-/-) hearts, they are normal in foetal SMGRKO hearts. These data demonstrate that structural, functional and biochemical maturation of the foetal heart is dependent on glucocorticoid signalling within cardiomyocytes and vascular smooth muscle, though some aspects of heart maturation (size, ANP expression) are independent of GR at these key sites.


Asunto(s)
Corazón Fetal/crecimiento & desarrollo , Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal , Animales , Corticosterona/sangre , Corticosterona/fisiología , Corazón Fetal/fisiología , Corazón/embriología , Corazón/fisiología , Ratones , Ratones Transgénicos , Músculo Liso Vascular/embriología , Músculo Liso Vascular/metabolismo , Contracción Miocárdica , Miocardio/ultraestructura , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miofibrillas/ultraestructura
16.
Hum Mol Genet ; 22(7): 1473-81, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23297363

RESUMEN

We conducted a genome-wide association study to search for risk alleles associated with Tetralogy of Fallot (TOF), using a northern European discovery set of 835 cases and 5159 controls. A region on chromosome 12q24 was associated (P = 1.4 × 10(-7)) and replicated convincingly (P = 3.9 × 10(-5)) in 798 cases and 2931 controls [per allele odds ratio (OR) = 1.27 in replication cohort, P = 7.7 × 10(-11) in combined populations]. Single nucleotide polymorphisms in the glypican 5 gene on chromosome 13q32 were also associated (P = 1.7 × 10(-7)) and replicated convincingly (P = 1.2 × 10(-5)) in 789 cases and 2927 controls (per allele OR = 1.31 in replication cohort, P = 3.03 × 10(-11) in combined populations). Four additional regions on chromosomes 10, 15 and 16 showed suggestive association accompanied by nominal replication. This study, the first genome-wide association study of a congenital heart malformation phenotype, provides evidence that common genetic variation influences the risk of TOF.


Asunto(s)
Cromosomas Humanos Par 12/genética , Cromosomas Humanos Par 13/genética , Estudio de Asociación del Genoma Completo , Tetralogía de Fallot/genética , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Sitios Genéticos , Humanos , Desequilibrio de Ligamiento , Masculino , Polimorfismo de Nucleótido Simple
17.
Am J Hum Genet ; 91(3): 489-501, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22939634

RESUMEN

Previous studies have shown that copy-number variants (CNVs) contribute to the risk of complex developmental phenotypes. However, the contribution of global CNV burden to the risk of sporadic congenital heart disease (CHD) remains incompletely defined. We generated genome-wide CNV data by using Illumina 660W-Quad SNP arrays in 2,256 individuals with CHD, 283 trio CHD-affected families, and 1,538 controls. We found association of rare genic deletions with CHD risk (odds ratio [OR] = 1.8, p = 0.0008). Rare deletions in study participants with CHD had higher gene content (p = 0.001) with higher haploinsufficiency scores (p = 0.03) than they did in controls, and they were enriched with Wnt-signaling genes (p = 1 × 10(-5)). Recurrent 15q11.2 deletions were associated with CHD risk (OR = 8.2, p = 0.02). Rare de novo CNVs were observed in ~5% of CHD trios; 10 out of 11 occurred on the paternally transmitted chromosome (p = 0.01). Some of the rare de novo CNVs spanned genes known to be involved in heart development (e.g., HAND2 and GJA5). Rare genic deletions contribute ~4% of the population-attributable risk of sporadic CHD. Second to previously described CNVs at 1q21.1, deletions at 15q11.2 and those implicating Wnt signaling are the most significant contributors to the risk of sporadic CHD. Rare de novo CNVs identified in CHD trios exhibit paternal origin bias.


Asunto(s)
Variaciones en el Número de Copia de ADN , Eliminación de Gen , Cardiopatías Congénitas/genética , Niño , Cromosomas Humanos Par 15 , Cromosomas Humanos Par 8 , Padre , Femenino , Dosificación de Gen , Humanos , Masculino , Polimorfismo de Nucleótido Simple
18.
J Biol Chem ; 288(41): 29836-45, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-23965989

RESUMEN

Tetrahydrobiopterin (BH4) is a required cofactor for the synthesis of NO by NOS. Bioavailability of BH4 is a critical factor in regulating the balance between NO and superoxide production by endothelial NOS (eNOS coupling). Crystal structures of the mouse inducible NOS oxygenase domain reveal a homologous BH4-binding site located in the dimer interface and a conserved tryptophan residue that engages in hydrogen bonding or aromatic stacking interactions with the BH4 ring. The role of this residue in eNOS coupling remains unexplored. We overexpressed human eNOS W447A and W447F mutants in novel cell lines with tetracycline-regulated expression of human GTP cyclohydrolase I, the rate-limiting enzyme in BH4 synthesis, to determine the importance of BH4 and Trp-447 in eNOS uncoupling. NO production was abolished in eNOS-W447A cells and diminished in cells expressing W447F, despite high BH4 levels. eNOS-derived superoxide production was significantly elevated in W447A and W447F versus wild-type eNOS, and this was sufficient to oxidize BH4 to 7,8-dihydrobiopterin. In uncoupled, BH4-deficient cells, the deleterious effects of W447A mutation were greatly exacerbated, resulting in further attenuation of NO and greatly increased superoxide production. eNOS dimerization was attenuated in W447A eNOS cells and further reduced in BH4-deficient cells, as demonstrated using a novel split Renilla luciferase biosensor. Reduction of cellular BH4 levels resulted in a switch from an eNOS dimer to an eNOS monomer. These data reveal a key role for Trp-447 in determining NO versus superoxide production by eNOS, by effects on BH4-dependent catalysis, and by modulating eNOS dimer formation.


Asunto(s)
Biopterinas/análogos & derivados , Óxido Nítrico Sintasa de Tipo III/metabolismo , Triptófano/metabolismo , Células 3T3 , Sustitución de Aminoácidos , Animales , Sitios de Unión/genética , Biocatálisis , Biopterinas/química , Biopterinas/metabolismo , Western Blotting , Dominio Catalítico , Humanos , Ratones , Modelos Moleculares , Mutación , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/química , Óxido Nítrico Sintasa de Tipo III/genética , Oxidación-Reducción , Multimerización de Proteína , Superóxidos/metabolismo , Triptófano/genética
19.
Hum Mol Genet ; 21(7): 1513-20, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22199024

RESUMEN

Recurrent rearrangements of chromosome 1q21.1 that occur via non-allelic homologous recombination have been associated with variable phenotypes exhibiting incomplete penetrance, including congenital heart disease (CHD). However, the gene or genes within the ~1 Mb critical region responsible for each of the associated phenotypes remains unknown. We examined the 1q21.1 locus in 948 patients with tetralogy of Fallot (TOF), 1488 patients with other forms of CHD and 6760 ethnically matched controls using single nucleotide polymorphism genotyping arrays (Illumina 660W and Affymetrix 6.0) and multiplex ligation-dependent probe amplification. We found that duplication of 1q21.1 was more common in cases of TOF than in controls [odds ratio (OR) 30.9, 95% confidence interval (CI) 8.9-107.6); P = 2.2 × 10(-7)], but deletion was not. In contrast, deletion of 1q21.1 was more common in cases of non-TOF CHD than in controls [OR 5.5 (95% CI 1.4-22.0); P = 0.04] while duplication was not. We also detected rare (n = 3) 100-200 kb duplications within the critical region of 1q21.1 in cases of TOF. These small duplications encompassed a single gene in common, GJA5, and were enriched in cases of TOF in comparison to controls [OR = 10.7 (95% CI 1.8-64.3), P = 0.01]. These findings show that duplication and deletion at chromosome 1q21.1 exhibit a degree of phenotypic specificity in CHD, and implicate GJA5 as the gene responsible for the CHD phenotypes observed with copy number imbalances at this locus.


Asunto(s)
Deleción Cromosómica , Duplicación Cromosómica , Cromosomas Humanos Par 1 , Conexinas/genética , Cardiopatías Congénitas/genética , Tetralogía de Fallot/genética , Duplicación de Gen , Humanos , Fenotipo , Proteína alfa-5 de Unión Comunicante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA