Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cardiovasc Dev Dis ; 8(8)2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34436232

RESUMEN

In congenital heart disease, the presence of structural defects affects blood flow in the heart and circulation. However, because the fetal circulation bypasses the lungs, fetuses with cyanotic heart defects can survive in utero but need prompt intervention to survive after birth. Tetralogy of Fallot and persistent truncus arteriosus are two of the most significant conotruncal heart defects. In both defects, blood access to the lungs is restricted or non-existent, and babies with these critical conditions need intervention right after birth. While there are known genetic mutations that lead to these critical heart defects, early perturbations in blood flow can independently lead to critical heart defects. In this paper, we start by comparing the fetal circulation with the neonatal and adult circulation, and reviewing how altered fetal blood flow can be used as a diagnostic tool to plan interventions. We then look at known factors that lead to tetralogy of Fallot and persistent truncus arteriosus: namely early perturbations in blood flow and mutations within VEGF-related pathways. The interplay between physical and genetic factors means that any one alteration can cause significant disruptions during development and underscore our need to better understand the effects of both blood flow and flow-responsive genes.

3.
J Vis Exp ; (91): 51911, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25285454

RESUMEN

Western blot analysis is a commonly employed technique for detecting and quantifying protein levels. However, for small tissue samples, this analysis method may not be sufficiently sensitive to detect a protein of interest. To overcome these difficulties, we examined protocols for obtaining protein from adult human cardiac valves and modified these protocols for the developing early embryonic mouse counterparts. In brief, the mouse embryonic aortic valve regions, including the aortic valve and surrounding aortic wall, are collected in the minimal possible volume of a Tris-based lysis buffer with protease inhibitors. If required based on the breeding strategy, embryos are genotyped prior to pooling four embryonic aortic valve regions for homogenization. After homogenization, an SDS-based sample buffer is used to denature the sample for running on an SDS-PAGE gel and subsequent western blot analysis. Although the protein concentration remains too low to quantify using spectrophotometric protein quantification assays and have sample remaining for subsequent analyses, this technique can be used to successfully detect and semi-quantify phosphorylated proteins via western blot from pooled samples of four embryonic day 13.5 mouse aortic valve regions, each of which yields approximately 1 µg of protein. This technique will be of benefit for studying cell signaling pathway activation and protein expression levels during early embryonic mouse valve development.


Asunto(s)
Válvulas Cardíacas/química , Proteínas Musculares/análisis , Animales , Western Blotting , Electroforesis en Gel de Poliacrilamida , Femenino , Válvulas Cardíacas/embriología , Válvulas Cardíacas/metabolismo , Ratones , Proteínas Musculares/metabolismo , Embarazo
4.
Trends Endocrinol Metab ; 25(9): 472-80, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24908616

RESUMEN

The bone morphogenetic protein (BMP) family of proteins has a multitude of roles throughout the body. In embryonic development, BMPs promote endothelial specification and subsequent venous differentiation. The BMP pathway also plays important roles in the adult vascular endothelium, promoting angiogenesis and mediating shear and oxidative stress. The canonical BMP pathway functions through the Smad transcription factors; however, other intracellular signaling cascades can be activated, and receptor complexes beyond the traditional type I and type II receptors add additional layers of regulation. Dysregulated BMP signaling has been linked to vascular diseases including pulmonary hypertension and atherosclerosis. This review addresses recent advances in the roles of BMP signaling in the endothelium and how BMPs affect endothelial dysfunction and human disease.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Endotelio Vascular/metabolismo , Modelos Biológicos , Transducción de Señal , Enfermedades Vasculares/metabolismo , Animales , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/agonistas , Receptores de Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/genética , Endotelio Vascular/citología , Humanos , Hipertensión/metabolismo , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/metabolismo , Ratones Transgénicos , Neovascularización Patológica/etiología , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica , Estrés Oxidativo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Resistencia al Corte , Estrés Fisiológico , Enfermedades Vasculares/etiología
5.
J Vis Exp ; (77)2013 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-23912902

RESUMEN

Cell culture has greatly enhanced our ability to assess individual populations of cells under myriad culture conditions. While immortalized cell lines offer significant advantages for their ease of use, these cell lines are unavailable for all potential cell types. By isolating primary cells from a specific region of interest, particularly from a transgenic mouse, more nuanced studies can be performed. The basic technique involves dissecting the organ or partial organ of interest (e.g. the heart or a specific region of the heart) and dissociating the organ to single cells. These cells are then incubated with magnetic beads conjugated to an antibody that recognizes the cell type of interest. The cells of interest can then be isolated with the use of a magnet, with a short trypsin incubation dissociating the cells from the beads. These isolated cells can then be cultured and analyzed as desired. This technique was originally designed for adult mouse organs but can be easily scaled down for use with embryonic organs, as demonstrated herein. Because our interest is in the developing coronary vasculature, we wanted to study this population of cells during specific embryonic stages. Thus, the original protocol had to be modified to be compatible with the small size of the embryonic ventricles and the low potential yield of endothelial cells at these developmental stages. Utilizing this scaled-down approach, we have assessed coronary plexus remodeling in transgenic embryonic ventricular endothelial cells.


Asunto(s)
Células Endoteliales/citología , Corazón/embriología , Separación Inmunomagnética/métodos , Miocardio/citología , Animales , Femenino , Ventrículos Cardíacos/embriología , Ratones , Ratones Transgénicos , Embarazo
6.
J Vis Exp ; (75): e50359, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23728379

RESUMEN

Developmental studies in the mouse are hampered by the inaccessibility of the embryo during gestation. Thus, protocols to isolate and culture individual organs of interest are essential to provide a method of both visualizing changes in development and allowing novel treatment strategies. To promote the long-term culture of the embryonic heart at late stages of gestation, we developed a protocol in which the excised heart is cultured in a semi-solid, dilute Matrigel. This substrate provides enough support to maintain the three-dimensional structure but is flexible enough to allow continued contraction. In brief, hearts are excised from the embryo and placed in a mixture of cold Matrigel diluted 1:1 with growth medium. After the diluted Matrigel solidifies, growth medium is added to the culture dish. Hearts excised as late as embryonic day 16.5 were viable for four days post-dissection. Analysis of the coronary plexus shows that this method does not disrupt coronary vascular development. Thus, we present a novel method for long-term culture of embryonic hearts.


Asunto(s)
Técnicas de Cultivo de Embriones/métodos , Corazón/embriología , Ratones/embriología , Animales , Colágeno , Combinación de Medicamentos , Embrión de Mamíferos , Femenino , Laminina , Embarazo , Proteoglicanos
7.
Molecules ; 18(5): 5594-610, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23676470

RESUMEN

Angiogenesis plays a key role in cancer progression and correlates with disease aggressiveness and poor clinical outcomes. Affinity ligands discovered by screening phage display random peptide libraries can be engineered to molecularly target tumor blood vessels for noninvasive imaging and early detection of tumor aggressiveness. In this study, we tested the ability of a phage-display-selected peptide sequence recognizing specifically bone marrow- derived pro-angiogenic tumor-homing cells, the QFP-peptide, radiolabeled with 64Cu radioisotope to selectively image tumor vasculature in vivo by positron emission tomography (PET). To prepare the targeted PET tracer we modified QFP-phage with the DOTA chelator and radiolabeled the purified QFP-phage-DOTA intermediate with 64Cu to obtain QFP-targeted radioconjugate with high radiopharmaceutical yield and specific activity. We evaluated the new PET tracer in vivo in a subcutaneous (s.c.) Lewis lung carcinoma (LLC) mouse model and conducted tissue distribution, small animal PET/CT imaging study, autoradiography, histology, fluorescence imaging, and dosimetry assessments. The results from this study show that, in the context of the s.c. LLC immunocompetent mouse model, the QFP-tracer can target tumor blood vessels selectively. However, further optimization of the biodistribution and dosimetry profile of the tracer is necessary to ensure efficient radiopharmaceutical applications enabled by the biological specificity of the QFP-peptide.


Asunto(s)
Carcinoma Pulmonar de Lewis , Neovascularización Patológica , Péptidos , Tomografía de Emisión de Positrones , Radiofármacos , Animales , Carcinoma Pulmonar de Lewis/diagnóstico por imagen , Carcinoma Pulmonar de Lewis/metabolismo , Cobre/química , Femenino , Isótopos/química , Ratones , Neovascularización Patológica/diagnóstico por imagen , Neovascularización Patológica/metabolismo , Péptidos/síntesis química , Péptidos/química , Péptidos/farmacología , Radiografía , Radiofármacos/síntesis química , Radiofármacos/química , Radiofármacos/farmacología
8.
Cardiovasc Pathol ; 22(3): 228-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23200275

RESUMEN

BACKGROUND: BMPER, an orthologue of Drosophila melanogaster Crossveinless-2, is a secreted factor that regulates bone morphogenetic protein activity in endothelial cell precursors and during early cardiomyocyte differentiation. Although previously described in the heart, the role of BMPER in cardiac development and function remain unknown. METHODS: BMPER-deficient hearts were phenotyped histologically and functionally using echocardiography and Doppler analysis. Since BMPER -/- mice die perinatally, adult BMPER +/- mice were challenged to pressure-overload-induced cardiac hypertrophy and hindlimb ischemia to determine changes in angiogenesis and regulation of cardiomyocyte size. RESULTS: We identify for the first time the cardiac phenotype associated with BMPER haploinsufficiency. BMPER messenger RNA and protein are present in the heart during cardiac development through at least E14.5 but is lost by E18.5. BMPER +/- ventricles are thinner and less compact than sibling wild-type hearts. In the adult, BMPER +/- hearts present with decreased anterior and posterior wall thickness, decreased cardiomyocyte size and an increase in cardiac vessel density. Despite these changes, BMPER +/- mice respond to pressure-overload-induced cardiac hypertrophy challenge largely to the same extent as wild-type mice. CONCLUSION: BMPER appears to play a role in regulating both vessel density and cardiac development in vivo; however, BMPER haploinsufficiency does not result in marked effects on cardiac function or adaptation to pressure overload hypertrophy.


Asunto(s)
Proteínas Portadoras/metabolismo , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/citología , Neovascularización Fisiológica/fisiología , Animales , Western Blotting , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Aumento de la Célula , Vasos Coronarios/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Arterioscler Thromb Vasc Biol ; 32(9): 2214-22, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22772758

RESUMEN

OBJECTIVE: Bone morphogenetic proteins (Bmps) are important mediators of inflammation and atherosclerosis, though their mechanism of action is not fully understood. To better understand the contribution of the Bmp signaling pathway in vascular inflammation, we investigated the role of Bmper (Bmp endothelial cell precursor-derived regulator), an extracellular Bmp modulator, in an induced in vivo model of inflammation and atherosclerosis. METHODS AND RESULTS: We crossed apolipoprotein E-deficient (ApoE(-/-)) mice with mice missing 1 allele of Bmper (Bmper(+/-) mice used in the place of Bmper(-/-) mice that die at birth) and measured the development of atherosclerosis in mice fed a high-fat diet. Bmper haploinsufficiency in ApoE(-/-) mice (Bmper(+/-);ApoE(-/-) mice) led to a more severe phenotype compared with Bmper(+/+);ApoE(-/-) mice. Bmper(+/-);ApoE(-/-) mice also exhibited increased Bmp activity in the endothelial cells in both the greater and lesser curvatures of the aortic arch, suggesting a role for Bmper in regulating Bmp-mediated inflammation associated with laminar and oscillatory shear stress. Small interfering RNA knockdown of Bmper in human umbilical vein endothelial cells caused a dramatic increase in the inflammatory markers intracellular adhesion molecule 1 and vascular cell adhesion molecule 1 at rest and after exposure to oscillatory and laminar shear stress. CONCLUSIONS: We conclude that Bmper is a critical regulator of Bmp-mediated vascular inflammation and that the fine-tuning of Bmp and Bmper levels is essential in the maintenance of normal vascular homeostasis.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Proteínas Portadoras/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/prevención & control , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Portadoras/genética , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/patología , Genotipo , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Estrés Mecánico , Factores de Tiempo , Transfección , Calcificación Vascular/inmunología , Calcificación Vascular/metabolismo , Calcificación Vascular/prevención & control , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
Circ Res ; 111(5): 564-74, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22777006

RESUMEN

RATIONALE: Among the extracellular modulators of Bmp (bone morphogenetic protein) signaling, Bmper (Bmp endothelial cell precursor-derived regulator) both enhances and inhibits Bmp signaling. Recently we found that Bmper modulates Bmp4 activity via a concentration-dependent, endocytic trap-and-sink mechanism. OBJECTIVE: To investigate the molecular mechanisms required for endocytosis of the Bmper/Bmp4 and signaling complex and determine the mechanism of Bmper's differential effects on Bmp4 signaling. METHODS AND RESULTS: Using an array of biochemical and cell biology techniques, we report that LRP1 (LDL receptor-related protein 1), a member of the LDL receptor family, acts as an endocytic receptor for Bmper and a coreceptor of Bmp4 to mediate the endocytosis of the Bmper/Bmp4 signaling complex. Furthermore, we demonstrate that LRP1-dependent Bmper/Bmp4 endocytosis is essential for Bmp4 signaling, as evidenced by the phenotype of lrp1-deficient zebrafish, which have abnormal cardiovascular development and decreased Smad1/5/8 activity in key vasculogenic structures. CONCLUSIONS: Together, these data reveal a novel role for LRP1 in the regulation of Bmp4 signaling by regulating receptor complex endocytosis. In addition, these data introduce LRP1 as a critical regulator of vascular development. These observations demonstrate Bmper's ability to fine-tune Bmp4 signaling at the single-cell level, unlike the spatial regulatory mechanisms applied by other Bmp modulators.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Proteínas Portadoras/metabolismo , Endocitosis/fisiología , Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Portadoras/genética , Línea Celular , Movimiento Celular/fisiología , Células Endoteliales/citología , Células HEK293 , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Fenotipo , ARN Interferente Pequeño/genética , Receptores de LDL/genética , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/genética , Pez Cebra , Proteínas de Pez Cebra/genética
11.
Dev Biol ; 348(2): 167-76, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20920499

RESUMEN

Sonic hedgehog signaling in the secondary heart field has a clear role in cardiac arterial pole development. In the absence of hedgehog signaling, proliferation is reduced in secondary heart field progenitors, and embryos predominantly develop pulmonary atresia. While it is expected that proliferation in the secondary heart field would be increased with elevated hedgehog signaling, this idea has never been tested. We hypothesized that up-regulating hedgehog signaling would increase secondary heart field proliferation, which would lead to arterial pole defects. In culture, secondary heart field explants proliferated up to 6-fold more in response to the hedgehog signaling agonist SAG, while myocardial differentiation and migration were unaffected. Treatment of chick embryos with SAG at HH14, just before the peak in secondary heart field proliferation, resulted unexpectedly in stenosis of both the aortic and pulmonary outlets. We examined proliferation in the secondary heart field and found that SAG-treated embryos exhibited a much milder increase in proliferation than was indicated by the in vitro experiments. To determine the source of other signaling factors that could modulate increased hedgehog signaling, we co-cultured secondary heart field explants with isolated pharyngeal endoderm or outflow tract and found that outflow tract co-cultures prevented SAG-induced proliferation. BMP2 is made and secreted by the outflow tract myocardium. To determine whether BMP signaling could prevent SAG-induced proliferation, we treated explants with SAG and BMP2 and found that BMP2 inhibited SAG-induced proliferation. In vivo, SAG-treated embryos showed up-regulated BMP2 expression and signaling. Together, these results indicate that BMP signaling from the outflow tract modulates hedgehog-induced proliferation in the secondary heart field.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Corazón/embriología , Proteínas Hedgehog/metabolismo , Transducción de Señal , Animales , Proteínas Morfogenéticas Óseas/genética , Proliferación Celular , Embrión de Pollo , Ciclohexilaminas/farmacología , Proteínas Hedgehog/genética , Miocardio/metabolismo , Organogénesis , Tiofenos/farmacología , Regulación hacia Arriba
12.
Sci Signal ; 3(122): pe17, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20484115

RESUMEN

A healthy vasculature is an essential component of development and is regulated by different signaling pathways. One of the most critical pathways involved is the vascular endothelial growth factor (VEGF) pathway. Components of this pathway serve as the first marker of primitive endothelial cells and are instrumental in inducing the initial differentiation of endothelial cells and later refining them into either arteries or veins. However, the regulation of VEGF signaling remains a mystery, with most studies focusing on the downstream components of this signaling cascade. New evidence shows that the protein cerebral cavernous malformation 3 (CCM3) is a key regulator of the VEGF pathway, bringing to light a previously unknown component of the VEGF signaling axis and opening the door to an exciting new era of vasculogenic research.


Asunto(s)
Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Ratones , Ratones Noqueados , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Semin Thromb Hemost ; 36(3): 227-35, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20490975

RESUMEN

The endothelium is composed of specialized epithelial cells that line the vasculature, the lymph vessels, and the heart. These endothelial cells are characterized by their stratification and are connected via intercellular junctions that confer specific permeability. Although all endothelium acts as a barrier, considerable heterogeneity exists among different organs and even within vessels. During development, the endothelial cells are specified before they migrate to their final destination, and then they commit to an arterial or venous fate. From the venous endothelial cell population, a subset of cells is further specified as lymphatic endothelium. The endothelium can be highly permeable, as in the lymph vessels, or impenetrable, as in the blood-brain barrier. These differences arise during development and are orchestrated through a series of signaling pathways. This review details how endothelial cells arise and are directed to their specific fate, specifically targeting what differentiates endothelial populations.


Asunto(s)
Endotelio/crecimiento & desarrollo , Diferenciación Celular , Endotelio/citología , Endotelio/embriología , Endotelio/fisiología , Endotelio/fisiopatología , Permeabilidad , Transducción de Señal
14.
Dev Biol ; 336(2): 137-44, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19835857

RESUMEN

Although de la Cruz and colleagues showed as early as 1977 that the outflow tract was added after the heart tube formed, the source of these secondarily added cells was not identified for nearly 25 years. In 2001, three pivotal publications described a secondary or anterior heart field that contributed to the developing outflow tract. This review details the history of the heart field, the discovery and continuing elucidation of the secondarily adding myocardial cells, and how the different populations identified in 2001 are related to the more recent lineage tracing studies that defined the first and second myocardial heart fields/lineages. Much recent work has focused on secondary heart field progenitors that give rise to the myocardium and smooth muscle at the definitive arterial pole. These progenitors are the last to be added to the arterial pole and are particularly susceptible to abnormal development, leading to conotruncal malformations in children. The major signaling pathways (Wnt, BMP, FGF8, Notch, and Shh) that control various aspects of secondary heart field progenitor behavior are discussed.


Asunto(s)
Corazón/embriología , Animales , Humanos , Transducción de Señal
15.
Dev Biol ; 330(2): 305-17, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19361493

RESUMEN

The Sonic hedgehog (Shh)-null mouse was initially described as a phenotypic mimic of Tetralogy of Fallot with pulmonary atresia (Washington Smoak, I., Byrd, N.A., Abu-Issa, R., Goddeeris, M.M., Anderson, R., Morris, J., Yamamura, K., Klingensmith, J., and Meyers, E.N. 2005. Sonic hedgehog is required for cardiac outflow tract and neural crest cell development. Dev. Biol. 283, 357-372.); however, subsequent reports describe only a single outflow tract, leaving the phenotype and its developmental mechanism unclear. We hypothesized that the phenotype that occurs in response to Shh knockdown is pulmonary atresia and is directly related to the abnormal development of the secondary heart field. We found that Shh was expressed by the pharyngeal endoderm adjacent to the secondary heart field and that its receptor Ptc2 was expressed in a gradient in the secondary heart field, with the most robust expression in the caudal secondary heart field, closest to the Shh expression. In vitro culture of secondary heart field with the hedgehog inhibitor cyclopamine significantly reduced proliferation. In ovo, cyclopamine treatment before the secondary heart field adds to the outflow tract reduced proliferation only in the caudal secondary heart field, which coincided with the region of high Ptc2 expression. After outflow tract septation should occur, embryos treated with cyclopamine exhibited pulmonary atresia, pulmonary stenosis, and persistent truncus arteriosus. In hearts with pulmonary atresia, cardiac neural crest-derived cells, which form the outflow tract septum, migrated into the outflow tract and formed a septum. However, this septum divided the outflow tract into two unequal sized vessels and effectively closed off the pulmonary outlet. These experiments show that Shh is necessary for secondary heart field proliferation, which is required for normal pulmonary trunk formation, and that embryos with pulmonary atresia have an outflow tract septum.


Asunto(s)
Proliferación Celular , Corazón/embriología , Proteínas Hedgehog/fisiología , Miocardio/citología , Animales , Secuencia de Bases , Cartilla de ADN , Corazón/efectos de los fármacos , Proteínas Hedgehog/genética , Hibridación in Situ , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alcaloides de Veratrum/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...