Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Development ; 147(17)2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32778568

RESUMEN

The role played by the Notch pathway in cardiac progenitor cell biology remains to be elucidated. Delta-like ligand 4 (Dll4), the arterial-specific Notch ligand, is expressed by second heart field (SHF) progenitors at time-points that are crucial in SHF biology. Dll4-mediated Notch signaling is required for maintaining an adequate pool of SHF progenitors, such that Dll4 knockout results in a reduction in proliferation and an increase in apoptosis. A reduced SHF progenitor pool leads to an underdeveloped right ventricle (RV) and outflow tract (OFT). In its most severe form, there is severe RV hypoplasia and poorly developed OFT resulting in early embryonic lethality. In its milder form, the OFT is foreshortened and misaligned, resulting in a double outlet right ventricle. Dll4-mediated Notch signaling maintains Fgf8 expression by transcriptional regulation at the promoter level. Combined heterozygous knockout of Dll4 and Fgf8 demonstrates genetic synergy in OFT alignment. Exogenous supplemental Fgf8 rescues proliferation in Dll4 mutants in ex-vivo culture. Our results establish a novel role for Dll4-mediated Notch signaling in SHF biology. More broadly, our model provides a platform for understanding oligogenic inheritance that results in clinically relevant OFT malformations.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proliferación Celular , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Ventrículos Cardíacos/embriología , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Unión al Calcio/genética , Factor 8 de Crecimiento de Fibroblastos/genética , Ratones , Ratones Noqueados , Receptores Notch/genética
2.
J Pediatr Urol ; 16(1): 41.e1-41.e10, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31718875

RESUMEN

INTRODUCTION: Fibroblast growth factors (FGFs) play a crucial role in early embryogenesis of the genital tubercle and are involved in the development of hypospadias, affecting both endo- and ectodermally derived tissues. It was hypothesized that expression of FGFs could be qualitatively or quantitatively altered in skin of children with hypospadias. OBJECTIVE: The objective of the study was to investigate expression patterns and transcription levels of FGF8, FGF10, and FGF Receptor 2 (FGFR2) in patients with hypospadias compared to normal controls. PATIENTS AND METHODS: Skin samples from the ventro-lateral aspect of the foreskin of 32 patients with hypospadias (17 distal and 15 proximal, mean age 25 months) and 10 normal foreskin samples (mean age 77 months) were analyzed by immunohistochemistry. Staining, localization, and distribution of positive cells in epidermis and dermis were categorized independently by two researchers. Complementary DNA (cDNA) samples prepared from messenger RNA (mRNA) isolates of the same samples were analyzed by quantitative polymerase chain reaction (qPCR), comparing expressions of FGF8, FGF10, and FGFR2 with loading controls. RESULTS: Patients with hypospadias consistently showed aberrant immunohistochemical staining patterns for FGF8/FGF10/FGFR2 in epidermis and dermis compared to patients without penile malformation (p < 0.01 for all markers). qPCR displayed no difference in expression levels on mRNA level (FGFR2 p = 0.44, FGF8 p = 0.77, and FGF10 p = 0.17) comparing normal foreskin with foreskin from patients with hypospadias. Figure. DISCUSSION: The results point at an impact of FGF signaling during embryological development of hypospadias on skin, as an ectodermally derived tissue. Similar to the urethral development, this might be a result of mesothelial-epithelial interactions. The differing expression patterns in immunohistochemistry are not matched by a quantitative difference in marker expression on the mRNA level, putatively caused by post-translational modifications or alterations of the downstream pathway. FGFs, particularly FGF10 and FGFR2, are critically involved in wound healing. CONCLUSIONS: There are significant differences in localization and distribution of FGF8, FGF10, and FGFR2 in comparisons of normal foreskin to foreskin of patients with hypospadias, whereas there is no difference in the quantitative expression of these markers on the mRNA level. This confirms the notion that penile skin is affected as well by the embryological aberrations during the embryogenesis of hypospadias.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/biosíntesis , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/genética , Prepucio/metabolismo , Hipospadias/genética , Hipospadias/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Transcripción Genética , Niño , Preescolar , Factor 10 de Crecimiento de Fibroblastos/análisis , Factor 8 de Crecimiento de Fibroblastos/análisis , Prepucio/química , Expresión Génica , Humanos , Hipospadias/patología , Inmunohistoquímica , Lactante , Masculino , Estudios Prospectivos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/análisis
3.
Cells ; 8(9)2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31527546

RESUMEN

Even though distinctive advances in the field of esophageal cancer therapy have occurred over the last few years, patients' survival rates remain poor. FGF8, FGF18, and FGFR4 have been identified as promising biomarkers in a number of cancers; however no data exist on expression of FGF8, FGF18, and FGFR4 in adenocarcinomas of the esophago-gastric junction (AEG). A preliminary analysis of the Cancer Genome Atlas (TCGA) database on FGF8, FGF18, and FGFR4 mRNA expression data of patients with AEG was performed. Furthermore, protein levels of FGF8, FGF18, and FGFR4 in diagnostic biopsies and post-operative specimens in neoadjuvantly treated and primarily resected patients using immunohistochemistry were investigated. A total of 242 patients was analyzed in this study: 87 patients were investigated in the TCGA data set analysis and 155 patients in the analysis of protein expression using immunohistochemistry. High protein levels of FGF8, FGF18, and FGFR4 were detected in 94 (60.7%), 49 (31.6%) and 84 (54.2%) patients, respectively. Multivariable Cox proportional hazard regression models revealed that high expression of FGF8 was an independent prognostic factor for diminished overall survival for all patients and for neoadjuvantly treated patients. By contrast, FGF18 overexpression was significantly associated with longer survival rates in neoadjuvantly treated patients. In addition, FGF8 protein level correlated with Mandard regression due to neoadjuvant therapy, indicating potential as a predictive marker. In summary, FGF8 and FGF18 are promising candidates for prognostic factors in adenocarcinomas of the esophago-gastric junction and new potential targets for new anti-cancer therapies.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Esofágicas/metabolismo , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factores de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Adenocarcinoma/diagnóstico , Adenocarcinoma/patología , Anciano , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/patología , Femenino , Factor 8 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/genética , Humanos , Masculino , Persona de Mediana Edad , Análisis Multivariante , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Tasa de Supervivencia
4.
J Comp Neurol ; 527(9): 1461-1477, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30689213

RESUMEN

In the mouse, two telencephalic signaling centers orchestrate embryonic patterning of the cerebral cortex. From the rostral patterning center in the telencephalon, the Fibroblast Growth Factor, FGF8, disperses as a morphogen to establish the rostral to caudal axis of the neocortical area map. FGF8 coordinates with Wnt3a from the cortical hem to regulate graded expression of transcription factors that position neocortical areas, and control hippocampal development. Whether similar signaling centers pattern the much larger cortices of carnivore and primate species, however, is unclear. The limited dispersion range of FGF8 and Wnt3a is inconsistent with patterning larger cortical primordia. Yet the implication that different mechanisms organize cortex in different mammals flies in the face of the tenet that developmental patterning mechanisms are conserved across vertebrate species. In the present study, both signaling centers were identified in the ferret telencephalon, as were expression gradients of the patterning transcription factor genes regulated by FGF8 and Wnt3a. Notably, at the stage corresponding to the peak period of FGF8 signaling in the mouse neocortical primordium (NP), the NP was the same size in ferret and mouse, which would allow morphogen patterning of the ferret NP. Subsequently, the size of ferret neocortex shot past that of the mouse. Images from online databases further suggest that NP growth in humans, too, is slowed in early cortical development. We propose that if early growth in larger brains is held back, mechanisms that pattern the neocortical area map in the mouse could be conserved across mammalian species.


Asunto(s)
Hurones/embriología , Lisencefalia/embriología , Neocórtex/embriología , Animales , Femenino , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Humanos , Hibridación in Situ , Lisencefalia/patología , Masculino , Ratones , Modelos Animales , Modelos Neurológicos , Neocórtex/patología , Tamaño de los Órganos , Organogénesis , Transducción de Señal/fisiología , Somitos/ultraestructura , Especificidad de la Especie , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Proteína Wnt3A/biosíntesis , Proteína Wnt3A/genética , Proteína Wnt3A/fisiología
5.
J Biol Chem ; 292(25): 10520-10533, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28461333

RESUMEN

Amputation of the proximal region in mammals is not followed by regeneration because blastema cells (BCs) and expression of regenerative genes, such as Msh homeobox (Msx) genes, are absent in this animal group. The lack of BCs and positional information in other cells is therefore the main obstacle to therapeutic approaches for limb regeneration. Hence, this study aimed to create blastema-like cells (BlCs) by overexpressing Msx1 and Msx2 genes in mouse bone marrow-derived mesenchymal stem cells (mBMSCs) to regenerate a proximally amputated digit tip. We transduced mBMSCs with Msx1 and Msx2 genes and compared osteogenic activity and expression levels of several Msx-regulated genes (Bmp4, Fgf8, and keratin 14 (K14)) in BlC groups, including MSX1, MSX2, and MSX1/2 (in a 1:1 ratio) with those in mBMSCs and BCs in vitro and in vivo following injection into the amputation site. We found that Msx gene overexpression increased expression of specific blastemal markers and enhanced the proliferation rate and osteogenesis of BlCs compared with mBMSCs and BCs via activation of Fgf8 and Bmp4 Histological analyses indicated full regrowth of digit tips in the Msx-overexpressing groups, particularly in MSX1/2, through endochondral ossification 6 weeks post-injection. In contrast, mBMSCs and BCs formed abnormal bone and nail. Full digit tip was regenerated only in the MSX1/2 group and was related to boosted Bmp4, Fgf8, and K14 gene expression and to limb-patterning properties resulting from Msx1 and Msx2 overexpression. We propose that Msx-transduced cells that can regenerate epithelial and mesenchymal tissues may potentially be utilized in limb regeneration.


Asunto(s)
Células de la Médula Ósea/metabolismo , Miembro Posterior/fisiología , Proteínas de Homeodominio/biosíntesis , Factor de Transcripción MSX1/biosíntesis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Regeneración , Aloinjertos , Animales , Proteína Morfogenética Ósea 4/biosíntesis , Proteína Morfogenética Ósea 4/genética , Proliferación Celular/genética , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/genética , Proteínas de Homeodominio/genética , Queratina-14/biosíntesis , Queratina-14/genética , Factor de Transcripción MSX1/genética , Ratones , Transducción Genética
6.
Dev Growth Differ ; 58(5): 437-45, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27273073

RESUMEN

In this paper, we review how midbrain and hindbrain are specified. Otx2 and Gbx2 are expressed from the early phase of development, and their expression abuts at the midbrain hindbrain boundary (MHB), where Fgf8 expression is induced, and functions as an organizing molecule for the midbrain and hindbrain. Fgf8 induces En1 and Pax2 expression at the region where Otx2 is expressed to specify midbrain. Fgf8 activates Ras-ERK pathway to specify hindbrain. Downstream of ERK, Pea3 specifies isthmus (rhombomere 0, r0), and Irx2 may specify r1, where the cerebellum is formed.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Regulación del Desarrollo de la Expresión Génica/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Mesencéfalo/embriología , Rombencéfalo/embriología , Animales , Factor 8 de Crecimiento de Fibroblastos/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX2/metabolismo
7.
Appl Microbiol Biotechnol ; 100(2): 625-35, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26411459

RESUMEN

Human fibroblast growth factor 8b (FGF8b) was expressed based on a baculovirus expression vector system (BEVS) and identified as having a protective effect on Parkinson's disease. Immunoblotting demonstrated that rhFGF8b proteins were recognized by a human anti-FGF8b antibody. The multiplicity of infection and timing of harvest had a significant effect on protein yield and protein quality. Our results indicated that the rhFGF8b was first detectable at 36 h postinfection and reached a maximum at 60 h. A multiplicity of infection (MOI) of 8 pfu/mL was suitable for harvest. The target protein was purified by heparin-affinity chromatography. In vitro methylthiazol tetrazolium (MTT) assays demonstrated that the purified rhFGF8b could significantly stimulate proliferation of NIH3T3 cells. Furthermore, to elucidate the effect of rhFGF8b on Parkinson's disease, we used FGF8b pretreatment on a cell model of Parkinson's disease. The results indicated that rhFGF8b prevented necrosis and apoptosis of 1-METHYL-4-phenyl pyridine (MPP(+)) treated PC12 cells. Moreover, the effect of FGF8b on messenger RNA (mRNA) levels of apoptosis and ERS genes was investigated to clarify the molecular mechanisms of FGF8b. The results suggest that FGF8b exerts neuroprotective effects by alleviating endoplasmic reticulum (ER) stress during PD. These results suggest that FGF8b may be a promising candidate therapeutic drug for neurodegenerative diseases related to ER stress.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Apoptosis/efectos de los fármacos , Baculoviridae/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromatografía de Afinidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/aislamiento & purificación , Humanos , Ratones , Células 3T3 NIH , Fármacos Neuroprotectores/aislamiento & purificación , Células PC12 , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Sales de Tetrazolio , Tiazoles
8.
Hum Mol Genet ; 24(15): 4185-97, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25911675

RESUMEN

The p63 transcription factor, homolog to the p53 tumor suppressor gene, plays a crucial role in epidermal and limb development, as its mutations are associated to human congenital syndromes characterized by skin, craniofacial and limb defects. While limb and skin-specific p63 transcriptional targets are being discovered, little is known of the post-translation modifications controlling ΔNp63α functions. Here we show that the p300 acetyl-transferase physically interacts in vivo with ΔNp63α and catalyzes its acetylation on lysine 193 (K193) inducing ΔNp63α stabilization and activating specific transcriptional functions. Furthermore we show that Fibroblast Growth Factor-8 (FGF8), a morphogenetic signaling molecule essential for embryonic limb development, increases the binding of ΔNp63α to the tyrosine kinase c-Abl as well as the levels of ΔNp63α acetylation. Notably, the natural mutant ΔNp63α-K193E, associated to the Split-Hand/Foot Malformation-IV syndrome, cannot be acetylated by this pathway. This mutant ΔNp63α protein displays promoter-specific loss of DNA binding activity and consequent altered expression of development-associated ΔNp63α target genes. Our results link FGF8, c-Abl and p300 in a regulatory pathway that controls ΔNp63α protein stability and transcriptional activity. Hence, limb malformation-causing p63 mutations, such as the K193E mutation, are likely to result in aberrant limb development via the combined action of altered protein stability and altered promoter occupancy.


Asunto(s)
Anomalías Congénitas/genética , Factor 8 de Crecimiento de Fibroblastos/genética , Proteínas Proto-Oncogénicas c-abl/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Factores de Transcripción p300-CBP/genética , Animales , Línea Celular , Anomalías Congénitas/embriología , Anomalías Congénitas/patología , Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Deformidades Congénitas de las Extremidades/genética , Deformidades Congénitas de las Extremidades/patología , Ratones , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-abl/biosíntesis , Proteínas Proto-Oncogénicas c-abl/metabolismo , Transducción de Señal , Factores de Transcripción/biosíntesis , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/metabolismo , Factores de Transcripción p300-CBP/biosíntesis , Factores de Transcripción p300-CBP/metabolismo
9.
Dev Dyn ; 244(6): 797-807, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25809880

RESUMEN

BACKGROUND: Vertebrate body axis extension occurs in a head-to-tail direction from a caudal progenitor zone that responds to interacting signals. Wnt/ß-catenin signaling is critical for generation of paraxial mesoderm, somite formation, and maintenance of the axial stem cell pool. Body axis extension requires Wnt8a in lower vertebrates, but in mammals Wnt3a is required, although the anterior trunk develops in the absence of Wnt3a. RESULTS: We examined mouse Wnt8a(-/-) and Wnt3a(-/-) single and double mutants to explore whether mammalian Wnt8a contributes to body axis extension and to determine whether a posterior growth function for Wnt8a is conserved throughout the vertebrate lineage. We find that caudal Wnt8a is expressed only during early somite stages and is required for normal development of the anterior trunk in the absence of Wnt3a. During this time, we show that Wnt8a and Wnt3a cooperate to maintain Fgf8 expression and prevent premature Sox2 up-regulation in the axial stem cell niche, critical for posterior growth. Similar to Fgf8, Wnt8a requires retinoic acid (RA) signaling to restrict its caudal expression boundary and possesses an upstream RA response element that binds RA receptors. CONCLUSIONS: These findings provide new insight into interaction of caudal Wnt-FGF-RA signals required for body axis extension.


Asunto(s)
Tipificación del Cuerpo/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Nicho de Células Madre/fisiología , Proteína Wnt3A/fisiología , Anomalías Múltiples/embriología , Anomalías Múltiples/genética , Oxidorreductasas de Alcohol/deficiencia , Oxidorreductasas de Alcohol/genética , Animales , Tipificación del Cuerpo/genética , Secuencia Conservada , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/genética , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados , Fenotipo , Receptores de Ácido Retinoico/fisiología , Elementos de Respuesta/genética , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción SOXB1/genética , Transducción de Señal/fisiología , Somitos/crecimiento & desarrollo , Somitos/metabolismo , Tretinoina/farmacología , Vertebrados/embriología , Proteínas Wnt , Proteína Wnt3A/deficiencia , Proteína Wnt3A/genética
10.
Dev Biol ; 387(1): 37-48, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24424161

RESUMEN

Isl1 expression marks progenitor populations in developing embryos. In this study, we investigated the contribution of Isl1-expressing cells that utilize the ß-catenin pathway to skeletal development. Inactivation of ß-catenin in Isl1-expressing cells caused agenesis of the hindlimb skeleton and absence of the lower jaw (agnathia). In the hindlimb, Isl1-lineages broadly contributed to the mesenchyme; however, deletion of ß-catenin in the Isl1-lineage caused cell death only in a discrete posterior domain of nascent hindlimb bud mesenchyme. We found that the loss of posterior mesenchyme, which gives rise to Shh-expressing posterior organizer tissue, caused loss of posterior gene expression and failure to expand chondrogenic precursor cells, leading to severe truncation of the hindlimb. In facial tissues, Isl1-expressing cells broadly contributed to facial epithelium. We found reduced nuclear ß-catenin accumulation and loss of Fgf8 expression in mandibular epithelium of Isl1(-/-) embryos. Inactivating ß-catenin in Isl1-expressing epithelium caused both loss of epithelial Fgf8 expression and death of mesenchymal cells in the mandibular arch without affecting epithelial proliferation and survival. These results suggest a Isl1→ß-catenin→Fgf8 pathway that regulates mesenchymal survival and development of the lower jaw in the mandibular epithelium. By contrast, activating ß-catenin signaling in Isl1-lineages caused activation of Fgf8 broadly in facial epithelium. Our results provide evidence that, despite its broad contribution to hindlimb mesenchyme and facial epithelium, the Isl1-ß-catenin pathway regulates skeletal development of the hindlimb and lower jaw through discrete populations of cells that give rise to Shh-expressing posterior hindlimb mesenchyme and Fgf8-expressing mandibular epithelium.


Asunto(s)
Miembro Posterior/embriología , Anomalías Maxilomandibulares/embriología , Proteínas con Homeodominio LIM/metabolismo , Osteogénesis/genética , Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Región Branquial/embriología , Linaje de la Célula/genética , Proliferación Celular , Supervivencia Celular , Regulación hacia Abajo , Fosfatasa 6 de Especificidad Dual/biosíntesis , Embrión de Mamíferos/metabolismo , Epitelio/embriología , Epitelio/metabolismo , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/deficiencia , Factor 8 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Miembro Posterior/anomalías , Proteínas de Homeodominio/biosíntesis , Anomalías Maxilomandibulares/genética , Factores de Transcripción de Tipo Kruppel/biosíntesis , Proteínas con Homeodominio LIM/genética , Mandíbula/embriología , Mesodermo/embriología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/biosíntesis , Transducción de Señal/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Regulación hacia Arriba , Proteína Gli3 con Dedos de Zinc , beta Catenina/genética
11.
Dev Biol ; 385(1): 41-51, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24479159

RESUMEN

Anorectal malformation (ARM) is a common birth defect but the developmental history and the underlying molecular mechanism are poorly understood. Using murine genetic models, we report here that a signaling molecule Dickkopf-1 (Dkk1) is a critical regulator. The anorectal and genitourinary tracts are major derivatives of caudal hindgut, or the cloaca.Dkk1 is highly expressed in the dorsal peri-cloacal mesenchymal (dPCM) progenitors. We show that the deletion of Dkk1 causes the imperforate anus with rectourinary fistula. Mutant genital tubercles exhibit a preputial hypospadias phenotype and premature urethral canalization.Dkk1 mutants have an ectopic expansion of the dPCM tissue, which correlates with an aberrant increase of cell proliferation and survival. This ectopic tissue is detectable before the earliest sign of the anus formation, suggesting that it is most likely the primary or early cause of the defect. Deletion of Dkk1 results in an elevation of the Wnt/ß-catenin activity. Signaling molecules Shh, Fgf8 and Bmp4 are also upregulated. Furthermore, genetic hyperactivation of Wnt/ß-catenin signal pathway in the cloacal mesenchyme partially recapitulates Dkk1 mutant phenotypes. Together, these findings underscore the importance ofDKK1 in regulating behavior of dPCM progenitors, and suggest that formation of anus and urethral depends on Dkk1-mediated dynamic inhibition of the canonical Wnt/ß-catenin signal pathway.


Asunto(s)
Canal Anal/embriología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mesodermo/embriología , Recto/embriología , Sistema Urogenital/embriología , Canal Anal/anomalías , Animales , Malformaciones Anorrectales , Ano Imperforado/embriología , Ano Imperforado/genética , Proteína Morfogenética Ósea 4/biosíntesis , Diferenciación Celular/genética , Proliferación Celular , Supervivencia Celular/genética , Activación Enzimática/genética , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Proteínas Hedgehog/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Recto/anomalías , Células Madre , Regulación hacia Arriba , Anomalías Urogenitales/embriología , Anomalías Urogenitales/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
12.
Development ; 141(1): 148-57, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24284205

RESUMEN

Non-canonical Wnt/planar cell polarity (PCP) signaling plays a primary role in the convergent extension that drives neural tube closure and body axis elongation. PCP signaling gene mutations cause severe neural tube defects (NTDs). However, the role of canonical Wnt/ß-catenin signaling in neural tube closure and NTDs remains poorly understood. This study shows that conditional gene targeting of ß-catenin in the dorsal neural folds of mouse embryos represses the expression of the homeobox-containing genes Pax3 and Cdx2 at the dorsal posterior neuropore (PNP), and subsequently diminishes the expression of the Wnt/ß-catenin signaling target genes T, Tbx6 and Fgf8 at the tail bud, leading to spina bifida aperta, caudal axis bending and tail truncation. We demonstrate that Pax3 and Cdx2 are novel downstream targets of Wnt/ß-catenin signaling. Transgenic activation of Pax3 cDNA can rescue the closure defect in the ß-catenin mutants, suggesting that Pax3 is a key downstream effector of ß-catenin signaling in the PNP closure process. Cdx2 is known to be crucial in posterior axis elongation and in neural tube closure. We found that Cdx2 expression is also repressed in the dorsal PNPs of Pax3-null embryos. However, the ectopically activated Pax3 in the ß-catenin mutants cannot restore Cdx2 mRNA in the dorsal PNP, suggesting that the presence of both ß-catenin and Pax3 is required for regional Cdx2 expression. Thus, ß-catenin signaling is required for caudal neural tube closure and elongation, acting through the transcriptional regulation of key target genes in the PNP.


Asunto(s)
Tipificación del Cuerpo/fisiología , Proteínas de Homeodominio/metabolismo , Tubo Neural/embriología , Factores de Transcripción Paired Box/metabolismo , Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Animales , Tipificación del Cuerpo/genética , Factor de Transcripción CDX2 , Adhesión Celular/genética , Polaridad Celular/fisiología , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Factor de Transcripción MSX1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tubo Neural/crecimiento & desarrollo , Tubo Neural/metabolismo , Defectos del Tubo Neural/genética , Neurulación , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/biosíntesis , Factores de Transcripción Paired Box/genética , Disrafia Espinal/genética , Proteínas de Dominio T Box , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transcripción Genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/genética
13.
Congenit Anom (Kyoto) ; 54(1): 54-62, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24344727

RESUMEN

Thalidomide (TM) induces limb defects in humans and some animal species including rabbits. Although the mechanism of TM-induced limb defects has been investigated for a long period, the limb development-related genes expressions have not been vigorously characterized in rabbits. In this study, we investigated the Fgf8, Bmp4 and Hoxa11 expressions in TM-treated JW rabbit embryos on gestation days (GDs) 10, 11 and 12 by whole mount in situ hybridization. On GDs 10 and 11, growth retardation of the embryo was induced by TM treatment. The Fgf8 expression lengths on GDs 10 and 11 in the forelimb bud were significantly or tended to be decreased in the TM-treated embryos, which was correlated to the growth retardation and was not considered to be directly relevant to the teratogenic effect of TM in the forelimb. The TM-induced characteristic changes in the expression pattern of Hoxa11 and Bmp4 on GDs 10 and/or 11 were not noted. On GD 12, TM-induced growth retardation was not noted and the Fgf8 and Bmp4 expressions were not changed. On the contrary, Hoxa11 expression was narrowed at the anterior region, which was located on the radial side, and was not changed at the middle and posterior regions in the forelimb bud and in all regions in the hindlimb bud. Because the radius malformations were induced by TM treatment, we concluded the decrease in the Hoxa11 expression was related to the TM-induced limb defects and can be a good marker for early prediction of the teratogenic effect of TM.


Asunto(s)
Proteína Morfogenética Ósea 4/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Proteínas de Homeodominio/biosíntesis , Talidomida/toxicidad , Anomalías Inducidas por Medicamentos/genética , Animales , Embrión de Mamíferos/anomalías , Desarrollo Embrionario/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Datos de Secuencia Molecular , Embarazo , Conejos
14.
Birth Defects Res B Dev Reprod Toxicol ; 98(6): 486-92, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24408660

RESUMEN

The kidney is one of the key organs in clearing foreign compounds. The effects of drugs on the developing kidney are relatively unknown. We studied the direct effect of furosemide, hydrochlorothiazide, ibuprofen, and indomethacin on kidney development in an ex vivo embryonic kidney model. At embryonic day 13, metanephroi were dissected from mice and cultured in control media or media supplemented with various clinically relevant concentrations of drugs. The ureteric tree was visualized by whole-mount staining and branching was evaluated by counting. Additionally, gene expression levels of Wt1, Sox9, Bmp7, Fgf8, and Gdnf were investigated. No distinct differences were noted on either ureteric tip development or gene expression analysis for each drug after 24 hr of exposure. Even though short-term exposure to clinically relevant concentrations seems not to disturb renal development, future research is needed to study prolonged or repeated exposures.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Diuréticos/farmacología , Riñón/embriología , Animales , Proteína Morfogenética Ósea 7/biosíntesis , Femenino , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Furosemida/farmacología , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Hidroclorotiazida/farmacología , Ibuprofeno/farmacología , Indometacina/farmacología , Ratones , Ratones Endogámicos ICR , Técnicas de Cultivo de Órganos , Embarazo , Factor de Transcripción SOX9/biosíntesis , Proteínas WT1/biosíntesis
15.
Development ; 139(6): 1213-24, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22354841

RESUMEN

Retinoic acid signaling is a major component of the neural posteriorizing process in vertebrate development. Here, we identify a new role for the retinoic acid receptor (RAR) in the anterior of the embryo, where RAR regulates Fgf8 expression and formation of the pre-placodal ectoderm (PPE). RARα2 signaling induces key pre-placodal genes and establishes the posterolateral borders of the PPE. RAR signaling upregulates two important genes, Tbx1 and Ripply3, during early PPE development. In the absence of RIPPLY3, TBX1 is required for the expression of Fgf8 and hence, PPE formation. In the presence of RIPPLY3, TBX1 acts as a transcriptional repressor, and functions to restrict the positional expression of Fgf8, a key regulator of PPE gene expression. These results establish a novel role for RAR as a regulator of spatial patterning of the PPE through Tbx1 and RIPPLY3. Moreover, we demonstrate that Ripply3, acting downstream of RAR signaling, is a key player in establishing boundaries in the PPE.


Asunto(s)
Ectodermo/fisiología , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Receptores de Ácido Retinoico/metabolismo , Proteínas de Dominio T Box/biosíntesis , Tretinoina/metabolismo , Proteínas de Xenopus/biosíntesis , Xenopus laevis/embriología , Animales , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Embrión no Mamífero/fisiología , Desarrollo Embrionario , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Sistema Nervioso/embriología , Receptor alfa de Ácido Retinoico , Transducción de Señal , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
16.
Cell Mol Life Sci ; 68(9): 1569-79, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21207098

RESUMEN

Fifty years ago, prescription of the sedative thalidomide caused a worldwide epidemic of multiple birth defects. The drug is now used in the treatment of leprosy and multiple myeloma. However, its use is limited due to its potent teratogenic activity. The mechanism by which thalidomide causes limb malformations and other developmental defects is a long-standing question. Multiple hypotheses exist to explain the molecular mechanism of thalidomide action. Among them, theories involving oxidative stress and anti-angiogenesis have been widely supported. Nevertheless, until recently, the direct target of thalidomide remained elusive. We identified a thalidomide-binding protein, cereblon (CRBN), as a primary target for thalidomide teratogenicity. Our data suggest that thalidomide initiates its teratogenic effects by binding to CRBN and inhibiting its ubiquitin ligase activity. In this review, we summarize the biology of thalidomide, focusing on the molecular mechanisms of its teratogenic effects. In addition, we discuss the questions still to be addressed.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Estrés Oxidativo , Péptido Hidrolasas/metabolismo , Teratógenos/toxicidad , Talidomida/toxicidad , Proteínas Adaptadoras Transductoras de Señales , Animales , Embrión de Pollo , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Humanos , Estrés Oxidativo/efectos de los fármacos , Conejos , Especificidad de la Especie , Teratógenos/química , Teratógenos/farmacocinética , Talidomida/química , Talidomida/farmacocinética , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Pez Cebra
17.
Angle Orthod ; 81(2): 270-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21208079

RESUMEN

OBJECTIVE: To evaluate cellular hypertrophic activities in the mandibular condylar cartilage (MCC) and the glenoid fossa (GF) during mandibular advancement in the temporomandibular joint (TMJ) of Sprague-Dawley rats, as evidenced by fibroblast growth factor 8 (FGF8). METHODS AND MATERIALS: Fifty-five female 24-day-old Sprague-Dawley rats were randomly divided into four experimental and control groups, with a mandibular advancement appliance on the experimental rats' lower incisors. The rats were euthanized on days 3, 14, 21, and 30 of the study, and their TMJ was prepared for a immunohistochemical staining procedure to detect FGF8. RESULTS: FGF8 expression was significantly higher among the experimental rats (P  =  .002). Patterns of ascension and descension of FGF8 expression were similar in experimental and control samples. The results show an overall enhanced osteogenic transition occurring in both the MCC and the GF in experimental rats in comparison with controls. The level of cellular changes in the MCC is remarkably higher than in the GF. CONCLUSION: In the MCC and the GF, cellular morphologic and hypertrophic differentiations increase significantly during mandibular advancement. It is also concluded that endochondral ossification in the MCC and intramembranous ossification in the GF occur during adaptive remodeling.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Avance Mandibular , Articulación Temporomandibular/metabolismo , Articulación Temporomandibular/patología , Adaptación Fisiológica , Animales , Remodelación Ósea , Cartílago Articular/metabolismo , Cartílago Articular/patología , Condrogénesis , Femenino , Hipertrofia/patología , Cóndilo Mandibular/metabolismo , Cóndilo Mandibular/patología , Osteogénesis , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Hueso Temporal/metabolismo , Hueso Temporal/patología
18.
BMC Cancer ; 10: 596, 2010 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-21034500

RESUMEN

BACKGROUND: Prostate tumours are commonly poorly oxygenated which is associated with tumour progression and development of resistance to chemotherapeutic drugs and radiotherapy. Fibroblast growth factor 8b (FGF8b) is a mitogenic and angiogenic factor, which is expressed at an increased level in human prostate tumours and is associated with a poor prognosis. We studied the effect of FGF8b on tumour oxygenation and growth parameters in xenografts in comparison with vascular endothelial growth factor (VEGF)-expressing xenografts, representing another fast growing and angiogenic tumour model. METHODS: Subcutaneous tumours of PC-3 cells transfected with FGF8b, VEGF or empty (mock) vectors were produced and studied for vascularity, cell proliferation, glucose metabolism and oxygenation. Tumours were evaluated by immunohistochemistry (IHC), flow cytometry, use of radiolabelled markers of energy metabolism ([18F]FDG) and hypoxia ([18F]EF5), and intratumoral polarographic measurements of pO2. RESULTS: Both FGF8b and VEGF tumours grew rapidly in nude mice and showed highly vascularised morphology. Perfusion studies, pO2 measurements, [18F]EF5 and [18F]FDG uptake as well as IHC staining for glucose transport protein (GLUT1) and hypoxia inducible factor (HIF) 1 showed that VEGF xenografts were well-perfused and oxygenised, as expected, whereas FGF8b tumours were as hypoxic as mock tumours. These results suggest that FGF8b-induced tumour capillaries are defective. Nevertheless, the growth rate of hypoxic FGF8b tumours was highly increased, as that of well-oxygenised VEGF tumours, when compared with hypoxic mock tumour controls. CONCLUSION: FGF8b is able to induce fast growth in strongly hypoxic tumour microenvironment whereas VEGF-stimulated growth advantage is associated with improved perfusion and oxygenation of prostate tumour xenografts.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Animales , Humanos , Hipoxia , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Oxígeno/química , Oxígeno/metabolismo , Pronóstico , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Biol Reprod ; 83(4): 514-24, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20592308

RESUMEN

Follicle-stimulating hormone (FSH) and oocyte-secreted factors influence granulosa cell differentiation and follicle development. Whereas FSH stimulates the expression of mural cell transcripts, oocyte-secreted factors regulate specific cumulus cell genes and suppress the appearance of mural cell transcripts. This study addresses the extent to which clinically relevant changes in FSH doses applied during antral follicle development in vitro could alter the expression of oocyte and cumulus cell transcripts. A 12-day culture system in which mouse ovarian preantral follicles can grow to preovulatory follicles was used. The following three FSH regimens were considered: 1) continuous exposure to an FSH level of 10 mIU/ml (control), 2) decreasing concentrations of FSH (low FSH), and 3) an FSH level of 25 mIU/ml (high FSH) as soon as the antrum is formed. Transcripts in oocytes (Gdf9, Bmp15, and Fgf8) and in cumulus cells (Amh, Lhcgr, Ar, and Pfkp) were quantified by real-time PCR. Under high FSH, the three oocyte transcripts were upregulated, while in cumulus cells a shutdown of the Amh signal and substantial increases in Lhcgr and Ar expression were measured. In contrast, low FSH tended to reduce Lhcgr to levels comparable to those in vivo. Levels of Pfkp were not affected by FSH doses. These results demonstrate that a 2.5-fold increase in FSH changes both oocyte and cumulus cell transcript levels. Conversely, a decrease in FSH does not affect transcript levels but seems to limit inappropriate Lhcgr expression. Modulating FSH within physiological ranges during the antral phase of culture alters cumulus cell differentiation.


Asunto(s)
Células del Cúmulo/fisiología , Hormona Folículo Estimulante/farmacología , Regulación del Desarrollo de la Expresión Génica , Oocitos/fisiología , Folículo Ovárico/fisiología , Animales , Hormona Antimülleriana/biosíntesis , Hormona Antimülleriana/genética , Proteína Morfogenética Ósea 15/biosíntesis , Proteína Morfogenética Ósea 15/genética , Cruzamientos Genéticos , Células del Cúmulo/citología , Células del Cúmulo/efectos de los fármacos , Femenino , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 9 de Diferenciación de Crecimiento/biosíntesis , Factor 9 de Diferenciación de Crecimiento/genética , L-Lactato Deshidrogenasa/biosíntesis , L-Lactato Deshidrogenasa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Oocitos/citología , Oocitos/efectos de los fármacos , Folículo Ovárico/citología , Folículo Ovárico/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética , Receptores de HL/biosíntesis , Receptores de HL/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
J Dent Res ; 89(9): 909-14, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20530729

RESUMEN

beta-Catenin signaling is required for embryonic tooth morphogenesis and promotes continuous tooth development when activated in embryos. To determine whether activation of this pathway in the adult oral cavity could promote tooth development, we induced mutation of epithelial beta-catenin to a stabilized form in adult mice. This caused increased proliferation of the incisor tooth cervical loop, outpouching of incisor epithelium, abnormal morphology of the epithelial-mesenchymal junction, and enhanced expression of genes associated with embryonic tooth development. Ectopic dental-like structures were formed from the incisor region following implantation into immunodeficient mice. Thus, forced activation of beta-catenin signaling can initiate an embryonic-like program of tooth development in adult rodent incisor teeth.


Asunto(s)
Células Madre Adultas/fisiología , Papila Dental/citología , Órgano del Esmalte/citología , Odontogénesis/genética , beta Catenina/fisiología , Animales , Células Epiteliales/citología , Femenino , Factor 8 de Crecimiento de Fibroblastos/biosíntesis , Factor 8 de Crecimiento de Fibroblastos/genética , Incisivo/citología , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Desnudos , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Transducción de Señal , Calcificación de Dientes , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...