Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Open Forum Infect Dis ; 7(6): ofaa180, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32550237

RESUMEN

In this work, we report 2 cases of vancomycin-resistant Enterococcus faecium bacteremia with development of daptomycin resistance in 2 patients with acute myeloid leukemia and myelodysplastic syndrome. Mutations related to daptomycin-nonsusceptible phenotype in liaSR genes were found in all strains of the study, including those with a minimum inhibitory concentration <1 µg/mL collected before daptomycin therapy. Epidemiological investigation using core genome single nucleotide polymorphism and core genome multilocus sequence typing revealed clonality of all the isolates. In this study, we conclude that real-time genome sequencing of clinical isolates can provide rapid access to timely information on daptomycin-resistant genotypes that would help clinicians speed up and optimize the selection of the antibiotic for treatment.

2.
Gen Dent ; 66(3): 68-74, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29714704

RESUMEN

The purpose of this study was to compare the depths of cure of a proximal box preparation filled in bulk with various approaches: filled with a bulk-fill or conventional composite; placed with a new perforated metal matrix, a traditional metal matrix, or a clear matrix; and polymerized with either occlusal-only or tri-sited light curing. After tri-sited curing, the use of the new perforated metal matrix band resulted in a depth of cure that was not significantly different from that achieved with the use of metal bands (removed during curing) or transparent matrix bands. Adequate polymerization was obtained at depths of more than 5.0 mm for the bulk-fill composite and more than 4.0 mm for the conventional composite when tri-sited light curing was used. Tri-sited light curing resulted in a significantly greater depth of cure than occlusal-only curing. The perforated metal band may be used as an alternative to the use of solid metal bands or transparent matrix bands to provide similar depths of cure for composite resins, with the possible benefits of malleability and the ability to leave the band in place during tri-sited light curing.


Asunto(s)
Resinas Compuestas/uso terapéutico , Restauración Dental Permanente/métodos , Preparación de la Cavidad Dental/métodos , Restauración Dental Permanente/instrumentación , Humanos , Técnicas In Vitro , Bandas de Matriz
3.
Gen Dent ; 65(2): e12-e16, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28253189

RESUMEN

A new fluoride-releasing sealant system is claimed to allow easier and faster placement while providing high bond strengths without the need for phosphoric acid etching. A study was designed to compare the microleakage and shear bond strength of a self-etching, Giomer-based sealant system with those of a traditional resin sealant. Group 1 received traditional sealant applied after use of a 35% phosphoric acid etchant; group 2 received Giomer sealant after use of a self-etching primer; and group 3 received Giomer sealant after the addition of an initial phosphoric acid etching step and placement of the primer. The sealants were placed in the occlusal pits and fissures of extracted human third molars, thermocycled, placed in dye, and sectioned. The extent of microleakage (dye penetration) was expressed as a percentage of the cross-sectional length of the sealed interface. The sealants were also bonded to the facial enamel of bovine incisors. Specimens were thermocycled and tested in shear mode in a universal testing machine. The new self-etching sealant demonstrated significantly greater microleakage (P < 0.017) and lower bond strength (P < 0.05) than both the traditional sealant system and the new system when placed with phosphoric acid etchant. Phosphoric acid etching significantly improved the shear bond strength and reduced the microleakage of the new sealant.


Asunto(s)
Resinas Acrílicas/uso terapéutico , Recubrimiento Dental Adhesivo/métodos , Filtración Dental/etiología , Selladores de Fosas y Fisuras/uso terapéutico , Dióxido de Silicio/uso terapéutico , Grabado Ácido Dental/métodos , Resinas Acrílicas/efectos adversos , Análisis del Estrés Dental , Humanos , Selladores de Fosas y Fisuras/efectos adversos , Resistencia al Corte , Dióxido de Silicio/efectos adversos
4.
Dent Mater ; 32(11): 1343-1351, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27613394

RESUMEN

OBJECTIVES: The purpose of this study was to evaluate the effect of various surface treatments on the mechanical properties and antibacterial activity of desiccated glass-ionomer (GI) and resin-modified glass-ionomer (RMGI) materials. METHODS: One hundred GI and RMGI specimens were fabricated in a mold, stored in 100% humidity for 24h, placed in air to desiccate for 24h, and then stored for one week in one of the five media [casein phosphopeptide-amorphous calcium phosphate (CPP-ACP), chlorhexidine (CHX), sodium fluoride (NaF), cetylpyridinium chloride (CPC), or 100% humidity (control)]. Fifty GI and RMGI specimens were tested in flexure to determine flexural strength and modulus, with the fragments used for Knoop hardness testing. The remaining 50 GI and RMGI specimens were covered with a suspension of Streptococcus mutans and incubated for 24h. The bacterial suspension was removed and the specimens were washed. Sterile saline was added, vortex mixed, serially diluted, and plated. CFU/mLs were calculated after 3days of incubation. RESULTS: Compared to the 100% humidity control group, surface treatment of the desiccated GI and RMGI materials had a variable effect on the mechanical properties. In general, NaF provided the greatest improvement in flexural strength and modulus. Surface treatment of the desiccated GI or RMGI specimens with CHX or CPC resulted in no growth of the S. mutans. NaF resulted in significantly lower CFU/mL than CPP-ACP, which was significantly lower than the control group. SIGNIFICANCE: Surface treatment with 5% NaF provides improved antimicrobial and strength properties of desiccated GI or RMGI materials.


Asunto(s)
Antiinfecciosos , Cementos de Ionómero Vítreo , Resinas Acrílicas , Ensayo de Materiales , Dióxido de Silicio , Propiedades de Superficie
5.
Gen Dent ; 64(3): 51-4, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27148657

RESUMEN

The study evaluated the effect of salivary-contaminant removal methods on the bond strength of resin cement to hydrofluoric acid-etched ceramic. Treatment with a new cleaning paste resulted in bond strengths not significantly different from those obtained in phosphoric acid-treated, hydrofluoric acid-treated, and uncontaminated control groups; thus the paste may be considered an alternative to phosphoric acid or hydrofluoric acid for removal of salivary contaminants.


Asunto(s)
Grabado Ácido Dental/métodos , Recubrimiento Dental Adhesivo/métodos , Cementos de Resina/uso terapéutico , Cerámica/uso terapéutico , Porcelana Dental/uso terapéutico , Análisis del Estrés Dental , Humanos , Saliva/metabolismo
6.
Development ; 135(21): 3599-610, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18832392

RESUMEN

In order to understand how secreted signals regulate complex morphogenetic events, it is crucial to identify their cellular targets. By conditional inactivation of Fgfr1 and Fgfr2 and overexpression of the FGF antagonist sprouty 2 in different cell types, we have dissected the role of FGF signaling during heart outflow tract development in mouse. Contrary to expectation, cardiac neural crest and endothelial cells are not primary paracrine targets. FGF signaling within second heart field mesoderm is required for remodeling of the outflow tract: when disrupted, outflow myocardium fails to produce extracellular matrix and TGFbeta and BMP signals essential for endothelial cell transformation and invasion of cardiac neural crest. We conclude that an autocrine regulatory loop, initiated by the reception of FGF signals by the mesoderm, regulates correct morphogenesis at the arterial pole of the heart. These findings provide new insight into how FGF signaling regulates context-dependent cellular responses during development.


Asunto(s)
Arterias/embriología , Comunicación Autocrina , Factores de Crecimiento de Fibroblastos/metabolismo , Corazón/embriología , Mesodermo/embriología , Mesodermo/metabolismo , Morfogénesis , Proteínas Adaptadoras Transductoras de Señales , Animales , Arterias/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Región Branquial/embriología , Región Branquial/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Epitelio/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Eliminación de Gen , Dosificación de Gen , Integrasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/metabolismo , Ratones , Miocardio/citología , Miocardio/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
7.
Development ; 135(10): 1887-95, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18441277

RESUMEN

Septation of the mammalian heart into four chambers requires the orchestration of multiple tissue progenitors. Abnormalities in this process can result in potentially fatal atrioventricular septation defects (AVSD). The contribution of extracardiac cells to atrial septation has recently been recognized. Here, we use a genetic marker and novel magnetic resonance microscopy techniques to demonstrate the origins of the dorsal mesenchymal protrusion in the dorsal mesocardium, and its substantial contribution to atrioventricular septation. We explore the functional significance of this tissue to atrioventricular septation through study of the previously uncharacterized AVSD phenotype of Shh(-/-) mutant mouse embryos. We demonstrate that Shh signaling is required within the dorsal mesocardium for its contribution to the atria. Failure of this addition results in severe AVSD. These studies demonstrate that AVSD can result from a primary defect in dorsal mesocardium, providing a new paradigm for the understanding of human AVSD.


Asunto(s)
Corazón Fetal/citología , Proteínas Hedgehog/metabolismo , Animales , Corazón Fetal/embriología , Atrios Cardíacos/citología , Atrios Cardíacos/embriología , Defectos del Tabique Interventricular/embriología , Proteínas Hedgehog/genética , Mesodermo/citología , Ratones , Ratones Mutantes , Mutación , Transducción de Señal
8.
Development ; 134(8): 1593-604, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17344228

RESUMEN

Cardiac outflow tract (OFT) septation is crucial to the formation of the aortic and pulmonary arteries. Defects in the formation of the OFT can result in serious congenital heart defects. Two cell populations, the anterior heart field (AHF) and cardiac neural crest cells (CNCCs), are crucial for OFT development and septation. In this study, we use a series of tissue-specific genetic manipulations to define the crucial role of the Hedgehog pathway in these two fields of cells during OFT development. These data indicate that endodermally-produced SHH ligand is crucial for several distinct processes, all of which are required for normal OFT septation. First, SHH is required for CNCCs to survive and populate the OFT cushions. Second, SHH mediates signaling to myocardial cells derived from the AHF to complete septation after cushion formation. Finally, endodermal SHH signaling is required in an autocrine manner for the survival of the pharyngeal endoderm, which probably produces a secondary signal required for AHF survival and for OFT lengthening. Disruption of any of these steps can result in a single OFT phenotype.


Asunto(s)
Arterias/embriología , Región Branquial/embriología , Corazón/embriología , Proteínas Hedgehog/fisiología , Miocardio/metabolismo , Cresta Neural/embriología , Animales , Región Branquial/irrigación sanguínea , Región Branquial/metabolismo , Endodermo/metabolismo , Proteínas Hedgehog/metabolismo , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Noqueados , Morfogénesis , Miocardio/citología , Miocitos Cardíacos/fisiología , Cresta Neural/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
9.
Circ Res ; 100(2): 220-8, 2007 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-17218603

RESUMEN

Bone morphogenetic proteins (BMPs) play many roles in mammalian cardiac development. Here we address the functions of Noggin, a dedicated BMP antagonist, in the developing mouse heart. In early cardiac tissues, the Noggin gene is mainly expressed in the myocardial cells of the outflow tract, atrioventricular canal, and future right ventricle. The major heart phenotypes of Noggin mutant embryos are thicker myocardium and larger endocardial cushions. Both defects result from increased cell number. Cell proliferation is increased and cell cycle exit is decreased in the myocardium. Although we find evidence of increased BMP signal transduction in the myocardium and endocardium, we show that the cardiac defects of Noggin mutants are rescued by halving the gene dosage of Bmp4. In culture, BMP increases the epithelial-to-mesenchymal transformation (EMT) of endocardial explant cells. Increased EMT likely accounts for the enlarged atrioventricular cushion. In the outflow tract cushion, we observed an increased contribution of cardiac neural crest cells to the mutant cushion mesenchyme, although many cells of the cushion were not derived from neural crest. Thus the enlarged outflow tract cushion of Noggin mutants likely arises by increased contributions both of endocardial cells that have undergone EMT as well as cells that have migrated from the neural crest. These data indicate that antagonism of BMP signaling by Noggin plays a critical role in ensuring proper levels of cell proliferation and EMT during cardiac morphogenesis in the mouse.


Asunto(s)
Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Portadoras/fisiología , Corazón/embriología , Morfogénesis/fisiología , Animales , Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Morfogenéticas Óseas/genética , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proliferación Celular , Endocardio/citología , Endocardio/embriología , Endocardio/patología , Corazón/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Morfogénesis/genética , Miocardio/citología , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Fenotipo , Transducción de Señal/genética
10.
Development ; 133(12): 2435-45, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16720880

RESUMEN

In the mouse embryo, the splanchnic mesodermal cells of the anterior heart field (AHF) migrate from the pharynx to contribute to the early myocardium of the outflow tract (OT) and right ventricle (RV). Recent studies have attempted to distinguish the AHF from other precardiac populations, and to determine the genetic and molecular mechanisms that regulate its development. Here, we have used an Fgf8lacZ allele to demonstrate that Fgf8 is expressed within the developing AHF. In addition, we use both a hypomorphic Fgf8 allele (Fgf8neo) and Cre-mediated gene ablation to show that Fgf8 is essential for the survival and proliferation of the AHF. Nkx2.5Cre is expressed in the AHF, primary heart tube and pharyngeal endoderm, while TnT-Cre is expressed only within the specified heart tube myocardium. Deletion of Fgf8 by Nkx2.5Cre results in a significant loss of the Nkx2.5Cre lineage and severe OT and RV truncations by E9.5, while the remaining heart chambers (left ventricle and atria) are grossly normal. These defects result from significant decreases in cell proliferation and aberrant cell death in both the pharyngeal endoderm and splanchnic mesoderm. By contrast, ablation of Fgf8 in the TnT-Cre domain does not result in OT or RV defects, providing strong evidence that Fgf8 expression is crucial in the pharyngeal endoderm and/or overlying splanchnic mesoderm of the AHF at a stage prior to heart tube elongation. Analysis of downstream signaling components, such as phosphorylated-Erk and Pea3, identifies the AHF splanchnic mesoderm itself as a target for Fgf8 signaling.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/metabolismo , Corazón/anatomía & histología , Corazón/embriología , Transducción de Señal/fisiología , Animales , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Región Branquial/anatomía & histología , Región Branquial/metabolismo , Anomalías Cardiovasculares , Anomalías Craneofaciales , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/patología , Embrión de Mamíferos/fisiología , Endodermo/citología , Endodermo/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 8 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Corazón/crecimiento & desarrollo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Cresta Neural/citología , Cresta Neural/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Development ; 132(17): 3847-57, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16049112

RESUMEN

During kidney morphogenesis, the formation of nephrons begins when mesenchymal nephron progenitor cells aggregate and transform into epithelial vesicles that elongate and assume an S-shape. Cells in different regions of the S-shaped body subsequently differentiate into the morphologically and functionally distinct segments of the mature nephron. Here, we have used an allelic series of mutations to determine the role of the secreted signaling molecule FGF8 in nephrogenesis. In the absence of FGF8 signaling, nephron formation is initiated, but the nascent nephrons do not express Wnt4 or Lim1, and nephrogenesis does not progress to the S-shaped body stage. Furthermore, the nephron progenitor cells that reside in the peripheral zone, the outermost region of the developing kidney, are progressively lost. When FGF8 signaling is severely reduced rather than eliminated, mesenchymal cells differentiate into S-shaped bodies. However, the cells within these structures that normally differentiate into the tubular segments of the mature nephron undergo apoptosis, resulting in the formation of kidneys with severely truncated nephrons consisting of renal corpuscles connected to collecting ducts by an abnormally short tubular segment. Thus, unlike other FGF family members, which regulate growth and branching morphogenesis of the collecting duct system, Fgf8 encodes a factor essential for gene regulation and cell survival at distinct steps in nephrogenesis.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Nefronas/citología , Nefronas/metabolismo , Alelos , Animales , Supervivencia Celular , Factor 8 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Nefronas/embriología , Fenotipo , Transducción de Señal , Médula Espinal/embriología , Médula Espinal/metabolismo , Médula Espinal/patología
12.
Dev Biol ; 284(1): 233-45, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15996652

RESUMEN

Several syndromes characterized by defects in cardiovascular and craniofacial development are associated with a hemizygous deletion of chromosome 22q11 in humans and involve defects in pharyngeal arch and neural crest cell development. Recent efforts have focused on identifying 22q11 deletion syndrome modifying loci. In this study, we show that mouse embryos deficient for Gbx2 display aberrant neural crest cell patterning and defects in pharyngeal arch-derived structures. Gbx2(-/-) embryos exhibit cardiovascular defects associated with aberrant development of the fourth pharyngeal arch arteries including interrupted aortic arch type B, right aortic arch, and retroesophageal right subclavian artery. Other developmental abnormalities include overriding aorta, ventricular septal defects, cranial nerve, and craniofacial skeletal patterning defects. Recently, Fgf8 has been proposed as a candidate modifier for 22q11 deletion syndromes. Here, we demonstrate that Fgf8 and Gbx2 expression overlaps in regions of the developing pharyngeal arches and that they interact genetically during pharyngeal arch and cardiovascular development.


Asunto(s)
Tipificación del Cuerpo/fisiología , Región Branquial/irrigación sanguínea , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Cresta Neural/embriología , Animales , Arterias/anomalías , Inmunohistoquímica , Hibridación in Situ , Ratones , Microscopía Confocal
13.
Mol Cell Biol ; 24(14): 6445-55, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15226444

RESUMEN

The mouse gene Zfp36L1 encodes zinc finger protein 36-like 1 (Zfp36L1), a member of the tristetraprolin (TTP) family of tandem CCCH finger proteins. TTP can bind to AU-rich elements within the 3'-untranslated regions of the mRNAs encoding tumor necrosis factor (TNF) and granulocyte-macrophage colony-stimulating factor (GM-CSF), leading to accelerated mRNA degradation. TTP knockout mice exhibit an inflammatory phenotype that is largely due to increased TNF secretion. Zfp36L1 has activities similar to those of TTP in cellular RNA destabilization assays and in cell-free RNA binding and deadenylation assays, suggesting that it may play roles similar to those of TTP in mammalian physiology. To address this question we disrupted Zfp36L1 in mice. All knockout embryos died in utero, most by approximately embryonic day 11 (E11). Failure of chorioallantoic fusion occurred in about two-thirds of cases. Even when fusion occurred, by E10.5 the affected placentas exhibited decreased cell division and relative atrophy of the trophoblast layers. Although knockout embryos exhibited neural tube abnormalities and increased apoptosis within the neural tube and also generalized runting, these and other findings may have been due to deficient placental function. Embryonic expression of Zfp36L1 at E8.0 was greatest in the allantois, consistent with a potential role in chorioallantoic fusion. Fibroblasts derived from knockout embryos had apparently normal levels of fully polyadenylated compared to deadenylated GM-CSF mRNA and normal rates of turnover of this mRNA species, both sensitive markers of TTP deficiency in cells. We postulate that lack of Zfp36L1 expression during mid-gestation results in the abnormal stabilization of one or more mRNAs whose encoded proteins lead directly or indirectly to abnormal placentation and fetal death.


Asunto(s)
Alantoides/metabolismo , Corion/metabolismo , Proteínas de Unión al ADN , Embrión de Mamíferos/fisiología , Proteínas Inmediatas-Precoces/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Dedos de Zinc , Alantoides/anatomía & histología , Alantoides/patología , Animales , Factor 1 de Respuesta al Butirato , Células Cultivadas , Corion/anatomía & histología , Corion/patología , Embrión de Mamíferos/anatomía & histología , Fibroblastos/citología , Fibroblastos/fisiología , Marcación de Gen , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Familia de Multigenes , Proteínas Nucleares , Fenotipo , Placenta/citología , Placenta/metabolismo , Placenta/patología , Estabilidad del ARN , ARN Mensajero/metabolismo , Distribución Tisular , Tristetraprolina
14.
Development ; 131(9): 2205-18, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15073157

RESUMEN

The neural crest is a multipotent, migratory cell population arising from the border of the neural and surface ectoderm. In mouse, the initial migratory neural crest cells occur at the five-somite stage. Bone morphogenetic proteins (BMPs), particularly BMP2 and BMP4, have been implicated as regulators of neural crest cell induction, maintenance, migration, differentiation and survival. Mouse has three known BMP2/4 type I receptors, of which Bmpr1a is expressed in the neural tube sufficiently early to be involved in neural crest development from the outset; however, earlier roles in other domains obscure its requirement in the neural crest. We have ablated Bmpr1a specifically in the neural crest, beginning at the five-somite stage. We find that most aspects of neural crest development occur normally; suggesting that BMPRIA is unnecessary for many aspects of early neural crest biology. However, mutant embryos display a shortened cardiac outflow tract with defective septation, a process known to require neural crest cells and to be essential for perinatal viability. Surprisingly, these embryos die in mid-gestation from acute heart failure, with reduced proliferation of ventricular myocardium. The myocardial defect may involve reduced BMP signaling in a novel, minor population of neural crest derivatives in the epicardium, a known source of ventricular myocardial proliferation signals. These results demonstrate that BMP2/4 signaling in mammalian neural crest derivatives is essential for outflow tract development and may regulate a crucial proliferation signal for the ventricular myocardium.


Asunto(s)
Embrión de Mamíferos/fisiología , Ventrículos Cardíacos/crecimiento & desarrollo , Morfogénesis/fisiología , Miocardio/metabolismo , Cresta Neural/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Factor de Crecimiento Transformador beta , Proteínas de Pez Cebra , Animales , Proteína Morfogenética Ósea 2 , Proteína Morfogenética Ósea 4 , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1 , Proteínas Morfogenéticas Óseas/metabolismo , Sistema Cardiovascular/anatomía & histología , Diferenciación Celular , Embrión de Mamíferos/anatomía & histología , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Cardiopatías Congénitas , Ventrículos Cardíacos/anatomía & histología , Ratones , Ratones Noqueados , Miocardio/citología , Cresta Neural/metabolismo , Pericardio/citología , Pericardio/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Factores de Crecimiento/genética , Transducción de Señal/fisiología , Somitos/citología , Somitos/metabolismo , Proteínas Wnt
15.
Dev Biol ; 267(1): 190-202, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14975726

RESUMEN

Tbx1 has been implicated as a candidate gene responsible for defective pharyngeal arch remodeling in DiGeorge/Velocardiofacial syndrome. Tbx1(+/-) mice mimic aspects of the DiGeorge phenotype with variable penetrance, and null mice display severe pharyngeal hypoplasia. Here, we identify enhancer elements in the Tbx1 gene that are conserved through evolution and mediate tissue-specific expression. We describe the generation of transgenic mice that utilize these enhancer elements to direct Cre recombinase expression in endogenous Tbx1 expression domains. We use these Tbx1-Cre mice to fate map Tbx1-expressing precursors and identify broad regions of mesoderm, including early cardiac mesoderm, which are derived from Tbx1-expressing cells. We test the hypothesis that fibroblast growth factor 8 (Fgf8) functions downstream of Tbx1 by performing tissue-specific inactivation of Fgf8 using Tbx1-Cre mice. Resulting newborn mice display DiGeorge-like congenital cardiovascular defects that involve the outflow tract of the heart. Vascular smooth muscle differentiation in the great vessels is disrupted. This data is consistent with a model in which Tbx1 induces Fgf8 expression in the pharyngeal endoderm, which is subsequently required for normal cardiovascular morphogenesis and smooth muscle differentiation in the aorta and pulmonary artery.


Asunto(s)
Sistema Cardiovascular/embriología , Factores de Crecimiento de Fibroblastos/genética , Proteínas de Dominio T Box/genética , Animales , Secuencia de Bases , Cartilla de ADN , Elementos de Facilitación Genéticos , Factor 8 de Crecimiento de Fibroblastos , Silenciador del Gen , Humanos , Ratones , Ratones Transgénicos
16.
Genes Dev ; 17(2): 269-81, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12533514

RESUMEN

Haploinsufficiency of Tbx1 is likely a major determinant of cardiac and craniofacial birth defects associated with DiGeorge syndrome. Although mice deficient in Tbx1 exhibit pharyngeal and aortic arch defects, the developmental program and mechanisms through which Tbx1 functions are relatively unknown. We identified a single cis-element upstream of Tbx1 that recognized winged helix/forkhead box (Fox)-containing transcription factors and was essential for regulation of Tbx1 transcription in the pharyngeal endoderm and head mesenchyme. The Tbx1 regulatory region was responsive to signaling by Sonic hedgehog (Shh) in vivo. We show that Shh is necessary for aortic arch development, similar to Tbx1, and is also required for expression of Foxa2 and Foxc2 in the pharyngeal endoderm and head mesenchyme, respectively. Foxa2, Foxc1, or Foxc2 could bind and activate transcription through the critical cis-element upstream of Tbx1, and Foxc proteins were required, within their expression domains, for Tbx1 transcription in vivo. We propose that Tbx1 is a direct transcriptional target of Fox proteins and that Fox proteins may serve an intermediary role in Shh regulation of Tbx1.


Asunto(s)
Proteínas Nucleares/fisiología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología , Transactivadores/genética , Factores de Transcripción/fisiología , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Síndrome de DiGeorge/embriología , Síndrome de DiGeorge/genética , Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog , Factor Nuclear 3-beta del Hepatocito , Humanos , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Modelos Biológicos , Proteínas Nucleares/genética , Especificidad de Órganos , Transducción de Señal , Proteínas de Dominio T Box/deficiencia , Factores de Transcripción/genética
17.
Development ; 129(19): 4605-11, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12223416

RESUMEN

Tbx1 haploinsufficiency causes aortic arch abnormalities in mice because of early growth and remodeling defects of the fourth pharyngeal arch arteries. The function of Tbx1 in the development of these arteries is probably cell non-autonomous, as the gene is not expressed in structural components of the artery but in the surrounding pharyngeal endoderm. We hypothesized that Tbx1 may trigger signals from the pharyngeal endoderm directed to the underlying mesenchyme. We show that the expression patterns of Fgf8 and Fgf10, which partially overlap with Tbx1 expression pattern, are altered in Tbx1(-/-) mutants. In particular, Fgf8 expression is abolished in the pharyngeal endoderm. To understand the significance of this finding for the pathogenesis of the mutant Tbx1 phenotype, we crossed Tbx1 and Fgf8 mutants. Double heterozygous Tbx1(+/-);Fgf8(+/-) mutants present with a significantly higher penetrance of aortic arch artery defects than do Tbx1(+/-);Fgf8(+/+) mutants, while Tbx1(+/+);Fgf8(+/-) animals are normal. We found that Fgf8 mutation increases the severity of the primary defect caused by Tbx1 haploinsufficiency, i.e. early hypoplasia of the fourth pharyngeal arch arteries, consistent with the time and location of the shared expression domain of the two genes. Hence, Tbx1 and Fgf8 interact genetically in the development of the aortic arch. Our data provide the first evidence of a genetic link between Tbx1 and FGF signaling, and the first example of a modifier of the Tbx1 haploinsufficiency phenotype. We speculate that the FGF8 locus might affect the penetrance of cardiovascular defects in individuals with chromosome 22q11 deletions involving TBX1.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Ligamiento Genético , Transducción de Señal/genética , Proteínas de Dominio T Box/genética , Animales , Aorta Torácica/anomalías , Aorta Torácica/embriología , Región Branquial/anomalías , Región Branquial/embriología , Femenino , Factor 10 de Crecimiento de Fibroblastos , Factor 8 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/fisiología , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Arteria Pulmonar/anomalías , Arteria Pulmonar/embriología , Proteínas de Dominio T Box/fisiología
18.
Development ; 129(19): 4613-25, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12223417

RESUMEN

We present here an analysis of cardiovascular and pharyngeal arch development in mouse embryos hypomorphic for Fgf8. Previously, we have described the generation of Fgf8 compound heterozygous (Fgf8(neo/-)) embryos. Although early analysis demonstrated that some of these embryos have abnormal left-right (LR) axis specification and cardiac looping reversals, the number and type of cardiac defects present at term suggested an additional role for Fgf8 in cardiovascular development. Most Fgf8(neo/-) mutant embryos survive to term with abnormal cardiovascular patterning, including outflow tract, arch artery and intracardiac defects. In addition, these mutants have hypoplastic pharyngeal arches, small or absent thymus and abnormal craniofacial development. Neural crest cells (NCCs) populate the pharyngeal arches and contribute to many structures of the face, neck and cardiovascular system, suggesting that Fgf8 may be required for NCC development. Fgf8 is expressed within the developing pharyngeal arch ectoderm and endoderm during NCC migration through the arches. Analysis of NCC development in Fgf8(neo/-) mutant embryos demonstrates that NCCs are specified and migrate, but undergo cell death in areas both adjacent and distal to where Fgf8 is normally expressed. This study defines the cardiovascular defects present in Fgf8 mutants and supports a role for Fgf8 in development of all the pharyngeal arches and in NCC survival.


Asunto(s)
Tipificación del Cuerpo , Región Branquial/embriología , Anomalías Cardiovasculares/metabolismo , Factores de Crecimiento de Fibroblastos/fisiología , Cardiopatías Congénitas/metabolismo , Animales , Aorta Torácica/embriología , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Biomarcadores , Región Branquial/anomalías , Sistema Cardiovascular/embriología , Recuento de Células , División Celular , Movimiento Celular , Vasos Coronarios/embriología , Proteínas de Unión al ADN/genética , Femenino , Factor 8 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Expresión Génica , Corazón/embriología , Cardiopatías Congénitas/embriología , Secuencias Hélice-Asa-Hélice , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Arteria Pulmonar/anomalías , Arteria Pulmonar/embriología , Receptores de Ácido Retinoico/genética , Proteínas de Dominio T Box/genética , Factor de Transcripción AP-2 , Factores de Transcripción/genética , Proteínas de Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...