Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Prenat Diagn ; 41(1): 15-20, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32920862

RESUMEN

OBJECTIVES: Agenesis of the ductus venosus (ADV) has been associated with additional anomalies in up to 83% of cases. We sought to investigate characteristics, co-morbidities and outcomes of ADV in the current era. We hypothesized that rates of cardiac and non-cardiac diagnoses and survival would be higher, due to advances in genetic testing, prenatal diagnosis and surveillance. METHODS: A retrospective series of cases diagnosed at our institution from 2007 to 2018 were identified by searching our database. Cardiac and obstetric charts were reviewed for cardiac and extra-cardiac anomalies, genetic results and outcomes. RESULTS: Fourteen cases were diagnosed at a mean gestational age of 23.9 weeks (range 13-33). All had associated genetic, cardiac or extra-cardiac anomalies. Eight (57%) had cardiac anomalies and one other developed cardiomyopathy by 6 months. Extra-cardiac anomalies were present in 93% (13/14) and genetic diagnoses made in 75% (6/8) of those tested. Cardiac output Z-scores were >2 in 60% (6/10) prior to delivery. Two had hydrops, there was one intra-uterine death, 13 live-births and two neonatal deaths. CONCLUSION: Our cohort had more associated diagnoses and a lower mortality than previously reported. In our experience, high output occurs frequently, however with a relatively low risk of hydrops and intrauterine death.


Asunto(s)
Anomalías Cardiovasculares/diagnóstico , Anomalías Cardiovasculares/embriología , Feto/irrigación sanguínea , Resultado del Embarazo , Diagnóstico Prenatal , Aneuploidia , Anomalías Cardiovasculares/genética , Comorbilidad , Femenino , Corazón Fetal/anomalías , Corazón Fetal/embriología , Pruebas Genéticas , Humanos , Recién Nacido , Mutación , Embarazo , Estudios Retrospectivos , Ultrasonografía Prenatal , Venas Umbilicales/anomalías
2.
Nature ; 474(7353): 645-8, 2011 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-21623370

RESUMEN

The four-chambered mammalian heart develops from two fields of cardiac progenitor cells distinguished by their spatiotemporal patterns of differentiation and contributions to the definitive heart. The first heart field differentiates earlier in lateral plate mesoderm, generates the linear heart tube and ultimately gives rise to the left ventricle. The second heart field (SHF) differentiates later in pharyngeal mesoderm, elongates the heart tube, and gives rise to the outflow tract and much of the right ventricle. Because hearts in lower vertebrates contain a rudimentary outflow tract but not a right ventricle, the existence and function of SHF-like cells in these species has remained a topic of speculation. Here we provide direct evidence from Cre/Lox-mediated lineage tracing and loss-of-function studies in zebrafish, a lower vertebrate with a single ventricle, that latent TGF-ß binding protein 3 (ltbp3) transcripts mark a field of cardiac progenitor cells with defining characteristics of the anterior SHF in mammals. Specifically, ltbp3(+) cells differentiate in pharyngeal mesoderm after formation of the heart tube, elongate the heart tube at the outflow pole, and give rise to three cardiovascular lineages in the outflow tract and myocardium in the distal ventricle. In addition to expressing Ltbp3, a protein that regulates the bioavailability of TGF-ß ligands, zebrafish SHF cells co-express nkx2.5, an evolutionarily conserved marker of cardiac progenitor cells in both fields. Embryos devoid of ltbp3 lack the same cardiac structures derived from ltbp3(+) cells due to compromised progenitor proliferation. Furthermore, small-molecule inhibition of TGF-ß signalling phenocopies the ltbp3-morphant phenotype whereas expression of a constitutively active TGF-ß type I receptor rescues it. Taken together, our findings uncover a requirement for ltbp3-TGF-ß signalling during zebrafish SHF development, a process that serves to enlarge the single ventricular chamber in this species.


Asunto(s)
Corazón/embriología , Proteínas de Unión a TGF-beta Latente/metabolismo , Miocardio/metabolismo , Pez Cebra/embriología , Animales , Anomalías Cardiovasculares/embriología , Linaje de la Célula , Técnicas de Silenciamiento del Gen , Proteína Homeótica Nkx-2.5 , Datos de Secuencia Molecular , Miocardio/citología , Fenotipo , Transducción de Señal , Factores de Transcripción/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
J Ultrasound Med ; 36(5): 1015-1019, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28258603

RESUMEN

OBJECTIVES: The aim of this study was to assess the incidence of an aberrant right subclavian artery (ARSA) among an unselected population during second-trimester sonography and to review the importance of this conotruncal variant as a marker of Down syndrome. METHODS: The presence or absence of an ARSA was assessed in an unselected population of 1913 second-trimester fetuses. RESULTS: Among the 1913 patients, an ARSA was detected in 20 fetuses (1.04%), all with a normal karyotype. Thirteen of 20 fetuses had an isolated ARSA, and 7 of them were nonisolated. Associated abnormal sonographic findings were an intracardiac echogenic focus (n = 3), a choroid plexus cyst (n = 1), pyelectasis (n = 1) and tetralogy of Fallot (n = 2). One of the cases of tetralogy of Fallot was also associated with a persistent left superior vena cava, a persistent right umbilical vein, hydrocephalus, rhombencephalosynapsis, and unilateral renal agenesis. There were only 2 fetuses with Down syndrome in this group, and both of them had a normal origin of the right subclavian artery. CONCLUSIONS: In an unselected population, an ARSA may be seen less frequently than in a high-risk population and may not be related to Down syndrome. An isolated ARSA is not a sufficient indication for karyotype analysis; it can be managed with noninvasive prenatal testing rather than invasive testing.


Asunto(s)
Aneurisma/complicaciones , Aneurisma/diagnóstico por imagen , Anomalías Cardiovasculares/complicaciones , Anomalías Cardiovasculares/diagnóstico por imagen , Síndrome de Down/complicaciones , Segundo Trimestre del Embarazo , Arteria Subclavia/anomalías , Ultrasonografía Prenatal/métodos , Adolescente , Adulto , Aneurisma/embriología , Anomalías Cardiovasculares/embriología , Femenino , Humanos , Incidencia , Persona de Mediana Edad , Embarazo , Estudios Retrospectivos , Arteria Subclavia/diagnóstico por imagen , Arteria Subclavia/embriología , Turquía , Adulto Joven
4.
J Ultrasound Med ; 36(4): 785-790, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28072476

RESUMEN

OBJECTIVES: To determine whether fetuses with an isolated aberrant course of the right subclavian artery (ARSA) have increased risk for chromosomal abnormalities, including trisomy 21 or 22q11 deletion. METHODS: We performed a retrospective chart review of all fetuses with antenatally diagnosed ARSA. Data were collected from fetal anatomic surveys, fetal echocardiograms, noninvasive trisomy 21 screening programs, invasive genetic studies, and neonatal records. RESULTS: Seventy-nine fetuses with ARSA were identified at 20.3 ± 3.8 weeks' gestation. Forty-eight fetuses underwent chromosomal evaluation. Of those, seven had trisomy 21. Four other fetuses had unusual karyotype abnormalities. All fetuses with genetic anomalies had additional aberrant ultrasound findings. There were three spontaneous fetal deaths (trisomy 21-2 and Wolf-Hirshhorn-1). Nine pregnancies were terminated because of abnormalities and one died as a result of hypoplastic left heart syndrome. No neonate was found or suspected to have 22q11.2 deletion. The ARSA was isolated in 43 fetuses; all had unremarkable neonatal outcomes, and none were readmitted within 6 months after discharge. CONCLUSIONS: As an apparently isolated finding, ARSA is benign and not associated with trisomy 21 or 22q11.2 deletion. The finding of ARSA, however, warrants a detailed fetal ultrasound. All fetuses with ARSA and genetic anomalies had additional ultrasound findings.


Asunto(s)
Aneurisma/diagnóstico por imagen , Aneurisma/embriología , Anomalías Cardiovasculares/diagnóstico por imagen , Anomalías Cardiovasculares/embriología , Síndrome de Down/diagnóstico , Ecocardiografía/métodos , Pruebas Genéticas/métodos , Arteria Subclavia/anomalías , Ultrasonografía Prenatal/métodos , Aneurisma/complicaciones , Anomalías Cardiovasculares/complicaciones , Síndrome de Down/complicaciones , Femenino , Humanos , Recién Nacido , Masculino , Embarazo , Estudios Retrospectivos , Arteria Subclavia/diagnóstico por imagen , Arteria Subclavia/embriología
5.
Circ Res ; 114(6): 957-65, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24478334

RESUMEN

RATIONALE: Our previous study has shown that yes-associated protein (YAP) plays a crucial role in the phenotypic modulation of vascular smooth muscle cells (SMCs) in response to arterial injury. However, the role of YAP in vascular SMC development is unknown. OBJECTIVE: The goal of this study was to investigate the functional role of YAP in cardiovascular development in mice and determine the mechanisms underlying YAP's actions. METHODS AND RESULTS: YAP was deleted in cardiomyocytes and vascular SMCs by crossing YAP flox mice with SM22α-Cre transgenic mice. Cardiac/SMC-specific deletion of YAP directed by SM22α-Cre resulted in perinatal lethality in mice because of profound cardiac defects including hypoplastic myocardium, membranous ventricular septal defect, and double outlet right ventricle. The cardiac/SMC-specific YAP knockout mice also displayed severe vascular abnormalities including hypoplastic arterial wall, short/absent brachiocephalic artery, and retroesophageal right subclavian artery. Deletion of YAP in mouse vascular SMCs induced expression of a subset of cell cycle arrest genes including G-protein-coupled receptor 132 (Gpr132). Silencing Gpr132 promoted SMC proliferation, whereas overexpression of Gpr132 attenuated SMC growth by arresting cell cycle in G0/G1 phase, suggesting that ablation of YAP-induced impairment of SMC proliferation was mediated, at least in part, by induction of Gpr132 expression. Mechanistically, YAP recruited the epigenetic repressor histone deacetylase-4 to suppress Gpr132 gene expression via a muscle CAT element in the Gpr132 gene. CONCLUSIONS: YAP plays a critical role in cardiac/SMC proliferation during cardiovascular development by epigenetically regulating expression of a set of cell cycle suppressors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Anomalías Cardiovasculares/genética , Corazón Fetal/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Miocitos Cardíacos/citología , Miocitos del Músculo Liso/citología , Fosfoproteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Aneurisma/genética , Animales , Tronco Braquiocefálico/anomalías , Anomalías Cardiovasculares/embriología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , División Celular , Células Cultivadas , Ventrículo Derecho con Doble Salida/embriología , Ventrículo Derecho con Doble Salida/genética , Regulación del Desarrollo de la Expresión Génica/genética , Genes Letales , Genes cdc , Defectos del Tabique Interventricular/embriología , Defectos del Tabique Interventricular/genética , Histona Desacetilasas/metabolismo , Histona Desacetilasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/patología , Miocitos Cardíacos/patología , Miocitos del Músculo Liso/patología , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Ratas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Arteria Subclavia/anomalías , Proteínas Señalizadoras YAP
6.
J Exp Biol ; 218(Pt 21): 3468-77, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26347567

RESUMEN

In this study, we show that high-salt exposure dramatically increases chick mortality during embryo development. As embryonic mortality at early stages mainly results from defects in cardiovascular development, we focused on heart formation and angiogenesis. We found that high-salt exposure enhanced the risk of abnormal heart tube looping and blood congestion in the heart chamber. In the presence of high salt, both ventricular cell proliferation and apoptosis increased. The high osmolarity induced by high salt in the ventricular cardiomyocytes resulted in incomplete differentiation, which might be due to reduced expression of Nkx2.5 and GATA4. Blood vessel density and diameter were suppressed by exposure to high salt in both the yolk sac membrane (YSM) and chorioallantoic membrane models. In addition, high-salt-induced suppression of angiogenesis occurred even at the vasculogenesis stage, as blood island formation was also inhibited by high-salt exposure. At the same time, cell proliferation was repressed and cell apoptosis was enhanced by high-salt exposure in YSM tissue. Moreover, the reduction in expression of HIF2 and FGF2 genes might cause high-salt-suppressed angiogenesis. Interestingly, we show that high-salt exposure causes excess generation of reactive oxygen species (ROS) in the heart and YSM tissues, which could be partially rescued through the addition of antioxidants. In total, our study suggests that excess generation of ROS might play an important role in high-salt-induced defects in heart and angiogenesis.


Asunto(s)
Anomalías Cardiovasculares/inducido químicamente , Desarrollo Embrionario/efectos de los fármacos , Corazón/efectos de los fármacos , Cloruro de Sodio/farmacología , Animales , Antioxidantes/farmacología , Apoptosis , Anomalías Cardiovasculares/embriología , Proliferación Celular , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Morfogénesis , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Saco Vitelino/irrigación sanguínea , Saco Vitelino/efectos de los fármacos
7.
Circ Res ; 113(1): 22-31, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23603510

RESUMEN

RATIONALE: The peptide ligand apelin and its receptor APJ constitute a signaling pathway with numerous effects on the cardiovascular system, including cardiovascular development in model organisms such as xenopus and zebrafish. OBJECTIVE: This study aimed to characterize the embryonic lethal phenotype of the Apj-/- mice and to define the involved downstream signaling targets. METHODS AND RESULTS: We report the first characterization of the embryonic lethality of the Apj-/- mice. More than half of the expected Apj-/- embryos died in utero because of cardiovascular developmental defects. Those succumbing to early embryonic death had markedly deformed vasculature of the yolk sac and the embryo, as well as poorly looped hearts with aberrantly formed right ventricles and defective atrioventricular cushion formation. Apj-/- embryos surviving to later stages demonstrated incomplete vascular maturation because of a deficiency of vascular smooth muscle cells and impaired myocardial trabeculation and ventricular wall development. The molecular mechanism implicates a novel, noncanonical signaling pathway downstream of apelin-APJ involving Gα13, which induces histone deacetylase (HDAC) 4 and HDAC5 phosphorylation and cytoplasmic translocation, resulting in activation of myocyte enhancer factor 2. Apj-/- mice have greater endocardial Hdac4 and Hdac5 nuclear localization and reduced expression of the myocyte enhancer factor 2 (MEF2) transcriptional target Krüppel-like factor 2. We identify a number of commonly shared transcriptional targets among apelin-APJ, Gα13, and MEF2 in endothelial cells, which are significantly decreased in the Apj-/- embryos and endothelial cells. CONCLUSIONS: Our results demonstrate a novel role for apelin-APJ signaling as a potent regulator of endothelial MEF2 function in the developing cardiovascular system.


Asunto(s)
Anomalías Cardiovasculares/embriología , Sistema Cardiovascular/embriología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Factores Reguladores Miogénicos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transporte Activo de Núcleo Celular , Adipoquinas , Animales , Apelina , Receptores de Apelina , Anomalías Cardiovasculares/genética , Endocardio/embriología , Endocardio/metabolismo , Endotelio Vascular/metabolismo , Femenino , Corazón Fetal/anomalías , Subunidades alfa de la Proteína de Unión al GTP G12-G13/fisiología , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Histona Desacetilasas/metabolismo , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción MEF2 , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Transcripción Genética
8.
Dev Biol ; 383(2): 239-52, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24056078

RESUMEN

Neural crest cells (NCCs) are physically responsible for craniofacial skeleton formation, pharyngeal arch artery remodeling and cardiac outflow tract septation during vertebrate development. Cdc42 (cell division cycle 42) is a Rho family small GTP-binding protein that works as a molecular switch to regulate cytoskeleton remodeling and the establishment of cell polarity. To investigate the role of Cdc42 in NCCs during embryonic development, we deleted Cdc42 in NCCs by crossing Cdc42 flox mice with Wnt1-cre mice. We found that the inactivation of Cdc42 in NCCs caused embryonic lethality with craniofacial deformities and cardiovascular developmental defects. Specifically, Cdc42 NCC knockout embryos showed fully penetrant cleft lips and short snouts. Alcian Blue and Alizarin Red staining of the cranium exhibited an unfused nasal capsule and palatine in the mutant embryos. India ink intracardiac injection analysis displayed a spectrum of cardiovascular developmental defects, including persistent truncus arteriosus, hypomorphic pulmonary arteries, interrupted aortic arches, and right-sided aortic arches. To explore the underlying mechanisms of Cdc42 in the formation of the great blood vessels, we generated Wnt1Cre-Cdc42-Rosa26 reporter mice. By beta-galactosidase staining, a subpopulation of Cdc42-null NCCs was observed halting in their migration midway from the pharyngeal arches to the conotruncal cushions. Phalloidin staining revealed dispersed, shorter and disoriented stress fibers in Cdc42-null NCCs. Finally, we demonstrated that the inactivation of Cdc42 in NCCs impaired bone morphogenetic protein 2 (BMP2)-induced NCC cytoskeleton remodeling and migration. In summary, our results demonstrate that Cdc42 plays an essential role in NCC migration, and inactivation of Cdc42 in NCCs impairs craniofacial and cardiovascular development in mice.


Asunto(s)
Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/enzimología , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/enzimología , Morfogénesis , Cresta Neural/patología , Proteína de Unión al GTP cdc42/metabolismo , Actinas/metabolismo , Animales , Proteína Morfogenética Ósea 2/farmacología , Anomalías Cardiovasculares/patología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Anomalías Craneofaciales/patología , Cruzamientos Genéticos , Citoesqueleto/metabolismo , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/patología , Activación Enzimática/efectos de los fármacos , Femenino , Eliminación de Gen , Genotipo , Masculino , Ratones , Ratones Noqueados , Morfogénesis/efectos de los fármacos , Cresta Neural/efectos de los fármacos , Cresta Neural/enzimología , Osteogénesis/efectos de los fármacos , Fenotipo , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Timo/anomalías , Timo/efectos de los fármacos , Timo/patología
9.
J Biol Chem ; 288(12): 8566-8574, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23386621

RESUMEN

WNK1 is a widely expressed serine/threonine protein kinase that regulates multiple cellular and organ functions via diverse mechanisms. We previously reported that endothelial-specific deletion of Wnk1 in mice results in embryonic lethality, with angiogenesis and cardiac defects beginning at embryonic day ∼10.5. Here, we further investigated the signaling mechanism by which WNK1 regulates embryonic cardiovascular development. We found that mice with a global deletion of Osr1, which encodes oxidative stress-responsive kinase-1, a protein kinase activated by WNK1, died in utero beginning at embryonic day ∼11. The defects in Osr1-null yolk sacs and embryos were virtually identical to those observed in Wnk1-knock-out mice: no mature large vessels in yolk sacs, defective angiogenesis in the brain and intersomitic vessels, and smaller chambers and reduced myocardial trabeculation in mutant hearts. Endothelial-specific deletion of Osr1 generated by crossing Osr1(flox/flox) mice with Tie2-Cre mice phenocopied defects caused by global Osr1 deletion. To investigate whether OSR1 acts downstream of WNK1 in embryonic angiogenesis, we generated a mouse line that carries a catalytically and constitutively active human OSR1 transgene in the ROSA26 locus under the control of a cassette of floxed transcription stop codons. We found that endothelial-specific expression of the constitutively active mutant OSR1, generated by Tie2-Cre-mediated excision of floxed stop codons in the mutated ROSA26 locus, rescued angiogenesis and cardiac defects in global Wnk1-null embryos. These results indicate that WNK1 activation of the OSR1 signaling cascade is an essential pathway that regulates angiogenesis and cardiac formation during mouse embryo development.


Asunto(s)
Corazón/embriología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Alelos , Animales , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Sistema Cardiovascular/embriología , Sistema Cardiovascular/enzimología , Sistema Cardiovascular/patología , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Femenino , Regulación de la Expresión Génica , Genes Letales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos de Histocompatibilidad Menor , Neovascularización Fisiológica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Proteína Quinasa Deficiente en Lisina WNK 1
10.
Development ; 138(10): 1925-34, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21490058

RESUMEN

Protein O-glucosylation is a conserved post-translational modification that occurs on epidermal growth factor-like (EGF) repeats harboring the C(1)-X-S-X-P-C(2) consensus sequence. The Drosophila protein O-glucosyltransferase (Poglut) Rumi regulates Notch signaling, but the contribution of protein O-glucosylation to mammalian Notch signaling and embryonic development is not known. Here, we show that mouse Rumi encodes a Poglut, and that Rumi(-/-) mouse embryos die before embryonic day 9.5 with posterior axis truncation and severe defects in neural tube development, somitogenesis, cardiogenesis and vascular remodeling. Rumi knockdown in mouse cell lines results in cellular and molecular phenotypes of loss of Notch signaling without affecting Notch ligand binding. Biochemical, cell culture and cross-species transgenic experiments indicate that a decrease in Rumi levels results in reduced O-glucosylation of Notch EGF repeats, and that the enzymatic activity of Rumi is key to its regulatory role in the Notch pathway. Genetic interaction studies show that removing one copy of Rumi in a Jag1(+/-) (jagged 1) background results in severe bile duct morphogenesis defects. Altogether, our data indicate that addition of O-glucose to EGF repeats is essential for mouse embryonic development and Notch signaling, and that Jag1-induced signaling is sensitive to the gene dosage of the protein O-glucosyltransferase Rumi. Given that Rumi(-/-) embryos show more severe phenotypes compared to those displayed by other global regulators of canonical Notch signaling, Rumi is likely to have additional important targets during mammalian development.


Asunto(s)
Desarrollo Embrionario/fisiología , Glucosiltransferasas/metabolismo , Receptores Notch/metabolismo , Anomalías Múltiples/embriología , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Animales , Conductos Biliares Intrahepáticos/anomalías , Conductos Biliares Intrahepáticos/metabolismo , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Línea Celular , Proteínas de Drosophila , Desarrollo Embrionario/genética , Factor de Crecimiento Epidérmico/genética , Femenino , Dosificación de Gen , Glucosiltransferasas/deficiencia , Glucosiltransferasas/genética , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Hígado/anomalías , Hígado/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Fenotipo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal
11.
Proc Natl Acad Sci U S A ; 108(1): 149-54, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21173240

RESUMEN

Dynamic signals linking the actin cytoskeleton and cell adhesion receptors are essential for morphogenesis during development and normal tissue homeostasis. Abi1 is a central regulator of actin polymerization through interactions with multiple protein complexes. However, the in vivo role of Abi1 remains to be defined. The α4 integrin adhesion receptor is associated with enhanced protrusive activity and regulation of directional cell migration. Among integrin subunits, α4 exhibits unique properties in that it predominantly accumulates at the leading edge of migrating cells; however, the pathways that link the actin-regulatory machinery to α4 at the leading edge have remained elusive. We generated Abi1 KO mice and found that loss of Abi1 phenocopies KO of α4. Mice lacking Abi1 or α4 exhibit midgestational lethality with abnormalities in placental and cardiovascular development. Notably, purified Abi1 protein binds directly to the α4 cytoplasmic tail and endogenous Abi1 colocalizes with phosphorylated α4 at the leading edge of spreading cells. Moreover, Abi1-deficient cells expressing α4 have impaired cell spreading, which is rescued by WT Abi1 but not an Abi1 mutant lacking the α4-binding site. These data reveal a direct link between the α4 integrin and actin polymerization and uncover a role for Abi1 in the regulation of morphogenesis in vivo. The Abi1-α4 interaction establishes a mechanistic paradigm for signaling between adhesion events and enhanced actin polymerization at the earliest stages of protrusion.


Asunto(s)
Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anomalías Cardiovasculares/embriología , Movimiento Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Integrina alfa4/metabolismo , Morfogénesis/genética , Placenta/embriología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Southern Blotting , Anomalías Cardiovasculares/genética , Proteínas del Citoesqueleto/genética , Cartilla de ADN/genética , Femenino , Genotipo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Placenta/anomalías , Polimerizacion , Embarazo
12.
Fetal Diagn Ther ; 36(1): 59-68, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24903004

RESUMEN

INTRODUCTION: Bicuspid aortic valve (BAV) is common in Turner syndrome (TS). In adult TS, 82-95% of BAVs have fusion of the right and left coronary leaflets. Data in fetal stages are scarce. The purpose of this study was to gain insight into aortic valve morphology and associated cardiovascular abnormalities in a fetal TS cohort with adverse outcome early in development. MATERIAL AND METHODS: We studied post-mortem heart specimens of 36 TS fetuses and 1 TS newborn. RESULTS: BAV was present in 28 (76%) hearts. BAVs showed fusion of the right and left coronary leaflet (type 1 BAV) in 61%, and fusion of the right coronary and non-coronary leaflet (type 2 BAV) in 39%. There were no significant differences in occurrence of additional cardiovascular abnormalities between type 1 and type 2 BAV. However, all type 2 BAV hearts showed ascending aorta hypoplasia and tubular hypoplasia of the B segment, as opposed to only 55 and 64% of type 1 BAV hearts, respectively. DISCUSSION: The proportion of type 2 BAV seems higher in TS fetuses than in adults. Fetal type 2 BAV hearts all had severe aortic pathology, possibly contributing to a worse prognosis of type 2 than type 1 BAV in TS.


Asunto(s)
Válvula Aórtica/anomalías , Corazón Fetal/anomalías , Corazón Fetal/patología , Enfermedades de las Válvulas Cardíacas/patología , Síndrome de Turner/patología , Válvula Aórtica/embriología , Válvula Aórtica/patología , Enfermedad de la Válvula Aórtica Bicúspide , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/patología , Femenino , Corazón Fetal/embriología , Corazón/embriología , Enfermedades de las Válvulas Cardíacas/embriología , Humanos , Recién Nacido , Síndrome de Turner/embriología
13.
Ginekol Pol ; 85(10): 742-7, 2014 Oct.
Artículo en Polaco | MEDLINE | ID: mdl-25546924

RESUMEN

OBJECTIVES: Presentation of our own, preliminary experiences in the assessment of the right subclavian artery's (RSA) position during the second trimester scan. MATERIAL AND METHODS: Since January 2012 our center has started to conduct the assessment of the position of the right subclavian artery in the second trimester scan. Patients who were diagnosed with an aberrant right subclavian artery (ARSA) were referred to invasive method of prenatal diagnosis. Abnormal karyotype and microdeletion 22q11 were analyzed. Detailed echocardiography was conducted in each case. RESULTS: Between January 2012 and September 2013 we diagnosed 19 cases of ARSA. There were three cases of congenital heart defect (15.8%; 3/19) (ventricular septal defect--VSD, n=2, atrioventricular septal defect--AVSD, n=1). Two out of 17 cases showed an abnormal karyotype (11.8%; 2/17)--46,XY del(5) (q15q31) and 47,XX+18. No 22q11.2 deletions were observed. Two patients did not consent to invasive methods of prenatal diagnosis. CONCLUSIONS: The position of the right subclavian artery (RSA) should be routinely assessed during the second trimester of ultrasound screening. The presence of ARSA increases the risk for abnormal karyotype in the fetus and therefore, all patients who are diagnosed with ARSA should be referred to the reference center.


Asunto(s)
Aneurisma/diagnóstico por imagen , Aneurisma/embriología , Anomalías Cardiovasculares/diagnóstico por imagen , Anomalías Cardiovasculares/embriología , Trastornos de Deglución/diagnóstico por imagen , Trastornos de Deglución/embriología , Arteria Subclavia/anomalías , Adulto , Deleción Cromosómica , Cromosomas Humanos Par 22/genética , Femenino , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Medida de Translucencia Nucal , Embarazo , Segundo Trimestre del Embarazo , Diagnóstico Prenatal/métodos , Arteria Subclavia/diagnóstico por imagen , Arteria Subclavia/embriología
14.
Cell Metab ; 7(3): 258-68, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18316031

RESUMEN

The cellular uptake of vitamin A from its RBP4-bound circulating form (holo-RBP4) is a homeostatic process that evidently depends on the multidomain membrane protein STRA6. In humans, mutations in STRA6 are associated with Matthew-Wood syndrome, manifested by multisystem developmental malformations. Here we addressed the metabolic basis of this inherited disease. STRA6-dependent transfer of retinol from RBP4 into cultured NIH 3T3 fibroblasts was enhanced by lecithin:retinol acyltransferase (LRAT). The retinol transfer was bidirectional, strongly suggesting that STRA6 acts as a retinol channel/transporter. Loss-of-function analysis in zebrafish embryos revealed that Stra6 deficiency caused vitamin A deprivation of the developing eyes. We provide evidence that, in the absence of Stra6, holo-Rbp4 provokes nonspecific vitamin A excess in several embryonic tissues, impairing retinoic acid receptor signaling and gene regulation. These fatal consequences of Stra6 deficiency, including craniofacial and cardiac defects and microphthalmia, were largely alleviated by reducing embryonic Rbp4 levels by morpholino oligonucleotide or pharmacological treatments.


Asunto(s)
Anomalías Múltiples/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Vitamina A/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Anomalías Múltiples/genética , Aciltransferasas/metabolismo , Animales , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/metabolismo , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/metabolismo , Modelos Animales de Enfermedad , Ojo/embriología , Ojo/enzimología , Ojo/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Homeostasis , Humanos , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/genética , Ratones , Morfolinas/metabolismo , Células 3T3 NIH , Oligonucleótidos Antisentido/metabolismo , Proteínas Plasmáticas de Unión al Retinol/genética , Síndrome , Factores de Tiempo , Transducción Genética , Tretinoina/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
15.
Artículo en Inglés | MEDLINE | ID: mdl-23716477

RESUMEN

BACKGROUND: Heart defects are the most common abnormalities in infants of diabetic mothers. Cardiac malformation is associated with altered expression of the genes in the transforming growth factor ß system, including inhibin ßA, which forms activin-A as a homodimer and functions through its effectors, Smad2 and Smad3. This study aimed to investigate the role of activin-A in diabetes-induced cardiac malformations. METHODS: Diabetes mellitus in female mice (C57BL/6J) was induced via intravenous injection of streptozotocin. The expression of inhibin ßA protein and phosphorylation of Smad2 and Smad3 in the embryonic hearts were examined using immunohistochemical, in situ proximity ligation, and immunoblot assays. Embryos and endocardial cushions of nondiabetic mice were cultured in a high concentration of glucose and treated with activin-A. Mitosis was examined using BrdU incorporation assay and immunohistochemistry of phosphorylated histone H3. Migration of the endocardial cells was assessed using a collagen-based cell migration assay. RESULTS: The levels of inhibin ßA expression and Smad2 and Smad3 activation were significantly reduced by maternal diabetes. Treatment with activin-A significantly increased cell proliferation in the myocardium and migration of endocardial cells, compared with those in vehicle-treated high glucose group, to the level in the euglycemic control group. CONCLUSIONS: Maternal diabetes suppresses the expression of inhibin ßA protein, as well as the activation of Smad2 and Smad3. Activin-A rescues cell proliferation in the myocardium and migration of the endocardial cells suppressed by hyperglycemia. The activin-Smad2/3 signaling system appears to play a role in cardiac malformation in diabetic embryopathy.


Asunto(s)
Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/etiología , Subunidades beta de Inhibinas/metabolismo , Embarazo en Diabéticas , Animales , Bromodesoxiuridina/metabolismo , Anomalías Cardiovasculares/tratamiento farmacológico , Anomalías Cardiovasculares/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Endocardio/metabolismo , Endocardio/patología , Femenino , Inmunohistoquímica , Subunidades beta de Inhibinas/farmacología , Subunidades beta de Inhibinas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Fosforilación , Embarazo , Embarazo en Diabéticas/metabolismo , Proteína smad3/metabolismo
16.
Prenat Diagn ; 32(10): 968-75, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22847746

RESUMEN

OBJECTIVES: To evaluate the feasibility of examining aberrant right subclavian artery (ARSA) at first and second trimester gestation, its prevalence and associations in an unselected population. METHODS: Right subclavian artery (RSA) was prospectively evaluated in 6617 routine patients. When ARSA was detected, fetal echocardiography was offered and fetal karyotyping was discussed. If invasive testing was performed with normal karyotype, fluorescence in situ hybridization for 22q11.2 microdeletion and additionally, in case of nuchal translucency (NT) measurement above the 99(th) centile, oligo array-based comparative genomic hybridization, were offered. In all aneuploidies, NT and first trimester additional ultrasonographic (US) markers assessment (nasal bone, tricuspid valve, ductus venosus) were recorded. RESULTS: RSA assessment was feasible in 85.3% and 98% of first and second trimester examinations, respectively (overall feasibility 94%). There were detected 89 ARSA (1.42% of the feasible cases), of which 66 in the first trimester. More than 20% were associated to other abnormalities: 10 aneuploidies; 2 microdeletions (15q11.2 and 22q11.2); in the euploid fetuses, 8 associated abnormalities were observed, 4 of which were cardiac defects. In the case of 22q11.2 microdeletion, ARSA was associated only with increased NT. CONCLUSION: Prenatal routine US assessment of the RSA is feasible by highly experienced operators in first trimester screening. There is an important association of ARSA detected in unselected population with fetal abnormalities, including aneuploidies, cardiac defects and genetic anomalies. In trisomy 21 fetuses, ARSA can be the only first trimester US marker or, when associated to increased NT, it can represent the only 'additional' marker.


Asunto(s)
Aneurisma/diagnóstico por imagen , Aneurisma/embriología , Anomalías Cardiovasculares/diagnóstico por imagen , Anomalías Cardiovasculares/embriología , Trastornos de Deglución/diagnóstico por imagen , Trastornos de Deglución/embriología , Edad Gestacional , Diagnóstico Prenatal/métodos , Ultrasonografía Prenatal , Aneuploidia , Deleción Cromosómica , Cromosomas Humanos Par 22/genética , Anomalías Congénitas/diagnóstico por imagen , Estudios de Factibilidad , Femenino , Cardiopatías Congénitas/diagnóstico por imagen , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Medida de Translucencia Nucal , Embarazo , Estudios Prospectivos , Arteria Subclavia/anomalías , Arteria Subclavia/diagnóstico por imagen , Arteria Subclavia/embriología
17.
Proc Natl Acad Sci U S A ; 106(52): 22305-10, 2009 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-19966217

RESUMEN

Von Recklinghausen neurofibromatosis is a common autosomal dominant genetic disorder characterized by benign and malignant tumors of neural crest origin. Significant progress in understanding the pathophysiology of this disease has occurred in recent years, largely aided by the development of relevant animal models. Von Recklinghausen neurofibromatosis is caused by mutations in the NF1 gene, which encodes neurofibromin, a large protein that modulates the activity of Ras. Here, we describe the identification and characterization of zebrafish nf1a and nf1b, orthologues of NF1, and show neural crest and cardiovascular defects resulting from morpholino knockdown, including vascular and cardiac valvular abnormalities. Development of a zebrafish model of von Recklinghausen neurofibromatosis will allow for structure-function analysis and genetic screens in this tractable vertebrate system.


Asunto(s)
Fenómenos Fisiológicos Cardiovasculares/genética , Genes de Neurofibromatosis 1 , Neurofibromatosis 1/genética , Pez Cebra/genética , Pez Cebra/fisiología , Animales , Secuencia de Bases , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Modelos Animales de Enfermedad , Humanos , Hibridación in Situ , Mutación , Neurofibromatosis 1/patología , Neurofibromatosis 1/fisiopatología , Oligodesoxirribonucleótidos Antisentido/genética , Filogenia , Especificidad de la Especie , Pez Cebra/embriología
18.
Dev Dyn ; 240(1): 271-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21181944

RESUMEN

Morphogenesis of the heart is regulated by various cues, including growth factors and extracellular matrix (ECM) proteins. The mechanisms by which cardiac cells properly integrate these cues to regulate growth, differentiation, and migration remain poorly understood. Here we have used genetic strategies in mice to identify αvß8 integrin and its cytoskeletal adaptor protein, Band 4.1B, as essential regulators of cardiac morphogenesis. We demonstrate that approximately 60% of mouse embryos genetically null for ß8 integrin and Band 4.1B display cardiovascular phenotypes and die by E11.5. This premature death is due, in part, to defective development of the cardiac outflow tract (OFT), with reduced expression of smooth muscle α-actin (SMAα-actin) in OFT cells derived from the cardiac neural crest. These data are the first to identify cell adhesion and signaling pathways regulated by αvß8 integrin and Band 4.1B as essential for normal formation and function of the heart during embryogenesis.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Corazón/embriología , Cadenas beta de Integrinas/fisiología , Proteínas de la Membrana/fisiología , Animales , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Sistema Nervioso Central/anomalías , Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Cadenas beta de Integrinas/genética , Cadenas beta de Integrinas/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
19.
Cells Tissues Organs ; 193(6): 393-403, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21123999

RESUMEN

Maldevelopment of outflow tract and aortic arch arteries is among the most common forms of human congenital heart diseases. Both Bmp4 and Tbx1 are known to play critical roles during cardiovascular development. Expression of these two genes partially overlaps in pharyngeal arch areas in mouse embryos. In this study, we applied a conditional gene inactivation approach to test the hypothesis that Bmp4 expressed from the Tbx1 expression domain plays a critical role for normal development of outflow tract and pharyngeal arch arteries. We showed that inactivation of Bmp4 from Tbx1-expressing cells leads to the spectrum of deformities resembling the cardiovascular defects observed in human DiGeorge syndrome patients. Inactivation of Bmp4 from the Tbx1 expression domain did not cause patterning defects, but affected remodeling of outflow tract and pharyngeal arch arteries. Our further examination revealed that Bmp4 is required for normal recruitment/differentiation of smooth muscle cells surrounding the PAA4 and survival of outflow tract cushion mesenchymal cells.


Asunto(s)
Arterias/anomalías , Proteína Morfogenética Ósea 4/genética , Región Branquial/anomalías , Región Branquial/irrigación sanguínea , Sistema Cardiovascular/embriología , Silenciador del Gen , Proteínas de Dominio T Box/genética , Animales , Aorta Torácica/anomalías , Aorta Torácica/embriología , Aorta Torácica/patología , Apoptosis , Arterias/patología , Biomarcadores/metabolismo , Región Branquial/patología , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/patología , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patología , Proliferación Celular , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/patología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/patología , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Humanos , Integrasas , Ratones , Penetrancia , Fenotipo
20.
Proc Natl Acad Sci U S A ; 105(18): 6638-43, 2008 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-18448675

RESUMEN

Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase that plays an important role in integrin-mediated signal transduction. To explore the role and mechanisms of FAK in cardiac development, we inactivated FAK in embryonic cardiomyocytes by crossing the floxed FAK mice with myosin light chain-2a (MLC2a) Cre mice, which expressed Cre as early as embryonic day 9.5 in the heart. The majority of conditional FAK knockout mice generated from MLC2a-Cre (CFKO-2a) died in the embryonic stage with thin ventricular wall and ventricular septal defects. A small fraction of CFKO-2a mice survived to adulthood with spontaneous eccentric right ventricle hypertrophy. Transmission electron microscopy analysis displayed swelling in the rough endoplasmic reticulum in CFKO-2a embryonic cardiomyocytes. We found that decreased cell proliferation, but not increased cell apoptosis or differentiation, is the reason for the thin ventricular wall in CFKO-2a mice. Microarray analysis suggests that myocyte enhancer factor 2a (MEF2a) can be regulated by FAK and that inactivation of FAK in the embryonic heart compromised MEF2a expression. Last, we found that Src, but not PI3K, is important in mediating signal transduction for the regulation of MEF2a by FAK. Together, these results identified the role and mechanisms of FAK in embryonic cardiac development.


Asunto(s)
Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/deficiencia , Hipertrofia Ventricular Derecha/enzimología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Animales , Proliferación Celular , Embrión de Mamíferos/enzimología , Desarrollo Embrionario , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Eliminación de Gen , Integrasas/metabolismo , Factores de Transcripción MEF2 , Ratones , Ratones Noqueados , Miocardio/enzimología , Miocardio/patología , Miocardio/ultraestructura , Factores Reguladores Miogénicos/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Especificidad de Órganos , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda