Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Birth Defects Res A Clin Mol Teratol ; 106(9): 749-60, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27296863

RESUMEN

BACKGROUND: Embryonic acute exposure to ethanol (EtOH), lithium, and homocysteine (HCy) induces cardiac defects at the time of exposure; folic acid (FA) supplementation protects normal cardiogenesis (Han et al., , ; Serrano et al., ). Our hypothesis is that EtOH exposure and FA protection relate to lipid and FA metabolism during mouse cardiogenesis and placentation. METHODS: On the morning of conception, pregnant C57BL/6J mice were placed on either of two FA-containing diets: a 3.3 mg health maintenance diet or a high FA diet of 10.5 mg/kg. Mice were injected a binge level of EtOH, HCy, or saline on embryonic day (E) 6.75, targeting gastrulation. On E15.5, cardiac and umbilical blood flow were examined by ultrasound. Embryonic cardiac tissues were processed for gene expression of lipid and FA metabolism; the placenta and heart tissues for neutral lipid droplets, or for medium chain acyl-dehydrogenase (MCAD) protein. RESULTS: EtOH exposure altered lipid-related gene expression on E7.5 in comparison to control or FA-supplemented groups and remained altered on E15.5 similarly to changes with HCy, signifying FA deficiency. In comparison to control tissues, the lipid-related acyl CoA dehydrogenase medium length chain gene and its protein MCAD were altered with EtOH exposure, as were neutral lipid droplet localization in the heart and placenta. CONCLUSION: EtOH altered gene expression associated with lipid and folate metabolism, as well as neutral lipids, in the E15.5 abnormally functioning heart and placenta. In comparison to controls, the high FA diet protected the embryo and placenta from these effects allowing normal development. Birth Defects Research (Part A) 106:749-760, 2016. © 2016 The Authors Birth Defects Research Part A: Clinical and Molecular Teratology Published by Wiley Periodicals, Inc.


Asunto(s)
Etanol/toxicidad , Ácido Fólico/farmacología , Gastrulación/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Cardiopatías Congénitas , Corazón/embriología , Metabolismo de los Lípidos/efectos de los fármacos , Placenta/metabolismo , Animales , Femenino , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/prevención & control , Ratones , Embarazo
2.
Am J Physiol Heart Circ Physiol ; 306(3): H414-21, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24271490

RESUMEN

Alcohol-induced congenital heart defects are frequently among the most life threatening and require surgical correction in newborns. The etiology of these defects, collectively known as fetal alcohol syndrome, has been the focus of much study, particularly involving cellular and molecular mechanisms. Few studies have addressed the influential role of altered cardiac function in early embryogenesis because of a lack of tools with the capability to assay tiny beating hearts. To overcome this gap in our understanding, we used optical coherence tomography (OCT), a nondestructive imaging modality capable of micrometer-scale resolution imaging, to rapidly and accurately map cardiovascular structure and hemodynamics in real time under physiological conditions. In this study, we exposed avian embryos to a single dose of alcohol/ethanol at gastrulation when the embryo is sensitive to the induction of birth defects. Late-stage hearts were analyzed using standard histological analysis with a focus on the atrio-ventricular valves. Early cardiac function was assayed using Doppler OCT, and structural analysis of the cardiac cushions was performed using OCT imaging. Our results indicated that ethanol-exposed embryos developed late-stage valvuloseptal defects. At early stages, they exhibited increased regurgitant flow and developed smaller atrio-ventricular cardiac cushions, compared with controls (uninjected and saline-injected embryos). The embryos also exhibited abnormal flexion/torsion of the body. Our evidence suggests that ethanol-induced alterations in early cardiac function have the potential to contribute to late-stage valve and septal defects, thus demonstrating that functional parameters may serve as early and sensitive gauges of cardiac normalcy and abnormalities.


Asunto(s)
Etanol/toxicidad , Cardiopatías Congénitas/etiología , Corazón/efectos de los fármacos , Animales , Coturnix , Ecocardiografía Doppler , Embrión no Mamífero/efectos de los fármacos , Etanol/farmacología , Gastrulación , Corazón/embriología , Corazón/fisiología , Hemodinámica , Tomografía de Coherencia Óptica
3.
J Cell Biol ; 178(4): 649-60, 2007 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-17698608

RESUMEN

The epiblast of the chick embryo contains cells that express MyoD mRNA but not MyoD protein. We investigated whether MyoD-positive (MyoDpos) epiblast cells are stably committed to the skeletal muscle lineage or whether their fate can be altered in different environments. A small number of MyoDpos epiblast cells were tracked into the heart and nervous system. In these locations, they expressed MyoD mRNA and some synthesized MyoD protein. No MyoDpos epiblast cells differentiated into cardiac muscle or neurons. Similar results were obtained when MyoDpos cells were isolated from the epiblast and microinjected into the precardiac mesoderm or neural plate. In contrast, epiblast cells lacking MyoD differentiated according to their environment. These results demonstrate that the epiblast contains both multipotent cells and a subpopulation of cells that are stably committed to the skeletal muscle lineage before the onset of gastrulation. Stable programming in the epiblast may ensure that MyoDpos cells express similar signaling molecules in a variety of environments.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Músculo Esquelético/citología , Proteína MioD/genética , Animales , Técnicas de Cultivo de Célula , Embrión de Pollo , Músculo Esquelético/metabolismo , ARN Mensajero/metabolismo
4.
Am J Obstet Gynecol ; 207(2): 140.e7-19, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22704764

RESUMEN

OBJECTIVE: Mouse embryonic exposure to alcohol, lithium, and homocysteine results in intrauterine growth restriction (IUGR) and cardiac defects. Our present study focused on the placental effects. We analyzed the hypothesis that expression of nonmuscle myosin (NMM)-II isoforms involved in cell motility, mechanosensing, and extracellular matrix assembly are altered by the 3 factors in human trophoblast (HTR8/SVneo) cells in vitro and in the mouse placenta in vivo. STUDY DESIGN: After exposure during gastrulation to alcohol, homocysteine, or lithium, ultrasonography defined embryos exhibiting abnormal placental blood flow. RESULTS: NMM-IIA/NMM-IIB are differentially expressed in trophoblasts and in mouse placental vascular endothelial cells under pathological conditions. Misexpression of NMM-IIA/NMM-IIB in the affected placentas continued stably to midgestation but can be prevented by folate and myoinositol supplementation. CONCLUSION: It is concluded that folate and myoinositol initiated early in mouse pregnancy can restore NMM-II expression, permit normal placentation/embryogenesis, and prevent IUGR induced by alcohol, lithium, and homocysteine.


Asunto(s)
Miosina Tipo IIA no Muscular/metabolismo , Miosina Tipo IIB no Muscular/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo , Animales , Línea Celular , Movimiento Celular , Depresores del Sistema Nervioso Central/efectos adversos , Células Endoteliales/metabolismo , Etanol/efectos adversos , Femenino , Ácido Fólico/farmacología , Homocisteína/efectos adversos , Humanos , Inositol/farmacología , Compuestos de Litio/efectos adversos , Exposición Materna/efectos adversos , Ratones , Placenta/irrigación sanguínea , Circulación Placentaria , Embarazo , Ultrasonografía Doppler , Cordón Umbilical/irrigación sanguínea , Cordón Umbilical/diagnóstico por imagen , Complejo Vitamínico B/farmacología
5.
Am J Physiol Heart Circ Physiol ; 300(3): H879-91, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21239637

RESUMEN

Analyses of form-function relationships during heart looping are directly related to technological advances. Recent advances in four-dimensional optical coherence tomography (OCT) permit observations of cardiac dynamics at high-speed acquisition rates and high resolution. Real-time observation of the avian stage 13 looping heart reveals that interactions between the endocardial and myocardial compartments are more complex than previously depicted. Here we applied four-dimensional OCT to elucidate the relationships of the endocardium, myocardium, and cardiac jelly compartments in a single cardiac cycle during looping. Six cardiac levels along the longitudinal heart tube were each analyzed at 15 time points from diastole to systole. Using image analyses, the organization of mechanotransducing molecules, fibronectin, tenascin C, α-tubulin, and nonmuscle myosin II was correlated with specific cardiac regions defined by OCT data. Optical coherence microscopy helped to visualize details of cardiac architectural development in the embryonic mouse heart. Throughout the cardiac cycle, the endocardium was consistently oriented between the midline of the ventral floor of the foregut and the outer curvature of the myocardial wall, with multiple endocardial folds allowing high-volume capacities during filling. The cardiac area fractional shortening is much higher than previously published. The in vivo profile captured by OCT revealed an interaction of the looping heart with the extra-embryonic splanchnopleural membrane providing outside-in information. In summary, the combined dynamic and imaging data show the developing structural capacity to accommodate increasing flow and the mechanotransducing networks that organize to effectively facilitate formation of the trabeculated four-chambered heart.


Asunto(s)
Corazón/fisiología , Mecanotransducción Celular/fisiología , Animales , Fibronectinas/fisiología , Corazón/embriología , Ratones , Contracción Miocárdica/fisiología , Miocardio/química , Miosina Tipo II/fisiología , Codorniz/fisiología , Tenascina/fisiología , Tomografía de Coherencia Óptica , Tubulina (Proteína)/fisiología
6.
Am J Obstet Gynecol ; 203(1): 75.e7-75.e15, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20451895

RESUMEN

OBJECTIVE: Alcohol (ethanol) consumption during pregnancy is linked to congenital heart defects that are associated with fetal alcohol syndrome. Recent reports have associated ethanol exposure with the Wnt/beta-catenin pathway. Therefore, we defined whether ethanol affects Wnt/beta-catenin signaling during cardiac cell specification. STUDY DESIGN: Pregnant mice on embryonic day 6.75 during gastrulation were exposed by an intraperitoneal injection to a binge-drinking dose of ethanol. Folic acid supplementation of mouse diet was tested for the prevention of ethanol-induced cardiac birth defects. RESULTS: Acute ethanol exposure induced myocardial wall changes and atrioventricular and semilunar valve defects, which was determined by echocardiography on embryonic day 15.5. A high folate diet prevented the ethanol-induced cardiac defects. Ethanol exposure in avian embryos suppressed 2 key Wnt-modulated genes that are involved in cardiac induction; folic acid rescued normal gene expression. CONCLUSION: Folic acid supplementation alone or with myoinositol prevented alcohol potentiation of Wnt/beta-catenin signaling that allowed normal gene activation and cardiogenesis.


Asunto(s)
Etanol/toxicidad , Trastornos del Espectro Alcohólico Fetal/fisiopatología , Ácido Fólico/farmacología , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/prevención & control , Corazón/embriología , Animales , Embrión de Pollo , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Femenino , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/fisiopatología , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Distribución Aleatoria , Ultrasonografía Doppler , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo , beta Catenina/antagonistas & inhibidores , beta Catenina/metabolismo
7.
Curr Opin Pediatr ; 22(5): 561-6, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20844350

RESUMEN

PURPOSE OF REVIEW: Environmental factors, such as drugs, chemicals, or abnormal concentrations of natural metabolites, induce birth defects. Environmental effects on cardiogenesis have been little studied in contrast to neurogenesis. This review presents evidence on three environmental factors: alcohol, the drug lithium, and the metabolite homocysteine, impacting the Wnt/ß-catenin pathway during cardiac development and folate protection. RECENT FINDINGS: Animal and epidemiological studies have shown that folate protects the embryo from birth defects. New animal studies demonstrate that folate prevents cardiovascular defects induced by the drug lithium, homocysteine, or alcohol, but protection occurs at a higher concentration than currently used in vitamin supplements. The data indicate that folate in combination with myo-inositol may further reduce the risk of birth defects. Discussion is presented of the cell specification stages that are impacted resulting in cardiac defects, how Wnt/ß-catenin signaling is involved, and how folate and myo-inositol additively may protect embryonic pathways. The possible epigenetic role of folate in Wnt/ß-catenin signaling is described. SUMMARY: This review will enable better counseling of women by defining, during early pregnancy, a susceptible window of embryonic exposure leading to a high risk of cardiac defects, and provides a therapeutic means and the necessary timing for prevention of environmentally induced birth defects.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Ácido Fólico/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Cardiopatías Congénitas/prevención & control , beta Catenina/genética , Animales , Femenino , Corazón/embriología , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Humanos , Exposición Materna/efectos adversos , Embarazo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Complejo Vitamínico B/farmacología , beta Catenina/metabolismo
8.
Cell Motil Cytoskeleton ; 65(5): 379-92, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18302173

RESUMEN

The expression of striated muscle proteins occurs early in the developing embryo in the somites and forming heart. A major component of the assembling myofibrils is the actin-binding protein tropomyosin. In vertebrates, there are four genes for tropomyosin (TM), each of which can be alternatively spliced. TPM1 can generate at least 10 different isoforms including the striated muscle-specific TPM1alpha and TPM1kappa. We have undertaken a detailed study of the expression of various TM isoforms in 2-day-old (stage HH 10-12; 33 h) heart and somites, the progenitor of future skeletal muscles. Both TPM1alpha and TPM1kappa are expressed transiently in embryonic heart while TPM1alpha is expressed in somites. Both RT-PCR and in situ hybridization data suggest that TPM1kappa is expressed in embryonic heart whereas TPM1alpha is expressed in embryonic heart, and also in the branchial arch region of somites, and in the somites. Photobleaching studies of Yellow Fluorescent Protein-TPM1alpha and -TPM1kappa expressed in cultured avian cardiomyocytes revealed that the dynamics of the two probes was the same in both premyofibrils and in mature myofibrils. This was in sharp contrast to skeletal muscle cells in which the fluorescent proteins were more dynamic in premyofibrils. We speculate that the differences in the two muscles is due to the appearance of nebulin in the skeletal myocytes premyofibrils transform into mature myofibrils.


Asunto(s)
Regulación de la Expresión Génica , Miocitos Cardíacos/metabolismo , Somitos/metabolismo , Tropomiosina/metabolismo , Animales , Pollos , Embrión no Mamífero/metabolismo , Corazón/embriología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Codorniz , Somitos/citología , Tropomiosina/genética
9.
Birth Defects Res A Clin Mol Teratol ; 82(7): 508-18, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18418887

RESUMEN

BACKGROUND: Lithium (Li) has been associated with cardiac teratogenicity in the developing fetus. We took advantage of the association of therapeutic administration of Li with an increase in heart defects to gain insight into both normal and pathological heart and valve development with GSK-3 inhibition. The objective of this study was to define whether Li mimicry of canonical Wnt/beta-catenin signaling induces cardiac valve defects. METHODS: Li was administered by a single intraperitoneal injection to the pregnant mouse on embryonic day E6.75, much earlier than heretofore analyzed. On E15.5 developing heart defects were defined by Doppler ultrasound. The embryonic hearts were analyzed for changes in patterning of active canonical Wnt expression and nuclear factor of the activated T cells-c1 (NFATc1), both key regulators of valve development. Li-exposed chick embryos were used to define the early cell populations during gastrulation that are susceptible to GSK-3 inhibition and may relate to valve formation. RESULTS: Li exposure during gastrulation decreased the number of prechordal plate (PP) cells that reached the anterior intestinal portal, a region associated with valve development. Li decreased expression of Hex, an endoderm cardiac inducing molecule, normally also expressed by the PP cells, and of Sox 4 at the anterior intestinal portal and NFAT, critical factors in valvulogenesis. CONCLUSIONS: Cells existing already during gastrulation are associated with valve formation days later. The Wnt/beta-catenin signaling in PP cells is normally repressed by Wnt antagonists and Hex is up-regulated. The antagonism occurring at the receptor level is bypassed by Li exposure by its intracellular inactivation of GSK-3 directly to augment Wnt signaling.


Asunto(s)
Antipsicóticos/efectos adversos , Gastrulación/efectos de los fármacos , Válvulas Cardíacas/anomalías , Litio/efectos adversos , Factores de Transcripción NFATC , Proteínas Wnt , Animales , Antipsicóticos/administración & dosificación , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Corazón/efectos de los fármacos , Corazón/embriología , Cardiopatías Congénitas/embriología , Válvulas Cardíacas/efectos de los fármacos , Válvulas Cardíacas/embriología , Humanos , Litio/administración & dosificación , Ratones , Miocardio/citología , Miocardio/metabolismo , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Cresta Neural/efectos de los fármacos , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
10.
ScientificWorldJournal ; 7: 280-98, 2007 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-17334619

RESUMEN

Over the past 10 years, key genes involved in specification of left-right laterality pathways in the embryo have been defined. The read-out for misexpression of laterality genes is usually the direction of heart looping. The question of how dextral looping direction occurred mechanistically and how the heart tube bends remains unknown. It is becoming clear from our experiments and those of others that left-right differences in cell proliferation in the second heart field (anterior heart field) drives the dextral direction. Evidence is accumulating that the cytoskeleton is at the center of laterality, and the bending and rotational forces associated with heart looping. If laterality pathways are modulated upstream, the cytoskeleton, including nonmuscle myosin II (NMHC-II), is altered downstream within the cardiomyocytes, leading to looping abnormalities. The cytoskeleton is associated with important mechanosensing and signaling pathways in cell biology and development. The initiation of blood flow during the looping period and the inherent stresses associated with increasing volumes of blood flowing into the heart may help to potentiate the process. In recent years, the steps involved in this central and complex process of heart development that is the basis of numerous congenital heart defects are being unraveled.


Asunto(s)
Tipificación del Cuerpo/fisiología , Proteínas del Citoesqueleto/fisiología , Citoesqueleto/fisiología , Corazón/embriología , Corazón/crecimiento & desarrollo , Mecanotransducción Celular/fisiología , Organogénesis/fisiología , Animales , Humanos , Estrés Mecánico
11.
Reprod Toxicol ; 61: 82-96, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26993217

RESUMEN

Embryonic exposures can increase the risk of congenital cardiac birth defects and adult disease. The present study identifies the predominant pathways modulated by an acute embryonic mouse exposure during gastrulation to lithium or homocysteine that induces cardiac defects. High dose periconceptional folate supplementation normalized development. Microarray bioinformatic analysis of gene expression demonstrated that primarily lipid metabolism is altered after the acute exposures. The lipid-related modulation demonstrated a gender bias with male embryos showing greater number of lipid-related Gene Ontology biological processes altered than in female embryos. RT-PCR analysis demonstrated significant change of the fatty acid oxidation gene Acadm with homocysteine exposure primarily in male embryos than in female. The perturbations resulting from the exposures resulted in growth-restricted placentas with disorganized cellular lipid droplet distribution indicating lipids have a critical role in cardiac-placental abnormal development. High folate supplementation protected normal heart-placental function, gene expression and lipid localization.


Asunto(s)
Cardiotónicos/farmacología , Ácido Fólico/farmacología , Corazón/efectos de los fármacos , Homocisteína/toxicidad , Metabolismo de los Lípidos/efectos de los fármacos , Cloruro de Litio/toxicidad , Placentación/efectos de los fármacos , Acil-CoA Deshidrogenasa/genética , Animales , Cadherinas/metabolismo , Biología Computacional , Embrión de Mamíferos , Femenino , Corazón/embriología , Masculino , Ratones Endogámicos C57BL , Miocardio/metabolismo , Placenta/efectos de los fármacos , Placenta/metabolismo , Embarazo , Transcriptoma
12.
Front Physiol ; 5: 390, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25426076

RESUMEN

Analyses of cardiovascular development have shown an important interplay between heart function, blood flow, and morphogenesis of heart structure during the formation of a four-chambered heart. It is known that changes in vitelline and placental blood flow seemingly contribute substantially to early cardiac hemodynamics. This suggests that in order to understand mammalian cardiac structure-hemodynamic functional relationships, blood flow from the extra-embryonic circulation needs to be taken into account and its possible impact on cardiogenesis defined. Previously published Doppler ultrasound analyses and data of utero-placental blood flow from human studies and those using the mouse model are compared to changes observed with environmental exposures that lead to cardiovascular anomalies. Use of current concepts and models related to mechanotransduction of blood flow and fluid forces may help in the future to better define the characteristics of normal and abnormal utero-placental blood flow and the changes in the biophysical parameters that may contribute to congenital heart defects. Evidence from multiple studies is discussed to provide a framework for future modeling of the impact of experimental changes in blood flow on the mouse heart during normal and abnormal cardiogenesis.

13.
J Matern Fetal Neonatal Med ; 27(14): 1431-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24392847

RESUMEN

OBJECTIVE: To determine the prenatal variables predicting the risk of perinatal death in congenital right heart defects. METHODS: Retrospective analysis of 28 fetuses with right heart defects was performed. Logistic regression analyses were performed to obtain odds ratios (OR) for the relationship between the risk of death and echocardiographic parameters. The parameters that correlated with the outcome were incorporated in an attempt to devise a disease-specific cardiovascular profile score. RESULTS: Fetal echocardiograms (143) from 28 patients were analyzed. The cardiovascular profile score predicted the risk of death. A lower right ventricle (RV) pressure was associated with mortality (OR 0.959; 95% confidence intervals (CI) 0.940-0.978). Higher peak aortic velocity through the aortic valve (OR 0.104; 95% CI 0.020-0.529) was associated with a better outcome. These cardiac function parameters were incorporated in a modified disease-specific CVP Score. Patients with a mean modified cardiovascular profile score of ≤ 6 were over 3.7 times more likely to die than those with scores of 7-10. CONCLUSIONS: The original Cardiovascular Profile Score predicted the risk of death in right heart defects. The modified score was not validated as a good prediction tool by this study. Fetal RV pressure estimate and peak aortic velocity can be used as independent prognostic predictors.


Asunto(s)
Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/etiología , Resultado del Embarazo , Peso al Nacer , Ecocardiografía/métodos , Femenino , Cardiopatías Congénitas/epidemiología , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/patología , Humanos , Recién Nacido , Embarazo , Resultado del Embarazo/epidemiología , Índice de Embarazo , Pronóstico , Proyectos de Investigación , Estudios Retrospectivos , Factores de Riesgo , Ultrasonografía Prenatal
14.
J Pregnancy ; 2013: 320413, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23691322

RESUMEN

Extrapolating from animal studies to human pregnancy, our studies showed that folate (FA) deficiency as well as one-time exposure to environmental factors in the first two to three weeks of human gestation can result in severe congenital heart defects (CHDs). Considering that approximately 49% of pregnancies are unplanned, this period of pregnancy can be considered high-risk for cardiac, as well as for neural, birth defects, as the woman usually is not aware of her pregnancy and may not yet be taking precautionary actions to protect the developing embryo. Using avian and mouse vertebrate models, we demonstrated that FA supplementation prevents CHD induced by alcohol, lithium, or elevation of the metabolite homocysteine, a marker for FA deficiency. All three factors affected the important Wnt signaling pathway by suppressing Wnt-mediated gene expression in the heart fields, resulting in a delay of cardiomyocyte migration, cardiomyogenesis, and CHD. Optimal protection of cardiogenesis was observed to occur with FA supplementation provided upon morning after conception and at higher doses than the presently available in prenatal vitamin supplementation. Our studies demonstrate pathways and cell processes that are involved with protection of one-carbon metabolism during heart development.


Asunto(s)
Ácido Fólico/farmacología , Cardiopatías Congénitas/prevención & control , Corazón/embriología , Placenta/embriología , Animales , Aves , Diferenciación Celular/fisiología , Suplementos Dietéticos , Modelos Animales de Enfermedad , Etanol/toxicidad , Femenino , Ácido Fólico/administración & dosificación , Deficiencia de Ácido Fólico/dietoterapia , Deficiencia de Ácido Fólico/embriología , Deficiencia de Ácido Fólico/prevención & control , Edad Gestacional , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/embriología , Homocisteína/toxicidad , Humanos , Compuestos de Litio/toxicidad , Exposición Materna , Ratones , Miocitos Cardíacos/citología , Embarazo , Efectos Tardíos de la Exposición Prenatal/prevención & control , Transducción de Señal , Proteínas Wnt/fisiología
15.
Semin Fetal Neonatal Med ; 18(5): 245-50, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23751925

RESUMEN

Although the mammalian embryo is well protected in the uterus, environmental chemicals, drugs, and maternal nutritional imbalances can interfere with regulatory pathways directing placental and embryonic development early in gestation. Embryonic cells are most susceptible to environmental influences during cellular specification and differentiation stages. Because biochemical differentiation precedes morphological outcome often by days, the period of susceptibility to environmental chemicals expectedly precedes visible morphogenic effects. The cellular mechanisms by which drugs and other environmental factors disrupt embryonic development and induce cardiac abnormalities have remained undefined.


Asunto(s)
Enfermedades Ambientales/etiología , Desarrollo Fetal , Corazón Fetal/fisiopatología , Cardiopatías/etiología , Placenta/fisiopatología , Animales , Suplementos Dietéticos , Enfermedades Ambientales/congénito , Enfermedades Ambientales/fisiopatología , Enfermedades Ambientales/prevención & control , Contaminantes Ambientales/toxicidad , Femenino , Desarrollo Fetal/efectos de los fármacos , Corazón Fetal/efectos de los fármacos , Ácido Fólico/uso terapéutico , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/etiología , Cardiopatías Congénitas/fisiopatología , Cardiopatías Congénitas/prevención & control , Cardiopatías/embriología , Cardiopatías/fisiopatología , Cardiopatías/prevención & control , Humanos , Recién Nacido , Masculino , Exposición Materna/efectos adversos , Placenta/efectos de los fármacos , Embarazo , Fenómenos Fisiologicos de la Nutrición Prenatal
17.
Stem Cells Dev ; 19(12): 1959-65, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20624035

RESUMEN

We suggest that characterization of processes involved in differentiation of the pluripotential cardiac precursor cells in their embryonic environment will permit identifying pathways important for induction of diverse stem cells toward the cardiac phenotype. Phenotypic characteristics of cardiac cells are their contractile and electrical properties. The objective of the present study was to define whether calcium (Ca(++)) has a regulatory role in the pluripotential precursor cell population during commitment into cardiomyocytes. We used the chick embryo model because of ease of staging the embryos and visibility of heart development. Using the Ca(++) indicator Fluo-3/acetoxymethyl and confocal microscopy, we demonstrated the existence of higher free Ca(++) levels in the cardiogenic precursor cells than in neighboring cell populations outside of the heart fields. Subsequently, gastrulation stage 4/5 chick embryos were set up in modified New cultures in the medium containing either the L-type Ca channel blocker, diltiazem, or the N-type Ca channel inhibitor, ω-conotoxin. The embryos were incubated for 22-24 h during which time the control embryos developed, beating looping hearts. At the end of incubation, exposure to the L-type channel blockade with diltiazem resulted in an inhibition of cardiomyogenesis in the most posterior, uncommitted, part of the heart fields. N-type channel blockade with ω-conotoxin was less intense. Cells in the most anterior cardiogenic regions that were already committed at time of exposure continued to differentiate. Thus, regulation and maintenance of normal cytosolic Ca levels are necessary for the early steps of cardiomyocyte specification and commitment leading to differentiation.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Corazón/embriología , Miocitos Cardíacos/citología , Células Madre/citología , Células Madre/metabolismo , Compuestos de Anilina , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo N/metabolismo , Embrión de Pollo , Diltiazem/farmacología , Corazón/crecimiento & desarrollo , Microscopía Confocal , Contracción Miocárdica , Miocitos Cardíacos/fisiología , Xantenos , omega-Conotoxinas/farmacología
18.
Dis Model Mech ; 2(9-10): 467-78, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19638421

RESUMEN

Elevated plasma homocysteine (HCy), which results from folate (folic acid, FA) deficiency, and the mood-stabilizing drug lithium (Li) are both linked to the induction of human congenital heart and neural tube defects. We demonstrated previously that acute administration of Li to pregnant mice on embryonic day (E)6.75 induced cardiac valve defects by potentiating Wnt-beta-catenin signaling. We hypothesized that HCy may similarly induce cardiac defects during gastrulation by targeting the Wnt-beta-catenin pathway. Because dietary FA supplementation protects from neural tube defects, we sought to determine whether FA also protects the embryonic heart from Li- or HCy-induced birth defects and whether the protection occurs by impacting Wnt signaling. Maternal elevation of HCy or Li on E6.75 induced defective heart and placental function on E15.5, as identified non-invasively using echocardiography. This functional analysis of HCy-exposed mouse hearts revealed defects in tricuspid and semilunar valves, together with altered myocardial thickness. A smaller embryo and placental size was observed in the treated groups. FA supplementation ameliorates the observed developmental errors in the Li- or HCy-exposed mouse embryos and normalized heart function. Molecular analysis of gene expression within the avian cardiogenic crescent determined that Li, HCy or Wnt3A suppress Wnt-modulated Hex (also known as Hhex) and Islet-1 (also known as Isl1) expression, and that FA protects from the gene misexpression that is induced by all three factors. Furthermore, myoinositol with FA synergistically enhances the protective effect. Although the specific molecular epigenetic control mechanisms remain to be defined, it appears that Li or HCy induction and FA protection of cardiac defects involve intimate control of the canonical Wnt pathway at a crucial time preceding, and during, early heart organogenesis.


Asunto(s)
Ácido Fólico/farmacología , Cardiopatías Congénitas/prevención & control , Proteínas Wnt/metabolismo , Animales , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Pollos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/diagnóstico por imagen , Embrión de Mamíferos/efectos de los fármacos , Embrión no Mamífero/anomalías , Embrión no Mamífero/efectos de los fármacos , Gastrulación/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/diagnóstico por imagen , Cardiopatías Congénitas/fisiopatología , Pruebas de Función Cardíaca/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homocisteína , Inositol/farmacología , Proteínas con Homeodominio LIM , Litio , Ratones , Miocardio/metabolismo , Miocardio/patología , Factores de Transcripción , Ultrasonografía , Proteína Wnt3 , Proteína Wnt3A
19.
Dev Dyn ; 237(12): 3577-90, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18697221

RESUMEN

Flectin, a protein previously described to be expressed in a left-dominant manner in the embryonic chick heart during looping, is a member of the nonmuscle myosin II (NMHC-II) protein class. During looping, both NMHC-IIA and NMHC-IIB are expressed in the mouse heart on embryonic day 9.5. The patterns of localization of NMHC-IIB, rather than NMHC-IIA in the mouse looping heart and in neural crest cells, are equivalent to what we reported previously for flectin. Expression of full-length human NMHC-IIA and -IIB in 10 T1/2 cells demonstrated that flectin antibody recognizes both isoforms. Electron microscopy revealed that flectin antibody localizes in short cardiomyocyte cell processes extending from the basal layer of the cardiomyocytes into the cardiac jelly. Flectin antibody also recognizes stress fibrils in the cardiac jelly in the mouse and chick heart; while NMHC-IIB antibody does not. Abnormally looping hearts of the Nodal(Delta 600) homozygous mouse embryos show decreased NMHC-IIB expression on both the mRNA and protein levels. These results document the characterization of flectin and extend the importance of NMHC-II and the cytoskeletal actomyosin complex to the mammalian heart and cardiac looping.


Asunto(s)
Corazón/embriología , Miocardio/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Miosina Tipo IIB no Muscular/metabolismo , Animales , Línea Celular , Embrión de Pollo , Proteínas de la Matriz Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros/genética , Humanos , Ratones , Ratones Noqueados , Proteína Nodal/genética , Proteína Nodal/metabolismo , Miosina Tipo IIA no Muscular/genética , Miosina Tipo IIB no Muscular/deficiencia , Miosina Tipo IIB no Muscular/genética , Unión Proteica , Proteómica , ARN Mensajero/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
Dev Dyn ; 235(1): 124-31, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16110503

RESUMEN

We have identified the presence of the hypoxia marker EF5 in the stage 4/5 chick heart fields. This suggests that cardiac cell differentiation occurs in a relatively anaerobic environment. Monocarboxylate transporter (MCT) studies in adult cardiac myocytes have demonstrated that MCTs catalyze proton-linked pyruvate and lactate transport activity. 5A11/Basigin is an ancillary protein that targets MCTs to the plasma membrane for their function. MCT-4 expression is most evident in cells with a high glycolytic rate associated with hypoxic energy production. Subsequent to the immunohistochemical localization of EF5 in the early heart field, we continued in our analysis during stages 5 to 12 for the expression of indicators of cellular glycolytic metabolism in the developing heart, such as MCT-4, MCT-1, and 5A11 (Basigin/CD147). Our observations indicate that MCT-4 and 5A11/Basigin are expressed early, in a differential left-right pattern, in the bi-lateral plate mesoderm, as the cardiac compartment is forming. At stage 11, MCT-4/5A11 continues to be highly expressed in the myocardial wall of the looping heart, but not in the dorsal mesocardium. RT-PCR analyses for MCT-1, -4, and 5A11 indicate that MCT-4 and 5A11 are expressed throughout precardiac, embryonic, and fetal stages in the heart. MCT-1 is first detected in the heart on embryonic day 3 and then remains expressed throughout development to hatching. These results indicate that cardiac precursor cells are equipped for differentiating in a hypoxic environment using anaerobic metabolism for energy production.


Asunto(s)
Proteínas Aviares/biosíntesis , Basigina/biosíntesis , Diferenciación Celular/fisiología , Etanidazol/análogos & derivados , Hidrocarburos Fluorados/metabolismo , Hipoxia/metabolismo , Transportadores de Ácidos Monocarboxílicos/biosíntesis , Miocitos Cardíacos/citología , Animales , Proteínas Aviares/genética , Basigina/genética , Biomarcadores , Embrión de Pollo , Etanidazol/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Miocitos Cardíacos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA