Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 174
Filtrar
1.
Gastroenterology ; 162(1): 179-192.e11, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34425092

RESUMEN

BACKGROUND AND AIMS: The enteric nervous system, which regulates many gastrointestinal functions, is derived from neural crest cells (NCCs). Defective NCC migration during embryonic development may lead to enteric neuropathies such as Hirschsprung's disease (hindgut aganglionosis). Sox10 is known to be essential for cell migration but downstream molecular events regulating early NCC migration have not been fully elucidated. This study aimed to determine how Sox10 regulates migration of sacral NCCs toward the hindgut using Dominant megacolon mice, an animal model of Hirschsprung's disease with a Sox10 mutation. METHODS: We used the following: time-lapse live cell imaging to determine the migration defects of mutant sacral NCCs; genome-wide microarrays, site-directed mutagenesis, and whole embryo culture to identify Sox10 targets; and liquid chromatography and tandem mass spectrometry to ascertain downstream effectors of Sox10. RESULTS: Sacral NCCs exhibited retarded migration to the distal hindgut in Sox10-null embryos with simultaneous down-regulated expression of cadherin-19 (Cdh19). Sox10 was found to bind directly to the Cdh19 promoter. Cdh19 knockdown resulted in retarded sacral NCC migration in vitro and ex vivo, whereas re-expression of Cdh19 partially rescued the retarded migration of mutant sacral NCCs in vitro. Cdh19 formed cadherin-catenin complexes, which then bound to filamentous actin of the cytoskeleton during cell migration. CONCLUSIONS: Cdh19 is a direct target of Sox10 during early sacral NCC migration toward the hindgut and forms cadherin-catenin complexes which interact with the cytoskeleton in migrating cells. Elucidation of this novel molecular pathway helps to provide insights into the pathogenesis of enteric nervous system developmental defects.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Sistema Nervioso Entérico/metabolismo , Enfermedad de Hirschsprung/metabolismo , Cresta Neural/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Factores de Transcripción SOXE/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Animales , Cadherinas/genética , Células Cultivadas , Modelos Animales de Enfermedad , Técnicas de Cultivo de Embriones , Sistema Nervioso Entérico/anomalías , Regulación del Desarrollo de la Expresión Génica , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Cresta Neural/anomalías , Células-Madre Neurales/patología , Unión Proteica , Factores de Transcripción SOXE/genética , Transducción de Señal , Factores de Tiempo
2.
Genesis ; 59(11): e23445, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34490995

RESUMEN

Mouse models provide opportunities to investigate genetic interactions that cause or modify the frequency of neural tube defects (NTDs). Mutation of the PAX3 transcription factor prevents neural tube closure, leading to cranial and spinal NTDs whose frequency is responsive to folate status. Canonical Wnt signalling is implicated both in regulation of Pax3 expression and as a target of PAX3. This study investigated potential interactions of Pax3 mutation and canonical Wnt signalling using conditional gain- and loss-of-function models of ß-catenin. We found an additive effect of ß-catenin gain of function and Pax3 loss of function on NTDs and neural crest defects. ß-catenin gain of function in the Pax3 expression domain led to significantly increased frequency of cranial but not spinal NTDs in embryos that are heterozygous for Pax3 mutation, while both cranial and spinal neural tube closure were exacerbated in Pax3 homozygotes. Similarly, deficits of migrating neural crest cells were exacerbated by ß-catenin gain of function, with almost complete ablation of spinal neural crest cells and derivatives in Pax3 homozygous mutants. Pax3 expression was not affected by ß-catenin gain of function, while we confirmed that loss of function led to reduced Pax3 transcription. In contrast to gain of function, ß-catenin knockout in the Pax3 expression domain lowered the frequency of cranial NTDs in Pax3 null embryos. However, loss of function of ß-catenin and Pax3 resulted in spinal NTDs, suggesting differential regulation of cranial and spinal neural tube closure. In summary, ß-catenin function modulates the frequency of PAX3-related NTDs in the mouse.


Asunto(s)
Cresta Neural/metabolismo , Defectos del Tubo Neural/genética , Factor de Transcripción PAX3/genética , Vía de Señalización Wnt , Animales , Heterocigoto , Ratones , Ratones Endogámicos C57BL , Mutación , Cresta Neural/anomalías , Cresta Neural/embriología , Factor de Transcripción PAX3/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
3.
Toxicology ; 458: 152843, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34186166

RESUMEN

Adverse outcome pathway (AOP) is a conceptual framework that links a molecular initiating event (MIE) via intermediate key events (KEs) with adverse effects (adverse outcomes, AO) relevant for risk assessment, through defined KE relationships (KERs). The aim of the present work is to describe a linear AOP, supported by experimental data, for skeletal craniofacial defects as the AO. This AO was selected in view of its relative high incidence in humans and the suspected relation to chemical exposure. We focused on inhibition of CYP26, a retinoic acid (RA) metabolizing enzyme, as MIE, based on robust previously published data. Conazoles were selected as representative stressors. Intermediate KEs are RA disbalance, aberrant HOX gene expression, disrupted specification, migration, and differentiation of neural crest cells, and branchial arch dysmorphology. We described the biological basis of the postulated events and conducted weight of evidence (WoE) assessments. The biological plausibility and the overall empirical evidence were assessed as high and moderate, respectively, the latter taking into consideration the moderate evidence for concordance of dose-response and temporal relationships. Finally, the essentiality assessment of the KEs, considered as high, supported the robustness of the presented AOP. This AOP, which appears of relevance to humans, thus contributes to mechanistic underpinning of selected test methods, thereby supporting their application in integrated new approach test methodologies and strategies and application in a regulatory context.


Asunto(s)
Rutas de Resultados Adversos , Anomalías Craneofaciales/metabolismo , Tretinoina/metabolismo , Animales , Azoles/toxicidad , Familia 26 del Citocromo P450/antagonistas & inhibidores , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Cresta Neural/anomalías , Cresta Neural/efectos de los fármacos , Medición de Riesgo
4.
Rev. medica electron ; 42(4): 2049-2065,
Artículo en Español | LILACS, CUMED | ID: biblio-1139295

RESUMEN

RESUMEN Las células de la cresta neural son pluripotenciales y son llamadas la cuarta hoja germinativa del embrión. Con el objetivo de estructurar los referentes teóricos actualizados que sustenten la afirmación precedente y que constituirá material de estudio para los estudiantes de las Ciencias Médicas, se realizó la revisión de 28 referencias bibliográficas, de ellas 89% actualizadas. Estas células aparecen durante la neurulación y pasado este proceso transitan de epitelial a mesenquimatosa; migran siguiendo señales de la matriz extracelular a todo el cuerpo del embrión diferenciándose en tejidos disimiles. Muy vinculados en su evolución a mecanismos epigenéticos, hacen a esta población celular vulnerables a ser dañadas invocándose en la etiología de diferentes defectos congénitos y enfermedades crónicas no trasmisibles como cáncer. Como conclusión por su pluripotencialidad y por los mecanismos moleculares que distinguen su evolución son consideradas por muchos autores la cuarta hoja germinativa del embrión (AU).


SUMMARY Neural crest cells are pluripotentials, and are called the fourth germinative leaf of the embryo. With the objective of structuring the updated theoretical referents backing up the precedent affirmation that will be study material for the students of Medical Sciences, the authors reviewed 28 bibliographic references, 89 % of them updated. These cells appear during neurulation and after this process they transit from epithelial to mesenchymal; following extracellular matrix signals, they migrate to the whole embryo body differentiating themselves in dissimilar tissues. Tightly related in their evolution to epigenetic mechanisms, this cell population is very likely to be damaged and so they are invoked in the etiology of different congenital defects and noncommunicable chronic diseases like cancer. In conclusion, due to their pluripotentiality and the molecular mechanisms distinguishing their evolution, many authors consider them the embryo´s fourth germinative leaf (AU).


Asunto(s)
Humanos , Masculino , Femenino , Células/metabolismo , Cresta Neural/patología , Estudiantes de Medicina , Vertebrados/genética , Neurulación/fisiología , Cresta Neural/anomalías , Cresta Neural/fisiología , Cresta Neural/fisiopatología
5.
Genet Med ; 22(3): 547-556, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31649276

RESUMEN

PURPOSE: Treacher Collins syndrome (TCS) is a rare autosomal dominant mandibulofacial dysostosis, with a prevalence of 0.2-1/10,000. Features include bilateral and symmetrical malar and mandibular hypoplasia and facial abnormalities due to abnormal neural crest cell (NCC) migration and differentiation. To date, three genes have been identified: TCOF1, POLR1C, and POLR1D. Despite a large number of patients with a molecular diagnosis, some remain without a known genetic anomaly. METHODS: We performed exome sequencing for four individuals with TCS but who were negative for pathogenic variants in the known causative genes. The effect of the pathogenic variants was investigated in zebrafish. RESULTS: We identified three novel pathogenic variants in POLR1B. Knockdown of polr1b in zebrafish induced an abnormal craniofacial phenotype mimicking TCS that was associated with altered ribosomal gene expression, massive p53-associated cellular apoptosis in the neuroepithelium, and reduced number of NCC derivatives. CONCLUSION: Pathogenic variants in the RNA polymerase I subunit POLR1B might induce massive p53-dependent apoptosis in a restricted neuroepithelium area, altering NCC migration and causing cranioskeletal malformations. We identify POLR1B as a new causative gene responsible for a novel TCS syndrome (TCS4) and establish a novel experimental model in zebrafish to study POLR1B-related TCS.


Asunto(s)
Anomalías Craneofaciales/genética , ARN Polimerasas Dirigidas por ADN/genética , Disostosis Mandibulofacial/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Animales , Apoptosis/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Anomalías Craneofaciales/patología , Predisposición Genética a la Enfermedad , Humanos , Disostosis Mandibulofacial/patología , Mutación , Cresta Neural/anomalías , Cresta Neural/patología , Proteína p53 Supresora de Tumor/genética , Secuenciación del Exoma , Pez Cebra/genética
6.
Pediatr Cardiol ; 40(7): 1339-1344, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31338559

RESUMEN

Hand1 is a basic Helix-loop-Helix transcription factor that exhibits post-translationally regulated dimer partner choice that allows for a diverse set of Hand1 transcriptional complexes. Indeed, when Hand1 phosphoregulation is altered, conditionally activated hypophorylation (Hand1PO4-) and phosphorylation mimic (Hand1PO4+) Hand1 alleles disrupt both craniofacial and limb morphogenesis with 100% penetrance. Interestingly, activation of conditional Hand1 Phosphomutant alleles within post-migratory neural crest cells produce heart defects that include ventricular septal defects, double-outlet right ventricle, persistent truncus arteriosus with partial penetrance. Single versus double-lobed thymus is a distinguishing feature between Wnt1-Cre;Hand1PO4-/+ and Wnt1-Cre;Hand1PO4+/+ mice. These data show that although Hand1 dimer regulation plays critical and consistent roles in disrupting craniofacial and limb morphogenesis, Hand1 dimer regulation during cardiac outflow track formation is less critical for normal morphogenesis. This review will present the OFT phenotypes observed in Hand1 Phosphomutant mice, and discuss possible mechanisms of how penetrance differences within the same tissues within the same embryos could be variable.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ventrículo Derecho con Doble Salida/genética , Cresta Neural/anomalías , Alelos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Morfogénesis , Fenotipo , Transcripción Genética
7.
J Neurosci Res ; 97(1): 7-15, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29660784

RESUMEN

Development of the eye is closely associated with neural crest cell migration and specification. Eye development is extremely complex, as it requires the working of a combination of local factors, receptors, inductors, and signaling interactions between tissues such as the optic cup and periocular mesenchyme (POM). The POM is comprised of neural crest-derived mesenchymal progenitor cells that give rise to numerous important ocular structures including those tissues that form the optic cup and anterior segment of the eye. A number of genes are involved in the migration and specification of the POM such as PITX2, PITX3, FOXC1, FOXE3, PAX6, LMX1B, GPR48, TFAP2A, and TFAP2B. In this review, we will discuss the relevance of these genes in the development of the POM and how mutations and defects result in rare ocular diseases.


Asunto(s)
Anomalías del Ojo/genética , Oftalmopatías/genética , Cresta Neural/anomalías , Cresta Neural/metabolismo , Enfermedades Raras/genética , Segmento Anterior del Ojo/anomalías , Oftalmopatías/patología , Humanos , Mutación , Segmento Posterior del Ojo/anomalías , Enfermedades Raras/patología , Factores de Transcripción
8.
Am J Med Genet C Semin Med Genet ; 175(4): 478-486, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29082625

RESUMEN

Neural crest cells are highly migratory pluripotent cells that give rise to diverse derivatives including cartilage, bone, smooth muscle, pigment, and endocrine cells as well as neurons and glia. Abnormalities in neural crest-derived tissues contribute to the etiology of CHARGE syndrome, a complex malformation disorder that encompasses clinical symptoms like coloboma, heart defects, atresia of the choanae, retarded growth and development, genital hypoplasia, ear anomalies, and deafness. Mutations in the chromodomain helicase DNA-binding protein 7 (CHD7) gene are causative of CHARGE syndrome and loss-of-function data in different model systems have firmly established a role of CHD7 in neural crest development. Here, we will summarize our current understanding of the function of CHD7 in neural crest development and discuss possible links of CHARGE syndrome to other developmental disorders.


Asunto(s)
Síndrome CHARGE/diagnóstico , Síndrome CHARGE/etiología , Cresta Neural/anomalías , Fenotipo , Adenosina Trifosfato/metabolismo , Animales , Síndrome CHARGE/metabolismo , Ensamble y Desensamble de Cromatina , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Humanos , Complejos Multiproteicos/metabolismo , Mutación , Unión Proteica
9.
Interv Neuroradiol ; 23(6): 572-576, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28814167

RESUMEN

A novel hypothesis proposes that "cardio-cephalic neural crest (NC) syndrome," i.e. cephalic NC including cardiac NC, contributes to the concurrent occurrence of vascular diseases in the cardio- and cerebrovascular regions. NC is a transient structure present in early embryogenesis. Cephalic NC provides mesenchymal cells to the vascular media in these regions. Concurrent cardio- and cerebrovascular lesions have been reported in PHACE syndrome, ACTA2 mutation syndrome, and less frequently in the spontaneous occlusion of the circle of Willis (so-called moyamoya disease). Cardiovascular lesions in these syndromes include coarctation of the aorta, persistent truncus arteriosus, patent ductus arteriosus, and coronary artery disease, and cerebrovascular lesions include agenesis and stenosis/occlusion of the internal carotid arteries, and moyamoya phenomenon. These concurrent vascular lesions both in the cardio- and cerebrovascular regions might be related to cephalic NC. This hypothesis, although not proven, may facilitate a better understanding of the above-mentioned NC-related vascular pathologies and lead to appropriate diagnostic and therapeutic approaches for clinicians and chart future direction for researchers.


Asunto(s)
Coartación Aórtica/diagnóstico por imagen , Trastornos Cerebrovasculares/diagnóstico por imagen , Anomalías del Ojo/diagnóstico por imagen , Enfermedad de Moyamoya/diagnóstico por imagen , Cresta Neural/anomalías , Síndromes Neurocutáneos/diagnóstico por imagen , Actinas , Adenosina Trifosfatasas , Coartación Aórtica/genética , Circulación Cerebrovascular , Trastornos Cerebrovasculares/genética , Anomalías del Ojo/genética , Humanos , Enfermedad de Moyamoya/genética , Mutación Missense , Síndromes Neurocutáneos/genética , Fenotipo , Ubiquitina-Proteína Ligasas
11.
Sci Rep ; 7: 42506, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28195132

RESUMEN

Birth defects affect 3% of children in the United States. Among the birth defects, congenital heart disease and craniofacial malformations are major causes of mortality and morbidity. Unfortunately, the genetic mechanisms underlying craniocardiac malformations remain largely uncharacterized. To address this, human genomic studies are identifying sequence variations in patients, resulting in numerous candidate genes. However, the molecular mechanisms of pathogenesis for most candidate genes are unknown. Therefore, there is a need for functional analyses in rapid and efficient animal models of human disease. Here, we coupled the frog Xenopus tropicalis with Optical Coherence Tomography (OCT) to create a fast and efficient system for testing craniocardiac candidate genes. OCT can image cross-sections of microscopic structures in vivo at resolutions approaching histology. Here, we identify optimal OCT imaging planes to visualize and quantitate Xenopus heart and facial structures establishing normative data. Next we evaluate known human congenital heart diseases: cardiomyopathy and heterotaxy. Finally, we examine craniofacial defects by a known human teratogen, cyclopamine. We recapitulate human phenotypes readily and quantify the functional and structural defects. Using this approach, we can quickly test human craniocardiac candidate genes for phenocopy as a critical first step towards understanding disease mechanisms of the candidate genes.


Asunto(s)
Anomalías Craneofaciales/diagnóstico por imagen , Cardiopatías Congénitas/diagnóstico por imagen , Tomografía de Coherencia Óptica , Xenopus/anomalías , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Cresta Neural/anomalías , Fenotipo
12.
Int. j. morphol ; 34(2): 489-494, June 2016. ilus
Artículo en Español | LILACS | ID: lil-787026

RESUMEN

Las cardiopatías congénitas corresponden al grupo de anomalías del desarrollo que se presentan con mayor frecuencia. Durante el desarrollo cardíaco participan distintos linajes celulares, donde destacan las Células de la Cresta Neural (CCN) por su amplia gama de derivados embriológicos y la susceptibilidad de afectar a múltiples sistemas si su función es alterada. El objetivo fue determinar el rol que cumplen las CCN durante el desarrollo cardíaco y las cardiopatías congénitas asociadas. Se diseñó un estudio descriptivo en base a una revisión sistemática de la literatura de las bases de datos MEDLINE y Scopus, utilizando la combinación de términos MeSH: ("Heart Diseases/congenital" OR "Heart Diseases/embriology" OR "Heart Diseases/etiology" OR "Heart Disesaes/epidemiology") AND ("Neural Crest/abnormalities"). Se restringió la búsqueda a artículos de los últimos 10 años. De un total de 35 artículos obtenidos, 22 fueron incluidos para su revisión por estar relacionados con los objetivos de este estudio, excluyéndose duplicados entre bases de datos. Posteriormente se hizo un análisis individual y en conjunto de la información obtenida de los artículos seleccionados. La evidencia indica la participación directa o indirecta de las CCN durante la formación de las estructuras derivadas del polo arterioso del corazón en desarrollo, los grandes vasos arteriales y sus ramas colaterales, así como en su inervación y sistema de conducción. La alteración del funcionamiento normal de las CCN produce fenotipos cardíacos alterados, siendo la persistencia del tronco arterioso, doble salida ventricular derecha, defectos septales interventriculares y malformación de los aparatos valvares aórtico y pulmonar, los más frecuentes.


Congenital heart defects are the group of most frequent anomalies of development. Cardiac development in different cell lines, which include the Neural crest cells (NCC) for their wide range of embryological derivatives and susceptibility to affect multiple systems if their function is altered participate. The objective was to determine the role of the NCC during heart development and associated congenital heart disease. A descriptive study was designed based on a systematic review of the literature from the MEDLINE and Scopus data, using a combination of MeSH terms ("Heart Diseases / congenital" OR "Heart Diseases / Embryology" OR "Heart Diseases/etiology "OR" Heart Diseases/epidemiology ") AND ("Neural Crest/abnormalities"). Search for articles in the last 10 years was restricted. From a total of 35 articles retrieved, 22 were included related to the objectives of this study for review, excluding duplicated between databases. Subsequently, an individual and joint analysis was realized with the information from the selected items. Evidence indicates the direct or indirect involvement of NCC during the formation of the structures derived from arterial pole of the developing heart, the large arterial vessels and their collateral branches, as well as its innervation and conduction system. The disruption of normal operation of the NCC produces altered cardiac phenotypes, with the Persistence Truncus Arteriosus, Double-Outlet Right Ventricle, ventricular Septal Defects and malformation of the most common valvular aortic and pulmonary devices.


Asunto(s)
Humanos , Cardiopatías Congénitas/embriología , Cresta Neural/anomalías , Cardiopatías Congénitas/etiología
13.
Hum Mol Genet ; 24(24): 7005-16, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26420840

RESUMEN

Mutations in subunits or regulators of cohesin cause a spectrum of disorders in humans known as the 'cohesinopathies'. Cohesinopathies, including the best known example Cornelia de Lange syndrome (CdLS), are characterized by broad spectrum, multifactorial developmental anomalies. Heart defects occur at high frequency and can reach up to 30% in CdLS. The mechanisms by which heart defects occur are enigmatic, but assumed to be developmental in origin. In this study, we depleted cohesin subunit Rad21 by 70-80% in a zebrafish cohesinopathy model. The hearts of Rad21-depleted animals were smaller, often failed to loop, and functioned less efficiently than size-matched controls. Functional deficiency was accompanied by valve defects and reduced ejection fraction. Interestingly, neural crest cells failed to populate the heart and instead exhibited a wandering behavior. Consequently, these cells also failed to condense correctly into pharyngeal arches. Transcriptome analysis revealed that Wnt pathway, chemokine and cadherin genes are dysregulated at the time of cardiac neural crest development. Our results give insight into the etiology of heart defects in the cohesinopathies, and raise the possibility that mild mutations in cohesin genes may be causative of a fraction of congenital heart disease in human populations.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cardiopatías Congénitas/embriología , Cresta Neural/anomalías , Proteínas de Pez Cebra/genética , Animales , Proteínas de Ciclo Celular/deficiencia , Movimiento Celular , Proteínas Cromosómicas no Histona/genética , Modelos Animales de Enfermedad , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Válvulas Cardíacas/anomalías , Pez Cebra , Proteínas de Pez Cebra/deficiencia , Cohesinas
14.
Dev Biol ; 406(2): 186-95, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26385750

RESUMEN

Ephrin-B2, a member of the Eph/ephrin family of cell signaling molecules, has been implicated in the guidance of cranial and trunk neural crest cells (NCC) and development of the branchial arches(BA), but detailed examination in mice has been hindered by embryonic lethality of Efnb2 null loss of function due to a requirement in angiogenic remodeling. To elucidate the developmental roles for Efnb2, we generated a conditional rescue knock-in allele that allows rescue of ephrin-B2 specifically in the vascular endothelium (VE), but is otherwise ephrin-B2 deficient. Restoration of ephrin-B2 expression specifically to the VE completely circumvents angiogenic phenotypes, indicating that the requirement of ephrin-B2 in angiogenesis is limited to the VE. Surprisingly, we find that expression of ephrin-B2 specifically in the VE is also sufficient for normal NCC migration and that conversely, embryos in which ephrin-B2 is absent specifically from the VE exhibit NCC migration and survival defects. Disruption of vascular development independent of loss of ephrin-B2 function also leads to defects in NCC and BA development. Together, these data indicate that direct ephrin-B2 signaling to NCCs is not required for NCC guidance, which instead depends on proper organization of the embryonic vasculature.


Asunto(s)
Vasos Sanguíneos/embriología , Endotelio Vascular/metabolismo , Efrina-B2/genética , Cresta Neural/anomalías , Cresta Neural/fisiología , Fenotipo , Animales , Movimiento Celular/fisiología , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Técnicas Histológicas , Hibridación in Situ , Ratones , Mutación/genética
15.
Toxicol Sci ; 148(1): 299-310, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26293553

RESUMEN

Copper-induced delayed hatching and dysfunctional movement had been reported previously, and unbalanced free copper was found in the body of humans with Alzheimer's disease and other neural diseases, but details of the underlying mechanisms are still unknown. In this study, zebrafish (Danio rerio) embryos exposed to over 3.9 µM of copper-exhibited delayed hatching and significantly dysfunctional movement. Using high-throughput in situ hybridization screening and by conducting an in-depth analysis of gene characterization in embryos exposed to copper, we found that copper caused neural crest defects from the initiation stage of neurogenesis, and embryos younger than the 70% epiboly stage were sensitive to copper toxicity. The myelination of Schwann cells, other than melanophores, cartilage, and neurons, was inhibited by copper during neurogenesis. In addition, axon guidance was blocked by copper. Downregulated cdx4-hox might have contributed to the neurogenesis-related defects. Moreover, copper inhibited the differentiation of muscle fibers and myotomes but not the specification of muscle progenitors. In summary, our data reveal a novel molecular mechanism for copper-inhibited locomotor behavior in embryos, in which copper blocks functional muscle fiber specification during myogenesis and inhibits the specification of axons and Schwann cell myelination during neurogenesis. A combination of these processes results in dysfunctional locomotor behavior in zebrafish embryos exposed to copper.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cobre/toxicidad , Embrión no Mamífero/efectos de los fármacos , Proteínas de Peces/metabolismo , Actividad Motora/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Biomarcadores/metabolismo , Embrión no Mamífero/anomalías , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Proteínas de Peces/genética , Hibridación in Situ , Desarrollo de Músculos/efectos de los fármacos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/anomalías , Cresta Neural/efectos de los fármacos , Cresta Neural/embriología , Neurogénesis/efectos de los fármacos , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Pez Cebra
16.
Neurobiol Dis ; 80: 1-14, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25959061

RESUMEN

Distinct classes of SOX10 mutations result in peripheral demyelinating neuropathy, central dysmyelinating leukodystrophy, Waardenburg syndrome, and Hirschsprung disease, collectively known as PCWH. Meanwhile, SOX10 haploinsufficiency caused by allelic loss-of-function mutations leads to a milder non-neurological disorder, Waardenburg-Hirschsprung disease. The cellular pathogenesis of more complex PCWH phenotypes in vivo has not been thoroughly understood. To determine the pathogenesis of PCWH, we have established a transgenic mouse model. A known PCWH-causing SOX10 mutation, c.1400del12, was introduced into mouse Sox10-expressing cells by means of bacterial artificial chromosome (BAC) transgenesis. By crossing the multiple transgenic lines, we examined the effects produced by various copy numbers of the mutant transgene. Within the nervous systems, transgenic mice revealed a delay in the incorporation of Schwann cells in the sciatic nerve and the terminal differentiation of oligodendrocytes in the spinal cord. Transgenic mice also showed defects in melanocytes presenting as neurosensory deafness and abnormal skin pigmentation, and a loss of the enteric nervous system. Phenotypes in each lineage were more severe in mice carrying higher copy numbers, suggesting a gene dosage effect for mutant SOX10. By uncoupling the effects of gain-of-function and haploinsufficiency in vivo, we have demonstrated that the effect of a PCWH-causing SOX10 mutation is solely pathogenic in each SOX10-expressing cellular lineage in a dosage-dependent manner. In both the peripheral and central nervous systems, the primary consequence of SOX10 mutations is hypomyelination. The complex neurological phenotypes in PCWH patients likely result from a combination of haploinsufficiency and additive dominant effect.


Asunto(s)
Enfermedades Desmielinizantes/genética , Enfermedad de Hirschsprung/genética , Factores de Transcripción SOXE/genética , Síndrome de Waardenburg/genética , Animales , Encéfalo/anomalías , Encéfalo/ultraestructura , Cuerpo Calloso/ultraestructura , Enfermedades Desmielinizantes/embriología , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Genes Dominantes , Haploinsuficiencia , Enfermedad de Hirschsprung/embriología , Enfermedad de Hirschsprung/patología , Humanos , Ratones , Ratones Transgénicos , Cresta Neural/anomalías , Fenotipo , Células de Schwann/patología , Nervio Ciático/ultraestructura , Síndrome de Waardenburg/embriología , Síndrome de Waardenburg/patología
17.
J Neurosurg Pediatr ; 16(1): 46-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25837885

RESUMEN

In infants, the presence of a cranial defect may be due to a variety of traumatic, inflammatory, neoplastic, and congenital abnormalities. Differentiation between these possible etiologies is facilitated by clinical presentation, patient history, and physical examination. Congenital cutaneous neural crest-derived lesions are unlikely to be considered in a patient presenting with an asymptomatic cranial defect without overlying mass or skin pigmentation. The authors present an unusual case of a 2-month-old infant with an asymptomatic calvarial defect with normal overlying skin. Pathology of the excised tissue showed features consistent with a congenital neurocristic tumor: a pigmented, neural crest-derived hamartomatous tumor that typically presents as a melanotic skin lesion.


Asunto(s)
Neoplasias Óseas/congénito , Neoplasias Óseas/diagnóstico , Hamartoma/congénito , Hamartoma/diagnóstico , Cresta Neural/anomalías , Cráneo/patología , Neoplasias Óseas/patología , Neoplasias Óseas/cirugía , Diagnóstico Diferencial , Hamartoma/patología , Hamartoma/cirugía , Humanos , Imagenología Tridimensional , Lactante , Masculino , Hueso Occipital/patología , Hueso Parietal/patología , Neoplasias Cutáneas/congénito , Neoplasias Cutáneas/diagnóstico , Tomografía Computarizada por Rayos X
18.
J Am Heart Assoc ; 5(1)2015 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-26722124

RESUMEN

BACKGROUND: The small GTPase Rac1 regulates diverse cellular functions, including both apicobasal and planar cell polarity pathways; however, its role in cardiac outflow tract (OFT) development remains unknown. In the present study, we aimed to examine the role of Rac1 in the anterior second heart field (SHF) splanchnic mesoderm and subsequent OFT development during heart morphogenesis. METHODS AND RESULTS: Using the Cre/loxP system, mice with an anterior SHF-specific deletion of Rac1 (Rac1(SHF)) were generated. Embryos were collected at various developmental time points for immunostaining and histological analysis. Intrauterine echocardiography was also performed to assess aortic valve blood flow in embryos at embryonic day 18.5. The Rac1(SHF) splanchnic mesoderm exhibited disruptions in SHF progenitor cellular organization and proliferation. Consequently, this led to a spectrum of OFT defects along with aortic valve defects in Rac1(SHF) embryos. Mechanistically, it was found that the ability of the Rac1(SHF) OFT myocardial cells to migrate into the proximal OFT cushion was severely reduced. In addition, expression of the neural crest chemoattractant semaphorin 3c was decreased. Lineage tracing showed that anterior SHF contribution to the OFT myocardium and aortic valves was deficient in Rac1(SHF) hearts. Furthermore, functional analysis with intrauterine echocardiography at embryonic day 18.5 showed aortic valve regurgitation in Rac1(SHF) hearts, which was not seen in control hearts. CONCLUSIONS: Disruptions of Rac1 signaling in the anterior SHF results in aberrant progenitor cellular organization and defects in OFT development. Our data show Rac1 signaling to be a critical regulator of cardiac OFT formation during embryonic heart development.


Asunto(s)
Insuficiencia de la Válvula Aórtica/enzimología , Válvula Aórtica/enzimología , Cardiopatías Congénitas/enzimología , Miocardio/enzimología , Neuropéptidos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Válvula Aórtica/anomalías , Insuficiencia de la Válvula Aórtica/diagnóstico , Insuficiencia de la Válvula Aórtica/genética , Linaje de la Célula , Movimiento Celular , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Edad Gestacional , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/genética , Ratones Noqueados , Morfogénesis , Miocardio/patología , Cresta Neural/anomalías , Cresta Neural/enzimología , Neuropéptidos/deficiencia , Neuropéptidos/genética , Fenotipo , Semaforinas/genética , Semaforinas/metabolismo , Transducción de Señal , Proteína de Unión al GTP rac1/deficiencia , Proteína de Unión al GTP rac1/genética
19.
Hum Mol Genet ; 24(2): 424-35, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25209980

RESUMEN

T-box transcription factor, TBX1, is the major candidate gene for 22q11.2 deletion syndrome (DiGeorge/ Velo-cardio-facial syndrome) characterized by facial defects, thymus hypoplasia, cardiovascular anomalies and cleft palates. Here, we report that the loss of Tbx1 in mouse (Tbx1(-/-)) results in skeletal abnormalities similar to those of cleidocranial dysplasia (CCD) in humans, which is an autosomal-dominant skeletal disease caused by mutations in RUNX2. Tbx1(-/-) mice display short stature, absence of hyoid bone, failed closure of fontanelle, bifid xiphoid process and hypoplasia of clavicle and zygomatic arch. A cell-type-specific deletion of Tbx1 in osteochondro-progenitor (Tbx1(OPKO)) or mesodermal (Tbx1(MKO)) lineage partially recapitulates the Tbx1(-/-) bone phenotypes. Although Tbx1 expression has not been previously reported in neural crest, inactivation of Tbx1 in the neural crest lineage (Tbx1(NCKO)) leads to an absence of the body of hyoid bone and postnatal lethality, indicating an unanticipated role of Tbx1 in neural crest development. Indeed, Tbx1 is expressed in the neural crest-derived hyoid bone primordium, in addition to mesoderm-derived osteochondral progenitors. Ablation of Tbx1 affected Runx2 expression in calvarial bones and overexpression of Tbx1 induced Runx2 expression in vitro. Taken together, our current studies reveal that Tbx1 is required for mesoderm- and neural crest-derived osteoblast differentiation and normal skeletal development. TBX1 mutation could lead to CCD-like bone phenotypes in human.


Asunto(s)
Huesos/anomalías , Displasia Cleidocraneal/metabolismo , Proteínas de Dominio T Box/deficiencia , Animales , Huesos/metabolismo , Diferenciación Celular , Displasia Cleidocraneal/embriología , Displasia Cleidocraneal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Cresta Neural/anomalías , Cresta Neural/embriología , Cresta Neural/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Fenotipo , Proteínas de Dominio T Box/genética
20.
Ann Biomed Eng ; 42(10): 2014-28, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25099772

RESUMEN

Aortopathy is characterized by vascular smooth muscle cell (VSMC) abnormalities and elastic fiber fragmentation. Elastin insufficient (Eln (+/-)) mice demonstrate latent aortopathy similar to human disease. We hypothesized that aortopathy manifests primarily in the aorto-pulmonary septal (APS) side of the thoracic aorta due to asymmetric cardiac neural crest (CNC) distribution. Anatomic (aortic root vs. ascending aorta) and molecular (APS vs. non-APS) regions of proximal aorta tissue were examined in adult and aged wild type (WT) and mutant (Eln (+/-)) mice. CNC, VSMCs, elastic fiber architecture, proteoglycan expression, morphometrics and biomechanical properties were examined using histology, 3D reconstruction, micropipette aspiration and in vivo magnetic resonance imaging (MRI). In the APS side of Eln (+/-) aorta, Sonic Hedgehog (SHH) is decreased while SM22 is increased. Elastic fiber architecture abnormalities are present in the Eln (+/-) aortic root and APS ascending aorta, and biglycan is increased in the aortic root while aggrecan is increased in the APS aorta. The Eln (+/-) ascending aorta is stiffer than the aortic root, the APS side is thicker and stiffer than the non-APS side, and significant differences in the individual aortic root sinuses are observed. Asymmetric structure-function abnormalities implicate regional CNC dysregulation in the development and progression of aortopathy.


Asunto(s)
Aorta/anomalías , Aorta/fisiología , Elastina/deficiencia , Envejecimiento/fisiología , Animales , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/fisiopatología , Fenómenos Biomecánicos , Niño , Módulo de Elasticidad , Elastina/genética , Elastina/fisiología , Humanos , Ratones Transgénicos , Miocitos del Músculo Liso/patología , Cresta Neural/anomalías , Proteoglicanos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...