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.
Eur J Pharmacol ; 838: 129-137, 2018 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-30196111

RESUMEN

Previous studies have demonstrated that fibroblast growth factor 8b (FGF8b) is up-regulated in a large proportion of prostate cancer patients, and plays a key role in the aggressive progress of prostate cancer. Herein, we investigated the effects of a short peptide derived from the gN helix domain of FGF8b on the metastatic behaviors of prostate cancer cells. The results demonstrated that the synthetic peptide might reverse the effects of FGF8b on cell proliferation, migration and invasion by suppressing the activation of MAPK and Akt signaling cascades, and reducing the expressions of the metastasis-related proteins, resulting in suppression of the aggressive phenotype of the prostate cancer cells. Collectively, these results underline the therapeutic potential of the FGF8b mimic peptide in advanced prostate cancer.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Péptidos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Factor 8 de Crecimiento de Fibroblastos/química , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Péptidos/química , Péptidos/uso terapéutico , Neoplasias de la Próstata/patología , Dominios Proteicos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo
2.
Dev Biol ; 418(1): 204-215, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27506116

RESUMEN

Retinoic acid (RA) repression of Fgf8 is required for many different aspects of organogenesis, however relatively little is known about how endogenous RA controls gene repression as opposed to gene activation. Here, we show that nuclear receptor corepressors NCOR1 and NCOR2 (SMRT) redundantly mediate the ability of RA to repress Fgf8. Ncor1;Ncor2 double mutants generated by CRISPR/Cas9 gene editing exhibited a small somite and distended heart phenotype similar to that of RA-deficient Raldh2-/- embryos, associated with increased Fgf8 expression and FGF signaling in caudal progenitors and heart progenitors. Embryo chromatin immunoprecipitation studies revealed that NCOR1/2 but not coactivators are recruited to the Fgf8 RA response element (RARE) in an RA-dependent manner, whereas coactivators but not NCOR1/2 are recruited RA-dependently to a RARE near Rarb that is activated by RA. CRISPR/Cas9-mediated genomic deletion of the Fgf8 RARE in mouse embryos often resulted in a small somite defect with Fgf8 derepression caudally, but no defect was observed in heart development or heart Fgf8 expression. This suggests the existence of another DNA element whose function overlaps with the Fgf8 RARE to mediate Fgf8 repression by RA and NCOR1/2. Our studies support a model in which NCOR1/2 mediates direct RA-dependent repression of Fgf8 in caudal progenitors in order to control somitogenesis.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/genética , Organogénesis/genética , Somitos/anomalías , Tretinoina/metabolismo , Aldehído Oxidorreductasas/genética , Animales , Secuencia de Bases , Proteínas Co-Represoras/genética , Desarrollo Embrionario , Edición Génica/métodos , Corazón/embriología , Ratones , Ratones Noqueados , Transducción de Señal , Somitos/embriología
3.
Dev Biol ; 406(1): 63-73, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26165600

RESUMEN

It is known that secreted proteins from the anterior lateral endoderm, FGF8 and BMP2, are involved in mesodermal cardiac differentiation, which determines the first cardiac field, defined by the expression of the earliest specific cardiac markers Nkx-2.5 and Gata4. However, the molecular mechanisms responsible for early cardiac development still remain unclear. At present, microRNAs represent a novel layer of complexity in the regulatory networks controlling gene expression during cardiovascular development. This paper aims to study the role of miR130 during early cardiac specification. Our model is focused on developing chick at gastrula stages. In order to identify those regulatory factors which are involved in cardiac specification, we conducted gain- and loss-of-function experiments in precardiac cells by administration of Fgf8, Bmp2 and miR130, through in vitro electroporation technique and soaked beads application. Embryos were subjected to in situ hybridization, immunohistochemistry and qPCR procedures. Our results reveal that Fgf8 suppresses, while Bmp2 induces, the expression of Nkx-2.5 and Gata4. They also show that Fgf8 suppresses Bmp2, and vice versa. Additionally, we observed that Bmp2 regulates miR-130 -a putative microRNA that targets Erk1/2 (Mapk1) 3'UTR, recognizing its expression in precardiac cells which overlap with Erk1/2 pattern. Finally, we evidence that miR-130 is capable to inhibit Erk1/2 and Fgf8, resulting in an increase of Bmp2, Nkx-2.5 and Gata4. Our data present miR-130 as a necessary linkage in the control of Fgf8 signaling, mediated by Bmp2, establishing a negative feed-back loop responsible to achieve early cardiac specification.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Gástrula/embriología , Corazón/embriología , MicroARNs/genética , Animales , Proteína Morfogenética Ósea 2/biosíntesis , Proteína Morfogenética Ósea 2/genética , Embrión de Pollo , Electroporación , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/genética , Factor de Transcripción GATA4/biosíntesis , Inmunohistoquímica , Hibridación in Situ , Reacción en Cadena de la Polimerasa , Transducción de Señal/genética , Factores de Transcripción/biosíntesis
4.
J Biosci Bioeng ; 119(3): 260-6, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25239070

RESUMEN

Expression of the LIM homeodomain transcription factor Lhx8 is restricted to and up-regulated in the mesenchyme of the upper face prominence before lip fusion. Msx1/2 acts in early development to control cell proliferation and differentiation. Deficiency of these genes is associated with nonsyndromic cleft lip with/without cleft palate. Since retinoid is a potential patterning influence on the developing face, we have examined whether retinoic acid (RA) signaling regulated Lhx8, Msx1 and Msx2 transcription through fibroblast growth factor (FGF) signals in the maxillary prominence. Application of exogenous RA caused severe defects of the maxilla. Citral also induced a specific loss of derivatives from the maxillary prominences by blocking RA synthesis. Real-time RT-PCR and semi-quantitative RT-PCR analysis of the maxillary mesenchyme revealed that the expressions of Lhx8, Msx1 and Msx2 were significantly down-regulated by RA as well as by citral. The downregulated Lhx8 was rescued by combined treatment with FGF-8b, which indicated a downstream of RA signaling. FGF-8b induced up-regulated Lhx8 expression whereas SU5402, a pan-FGF family antagonist, down-regulated and caused defective maxillary morphogenesis and cleft lip. Our data suggest that Lhx8 is regulated by RA signaling through FGF signals and the level window of RA and FGF-8b could control the upper jaw morphogenesis.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/metabolismo , Maxilares/efectos de los fármacos , Maxilares/embriología , Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción/metabolismo , Tretinoina/farmacología , Monoterpenos Acíclicos , Animales , Diferenciación Celular/efectos de los fármacos , Embrión de Pollo , Regulación hacia Abajo/efectos de los fármacos , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/farmacología , Proteínas de Homeodominio/metabolismo , Maxilares/metabolismo , Factor de Transcripción MSX1/metabolismo , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Monoterpenos/farmacología , Morfogénesis/efectos de los fármacos , Pirroles/farmacología , Transducción de Señal/efectos de los fármacos
5.
Brain Res ; 1529: 39-45, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23891716

RESUMEN

Many neurons degenerate after injuries resulting from overstimulation, drugs, genetic mutations, and aging. Although several growth factors and neurotrophins delay degeneration and promote regrowth of neural processes, the role of fibroblast growth factor 8 (FGF8) in mammalian spiral ganglion neurons (SGN) neurite outgrowth has not been examined. This study develops and uses SGN cell cultures suitable for experimental analysis, it investigates whether FGF8a and FGF8b isoforms affect the neurite outgrowth from SGN cultured in vitro. We found that both FGF8a and FGF8b promoted the outgrowth of neurites from cultured SGN. This response is mediated by FGF receptors and involves the activation of IκBα-mediated NFκB signaling pathway. These findings suggest that, besides its morphogenetic role during development, FGF8 may have trophic functions in the adult which are relevant to regeneration.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/farmacología , Neuritas/efectos de los fármacos , Neuronas/citología , Ganglio Espiral de la Cóclea/citología , Análisis de Varianza , Animales , Animales Recién Nacidos , Células Cultivadas , Proteínas de Dominio Doblecortina , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Ratones , Ratones Endogámicos ICR , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Isoformas de Proteínas/farmacología , Pirimidinas/farmacología , Tubulina (Proteína)/metabolismo
6.
Mol Cancer Ther ; 10(9): 1600-10, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21764903

RESUMEN

Fibroblast growth factor-8b (FGF8b) exerts nonredundant autocrine/paracrine functions in steroid hormone-regulated tumors. Previous observations had shown that the soluble pattern recognition receptor long pentraxin-3 (PTX3) is a natural selective antagonist for a restricted number of FGF family members, inhibiting FGF2 but not FGF1 and FGF4 activity. Here, we assessed the capacity of PTX3 to antagonize FGF8b and to inhibit the vascularization and growth of steroid hormone-regulated tumors. Surface plasmon resonance analysis shows that PTX3 binds FGF8b with high affinity (K(d) = 30-90 nmol/L). As a consequence, PTX3 prevents the binding of FGF8b to its receptors, inhibits FGF8b-driven ERK1/2 activation, cell proliferation, and chemotaxis in endothelial cells, and suppresses FGF8b-induced neovascularization in vivo. Also, PTX3 inhibits dihydrotestosterone (DHT)- and FGF8b-driven proliferation of androgen-regulated Shionogi 115 (S115) mouse breast tumor cells. Furthermore, DHT-treated, PTX3 overexpressing hPTX3_S115 cell transfectants show a reduced proliferation rate in vitro and a limited angiogenic activity in the chick embryo chorioallantoic membrane and murine s.c. Matrigel plug assays. Accordingly, hPTX3_S115 cells show a dramatic decrease of their tumorigenic activity when grafted in immunodeficient male mice. These results identify PTX3 as a novel FGF8b antagonist endowed with antiangiogenic and antineoplastic activity with possible implications for the therapy of hormonal tumors.


Asunto(s)
Andrógenos/fisiología , Inhibidores de la Angiogénesis/farmacología , Proteína C-Reactiva/farmacología , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Componente Amiloide P Sérico/farmacología , Inhibidores de la Angiogénesis/metabolismo , Animales , Proteína C-Reactiva/genética , Proteína C-Reactiva/metabolismo , Células CHO , Bovinos , Línea Celular , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Cricetinae , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Neoplasias/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Unión Proteica , Componente Amiloide P Sérico/genética , Componente Amiloide P Sérico/metabolismo
7.
Oncogene ; 30(13): 1518-30, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21119603

RESUMEN

The fibroblast growth factor 8b (FGF8b) oncogene is known to be primarily involved in the tumorigenesis and progression of hormone-related cancers. Its role in other epithelial cancers has not been investigated, except for esophageal cancer, in which FGF8b overexpression was mainly found in tumor biopsies of male patients. These observations were consistent with previous findings in these cancer types that the male sex-hormone androgen is responsible for FGF8b expression. Nasopharyngeal carcinoma (NPC) is a highly metastatic cancer of head and neck commonly found in Asia. It is etiologically associated with Epstein-Barr Virus (EBV) infection, inflammatory tumor microenvironment and relatively higher male predominance. Here, we reported for the first time that FGF8b is overexpressed in this EBV-associated non-hormone-related cancer of the head and neck, NPC. More importantly, overexpression of FGF8b mRNA and protein was detected in a large majority of NPC tumors from both male and female genders, in addition to multiple NPC cell lines. We hypothesized that FGF8b overexpression may contribute to NPC tumorigenesis. Using EBV-associated NPC cell lines, we demonstrated that specific knockdown of FGF8b by small interfering RNA inhibited cell proliferation, migration and invasion, whereas exogenous FGF8b stimulated these multiple phenotypes. Further mechanistic investigation revealed that in addition to NF-κB signaling (a major inflammatory signaling pathway known to be activated in NPC), an important EBV oncoprotein, the latent membrane protein 1 (LMP1), was found to be a direct inducer of FGF8b overexpression in NPC cells, whereas androgen (testosterone) has minimal effect on FGF8b expression in EBV-associated NPC cells. In summary, our study has identified LMP1 as the first viral oncogene capable of directly inducing FGF8b (an important cellular oncogene) expression in human cancer cells. This novel mechanism of viral-mediated FGF8 upregulation may implicate a new role of oncoviruses in human carcinogenesis.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica , Herpesvirus Humano 4/patogenicidad , Oncogenes , Carcinoma , Movimiento Celular , Proliferación Celular , Femenino , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/genética , Humanos , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/fisiología , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/virología , Invasividad Neoplásica , ARN Mensajero/análisis , ARN Interferente Pequeño/genética , Proteínas de la Matriz Viral/fisiología
8.
Proc Natl Acad Sci U S A ; 106(37): 15774-9, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19720987

RESUMEN

Emergence of the neural crest (NC) is considered an essential asset in the evolution of the chordate phylum, as specific vertebrate traits such as peripheral nervous system, cephalic skeletal tissues, and head development are linked to the NC and its derivatives. It has been proposed that the emergence of the NC was responsible for the formation of a "new head" characterized by the spectacular development of the forebrain and associated sense organs. It was previously shown that removal of the cephalic NC (CNC) prevents the formation of the facial structures but also results in anencephaly. This article reports on the molecular mechanisms whereby the CNC controls cephalic neurulation and brain morphogenesis. This study demonstrates that molecular variations of Gremlin and Noggin level in CNC account for morphological changes in brain size and development. CNC cells act in these processes through a multi-step control and exert cumulative effects counteracting bone morphogenetic protein signaling produced by the neighboring tissues (e.g., adjacent neuroepithelium, ventro-medial mesoderm, superficial ectoderm). These data provide an explanation for the fact that acquisition of the NC during the protochordate-to-vertebrate transition has coincided with a large increase of brain vesicles.


Asunto(s)
Encéfalo/embriología , Cresta Neural/embriología , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 7/genética , Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Embrión de Pollo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Modelos Neurológicos , Cresta Neural/metabolismo , Cresta Neural/cirugía , Codorniz , Interferencia de ARN
9.
Int J Vitam Nutr Res ; 78(1): 21-6, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18654950

RESUMEN

SC-3 cells, an androgen-dependent mouse mammary carcinoma cell line, in response to androgen stimuli, induces the secretion of fibroblast growth factor (FGF-8), which in turn increases the proliferation of these cells. We have shown previously that methylcobalamin (MeCbl) decreases the levels of FGF-8 mRNA in SC-3 cells stimulated by testosterone, inhibiting the proliferation of SC-3 cells and inducing apoptosis. In the present study, we analyzed the effects of MeCbl on SC-3 cell proliferation in response to exogenous addition of FGF-8. Thymidine incorporation showed a significant decrease in SC-3 cells cultured with MeCbl. Immunocytochemistry for single-stranded DNA (ssDNA) and DNA fragmentation analysis demonstrated that MeCbl induced apoptosis in SC-3 cells, even in the presence of FGF-8. These results show that the addition of FGF-8 stimulates the proliferation of SC-3 cells under the androgen-depleted condition and that MeCbl might be able to interfere with FGF-8 action.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor 8 de Crecimiento de Fibroblastos/farmacología , Inhibidores de Crecimiento/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Vitamina B 12/análogos & derivados , Animales , División Celular , ADN/efectos de los fármacos , ADN/metabolismo , Fragmentación del ADN/efectos de los fármacos , Electroforesis en Gel de Agar , Femenino , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Timidina/metabolismo , Células Tumorales Cultivadas , Vitamina B 12/farmacología
10.
Dev Dyn ; 237(12): 3516-27, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18553642

RESUMEN

In vertebrate gastrula/neurula embryos, a cilia-driven leftward flow asymmetrically activates the Nodal cascade in the left lateral plate mesoderm (LPM). In frog embryos left-right axis formation was postulated to depend on gap junctions (GJs) during cleavage. Here, we show that GJs cooperate with fibroblast growth factor-8 (FGF8) to specify asymmetric Nodal in the rabbit embryo at gastrula/neurula. GJs and FGF signaling were manipulated in whole embryo and explant cultures of rabbit blastodiscs. These experiments demonstrate that right-sided inhibition of Nodal by FGF8 depended on intercellular communication by means of GJs, and that left-sided induction of Nodal required attenuation of gap junctional communication (GJC). Before flow, the left and right side were equally competent but actively prevented from Nodal induction through FGF8/GJ. Our data suggest that flow unilaterally attenuates FGF8/GJ-mediated repression of Nodal on the left side, integrating GJC and FGF8 into the flow-based mechanism of symmetry breakage in the vertebrate embryo.


Asunto(s)
Tipificación del Cuerpo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Uniones Comunicantes/metabolismo , Proteína Nodal/metabolismo , Animales , Conexina 43/metabolismo , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Células Epiteliales/metabolismo , Femenino , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Heptanol/farmacología , Mesodermo/embriología , Mesodermo/metabolismo , Conejos
11.
Lancet Oncol ; 8(10): 933-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17913662

RESUMEN

Many attempts have been made to derive genetic signatures for progressive prostate cancer for both prognostic and therapeutic purposes. These investigations have resulted in the discovery of many pathways, but the signatures exhibit heterogeneity and restricted reproducibility. A thorough and disciplined analysis of genes with androgen-response elements that are expressed in progressive, castration-resistant prostate cancer is an integral step towards the development of new therapeutic or diagnostic targets. We discuss the effects of bona-fide downstream targets of the androgen receptor on cellular proliferation, evasion of apoptosis, and angiogenesis, and consider the clinical potential of these targets.


Asunto(s)
Andrógenos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Elementos de Respuesta/fisiología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/antagonistas & inhibidores , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/fisiología , Neoplasias de la Próstata/patología , Receptor PAR-1/antagonistas & inhibidores , Receptor PAR-1/fisiología , Receptores Androgénicos/fisiología
12.
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
13.
Dev Biol ; 306(1): 170-8, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17442299

RESUMEN

Anuran (frog) tadpoles and urodeles (newts and salamanders) are the only vertebrates capable of fully regenerating amputated limbs. During the early stages of regeneration these amphibians form a "blastema", a group of mesenchymal progenitor cells that specifically directs the regrowth of the limb. We report that wnt-3a is expressed in the apical epithelium of regenerating Xenopus laevis limb buds, at the appropriate time and place to play a role during blastema formation. To test whether Wnt/beta-catenin signaling is required for limb regeneration, we created transgenic X. laevis tadpoles that express Dickkopf-1 (Dkk1), a specific inhibitor of Wnt/beta-catenin signaling, under the control of a heat-shock promoter. Heat-shock immediately before limb amputation or during early blastema formation blocked limb regeneration but did not affect the development of contralateral, un-amputated limb buds. When the transgenic tadpoles were heat-shocked following the formation of a blastema, however, they retained the ability to regenerate partial hindlimb structures. Furthermore, heat-shock induced Dkk1 blocked fgf-8 but not fgf-10 expression in the blastema. We conclude that Wnt/beta-catenin signaling has an essential role during the early stages of limb regeneration, but is not absolutely required after blastema formation.


Asunto(s)
Extremidades/fisiología , Regeneración , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Animales Modificados Genéticamente , Factor 10 de Crecimiento de Fibroblastos/análisis , Factor 10 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 8 de Crecimiento de Fibroblastos/análisis , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Esbozos de los Miembros , Regiones Promotoras Genéticas , Transducción de Señal , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo , Proteína Wnt3 , Proteína Wnt3A , Proteínas de Xenopus/genética , Xenopus laevis , beta Catenina/antagonistas & inhibidores , beta Catenina/metabolismo
14.
Dev Dyn ; 236(2): 564-71, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17195184

RESUMEN

In vertebrates, cranial sensory ganglia are mainly derived from ectodermal placodes, which are focal thickenings at characteristic positions in the embryonic head. Here, we provide the first description of the early development of the epibranchial placode in zebrafish embryos using sox3 as a molecular marker. By the one-somite stage, we saw a pair of single sox3-expressing domains appear lateral to the future hindbrain. The sox3 domain, which is referred to here as the early lateral placode, is segregated during the early phase of segmentation to form a pax2a-positive medial area and a pax2a-negative lateral area. The medial area subsequently developed to form the otic placode, while the lateral area was further segregated along the anteroposterior axis, giving rise to four sox3-positive subdomains by 26 hr postfertilization. Given their spatial relationship with the expression of the markers for the epibranchial ganglion, as well as their positions and temporal changes, we propose that these four domains correspond to the facial, glossopharyngeal, vagal, and posterior lateral line placodes in an anterior-to-posterior order. The expression of sox3 in the early lateral placode was absent in mutants lacking functional fgf8, while implantation of fibroblast growth factor (FGF) beads restored the sox3 expression. Using SU5402, which inhibits the FGF signal, we were able to demonstrate that formation of both the early lateral domains and later epibranchial placodes depends on the FGF signal operating at the beginning of somitogenesis. Together, these data provide evidence for the essential role of FGF signals in the development of the epibranchial placodes.


Asunto(s)
Tipificación del Cuerpo/fisiología , Diferenciación Celular/fisiología , Ectodermo/fisiología , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Ganglios Sensoriales/embriología , Transducción de Señal/fisiología , Pez Cebra/embriología , Animales , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/fisiología , Proteínas del Grupo de Alta Movilidad/metabolismo , Hibridación in Situ , Pirroles/farmacología , Factores de Transcripción SOXB1 , Factores de Transcripción/metabolismo
15.
Development ; 133(16): 3265-76, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16873584

RESUMEN

We have used the maternal effect mutant ichabod, which is deficient in maternal beta-catenin signaling, to test for the epistatic relationship between beta-catenin activation, FGF signaling and bozozok, squint and chordin expression. Injection of beta-catenin RNA into ichabod embryos can completely rescue normal development. By contrast, when FGF signaling is inhibited, beta-catenin did not induce goosecoid and chordin, repress bmp4 expression or induce a dorsal axis. These results demonstrate that FGF signaling is necessary for beta-catenin induction of the zebrafish organizer. We show that FGFs function downstream of squint and bozozok to turn on chordin expression. Full rescue of ichabod by Squint is dependent on FGF signaling, and partial rescue by FGFs is completely dependent on chordin. By contrast, Bozozok can rescue the complete anteroposterior axis, but not notochord, in embryos blocked in FGF signaling. Surprisingly, accumulation of bozozok transcript in beta-catenin RNA-injected ichabod embryos is also dependent on FGF signaling, indicating a role of FGFs in maintenance of bozozok RNA. These experiments show that FGF-dependent organizer function operates through both bozozok RNA accumulation and a pathway consisting of beta-catenin-->Squint-->FGF-->Chordin, in which each component is sufficient for expression of the downstream factors of the pathway, and in which Nodal signaling is required for FGF gene expression and FGF signaling is required for Squint induction of chordin.


Asunto(s)
Factor 3 de Crecimiento de Fibroblastos/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Organizadores Embrionarios/metabolismo , Pez Cebra/embriología , beta Catenina/metabolismo , Animales , Tipificación del Cuerpo , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/metabolismo , Embrión no Mamífero/metabolismo , Factor 3 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 3 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/genética , Glicoproteínas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ligandos de Señalización Nodal , Transducción de Señal , Transcripción Genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , beta Catenina/agonistas
16.
Dev Biol ; 295(2): 486-97, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16765936

RESUMEN

Morphogenesis of the cardiac arterial pole is dependent on addition of myocardium and smooth muscle from the secondary heart field and septation by cardiac neural crest cells. Cardiac neural crest ablation results in persistent truncus arteriosus and failure of addition of myocardium from the secondary heart field leading to malalignment of the arterial pole with the ventricles. Previously, we have shown that elevated FGF signaling after neural crest ablation causes depressed Ca2+ transients in the primary heart tube. We hypothesized that neural crest ablation results in elevated FGF8 signaling in the caudal pharynx that disrupts secondary heart field development. In this study, we show that FGF8 signaling is elevated in the caudal pharynx after cardiac neural crest ablation. In addition, treatment of cardiac neural crest-ablated embryos with FGF8b blocking antibody or an FGF receptor blocker rescues secondary heart field myocardial development in a time- and dose-dependent manner. Interestingly, reduction of FGF8 signaling in normal embryos disrupts myocardial secondary heart field development, resulting in arterial pole malalignment. These results indicate that the secondary heart field myocardium is particularly sensitive to FGF8 signaling for normal conotruncal development, and further, that cardiac neural crest cells modulate FGF8 signaling in the caudal pharynx.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/fisiología , Corazón/embriología , Morfogénesis , Faringe/embriología , Transducción de Señal , Animales , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Embrión de Mamíferos , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Corazón/crecimiento & desarrollo , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/etiología , Ratones , Cresta Neural/anomalías , Faringe/metabolismo , Tronco Arterial Persistente/embriología , Tronco Arterial Persistente/etiología
17.
Mol Biol Cell ; 17(7): 3075-84, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16641368

RESUMEN

FGF8, a member of the fibroblast growth factor (FGF) family, has been shown to play important roles in different developing systems. Mouse embryonic carcinoma P19 cells could be induced by retinoic acid (RA) to differentiate into neuroectodermal cell lineages, and this process is cell aggregation dependent. In this report, we show that FGF8 expression is transiently up-regulated upon P19 cell aggregation, and the aggregation-dependent FGF8 elevation is pluripotent stem cell related. Overexpressing FGF8 promotes RA-induced monolayer P19 cell neural differentiation. Inhibition of FGF8 expression by RNA interference or blocking FGF signaling by the FGF receptor inhibitor, SU5402, attenuates neural differentiation of the P19 cell. Blocking the bone morphogenetic protein (BMP) pathway by overexpressing Smad6 in P19 cells, we also show that FGF signaling plays a BMP inhibition-independent role in P19 cell neural differentiation.


Asunto(s)
Diferenciación Celular , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Neuronas/citología , Células Madre Pluripotentes/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Carcinoma Embrionario , Agregación Celular , Línea Celular Tumoral , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/genética , Ratones , Neuronas/metabolismo , Células Madre Pluripotentes/citología , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal , Proteína smad6/metabolismo , Regulación hacia Arriba
18.
Dev Biol ; 292(1): 79-89, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16480709

RESUMEN

Tetrapod limbs, forelimbs and hindlimbs, emerge as limb buds during development from appropriate positions along the rostro-caudal axis of the main body. In this study, tissue interactions by which rostro-caudal level-specific limb initiation is established were analyzed. The limb bud originates from the lateral plate located laterally to the paraxial mesoderm, and we obtained evidence that level-specific tissue interactions between the paraxial mesoderm and the lateral plate mesoderm are important for the determination of the limb-type-specific gene expression and limb outgrowth. When the wing-level paraxial mesoderm was transplanted into the presumptive leg region, the wing-level paraxial mesoderm upregulated the expression of Tbx5, a wing marker gene, and down regulated the expression of Tbx4 and Pitx1, leg marker genes, in the leg-level lateral plate. The wing-level paraxial mesoderm relocated into the leg level also inhibited outgrowth of the hindlimb bud and down regulated Fgf10 and Fgf8 expression, demonstrating that the wing-level paraxial mesoderm cannot substitute for the function of the leg-level paraxial mesoderm in initiation and outgrowth of the hindlimb. The paraxial mesoderm taken from the neck- and flank-level regions also had effects on Tbx5/Tbx4 expression with different efficiencies. These findings suggest that the paraxial mesoderm has level-specific abilities along the rostro-caudal axis in the limb-type-specific mechanism for limb initiation.


Asunto(s)
Proteínas Aviares/biosíntesis , Extremidad Inferior/embriología , Mesodermo/fisiología , Proteínas de Dominio T Box/biosíntesis , Alas de Animales/embriología , Animales , Proteínas Aviares/genética , Embrión de Pollo , Regulación hacia Abajo/genética , Factor 10 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 8 de Crecimiento de Fibroblastos/genética , Miembro Posterior/embriología , Miembro Posterior/crecimiento & desarrollo , Extremidad Inferior/crecimiento & desarrollo , Mesodermo/trasplante , Técnicas de Cultivo de Órganos , Factores de Transcripción Paired Box/antagonistas & inhibidores , Factores de Transcripción Paired Box/genética , Proteínas de Dominio T Box/genética , Alas de Animales/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...