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










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 31(6): 107626, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32402288

RESUMEN

The shortage of donor lungs hinders lung transplantation, the only definitive option for patients with end-stage lung disease. Blastocyst complementation enables the generation of transplantable organs from pluripotent stem cells (PSCs) in animal models. Pancreases and kidneys have been generated from PSCs by blastocyst complementation in rodent models. Here, we report the generation of lungs using mouse embryonic stem cells (ESCs) in apneumic Fgf10 Ex1mut/Ex3mutmice by blastocyst complementation. Complementation with ESCs enables Fgf10-deficient mice to survive to adulthood without abnormalities. Both the generated lung alveolar parenchyma and the interstitial portions, including vascular endothelial cells, vascular and parabronchial smooth muscle cells, and connective tissue, largely originate from the injected ESCs. These data suggest that Fgf10 Ex1mut/Ex3mutblastocysts provide an organ niche for lung generation and that blastocyst complementation could be a viable approach for generating whole lungs.


Asunto(s)
Blastocisto/metabolismo , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Pulmón/fisiopatología , Animales , Quimerismo , Modelos Animales de Enfermedad , Ratones
2.
J Biol Chem ; 294(41): 15052-15067, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31431501

RESUMEN

Peritoneal fibrosis is a common complication of long-term peritoneal dialysis (PD) and the principal cause of ultrafiltration failure during PD. The initial and reversible step in PD-associated peritoneal fibrosis is the epithelial-mesenchymal transition (EMT). Although the mechanisms in the EMT have been the focus of many studies, only limited information is currently available concerning microRNA (miRNA) regulation in peritoneal fibrosis. In this study, we aimed to characterize the roles of microRNA-145 (miR-145) and fibroblast growth factor 10 (FGF10) in peritoneal fibrosis. After inducing EMT with transforming growth factor-ß1 (TGF-ß1) in vitro, we found that miR-145 is significantly up-regulated, whereas FGF10 is markedly down-regulated, suggesting a close link between miR-145 and FGF10 in peritoneal fibrosis, further confirmed in luciferase reporter experiments. Furthermore, in human peritoneal mesothelial cells (i.e. HMrSV5 cells), miR-145 mimics induced EMT, whereas miR-145 inhibition suppressed EMT, and we also observed that miR-145 suppressed FGF10 expression. In vivo, we found that the exogenous delivery of an miR-145 expression plasmid both blocked FGF10 and intensified the EMT, whereas miR-145 inhibition promoted the expression of FGF10 and reversed the EMT. In conclusion, miR-145 promotes the EMT during the development of peritoneal fibrosis by suppressing FGF10 activity, suggesting that miR-145 represents a potential therapeutic target for managing peritoneal fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Factor 10 de Crecimiento de Fibroblastos/genética , MicroARNs/genética , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/patología , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Línea Celular , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
3.
Dev Biol ; 451(2): 158-166, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30965042

RESUMEN

Mucus secretion and mucociliary clearance are crucial processes required to maintain pulmonary homeostasis. In the trachea and nasal passages, mucus is secreted by submucosal glands (SMGs) that line the airway, with an additional contribution from goblet cells of the surface airway epithelium. The SMG mucus is rich in mucins and antimicrobial enzymes. Defective tracheal SMGs contribute to hyper-secretory respiratory diseases, such as cystic fibrosis, asthma, and chronic obstructive pulmonary disease, however little is known about the signals that regulate their morphogenesis and patterning. Here, we show that Fgf10 is essential for the normal development of murine tracheal SMGs, with gland development arresting at the early bud stage in the absence of FGF10 signalling. As Fgf10 knockout mice are lethal at birth, inducible knockdown of Fgf10 at late embryonic stages was used to follow postnatal gland formation, confirming the essential role of FGF10 in SMG development. In heterozygous Fgf10 mice the tracheal glands formed but with altered morphology and restricted distribution. The reduction in SMG branching in Fgf10 heterozygous mice was not rescued with time and resulted in a reduction in overall tracheal mucus secretion. Fgf10 is therefore a key signal in SMG development, influencing both the number of glands and extent of branching morphogenesis, and is likely, therefore, to play a role in aspects of SMG-dependent respiratory health.


Asunto(s)
Glándulas Exocrinas/embriología , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Mucosa Respiratoria/embriología , Tráquea/embriología , Animales , Cruzamientos Genéticos , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Masculino , Ratones , Morfogénesis , Moco/metabolismo , Tráquea/metabolismo
4.
Hum Mol Genet ; 28(9): 1429-1444, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30566624

RESUMEN

Bronchopulmonary dysplasia (BPD), characterized by alveoli simplification and dysmorphic pulmonary microvasculature, is a chronic lung disease affecting prematurely born infants. Pulmonary hypertension (PH) is an important BPD feature associated with morbidity and mortality. In human BPD, inflammation leads to decreased fibroblast growth factor 10 (FGF10) expression but the impact on the vasculature is so far unknown. We used lungs from Fgf10+/- versus Fgf10+/+ pups to investigate the effect of Fgf10 deficiency on vascular development in normoxia (NOX) and hyperoxia (HOX, BPD mouse model). To assess the role of fibroblast growth factor receptor 2b (Fgfr2b) ligands independently of early developmentaldefects, we used an inducible double transgenic system in mice allowing inhibition of Fgfr2b ligands activity. Using vascular morphometry, we quantified the pathological changes. Finally, we evaluated changes in FGF10, surfactant protein C (SFTPC), platelet endothelial cell adhesion molecule (PECAM) and alpha-smooth muscle actin 2 (α-SMA) expression in human lung samples from patients suffering from BPD. In NOX, no major difference in the lung vasculature between Fgf10+/- and control pups was detected. In HOX, a greater loss of blood vessels in Fgf10+/- lungs is associated with an increase of poorly muscularized vessels. Fgfr2b ligands inhibition postnatally in HOX is sufficient to decrease the number of blood vessels while increasing the level of muscularization, suggesting a PH phenotype. BPD lungs exhibited decreased FGF10, SFTPC and PECAM but increased α-SMA. Fgf10 deficiency-associated vascular defects are enhanced in HOX and could represent an additional cause of morbidity in human patients with BPD.


Asunto(s)
Displasia Broncopulmonar/etiología , Displasia Broncopulmonar/patología , Susceptibilidad a Enfermedades , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Animales , Biomarcadores , Displasia Broncopulmonar/metabolismo , Biología Computacional/métodos , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Genotipo , Hipoxia , Pulmón/patología , Ratones , Mutación , Neovascularización Fisiológica/genética , Consumo de Oxígeno , Fosforilación , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
5.
Development ; 144(12): 2212-2221, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28506989

RESUMEN

The patterning of repeated structures is a major theme in developmental biology, and the inter-relationship between spacing and size of such structures is an unresolved issue. Fungiform papillae are repeated epithelial structures that house taste buds on the anterior tongue. Here, we report that FGF signaling is a crucial regulator of fungiform papillae development. We found that mesenchymal FGF10 controls the size of the papillary area, while overall patterning remains unchanged. Our results show that FGF signaling negatively affects the extent of canonical Wnt signaling, which is the main activation pathway during fungiform papillae development; however, this effect does not occur at the level of gene transcription. Rather, our experimental data, together with computational modeling, indicate that FGF10 modulates the range of Wnt effects, likely via induction of Sostdc1 expression. We suggest that modification of the reach of Wnt signaling could be due to local changes in morphogen diffusion, representing a novel mechanism in this tissue context, and we propose that this phenomenon might be involved in a broader array of mammalian developmental processes.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/metabolismo , Papilas Gustativas/embriología , Papilas Gustativas/metabolismo , Vía de Señalización Wnt , Proteínas Adaptadoras Transductoras de Señales , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Simulación por Computador , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Embarazo , Proteínas Serina-Treonina Quinasas
6.
J Pathol ; 241(1): 91-103, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27770432

RESUMEN

Inflammation-induced FGF10 protein deficiency is associated with bronchopulmonary dysplasia (BPD), a chronic lung disease of prematurely born infants characterized by arrested alveolar development. So far, experimental evidence for a direct role of FGF10 in lung disease is lacking. Using the hyperoxia-induced neonatal lung injury as a mouse model of BPD, the impact of Fgf10 deficiency in Fgf10+/- versus Fgf10+/+ pups was investigated. In normoxia, no lethality of Fgf10+/+ or Fgf10+/- pups was observed. By contrast, all Fgf10+/- pups died within 8 days of hyperoxic injury, with lethality starting at day 5, whereas Fgf10+/+ pups were all alive. Lungs of pups from the two genotypes were collected on postnatal day 3 following normoxia or hyperoxia exposure for further analysis. In hyperoxia, Fgf10+/- lungs exhibited increased hypoalveolarization. Analysis by FACS of the Fgf10+/- versus control lungs in normoxia revealed a decreased ratio of alveolar epithelial type II (AECII) cells over total Epcam-positive cells. In addition, gene array analysis indicated reduced AECII and increased AECI transcriptome signatures in isolated AECII cells from Fgf10+/- lungs. Such an imbalance in differentiation is also seen in hyperoxia and is associated with reduced mature surfactant protein B and C expression. Attenuation of the activity of Fgfr2b ligands postnatally in the context of hyperoxia also led to increased lethality with decreased surfactant expression. In summary, decreased Fgf10 mRNA levels lead to congenital lung defects, which are compatible with postnatal survival, but which compromise the ability of the lungs to cope with sub-lethal hyperoxic injury. Fgf10 deficiency affects quantitatively and qualitatively the formation of AECII cells. In addition, Fgfr2b ligands are also important for repair after hyperoxia exposure in neonates. Deficient AECII cells could be an additional complication for patients with BPD. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/etiología , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Surfactantes Pulmonares/metabolismo , ARN Mensajero/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo
7.
Dev Biol ; 419(2): 348-356, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27590203

RESUMEN

Hypertrophy, hyperplasia and altered mucus secretion from the respiratory submucosal glands (SMG) are characteristics of airway diseases such as cystic fibrosis, asthma and chronic bronchitis. More commonly, hyper-secretion of the nasal SMGs contributes to allergic rhinitis and upper airway infection. Considering the role of these glands in disease states, there is a significant dearth in understanding the molecular signals that regulate SMG development and patterning. Due to the imperative role of FGF signalling during the development of other branched structures, we investigated the role of Fgf10 during initiation and branching morphogenesis of murine nasal SMGs. Fgf10 is expressed in the mesenchyme around developing SMGs while expression of its receptor Fgfr2 is seen within glandular epithelial cells. In the Fgf10 null embryo, Steno's gland and the maxillary sinus gland were completely absent while other neighbouring nasal glands showed normal duct elongation but defective branching. Interestingly, the medial nasal glands were present in Fgf10 homozygotes but missing in Fgfr2b mutants, with expression of Fgf7 specifically expressed around these developing glands, indicating that Fgf7 might compensate for loss of Fgf10 in this group of glands. Intriguingly the lateral nasal glands were only mildly affected by loss of FGF signalling, while these glands were missing in Eda mutant mice, where the Steno's and maxillary sinus gland developed as normal. This analysis reveals that regulation of nasal gland development is complex with different subsets of glands being regulated by different signalling pathways. This analysis helps shed light on the nasal gland defects observed in patients with hypohidrotic ectodermal dysplasia (HED) (defect EDA pathway) and LADD syndrome (defect FGFR2b pathway).


Asunto(s)
Ectodisplasinas/fisiología , Glándulas Exocrinas/embriología , Factor 10 de Crecimiento de Fibroblastos/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/fisiología , Animales , Ectodisplasinas/deficiencia , Ectodisplasinas/genética , Resección Endoscópica de la Mucosa , Glándulas Exocrinas/metabolismo , Glándulas Exocrinas/ultraestructura , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 7 de Crecimiento de Fibroblastos/fisiología , Masculino , Seno Maxilar/embriología , Seno Maxilar/ultraestructura , Mesodermo/metabolismo , Ratones , Morfogénesis , Mucosa Nasal/embriología , Mucosa Nasal/ultraestructura , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/deficiencia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética
8.
Development ; 140(18): 3731-42, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23924632

RESUMEN

Localized Fgf10 expression in the distal mesenchyme adjacent to sites of lung bud formation has long been thought to drive stereotypic branching morphogenesis even though isolated lung epithelium branches in the presence of non-directional exogenous Fgf10 in Matrigel. Here, we show that lung agenesis in Fgf10 knockout mice can be rescued by ubiquitous overexpression of Fgf10, indicating that precisely localized Fgf10 expression is not required for lung branching morphogenesis in vivo. Fgf10 expression in the mesenchyme itself is regulated by Wnt signaling. Nevertheless, we found that during lung initiation simultaneous overexpression of Fgf10 is not sufficient to rescue the absence of primary lung field specification in embryos overexpressing Dkk1, a secreted inhibitor of Wnt signaling. However, after lung initiation, simultaneous overexpression of Fgf10 in lungs overexpressing Dkk1 is able to rescue defects in branching and proximal-distal differentiation. We also show that Fgf10 prevents the differentiation of distal epithelial progenitors into Sox2-expressing airway epithelial cells in part by activating epithelial ß-catenin signaling, which negatively regulates Sox2 expression. As such, these findings support a model in which the main function of Fgf10 during lung development is to regulate proximal-distal differentiation. As the lung buds grow out, proximal epithelial cells become further and further displaced from the distal source of Fgf10 and differentiate into bronchial epithelial cells. Interestingly, our data presented here show that once epithelial cells are committed to the Sox2-positive airway epithelial cell fate, Fgf10 prevents ciliated cell differentiation and promotes basal cell differentiation.


Asunto(s)
Diferenciación Celular , Células Epiteliales/patología , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Morfogénesis , Células Madre/patología , Animales , Regulación hacia Abajo , Activación Enzimática , Células Epiteliales/metabolismo , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Deformidades Congénitas de las Extremidades/patología , Pulmón/anomalías , Pulmón/patología , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Respiración , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Células Madre/enzimología , Tráquea/patología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
9.
Dev Biol ; 369(2): 340-8, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22819677

RESUMEN

Fibroblast growth factor (FGF) signaling to the epithelium and mesenchyme mediated by FGF10 and FGF9, respectively, controls cecal formation during embryonic development. In particular, mesenchymal FGF10 signals to the epithelium via FGFR2b to induce epithelial cecal progenitor cell proliferation. Yet the precise upstream mechanisms controlling mesenchymal FGF10 signaling are unknown. Complete deletion of Fgf9 as well as of Pitx2, a gene encoding a homeobox transcription factor, both lead to cecal agenesis. Herein, we used mouse genetic approaches to determine the precise contribution of the epithelium and/or mesenchyme tissue compartments in this process. Using tissue compartment specific Fgf9 versus Pitx2 loss of function approaches in the gut epithelium and/or mesenchyme, we determined that FGF9 signals to the mesenchyme via Pitx2 to induce mesenchymal Fgf10 expression, which in turn leads to epithelial cecal bud formation.


Asunto(s)
Ciego/embriología , Ciego/metabolismo , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Ciego/anomalías , Proliferación Celular , Cartilla de ADN/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 9 de Crecimiento de Fibroblastos/deficiencia , Factor 9 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Masculino , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Modelos Biológicos , Embarazo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteína del Homeodomínio PITX2
10.
Artículo en Inglés | MEDLINE | ID: mdl-21696361

RESUMEN

The FGF family comprises twenty-two evolutionarily related members with diverse functions in development, metabolism, and neuronal activities. FGF10 and FGF21 play unique roles in adipocyte development and metabolism, respectively. FGF10 mediates biological responses by activating FGF receptor 2b (FGFR2b) with heparin/heparan sulfate in a paracrine manner. In contrast, FGF21 mediates biological responses by activating FGFRs with ßKlotho in cultured cells. However, FGF21 acts in an autocrine manner via a ß Klotho-independent signaling pathway in mice. Fgf10 knockout mice die shortly after birth. Preadipocyte proliferation and adipogenesis are greatly impaired in Fgf10 knockout mouse embryos. FGF10 stimulates preadipocyte proliferation through the Ras/MAPK pathway followed by the cyclin D2-dependent phosphorylation of p130. FGF10 also stimulates adipogenesis by inducing the expression of pRb through the Ras/MAPK pathway. pRb binds C/EBPα. The pRb-C/EBPα complex induces adipogenesis. Fgf21 is abundantly expressed in the liver. Hepatic Fgf21 expression is markedly induced in mice by fasting. FGF21 exerts pharmacological effects on glucose and lipid metabolism in hepatocytes and adipocytes. However, the phenotypes of Fgf21 knockout mice, which are apparently normal and fertile, indicate FGF21 not to be a physiological regulator for hepatic functions. Hepatic FGF21 inhibits lipolysis in adipocytes, and so is a negative regulator of lipolysis during fasting. FGF21 may be a "thrifty factor". Serum FGF21 levels are increased in patients with metabolic diseases related with obesity, indicating potential roles of FGF21 in adipocyte metabolism.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/fisiología , Diferenciación Celular/fisiología , Factor 10 de Crecimiento de Fibroblastos/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Adipocitos/citología , Adipocitos/patología , Adipogénesis/genética , Animales , Diferenciación Celular/genética , Proliferación Celular , Células Cultivadas , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Humanos , Comunicación Paracrina/genética , Comunicación Paracrina/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Células Madre/citología , Células Madre/metabolismo , Células Madre/patología
11.
Development ; 138(2): 273-82, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21148187

RESUMEN

During embryonic development, appropriate dorsoventral patterning of the trachea leads to the formation of periodic cartilage rings from the ventral mesenchyme and continuous smooth muscle from the dorsal mesenchyme. In this work, we have investigated the role of two crucial morphogens, fibroblast growth factor 10 and sonic hedgehog, in the formation of periodically alternating cartilaginous and non-cartilaginous domains in the ventral mesenchyme. Using a combination of gain- and loss-of-function approaches for FGF10 and SHH, we demonstrate that precise spatio-temporal patterns and appropriate levels of expression of these two signaling molecules in the ventral area are crucial between embryonic day 11.5 and 13.5 for the proper patterning of the cartilage rings. We conclude that the expression level of FGF10 in the mesenchyme has to be within a critical range to allow for periodic expression of Shh in the ventral epithelium, and consequently for the correct patterning of the cartilage rings. We propose that disturbed balances of Fgf10 and Shh may explain a subset of human tracheomalacia without tracheo-esophageal fistula or tracheal atresia.


Asunto(s)
Cartílago/embriología , Factor 10 de Crecimiento de Fibroblastos/fisiología , Proteínas Hedgehog/fisiología , Tráquea/embriología , Animales , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Cartílago/anomalías , Cartílago/metabolismo , Diferenciación Celular , Proliferación Celular , Epitelio/embriología , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Humanos , Hibridación in Situ , Mesodermo/embriología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Embarazo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Transducción de Señal , Tráquea/anomalías , Tráquea/metabolismo
12.
Am J Med Genet A ; 152A(12): 2947-61, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21108385

RESUMEN

Retinoic acid (RA) is a biologically active derivative of vitamin A that is indispensable for inner ear development. The normal function of RA is achieved only at optimal homeostatic concentrations, with an excess or deficiency in RA leading to inner ear dysmorphogenesis. We present an overview of the role of RA in the developing mammalian inner ear, discussing both how and when RA may act to critically control a program of inner ear development. Molecular mechanisms of otic teratogenicity involving two members of the fibroblast growth factor family, FGF3 and FGF10, and their downstream targets, Dlx5 and Dlx6, are examined under conditions of both RA excess and deficiency. We term the effect of too little or too much RA on FGF/Dlx signaling a Goldilocks phenomenon. We demonstrate that in each case (RA excess, RA deficiency), RA can directly affect FGF3/FGF10 signaling within the otic epithelium, leading to downregulated expression of these essential signaling molecules, which in turn, leads to diminution in Dlx5/Dlx6 expression. Non-cell autonomous affects of the otic epithelium subsequently occur, altering transforming growth factor-beta (TGFß) expression in the neighboring periotic mesenchyme and serving as a putative explanation for RA-mediated otic capsule defects. We conclude that RA coordinates inner ear morphogenesis by controlling an FGF/Dlx signaling cascade, whose perturbation by deviations in local retinoid concentrations can lead to inner ear dysmorphogenesis.


Asunto(s)
Oído Interno/embriología , Organogénesis/genética , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Tretinoina/metabolismo , Animales , Comunicación Celular/genética , Oído Interno/anomalías , Embrión de Mamíferos , Epitelio/metabolismo , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/deficiencia , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Mesodermo/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología , Tretinoina/farmacología
13.
Circ Res ; 106(3): 495-503, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20035084

RESUMEN

RATIONALE: The genes encoding fibroblast growth factor (FGF) 8 and 10 are expressed in the anterior part of the second heart field that constitutes a population of cardiac progenitor cells contributing to the arterial pole of the heart. Previous studies of hypomorphic and conditional Fgf8 mutants show disrupted outflow tract (OFT) and right ventricle (RV) development, whereas Fgf10 mutants do not have detectable OFT defects. OBJECTIVES: Our aim was to investigate functional overlap between Fgf8 and Fgf10 during formation of the arterial pole. METHODS AND RESULTS: We generated mesodermal Fgf8; Fgf10 compound mutants with MesP1Cre. The OFT/RV morphology in these mutants was affected with variable penetrance; however, the incidence of embryos with severely affected OFT/RV morphology was significantly increased in response to decreasing Fgf8 and Fgf10 gene dosage. Fgf8 expression in the pharyngeal arch ectoderm is important for development of the pharyngeal arch arteries and their derivatives. We now show that Fgf8 deletion in the mesoderm alone leads to pharyngeal arch artery phenotypes and that these vascular phenotypes are exacerbated by loss of Fgf10 function in the mesodermal core of the arches. CONCLUSIONS: These results show functional overlap of FGF8 and FGF10 signaling from second heart field mesoderm during development of the OFT/RV, and from pharyngeal arch mesoderm during pharyngeal arch artery formation, highlighting the sensitivity of these key aspects of cardiovascular development to FGF dosage.


Asunto(s)
Región Branquial/irrigación sanguínea , Corazón Fetal/crecimiento & desarrollo , Factor 10 de Crecimiento de Fibroblastos/fisiología , Factor 8 de Crecimiento de Fibroblastos/fisiología , Cardiopatías Congénitas/embriología , Animales , Región Branquial/anomalías , Región Branquial/embriología , Cruzamientos Genéticos , Factor 10 de Crecimiento de Fibroblastos/biosíntesis , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/deficiencia , Factor 8 de Crecimiento de Fibroblastos/genética , Eliminación de Gen , Dosificación de Gen , Genotipo , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Ventrículos Cardíacos/anomalías , Ventrículos Cardíacos/embriología , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Mutantes , Obstrucción del Flujo Ventricular Externo/embriología , Obstrucción del Flujo Ventricular Externo/genética
14.
Neuron ; 63(1): 48-62, 2009 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-19607792

RESUMEN

Radial glia (RG), the progenitors of cortical neurons and basal progenitors (BPs), differentiate from neuroepithelial cells (NCs) with stem cell properties. We show that the morphogen Fgf10 is transiently expressed by NCs coincident with the transition period of NC differentiation into RG. Targeted deletion of Fgf10 delays RG differentiation, whereas overexpression has opposing effects. Delayed RG differentiation in Fgf10 mutants occurs selectively in rostral cortex, paralleled by an extended period of symmetric NC divisions increasing progenitor number, coupled with delayed and initially diminished production of neurons and BPs. RG eventually differentiate in excess number and overproduce neurons and BPs rostrally resulting in tangential expansion of frontal areas and increased laminar thickness. Thus, transient Fgf10 expression regulates timely differentiation of RG, and through this function, determines both length of the early progenitor expansion phase and onset of neurogenesis and ultimately the number of progenitors and neurons fated to specific cortical areas.


Asunto(s)
Diferenciación Celular/genética , Corteza Cerebral/citología , Células Madre Embrionarias/fisiología , Factor 10 de Crecimiento de Fibroblastos/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Ciclo Celular/genética , Proliferación Celular , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Electroporación/métodos , Embrión de Mamíferos , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis , Embarazo
15.
Dev Dyn ; 238(8): 1999-2013, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19618463

RESUMEN

Fibroblast growth factor 10 (FGF10) signaling through FGF receptor 2 (FGFR2) is required for lung initiation. While studies indicate that Fgf10 and Fgfr2 are also important at later stages of lung development, their roles in early branching events remain unclear. We addressed this question through conditional inactivation of both genes in mouse subsequent to lung initiation. Inactivation of Fgf10 in lung mesenchyme resulted in smaller lobes with a reduced number of branches. Inactivation of Fgfr2 in lung epithelium resulted in disruption of lobes and small epithelial outgrowths that arose arbitrarily along the main bronchi. In both mutants, there was an increase in cell death. Also, the expression patterns of key signaling molecules implicated in branching morphogenesis were altered and a proximal lung marker was expanded distally. Our results indicate that both Fgf10 and Fgfr2 are required for a normal branching program and for proper proximal-distal patterning of the lung.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/fisiología , Pulmón/embriología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Animales , Secuencia de Bases , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Muerte Celular/genética , Muerte Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Cartilla de ADN/genética , Epitelio/embriología , Epitelio/fisiología , Femenino , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Pulmón/anomalías , Pulmón/fisiología , Mesodermo/embriología , Mesodermo/fisiología , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Embarazo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/deficiencia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/fisiología , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/fisiología , Transducción de Señal
16.
Endocr Dev ; 12: 46-54, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17923768

RESUMEN

Over the last decades, pancreas development has been widely investigated. Understanding the mechanisms that control beta-cell development should allow progress towards the regeneration of these cells in humans. Particularly, it is well established that inductive signals from the mesenchyme play an essential role in the proliferation of precursor cells. In the present review, we focused on the roles of fibroblast growth factors (FGFs) in pancreas development. Improvements of the in vivo and in vitro techniques were used to define the function of FGF10. Experiments on FGF10 knockout mice showed that FGF10 is required for the proliferation of precursor cells and the pancreas development. Several laboratories used different in vitro techniques to study the effect of FGF10 on beta-cell differentiation. These methods of investigation are described here. In our experiments, pancreases were placed at the air-liquid interface to define the precise mechanism of action of FGF10. We showed that FGF10 positively regulates the beta-cell mass by increasing the proliferation of the early precursors and by extending the window of expression of the endocrine precursor marker Ngn3. These data are compared with studies performed with other culture systems. Finally, the role of other FGFs is discussed.


Asunto(s)
Islotes Pancreáticos/fisiología , Páncreas/embriología , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/fisiología , Inmunohistoquímica , Hibridación in Situ , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/citología , Islotes Pancreáticos/embriología , Técnicas de Cultivo de Órganos , Ratas , Ratas Mutantes
17.
Dev Biol ; 308(2): 379-91, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17601531

RESUMEN

FGF signaling is required during multiple stages of inner ear development in many different vertebrates, where it is involved in induction of the otic placode, in formation and morphogenesis of the otic vesicle as well as for cellular differentiation within the sensory epithelia. In this study we have looked to define the redundant and conserved roles of FGF3, FGF8 and FGF10 during the development of the murine and avian inner ear. In the mouse, hindbrain-derived FGF10 ectopically induces FGF8 and rescues otic vesicle formation in Fgf3 and Fgf10 homozygous double mutants. Conditional inactivation of Fgf8 after induction of the placode does not interfere with otic vesicle formation and morphogenesis but affects cellular differentiation in the inner ear. In contrast, inactivation of Fgf8 during induction of the placode in a homozygous Fgf3 null background leads to a reduced size otic vesicle or the complete absence of otic tissue. This latter phenotype is more severe than the one observed in mutants carrying null mutations for both Fgf3 and Fgf10 that develop microvesicles. However, FGF3 and FGF10 are redundantly required for morphogenesis of the otic vesicle and the formation of semicircular ducts. In the chicken embryo, misexpression of Fgf3 in the hindbrain induces ectopic otic vesicles in vivo. On the other hand, Fgf3 expression in the hindbrain or pharyngeal endoderm is required for formation of the otic vesicle from the otic placode. Together these results provide important insights into how the spatial and temporal expression of various FGFs controls different steps of inner ear formation during vertebrate development.


Asunto(s)
Oído Interno/embriología , Oído Interno/metabolismo , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 3 de Crecimiento de Fibroblastos/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Animales , Secuencia de Bases , Embrión de Pollo , ADN/genética , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 3 de Crecimiento de Fibroblastos/deficiencia , Factor 3 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/deficiencia , Factor 8 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
Dev Biol ; 299(1): 52-62, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16989802

RESUMEN

Fibroblast growth factor (FGF) signaling has been shown to regulate lung epithelial development but its influence on mesenchymal differentiation has been poorly investigated. To study the role of mesenchymal FGF signaling in the differentiation of the mesenchyme and its impact on epithelial morphogenesis, we took advantage of Fgfr2c(+/Delta) mice, which due to a splicing switch express Fgfr2b in mesenchymal tissues and manifest Apert syndrome-like phenotypes. Using a set of in vivo and in vitro studies, we show that an autocrine FGF10-FGFR2b signaling loop is established in the mutant lung mesenchyme, which has several consequences. It prevents the entry of the smooth muscle progenitors into the smooth muscle cell (SMC) lineage and results in reduced fibronectin and elastin deposition. Levels of Fgf10 expression are raised within the mutant mesenchyme itself. Epithelial branching as well as epithelial levels of FGF and canonical Wnt signaling is dramatically reduced. These defects result in arrested development of terminal airways and an "emphysema like" phenotype in postnatal lungs. Our work unravels part of the complex interactions that govern normal lung development and may be pertinent to understanding the basis of respiratory defects in Apert syndrome.


Asunto(s)
Diferenciación Celular , Pulmón/citología , Mesodermo/metabolismo , Miocitos del Músculo Liso/citología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Células Madre/citología , Animales , Animales Recién Nacidos , Comunicación Autocrina/fisiología , Diferenciación Celular/fisiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Fibronectinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Pulmón/embriología , Ratones , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Wnt/metabolismo
19.
Mech Dev ; 123(1): 17-23, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16326081

RESUMEN

Fibroblast growth factors (FGFs) have been shown to control formation and differentiation of multiple organ systems in the developing vertebrate embryo. The analysis of differential gene expression during embryogenesis is, therefore, a potent tool to identify novel target genes regulated by FGF signalling. Here, we have applied microarray analysis to identify differentially regulated genes in FGF mutant mouse embryos. Surprisingly, transcripts corresponding to vomeronasal receptors (VRs), which so far have been only detected in the vomeronasal organ (VNO), were found to be downregulated in FGF mutant embryos. VR expression was detected in the developing olfactory pit and the anlage of the VNO. Interestingly, several FGFs can be detected in the developing olfactory pit during mouse embryogenesis [Bachler, M., Neubuser, A. 2001. Expression of members of the Fgf family and their receptors during midfacial development. Mech. Dev. 100, 313-316]. FGF signalling may thus control expression of VRs in the olfactory pit and formation of the VNO. Moreover, VR expression was detected in unexpected locations within the developing embryo including retina, dorsal root ganglia and neural tube. The relevance of VR expression in these structures and for formation of the VNO is discussed.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 3 de Crecimiento de Fibroblastos/metabolismo , Receptores Odorantes/genética , Órgano Vomeronasal/embriología , Órgano Vomeronasal/metabolismo , Animales , Secuencia de Bases , ADN Complementario/genética , Regulación hacia Abajo , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 3 de Crecimiento de Fibroblastos/deficiencia , Factor 3 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
20.
Development ; 132(22): 4963-73, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16221725

RESUMEN

Heparan sulphate proteoglycans (HSPGs) are known to be crucial for signalling by the secreted Wnt, Hedgehog, Bmp and Fgf proteins during invertebrate development. However, relatively little is known about their effect on developmental signalling in vertebrates. Here, we report the analysis of daedalus, a novel zebrafish pectoral fin mutant. Positional cloning identified fgf10 as the gene disrupted in daedalus. We find that fgf10 mutants strongly resemble zebrafish ext2 and extl3 mutants, which encode glycosyltransferases required for heparan sulphate biosynthesis. This suggests that HSPGs are crucial for Fgf10 signalling during limb development. Consistent with this proposal, we observe a strong genetic interaction between fgf10 and extl3 mutants. Furthermore, application of Fgf10 protein can rescue target gene activation in fgf10, but not in ext2 or extl3 mutants. By contrast, application of Fgf4 protein can activate target genes in both ext2 and extl3 mutants, indicating that ext2 and extl3 are differentially required for Fgf10, but not Fgf4, signalling during limb development. This reveals an unexpected specificity of HSPGs in regulating distinct vertebrate Fgfs.


Asunto(s)
Extremidades/embriología , Factor 10 de Crecimiento de Fibroblastos/fisiología , Proteoglicanos de Heparán Sulfato/biosíntesis , N-Acetilglucosaminiltransferasas/fisiología , Transducción de Señal/fisiología , Pez Cebra/embriología , Animales , Factor 10 de Crecimiento de Fibroblastos/deficiencia , Factor 10 de Crecimiento de Fibroblastos/genética , Mutación , Fenotipo , Transducción de Señal/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...