Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 320(5): H1873-H1886, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33739154

RESUMEN

Compared with acyanotic congenital heart disease (CHD), cyanotic CHD has an increased risk of lifelong mortality and morbidity. These adverse outcomes may be attributed to delayed cardiomyocyte maturation, since the transition from a hypoxic fetal milieu to oxygen-rich postnatal environment is disrupted. We established a rodent model to replicate hypoxic myocardial conditions spanning perinatal development, and tested the hypothesis that chronic hypoxia impairs cardiac development. Pregnant mice were housed in hypoxia beginning at embryonic day 16. Pups stayed in hypoxia until postnatal day (P)8 when cardiac development is nearly complete. Global gene expression was quantified at P8 and at P30, after recovering in normoxia. Phenotypic testing included electrocardiogram, echocardiogram, and ex vivo electrophysiology study. Hypoxic P8 animals were 47% smaller than controls with preserved heart size. Gene expression was grossly altered by hypoxia at P8 (1,427 genes affected), but normalized after recovery (P30). Electrocardiograms revealed bradycardia and slowed conduction velocity in hypoxic animals at P8, with noticeable resolution after recovery (P30). Notable differences that persisted after recovery (P30) included a 65% prolongation in ventricular effective refractory period, sinus node dysfunction, 23% reduction in ejection fraction, and 16% reduction in fractional shortening in animals exposed to hypoxia. We investigated the impact of chronic hypoxia on the developing heart. Perinatal hypoxia was associated with changes in gene expression and cardiac function. Persistent changes to the electrophysiological substrate and contractile function warrant further investigation and may contribute to adverse outcomes observed in the cyanotic CHD population.NEW & NOTEWORTHY We utilized a new mouse model of chronic perinatal hypoxia to simulate the hypoxic myocardial conditions present in cyanotic congenital heart disease. Hypoxia caused numerous abnormalities in cardiomyocyte gene expression, the electrophysiologic substrate of the heart, and contractile function. Taken together, alterations observed in the neonatal period suggest delayed cardiac development immediately following hypoxia.


Asunto(s)
Cianosis/etiología , Corazón Fetal/crecimiento & desarrollo , Cardiopatías Congénitas/etiología , Hipoxia/complicaciones , Factores de Edad , Animales , Animales Recién Nacidos , Enfermedad Crónica , Cianosis/genética , Cianosis/metabolismo , Cianosis/fisiopatología , Modelos Animales de Enfermedad , Femenino , Corazón Fetal/metabolismo , Hipoxia Fetal/complicaciones , Hipoxia Fetal/genética , Hipoxia Fetal/metabolismo , Hipoxia Fetal/fisiopatología , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/fisiopatología , Frecuencia Cardíaca , Hipoxia/genética , Hipoxia/metabolismo , Hipoxia/fisiopatología , Ratones , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Organogénesis , Embarazo , Efectos Tardíos de la Exposición Prenatal
2.
Nature ; 521(7553): 520-4, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25807483

RESUMEN

Congenital heart disease (CHD) is the most prevalent birth defect, affecting nearly 1% of live births; the incidence of CHD is up to tenfold higher in human fetuses. A genetic contribution is strongly suggested by the association of CHD with chromosome abnormalities and high recurrence risk. Here we report findings from a recessive forward genetic screen in fetal mice, showing that cilia and cilia-transduced cell signalling have important roles in the pathogenesis of CHD. The cilium is an evolutionarily conserved organelle projecting from the cell surface with essential roles in diverse cellular processes. Using echocardiography, we ultrasound scanned 87,355 chemically mutagenized C57BL/6J fetal mice and recovered 218 CHD mouse models. Whole-exome sequencing identified 91 recessive CHD mutations in 61 genes. This included 34 cilia-related genes, 16 genes involved in cilia-transduced cell signalling, and 10 genes regulating vesicular trafficking, a pathway important for ciliogenesis and cell signalling. Surprisingly, many CHD genes encoded interacting proteins, suggesting that an interactome protein network may provide a larger genomic context for CHD pathogenesis. These findings provide novel insights into the potential Mendelian genetic contribution to CHD in the fetal population, a segment of the human population not well studied. We note that the pathways identified show overlap with CHD candidate genes recovered in CHD patients, suggesting that they may have relevance to the more complex genetics of CHD overall. These CHD mouse models and >8,000 incidental mutations have been sperm archived, creating a rich public resource for human disease modelling.


Asunto(s)
Cilios/patología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Animales , Cilios/diagnóstico por imagen , Cilios/genética , Cilios/fisiología , Análisis Mutacional de ADN , Electrocardiografía , Exoma/genética , Genes Recesivos , Pruebas Genéticas , Cardiopatías Congénitas/diagnóstico por imagen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Transducción de Señal , Ultrasonografía
3.
PLoS Genet ; 12(2): e1005821, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26918822

RESUMEN

Heterotaxy, a birth defect involving left-right patterning defects, and primary ciliary dyskinesia (PCD), a sinopulmonary disease with dyskinetic/immotile cilia in the airway are seemingly disparate diseases. However, they have an overlapping genetic etiology involving mutations in cilia genes, a reflection of the common requirement for motile cilia in left-right patterning and airway clearance. While PCD is a monogenic recessive disorder, heterotaxy has a more complex, largely non-monogenic etiology. In this study, we show mutations in the novel dynein gene DNAH6 can cause heterotaxy and ciliary dysfunction similar to PCD. We provide the first evidence that trans-heterozygous interactions between DNAH6 and other PCD genes potentially can cause heterotaxy. DNAH6 was initially identified as a candidate heterotaxy/PCD gene by filtering exome-sequencing data from 25 heterotaxy patients stratified by whether they have airway motile cilia defects. dnah6 morpholino knockdown in zebrafish disrupted motile cilia in Kupffer's vesicle required for left-right patterning and caused heterotaxy with abnormal cardiac/gut looping. Similarly DNAH6 shRNA knockdown disrupted motile cilia in human and mouse respiratory epithelia. Notably a heterotaxy patient harboring heterozygous DNAH6 mutation was identified to also carry a rare heterozygous PCD-causing DNAI1 mutation, suggesting a DNAH6/DNAI1 trans-heterozygous interaction. Furthermore, sequencing of 149 additional heterotaxy patients showed 5 of 6 patients with heterozygous DNAH6 mutations also had heterozygous mutations in DNAH5 or other PCD genes. We functionally assayed for DNAH6/DNAH5 and DNAH6/DNAI1 trans-heterozygous interactions using subthreshold double-morpholino knockdown in zebrafish and showed this caused heterotaxy. Similarly, subthreshold siRNA knockdown of Dnah6 in heterozygous Dnah5 or Dnai1 mutant mouse respiratory epithelia disrupted motile cilia function. Together, these findings support an oligogenic disease model with broad relevance for further interrogating the genetic etiology of human ciliopathies.


Asunto(s)
Síndrome de Heterotaxia/genética , Síndrome de Kartagener/genética , Animales , Dineínas Axonemales/genética , Dineínas Axonemales/metabolismo , Tipificación del Cuerpo , Cilios/fisiología , Embrión no Mamífero , Técnicas de Silenciamiento del Gen , Heterocigoto , Humanos , Macrófagos del Hígado/patología , Ratones Noqueados , Mutación , ARN Interferente Pequeño/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Pediatr Res ; 76(6): 500-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25167202

RESUMEN

BACKGROUND: Mouse mutants are used to model human congenital cardiovascular disease. Few studies exist comparing normal cardiovascular development in mice vs. humans. We carried out a systematic comparative analysis of mouse and human fetal cardiovascular development. METHODS: Episcopic fluorescence image capture (EFIC) was performed on 66 wild-type mouse embryos from embryonic day (E) 9.5 to birth; 2-dimensional and 3-dimensional datasets were compared with EFIC and magnetic resonance images from a study of 52 human fetuses (Carnegie stage 13-23). RESULTS: Time course of atrial, ventricular, and outflow septation were outlined and followed a similar sequence in both species. Bilateral venae cavae and prominent atrial appendages were seen in the mouse fetus; in human fetuses, atrial appendages were small, and a single right superior vena cava was present. In contrast to humans with separate pulmonary vein orifices, a pulmonary venous confluence with one orifice enters the left atrium in mice. CONCLUSION: The cardiac developmental sequences observed in mouse and human fetuses are comparable, with minor differences in atrial and venous morphology. These comparisons of mouse and human cardiac development strongly support that mouse morphogenesis is a good model for human development.


Asunto(s)
Corazón Fetal/embriología , Corazón/embriología , Animales , Apéndice Atrial/embriología , Tabique Interatrial/embriología , Edad Gestacional , Válvulas Cardíacas/embriología , Ventrículos Cardíacos/embriología , Humanos , Imagen por Resonancia Magnética , Ratones , Morfogénesis , Imagen Óptica , Especificidad de la Especie , Tabique Interventricular/embriología
5.
Circulation ; 125(18): 2232-42, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22499950

RESUMEN

BACKGROUND: Patients with congenital heart disease (CHD) and heterotaxy show high postsurgical morbidity/mortality, with some developing respiratory complications. Although this finding is often attributed to the CHD, airway clearance and left-right patterning both require motile cilia function. Thus, airway ciliary dysfunction (CD) similar to that of primary ciliary dyskinesia (PCD) may contribute to increased respiratory complications in heterotaxy patients. METHODS AND RESULTS: We assessed 43 CHD patients with heterotaxy for airway CD. Videomicrocopy was used to examine ciliary motion in nasal tissue, and nasal nitric oxide (nNO) was measured; nNO level is typically low with PCD. Eighteen patients exhibited CD characterized by abnormal ciliary motion and nNO levels below or near the PCD cutoff values. Patients with CD aged >6 years show increased respiratory symptoms similar to those seen in PCD. Sequencing of all 14 known PCD genes in 13 heterotaxy patients with CD, 12 without CD, 10 PCD disease controls, and 13 healthy controls yielded 0.769, 0.417, 1.0, and 0.077 novel variants per patient, respectively. One heterotaxy patient with CD had the PCD causing DNAI1 founder mutation. Another with hyperkinetic ciliary beat had 2 mutations in DNAH11, the only PCD gene known to cause hyperkinetic beat. Among PCD patients, 2 had known PCD causing CCDC39 and CCDC40 mutations. CONCLUSIONS: Our studies show that CHD patients with heterotaxy have substantial risk for CD and increased respiratory disease. Heterotaxy patients with CD were enriched for mutations in PCD genes. Future studies are needed to assess the potential benefit of prescreening and prophylactically treating heterotaxy patients for CD.


Asunto(s)
Trastornos de la Motilidad Ciliar/epidemiología , Cardiopatías Congénitas/epidemiología , Síndrome de Heterotaxia/epidemiología , Anomalías del Sistema Respiratorio/epidemiología , Adolescente , Adulto , Dineínas Axonemales/genética , Pruebas Respiratorias , Niño , Preescolar , Trastornos de la Motilidad Ciliar/genética , Proteínas del Citoesqueleto , Femenino , Cardiopatías Congénitas/genética , Síndrome de Heterotaxia/genética , Humanos , Lactante , Masculino , Microscopía por Video , Persona de Mediana Edad , Mutación , Óxido Nítrico/análisis , Prevalencia , Proteínas/genética , Anomalías del Sistema Respiratorio/genética , Adulto Joven
6.
Ultrasound Med Biol ; 47(3): 751-758, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33293111

RESUMEN

In vivo micro-imaging of mice is useful in studying the genetic basis of cardiac development in mutant embryos. We examined Phox2b-/- mutant mice, which lack autonomic innervation to the heart and die in utero, and investigated whether this lack of innervation causes cardiac dysfunction during embryogenesis. A VisualSonics Vevo 2100 ultrahigh-frequency linear array ultrasound machine with 30- and 40-MHz probes was used to analyze embryo size, gross characteristics, ventricular contractility and rhythm. Phox2b-/- mutant embryos underwent cessation of heartbeat and death at a greater rate than wild-type controls. We did not observe a hydrops phenotype or congenital heart defects in Phox2b-/- mutants. Analysis of heart rhythm revealed no significant correlation with genotype. Absent these signs of a progressive pathology, we suggest that Phox2b-/- mutant embryos likely die of sudden death secondary to acute arrhythmia. These data provide insight into the role of cardiac autonomic innervation during development.


Asunto(s)
Ecocardiografía/métodos , Pérdida del Embrión/genética , Corazón/diagnóstico por imagen , Corazón/embriología , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Animales , Homocigoto , Ratones , Ratones Endogámicos C57BL , Fenotipo
7.
J Clin Invest ; 117(12): 3742-52, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18037990

RESUMEN

Primary ciliary dyskinesia (PCD) is a genetically heterogeneous disorder associated with ciliary defects and situs inversus totalis, the complete mirror image reversal of internal organ situs (positioning). A variable incidence of heterotaxy, or irregular organ situs, also has been reported in PCD patients, but it is not known whether this is elicited by the PCD-causing genetic lesion. We studied a mouse model of PCD with a recessive mutation in Dnahc5, a dynein gene commonly mutated in PCD. Analysis of homozygous mutant embryos from 18 litters yielded 25% with normal organ situs, 35% with situs inversus totalis, and 40% with heterotaxy. Embryos with heterotaxy had complex structural heart defects that included discordant atrioventricular and ventricular outflow situs and atrial/pulmonary isomerisms. Variable combinations of a distinct set of cardiovascular anomalies were observed, including superior-inferior ventricles, great artery alignment defects, and interrupted inferior vena cava with azygos continuation. The surprisingly high incidence of heterotaxy led us to evaluate the diagnosis of PCD. PCD was confirmed by EM, which revealed missing outer dynein arms in the respiratory cilia. Ciliary dyskinesia was observed by videomicroscopy. These findings show that Dnahc5 is required for the specification of left-right asymmetry and suggest that the PCD-causing Dnahc5 mutation may also be associated with heterotaxy.


Asunto(s)
Trastornos de la Motilidad Ciliar/patología , Dineínas/genética , Cardiopatías Congénitas/ultraestructura , Mutación , Situs Inversus/ultraestructura , Animales , Cilios/genética , Cilios/ultraestructura , Trastornos de la Motilidad Ciliar/genética , Trastornos de la Motilidad Ciliar/fisiopatología , Modelos Animales de Enfermedad , Genes Recesivos , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Humanos , Pulmón/fisiopatología , Pulmón/ultraestructura , Ratones , Ratones Mutantes , Miocardio/ultraestructura , Situs Inversus/genética , Situs Inversus/fisiopatología , Vena Cava Inferior/fisiopatología , Vena Cava Inferior/ultraestructura
8.
Dis Model Mech ; 12(1)2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30578278

RESUMEN

The development of the aortic arch is a complex process that involves remodeling of the bilaterally symmetrical pharyngeal arch arteries (PAAs) into the mature asymmetric aortic arch. Retinoic acid signaling is a key regulator of this process by directing patterning of the second heart field (SHF), formation of the caudal PAAs and subsequent remodeling of the PAAs to form the aortic arch. Here, we identify the HECTD1 ubiquitin ligase as a novel modulator of retinoic acid signaling during this process. Hectd1opm/opm homozygous mutant embryos show a spectrum of aortic arch abnormalities that occur following loss of 4th PAAs and increased SHF marker expression. This sequence of defects is similar to phenotypes observed in mutant mouse models with reduced retinoic acid signaling. Importantly, HECTD1 binds to and influences ubiquitination of the retinoic acid receptor, alpha (RARA). Furthermore, reduced activation of a retinoic acid response element (RARE) reporter is detected in Hectd1 mutant cells and embryos. Interestingly, Hectd1opm/+ heterozygous embryos exhibit reduced retinoic acid signaling, along with intermediate increased expression of SHF markers; however, heterozygotes show normal development of the aortic arch. Decreasing retinoic acid synthesis by reducing Raldh2 (also known as Aldh1a2) gene dosage in Hectd1opm/+ heterozygous embryos reveals a genetic interaction. Double heterozygous embryos show hypoplasia of the 4th PAA and increased incidence of a benign aortic arch variant, in which the transverse arch between the brachiocephalic and left common carotid arteries is shortened. Together, our data establish that HECTD1 is a novel regulator of retinoic acid signaling required for proper aortic arch development.


Asunto(s)
Aorta Torácica/embriología , Aorta Torácica/metabolismo , Transducción de Señal , Tretinoina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Aldehído Oxidorreductasas/genética , Animales , Aorta Torácica/anomalías , Aorta Torácica/patología , Tipificación del Cuerpo , Región Branquial/irrigación sanguínea , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Femenino , Dosificación de Gen , Corazón/embriología , Ratones , Mutación/genética , Fenotipo , Unión Proteica , Receptor alfa de Ácido Retinoico/metabolismo , Ubiquitinación
9.
Ultrasound Med Biol ; 34(5): 741-52, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18328616

RESUMEN

To establish a developmental profile of fetal mouse cardiovascular parameters, we analyzed a large body of ultrasound measurements obtained by in utero echocardiography of C57BL/6J fetal mice from embryonic day 12.5 to 19.5 (term). Measurements were obtained using two-dimensional (2D), spectral Doppler and M-mode imaging with standard clinical cardiac ultrasound imaging planes. As these studies were conducted as part of a large scale mouse mutagenesis screen, stringent filtering criteria were used to eliminate potentially abnormal fetuses. Our analysis showed heart rate increased from 190 to 245 beats per minute as the mouse fetus grew from 8 mm at embryonic day 12.5 to 18.7 mm at term. This was accompanied by increases in peak outflow velocity, E-wave, E/A ratio and ventricular dimensions. In contrast, the A-wave, myocardial performance index and isovolemic contraction time decreased gradually. Systolic function remained remarkably stable at 80% ejection fraction. Analysis of intra- and interobserver variabilities showed these parameters were reproducible, with most comparing favorably to clinical ultrasound measurements in human fetuses. A comprehensive database was generated comprising 23 echocardiographic parameters delineating fetal mouse cardiovascular function from embryonic day 12.5 to term. This database can serve as a standard for evaluating cardiovascular pathophysiology in genetically altered and mutant mouse models.


Asunto(s)
Corazón Fetal/diagnóstico por imagen , Ultrasonografía Prenatal/métodos , Animales , Ecocardiografía Doppler , Femenino , Corazón Fetal/embriología , Edad Gestacional , Frecuencia Cardíaca Fetal , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Embarazo , Flujo Sanguíneo Regional , Procesamiento de Señales Asistido por Computador , Volumen Sistólico , Sístole
10.
Nat Genet ; 49(7): 1152-1159, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28530678

RESUMEN

Congenital heart disease (CHD) affects up to 1% of live births. Although a genetic etiology is indicated by an increased recurrence risk, sporadic occurrence suggests that CHD genetics is complex. Here, we show that hypoplastic left heart syndrome (HLHS), a severe CHD, is multigenic and genetically heterogeneous. Using mouse forward genetics, we report what is, to our knowledge, the first isolation of HLHS mutant mice and identification of genes causing HLHS. Mutations from seven HLHS mouse lines showed multigenic enrichment in ten human chromosome regions linked to HLHS. Mutations in Sap130 and Pcdha9, genes not previously associated with CHD, were validated by CRISPR-Cas9 genome editing in mice as being digenic causes of HLHS. We also identified one subject with HLHS with SAP130 and PCDHA13 mutations. Mouse and zebrafish modeling showed that Sap130 mediates left ventricular hypoplasia, whereas Pcdha9 increases penetrance of aortic valve abnormalities, both signature HLHS defects. These findings show that HLHS can arise genetically in a combinatorial fashion, thus providing a new paradigm for the complex genetics of CHD.


Asunto(s)
Heterogeneidad Genética , Síndrome del Corazón Izquierdo Hipoplásico/genética , Secuencia de Aminoácidos , Animales , Aorta/embriología , Sistemas CRISPR-Cas , Mapeo Cromosómico , Cromosomas Humanos/genética , Modelos Animales de Enfermedad , Exoma , Femenino , Edición Génica , Técnicas de Inactivación de Genes , Ventrículos Cardíacos/embriología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Mutación Missense , Miocitos Cardíacos/patología , Penetrancia , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Obstrucción del Flujo Ventricular Externo/genética , Pez Cebra/genética
11.
J Thorac Cardiovasc Surg ; 151(4): 1126-35.e2, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26611747

RESUMEN

OBJECTIVES: Patients with single ventricle congenital heart disease often form aortopulmonary collateral vessels via an unclear mechanism. To gain insights into the pathogenesis of aortopulmonary collateral vessels, we correlated angiogenic factor levels with in vitro activity and angiographic aortopulmonary collateral assessment and examined whether patients with single ventricle physiology have increased angiogenic factors that can stimulate endothelial cell sprouting in vitro. METHODS: In patients with single ventricle physiology (n = 27) and biventricular acyanotic control patients (n = 21), hypoxia-inducible angiogenic factor levels were measured in femoral venous and arterial plasma at cardiac catheterization. To assess plasma angiogenic activity, we used a 3-dimensional in vitro cell sprouting assay that recapitulates angiogenic sprouting. Aortopulmonary collateral angiograms were graded using a 4-point scale. RESULTS: Compared with controls, patients with single ventricle physiology had increased vascular endothelial growth factor (artery: 58.7 ± 1.2 pg/mL vs 35.3 ± 1.1 pg/mL, P < .01; vein: 34.8 ± 1.1 pg/mL vs 21 ± 1.2 pg/mL, P < .03), stromal-derived factor 1-alpha (artery: 1901.6 ± 1.1 pg/mL vs 1542.6 ± 1.1 pg/mL, P < .03; vein: 2092.8 pg/mL ± 1.1 vs 1752.9 ± 1.1 pg/mL, P < .02), and increased arterial soluble fms-like tyrosine kinase-1, a regulatory vascular endothelial growth factor receptor (612.3 ± 1.2 pg/mL vs 243.1 ± 1.2 pg/mL, P < .003). Plasma factors and sprout formation correlated poorly with aortopulmonary collateral severity. CONCLUSIONS: We are the first to correlate plasma angiogenic factor levels with angiography and in vitro angiogenic activity in patients with single ventricle disease with aortopulmonary collaterals. Patients with single ventricle disease have increased stromal-derived factor 1-alpha and soluble fms-like tyrosine kinase-1, and their roles in aortopulmonary collateral formation require further investigation. Plasma factors and angiogenic activity correlate poorly with aortopulmonary collateral severity in patients with single ventricles, suggesting complex mechanisms of angiogenesis.


Asunto(s)
Proteínas Angiogénicas/sangre , Aorta/fisiopatología , Circulación Colateral , Células Endoteliales/metabolismo , Cardiopatías Congénitas/sangre , Neovascularización Fisiológica , Arteria Pulmonar/fisiopatología , Circulación Pulmonar , Adolescente , Aortografía , Estudios de Casos y Controles , Células Cultivadas , Quimiocina CXCL12/sangre , Niño , Preescolar , Femenino , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/fisiopatología , Humanos , Lactante , Masculino , Factor A de Crecimiento Endotelial Vascular/sangre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre
12.
Circulation ; 106(4): 504-10, 2002 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-12135953

RESUMEN

BACKGROUND: Congenital conotruncal malformations frequently involve dextroposed aorta. The pathogenesis of dextroposed aorta is not known but is thought to be due to abnormal looping and/or wedging of the outflow tract during early heart development. We examined the stage of cardiac looping in an experimental model of dextroposed aorta to determine the embryogenesis of this conotruncal malformation. METHODS AND RESULTS: Hearts were examined from neural crest-ablated embryos by using videocinephotography, scanning electron microscopy, and histological sections. The inflow and outflow limbs of the looped cardiac tube were malpositioned with respect to each other, the inner curvature was diminished, and the outflow limb was straighter and displaced cranially in a manner consistent with diminished length. The altered length could be explained by a significant reduction in the number of cells added to the myocardium of the distal outflow tract from the secondary heart field. CONCLUSIONS: The data are consistent with research showing that normal looping and wedging are essential for normal alignment of the aorta with the left ventricle. These processes are abnormal in neural crest-ablated embryos because of a failure of the outflow tract to lengthen by the addition of myocardial cells from the secondary heart field.


Asunto(s)
Aorta/anomalías , Cardiopatías Congénitas/etiología , Cresta Neural , Animales , Aorta/embriología , Embrión de Pollo , Corazón/embriología , Cardiopatías Congénitas/patología , Cinética , Microscopía por Video , Miocardio/patología , Miocardio/ultraestructura , Cresta Neural/cirugía
14.
Respir Med ; 109(9): 1126-30, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26233707

RESUMEN

INTRODUCTION: Primary ciliary dyskinesia (PCD), a disease of impaired respiratory cilia motility, is often difficult to diagnose. Recent studies show low nasal nitric oxide (nNO) is closely linked to PCD, allowing the use of nNO measurement for PCD assessments. Nasal NO cutoff values for PCD are stratified by age, given nNO levels normally increase with age. However, normative values for nNO have not been established for infants less than 1 year old. In this study, we aim to establish normative values for nNO in infants and determine their utility in guiding infant PCD assessment. METHODS AND RESULTS: We obtained 42 nNO values from infants less than 1 year old without a history of PCD or recurrent sinopulmonary disease. Using regression analysis, we estimated the mean age-adjusted nNO values and established a 95% prediction interval (PI) for normal nNO. Using these findings, we were able to show 14 of 15 infant PCD patients had abnormally low nNO with values below the 95% PI. CONCLUSIONS: In this study we determined a regression model that best fits normative nNO values for infants less than 1 year old. This model identified the majority of PCD infants as having abnormally low nNO. These findings suggest nNO measurement can help guide PCD assessment in infants, and perhaps other pulmonary diseases with a link to low nNO. With early assessments, earlier clinical intervention may be possible to slow disease progression and help reduce pulmonary morbidity.


Asunto(s)
Síndrome de Kartagener/diagnóstico , Óxido Nítrico/metabolismo , Envejecimiento/metabolismo , Pruebas Respiratorias/métodos , Estudios de Factibilidad , Humanos , Lactante , Recién Nacido , Cavidad Nasal , Valores de Referencia
15.
J Am Soc Echocardiogr ; 17(8): 893-900, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15282495

RESUMEN

BACKGROUND: High-frequency ultrasound is effective for noninvasive phenotypic analysis of cardiovascular development and function in mutagenized fetal mice. However, lacking is a normative database of echocardiographic variables for monitoring growth and cardiovascular function. METHODS: C57BL/6J fetal mice were scanned in utero using an ultrasound system with a 15-MHz linear phased-array transducer. Pregnant mothers were anesthetized with 1% isoflurane mixed with 100% oxygen. Quantitative variables for monitoring fetal growth and cardiac function were obtained in several imaging planes and modalities. RESULTS: Fetal growth measurements increased linearly. Inflow velocities showed significant A-wave dominance. The E wave progressively increased during development. Cardiac function was best assessed through M-mode analysis, but short-axis images were difficult to obtain. Spectral Doppler was readily obtained and the myocardial performance index was calculated. CONCLUSIONS: These results provide an essential foundation for the evaluation of cardiovascular defects in mutagenized and transgenic mice.


Asunto(s)
Ecocardiografía , Desarrollo Embrionario y Fetal , Corazón Fetal/diagnóstico por imagen , Cardiopatías Congénitas/diagnóstico por imagen , Ultrasonografía Prenatal , Animales , Femenino , Cardiopatías Congénitas/embriología , Ratones , Ratones Endogámicos C57BL , Embarazo
16.
Ann Am Thorac Soc ; 11(9): 1426-32, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25302410

RESUMEN

RATIONALE: Patients with congenital heart disease with heterotaxy exhibit a high prevalence of abnormal airway ciliary motion and low nasal nitric oxide, characteristics associated with primary ciliary dyskinesia, a reflection of the role of motile cilia in airway clearance and left-right patterning. OBJECTIVES: To assess the potential broader clinical significance of airway ciliary dysfunction in congenital heart disease, we assessed the prevalence of ciliary dysfunction versus respiratory symptoms in patients with congenital heart disease with or without heterotaxy. METHODS: Patients with a broad spectrum of congenital heart disease were recruited (n = 218), 39 with heterotaxy. Nasal nitric oxide measurements and nasal biopsies for ciliary motion video microscopy were conducted. Sinopulmonary symptoms were reviewed by questionnaire. MEASUREMENTS AND MAIN RESULTS: A high prevalence of ciliary motion defects (51.8%) and low or borderline low nasal nitric oxide levels (35.5%) were observed in patients with congenital heart disease with or without heterotaxy. Patients with ciliary motion defects or low nasal nitric oxide showed increased sinopulmonary symptoms, with most respiratory symptoms seen in those with both abnormal ciliary motion and low nitric oxide. Multivariate analysis showed that abnormal ciliary motion and low nasal nitric oxide were more important in determining risk of sinopulmonary symptoms than heterotaxy status. CONCLUSIONS: Patients with congenital heart disease without heterotaxy exhibit a high prevalence of abnormal ciliary motion and low nasal nitric oxide. This was associated with more sinopulmonary symptoms. These findings suggest that patients with a broad spectrum of congenital heart disease and respiratory symptoms may benefit from screening for ciliary dysfunction and implementation of medical interventions to reduce sinopulmonary morbidities.


Asunto(s)
Trastornos de la Motilidad Ciliar/diagnóstico , Cardiopatías Congénitas/complicaciones , Síndrome de Heterotaxia/complicaciones , Adolescente , Adulto , Pruebas Respiratorias , Niño , Trastornos de la Motilidad Ciliar/complicaciones , Femenino , Humanos , Masculino , Microscopía por Video , Persona de Mediana Edad , Análisis Multivariante , Óxido Nítrico/metabolismo , Otitis Media/complicaciones , Hipersensibilidad Respiratoria/complicaciones , Enfermedades Respiratorias/complicaciones , Adulto Joven
17.
J Thorac Cardiovasc Surg ; 147(4): 1291-1298.e2, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23886032

RESUMEN

OBJECTIVE(S): Congenital heart disease (CHD) and heterotaxy patients have increased postoperative and respiratory complications. We recently showed CHD-heterotaxy patients can have respiratory ciliary dysfunction (CD) similar to that associated with primary ciliary dyskinesia, including low nasal nitric oxide and abnormal ciliary motion. In this study, we investigated whether CHD-heterotaxy patients with CD may have worse postsurgical outcomes. METHODS: We examined postsurgical outcome in 13 heterotaxy-CHD patients with CD (25 surgeries), compared with 14 heterotaxy-CHD patients without CD (27 surgeries). Outcome data were collected for each surgery, including respiratory complications, tracheostomy, use of inhaled ß-agonists or nitric oxide, length of hospital stay, days on ventilator, and death. RESULTS: The CD versus the no-CD CHD cohorts had similar Risk Adjustment in Congenital Heart Surgery-1 risk categories, repair track, age at surgery, and follow-up evaluation times. Respiratory complications (76% vs 37%; P = .006), need for tracheostomy (16% vs 0%; P = .047), and use of inhaled ß-agonists (64% vs 11%; P = .0001) all were increased significantly in heterotaxy-CHD patients with CD. No significant differences were detected in postoperative hospital stay, days on mechanical ventilation, or surgical mortality. A trend toward increased mortality for the CD group beyond the postoperative period was observed (33% vs 0%; P = .055) in patients younger than age 10 years. CONCLUSIONS: Our findings showed that heterotaxy-CHD patients with CD may have increased risks for respiratory deficiencies. Overall, there was a trend toward increased mortality in CD patients with intermediate follow-up evaluation. Because ß-agonists are known to increase ciliary beat frequency, presurgical screening for CD and perioperative treatment of CD patients with inhaled ß-agonists may improve postoperative outcomes and survival.


Asunto(s)
Trastornos de la Motilidad Ciliar/complicaciones , Síndrome de Heterotaxia/complicaciones , Síndrome de Heterotaxia/cirugía , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Trastornos Respiratorios/epidemiología , Trastornos Respiratorios/etiología , Adolescente , Adulto , Niño , Preescolar , Femenino , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/cirugía , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Resultado del Tratamiento , Adulto Joven
18.
Circ Cardiovasc Imaging ; 7(1): 31-42, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24319090

RESUMEN

BACKGROUND: Congenital heart disease (CHD) has a multifactorial pathogenesis, but a genetic contribution is indicated by heritability studies. To investigate the spectrum of CHD with a genetic pathogenesis, we conducted a forward genetic screen in inbred mice using fetal echocardiography to recover mutants with CHD. Mice are ideally suited for these studies given that they have the same four-chamber cardiac anatomy that is the substrate for CHD. METHODS AND RESULTS: Ethylnitrosourea mutagenized mice were ultrasound-interrogated by fetal echocardiography using a clinical ultrasound system, and fetuses suspected to have cardiac abnormalities were further interrogated with an ultrahigh-frequency ultrasound biomicroscopy. Scanning of 46 270 fetuses revealed 1722 with cardiac anomalies, with 27.9% dying prenatally. Most of the structural heart defects can be diagnosed using ultrasound biomicroscopy but not with the clinical ultrasound system. Confirmation with analysis by necropsy and histopathology showed excellent diagnostic capability of ultrasound biomicroscopy for most CHDs. Ventricular septal defect was the most common CHD observed, whereas outflow tract and atrioventricular septal defects were the most prevalent complex CHD. Cardiac/visceral organ situs defects were observed at surprisingly high incidence. The rarest CHD found was hypoplastic left heart syndrome, a phenotype never seen in mice previously. CONCLUSIONS: We developed a high-throughput, 2-tier ultrasound phenotyping strategy for efficient recovery of even rare CHD phenotypes, including the first mouse models of hypoplastic left heart syndrome. Our findings support a genetic pathogenesis for a wide spectrum of CHDs and suggest that the disruption of left-right patterning may play an important role in CHD.


Asunto(s)
Ecocardiografía Doppler , Corazón Fetal/diagnóstico por imagen , Pruebas Genéticas , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/genética , Microscopía Acústica , Mutación , Ultrasonografía Prenatal/métodos , Animales , Modelos Animales de Enfermedad , Ecocardiografía Doppler en Color , Etilnitrosourea/toxicidad , Femenino , Corazón Fetal/anomalías , Predisposición Genética a la Enfermedad , Cardiopatías Congénitas/embriología , Herencia , Ensayos Analíticos de Alto Rendimiento , Masculino , Ratones , Ratones Endogámicos C57BL , Linaje , Fenotipo
19.
Circ Cardiovasc Imaging ; 6(4): 551-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23759365

RESUMEN

BACKGROUND: Mice are well suited for modeling human congenital heart disease (CHD), given their 4-chamber cardiac anatomy. However, mice with CHD invariably die prenatally/neonatally, causing CHD phenotypes to be missed. Therefore, we investigated the efficacy of noninvasive microcomputed tomography (micro-CT) to screen for CHD in stillborn/fetal mice. These studies were performed using chemically mutagenized mice expected to be enriched for birth defects, including CHD. METHODS AND RESULTS: Stillborn/fetal mice obtained from the breeding of N-ethyl-N-nitrosourea mutagenized mice were formalin-fixed and stained with iodine, then micro-CT scanned. Those diagnosed with CHD and some CHD-negative pups were necropsied. A subset of these were further analyzed by histopathology to confirm the CHD/no-CHD diagnosis. Micro-CT scanning of 2105 fetal/newborn mice revealed an abundance of ventricular septal defects (n=307). Overall, we observed an accuracy of 89.8% for ventricular septal defect diagnosis. Outflow tract anomalies identified by micro-CT included double outlet right ventricle (n=36), transposition of the great arteries (n=14), and persistent truncus arteriosus (n=3). These were diagnosed with a 97.4% accuracy. Aortic arch anomalies also were readily detected with an overall 99.6% accuracy. This included right aortic arch (n=28) and coarctation/interrupted aortic arch (n=12). Also detected by micro-CT were atrioventricular septal defects (n=22), tricuspid hypoplasia/atresia (n=13), and coronary artery fistulas (n=16). They yielded accuracies of 98.9%, 100%, and 97.8%, respectively. CONCLUSIONS: Contrast enhanced micro-CT imaging in neonatal/fetal mice can reliably detect a wide spectrum of CHD. We conclude that micro-CT imaging can be used for routine rapid assessments of structural heart defects in fetal/newborn mice.


Asunto(s)
Corazón Fetal/anomalías , Corazón Fetal/diagnóstico por imagen , Cardiopatías Congénitas/diagnóstico por imagen , Diagnóstico Prenatal/métodos , Microtomografía por Rayos X , Animales , Animales Recién Nacidos , Medios de Contraste , Modelos Animales de Enfermedad , Corazón Fetal/patología , Genotipo , Edad Gestacional , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Variaciones Dependientes del Observador , Fenotipo , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA