Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Tipo del documento
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.
Stem Cell Res ; 40: 101572, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31539858

RESUMEN

High differentiation efficiency is one of the most important factors in developing an in vitro model from pluripotent stem cells. In this report, we improved the handling technique applied to mouse-induced pluripotent stem (iPS) cells, resulting in better differentiation into hypothalamic vasopressin (AVP) neurons. We modified the culture procedure to make the maintenance of iPS cells in an undifferentiated state much easier. Three-dimensional floating culture was demonstrated to be effective for mouse iPS cells. We also improved the differentiation method with regards to embryology, resulting in a greater number of bigger colonies of AVP neurons differentiating from mouse iPS cells. Fgf8, which was not used in the original differentiation method, increased iPS differentiation into AVP neurons. These refinements will be useful as a valuable tool for the modeling of degenerative disease in AVP neurons in vitro using disease-specific iPS cells in future studies.


Asunto(s)
Diferenciación Celular , Línea Celular/citología , Hipotálamo/citología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/citología , Animales , Línea Celular/metabolismo , Células Cultivadas , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Hipotálamo/metabolismo , Células Madre Pluripotentes Inducidas/citología , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Vasopresinas/metabolismo
3.
Brain Struct Funct ; 224(2): 661-679, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30470893

RESUMEN

Thalamic neurons are distributed between different nuclear groups of the thalamic multinuclear complex; they develop topologically ordered specific projections that convey information on voluntary motor programs and sensory modalities to functional areas in the cerebral cortex. Since thalamic neurons present a homogeneous morphology, their functional specificity is derived from their afferent and efferent connectivity. Adequate development of thalamic afferent and efferent connections depends on guide signals that bind receptors in nuclear neuropils and axonal growth cones, respectively. These are finally regulated by regionalization processes in the thalamic neurons, codifying topological information. In this work, we studied the role of Fgf8 morphogenetic signaling in establishing the molecular thalamic protomap, which was revealed by Igsf21, Pde10a and Btbd3 gene expression in the thalamic mantle layer. Fgf8 signaling activity was evidenced by pERK expression in radial glia cells and fibers, which may represent a scaffold that translates neuroepithelial positional information to the mantle layer. In this work, we describe the fact that Fgf8-hypomorphic mice did not express pERK in radial glia cells and fibers and presented disorganized thalamic regionalization, increasing neuronal death in the ventro-lateral thalamus and strong disruption of thalamocortical projections. In conclusion, Fgf8 encodes the positional information required for thalamic nuclear regionalization and the development of thalamocortical projections.


Asunto(s)
Células Ependimogliales/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Tálamo/metabolismo , Animales , Apoptosis/fisiología , Axones/metabolismo , Mapeo Encefálico/métodos , Proliferación Celular/fisiología , Factor 8 de Crecimiento de Fibroblastos/genética , Ratones , Ratones Noqueados , Fibras Nerviosas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Vías Nerviosas/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Fosforilación
5.
Zhonghua Nan Ke Xue ; 23(12): 1063-1068, 2017 Dec.
Artículo en Chino | MEDLINE | ID: mdl-29738174

RESUMEN

OBJECTIVE: To induce hypospadias in male rat offspring by maternal exposure to di-n-butyl phthalate (DBP) during late pregnancy and further investigate its mechanisms. METHODS: We randomly divided 20 pregnant rats into a DBP exposure and a control group, the former treated intragastrically with DBP while the latter with soybean oil at 750 mg per kilogram of the body weight per day from gestation days (GD) 14 to 18. On postnatal day (PND) 1, we recorded the incidence rate of hypospadias and observed the histopathological changes in the genital tubercle of the hypospadiac rats. We also measured the level of serum testosterone (T) by radioimmunoassay and determined the mRNA and protein expressions of the androgen receptor (AR), sonic hedgehog (Shh), bone morphogenetic protein 4 (Bmp4) and fibroblast growth factor 8 (Fgf8) in the genital tubercle by real-time PCR and Western blot. RESULTS: No hypospadiac male rats were found in the control group. The incidence rate of hypospadias in male offspring was 43.6% in the DBP-treatment group. Histological analysis confirmed hypospadiac malformation. The serum testosterone concentration was decreased in the hypospadiac male rats as compared with the controls (ï¼»0.49 ± 0.05ï¼½ vs ï¼»1.12 ± 0.05ï¼½ ng/ml, P <0.05). The mRNA expressions of AR, Shh, Bmp4 and Fgf8 in the genital tubercle were significantly lower in the hypospadiac male rats than in the controls (AR: 0.50 ± 0.05 vs 1.00 ± 0.12, P <0.05; Shh: 0.65 ± 0.07 vs 1.00 ± 0.15, P <0.05; Bmp4: 0.42 ± 0.05 vs 1.00 ± 0.13, P <0.05; Fgf8: 0.46 ± 0.04 vs 1.00 ± 0.12, P <0.05), and so were their protein expressions (AR: 0.34 ± 0.05 vs 1.00 ± 0.09, P <0.05; Shh: 0.51 ± 0.07 vs 1.00 ± 0.12, P <0.05; Bmp4: 0.43 ± 0.05 vs 1.00 ± 0.11, P <0.05; Fgf8: 0.57 ± 0.04 vs 1.00 ± 0.13, P <0.05). CONCLUSIONS: Maternal exposure to DBP during late pregnancy can induce hypospadias in the male rat offspring. DBP affects the development of the genital tubercle by reducing the serum T concentration and expressions of AR, Shh, Bmp4 and Fgf8 in the genital tubercle, which might underlie the mechanism of DBP inducing hypospadias.


Asunto(s)
Dibutil Ftalato/toxicidad , Hipospadias/inducido químicamente , Exposición Materna , Plastificantes/toxicidad , Animales , Peso Corporal , Proteína Morfogenética Ósea 4/sangre , Femenino , Factor 8 de Crecimiento de Fibroblastos/sangre , Proteínas Hedgehog/sangre , Hipospadias/sangre , Hipospadias/patología , Masculino , Embarazo , ARN Mensajero/sangre , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/sangre , Aceite de Soja , Testosterona/sangre
6.
National Journal of Andrology ; (12): 1063-1068, 2017.
Artículo en Chino | WPRIM | ID: wpr-812834

RESUMEN

Objective@#To induce hypospadias in male rat offspring by maternal exposure to di-n-butyl phthalate (DBP) during late pregnancy and further investigate its mechanisms.@*METHODS@#We randomly divided 20 pregnant rats into a DBP exposure and a control group, the former treated intragastrically with DBP while the latter with soybean oil at 750 mg per kilogram of the body weight per day from gestation days (GD) 14 to 18. On postnatal day (PND) 1, we recorded the incidence rate of hypospadias and observed the histopathological changes in the genital tubercle of the hypospadiac rats. We also measured the level of serum testosterone (T) by radioimmunoassay and determined the mRNA and protein expressions of the androgen receptor (AR), sonic hedgehog (Shh), bone morphogenetic protein 4 (Bmp4) and fibroblast growth factor 8 (Fgf8) in the genital tubercle by real-time PCR and Western blot.@*RESULTS@#No hypospadiac male rats were found in the control group. The incidence rate of hypospadias in male offspring was 43.6% in the DBP-treatment group. Histological analysis confirmed hypospadiac malformation. The serum testosterone concentration was decreased in the hypospadiac male rats as compared with the controls ([0.49 ± 0.05] vs [1.12 ± 0.05] ng/ml, P <0.05). The mRNA expressions of AR, Shh, Bmp4 and Fgf8 in the genital tubercle were significantly lower in the hypospadiac male rats than in the controls (AR: 0.50 ± 0.05 vs 1.00 ± 0.12, P <0.05; Shh: 0.65 ± 0.07 vs 1.00 ± 0.15, P <0.05; Bmp4: 0.42 ± 0.05 vs 1.00 ± 0.13, P <0.05; Fgf8: 0.46 ± 0.04 vs 1.00 ± 0.12, P <0.05), and so were their protein expressions (AR: 0.34 ± 0.05 vs 1.00 ± 0.09, P <0.05; Shh: 0.51 ± 0.07 vs 1.00 ± 0.12, P <0.05; Bmp4: 0.43 ± 0.05 vs 1.00 ± 0.11, P <0.05; Fgf8: 0.57 ± 0.04 vs 1.00 ± 0.13, P <0.05).@*CONCLUSIONS@#Maternal exposure to DBP during late pregnancy can induce hypospadias in the male rat offspring. DBP affects the development of the genital tubercle by reducing the serum T concentration and expressions of AR, Shh, Bmp4 and Fgf8 in the genital tubercle, which might underlie the mechanism of DBP inducing hypospadias.


Asunto(s)
Animales , Femenino , Masculino , Embarazo , Ratas , Peso Corporal , Proteína Morfogenética Ósea 4 , Sangre , Dibutil Ftalato , Toxicidad , Factor 8 de Crecimiento de Fibroblastos , Sangre , Proteínas Hedgehog , Sangre , Hipospadias , Sangre , Patología , Exposición Materna , Plastificantes , Toxicidad , ARN Mensajero , Sangre , Distribución Aleatoria , Ratas Sprague-Dawley , Receptores Androgénicos , Sangre , Aceite de Soja , Testosterona , Sangre
7.
Brain Struct Funct ; 221(7): 3709-27, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26459142

RESUMEN

The mammalian eminentia thalami (EmT) (or thalamic eminence) is an embryonic forebrain structure of unknown function. Here, we examined the molecular and cellular properties of the mouse EmT. We first studied mRNA expression of signalling molecules and found that the EmT is a structure, rich in expression of secreted factors, with Wnts being the most abundantly detected. We then examined whether EmT tissue could induce cell fate changes when grafted ectopically. For this, we transplanted EmT tissue from a tau-GFP mouse to the ventral telencephalon of a wild type host, a telencephalic region where Wnt signalling is not normally active but which we showed in culture experiments is competent to respond to Wnts. We observed that the EmT was able to induce in adjacent ventral telencephalic cells ectopic expression of Lef1, a transcriptional activator and a target gene of the Wnt/ß-catenin pathway. These Lef1-positive;GFP-negative cells expressed the telencephalic marker Foxg1 but not Ascl1, which is normally expressed by ventral telencephalic cells. These results suggest that the EmT has the capacity to activate Wnt/ß-catenin signalling in the ventral telencephalon and to suppress ventral telencephalic gene expression. Altogether, our data support a role of the EmT as a signalling centre in the developing mouse forebrain.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Tálamo/embriología , Tálamo/metabolismo , Vía de Señalización Wnt , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína Morfogenética Ósea 4 , Proteína Morfogenética Ósea 6 , Células Cultivadas , Factor 8 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos , Ratones , ARN Mensajero/metabolismo , Transducción de Señal
8.
Brain Struct Funct ; 221(6): 3095-109, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26311466

RESUMEN

The establishment of the brain structural complexity requires a precisely orchestrated interplay between extrinsic and intrinsic signals modulating cellular mechanisms to guide neuronal differentiation. However, little is known about the nature of these signals in the diencephalon, a complex brain region that processes and relays sensory and motor information to and from the cerebral cortex and subcortical structures. Morphogenetic signals from brain organizers regulate histogenetic processes such as cellular proliferation, migration, and differentiation. Sonic hedgehog (Shh) in the key signal of the ZLI, identified as the diencephalic organizer. Fgf15, the mouse gene orthologous of human, chick, and zebrafish Fgf19, is induced by Shh signal and expressed in the diencephalic alar plate progenitors during histogenetic developmental stages. This work investigates the role of Fgf15 signal in diencephalic development. In the absence of Fgf15, the complementary expression pattern of proneural genes: Ascl1 and Nng2, is disrupted and the GABAergic thalamic cells do not differentiate; in addition dorsal thalamic progenitors failed to exit from the mitotic cycle and to differentiate into neurons. Therefore, our findings indicate that Fgf15 is the Shh downstream signal to control thalamic regionalization, neurogenesis, and neuronal differentiation by regulating the expression and mutual segregation of neurogenic and proneural regulatory genes.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Tálamo/embriología , Tálamo/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Proliferación Celular , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Proteínas Hedgehog/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurogénesis , Neuronas/metabolismo , Neuronas/fisiología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteína Wnt1/metabolismo
9.
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
10.
J Neuroendocrinol ; 22(8): 944-50, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20553372

RESUMEN

Gonadotrophin-releasing hormone (GnRH) neurones control the onset and maintenance of fertility. Aberrant development of the GnRH system underlies infertility in Kallmann syndrome [KS; idiopathic hypogonadotropic hypogonadism (IHH) and anosmia]. Some KS patients harbour mutations in the fibroblast growth factor receptor 1 (Fgfr1) and Fgf8 genes. The biological significance of these two genes in GnRH neuronal development was corroborated by the observation that GnRH neurones were severely reduced in newborn transgenic mice deficient in either gene. In the present study, we hypothesised that the compound deficiency of Fgf8 and its cognate receptors, Fgfr1 and Fgfr3, may lead to more deleterious effects on the GnRH system, thereby resulting in a more severe reproductive phenotype in patients harbouring these mutations. This hypothesis was tested by counting the number of GnRH neurones in adult transgenic mice with digenic heterozygous mutations in Fgfr1/Fgf8, Fgfr3/Fgf8 or Fgfr1/Fgfr3. Monogenic heterozygous mutations in Fgfr1, Fgf8 or Fgfr3 caused a 30-50% decrease in the total number of GnRH neurones. Interestingly, mice with digenic mutations in Fgfr1/Fgf8 showed a greater decrease in GnRH neurones compared to mice with a heterozygous defect in the Fgfr1 or Fgf8 alone. This compounding effect was not detected in mice with digenic heterozygous mutations in Fgfr3/Fgf8 or Fgfr1/Fgfr3. These results support the hypothesis that IHH/KS patients with digenic mutations in Fgfr1/Fgf8 may have a further reduction in the GnRH neuronal population compared to patients harbouring monogenic haploid mutations in Fgfr1 or Fgf8. Because only Fgfr1/Fgf8 compound deficiency leads to greater GnRH system defect, this also suggests that these fibroblast growth factor signalling components interact in a highly specific fashion to support GnRH neuronal development.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Animales , Factor 8 de Crecimiento de Fibroblastos/genética , Humanos , Hipogonadismo/fisiopatología , Hipotálamo/citología , Hipotálamo/metabolismo , Síndrome de Kallmann/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética
11.
PLoS One ; 5(4): e10143, 2010 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-20405041

RESUMEN

Fibroblast growth factors (FGFs) mediate a vast range of CNS developmental processes including neural induction, proliferation, migration, and cell survival. Despite the critical role of FGF signaling for normal CNS development, few reports describe the mechanisms that regulate FGF receptor gene expression in the brain. We tested whether FGF8 could autoregulate two of its cognate receptors, Fgfr1 and Fgfr3, in three murine cell lines with different lineages: fibroblast-derived cells (3T3 cells), neuronal cells derived from hippocampus (HT-22 cells), and neuroendocrine cells derived from hypothalamic gonadotropin-releasing hormone (GnRH) neurons (GT1-7 cells). GnRH is produced by neurons in the hypothalamus and is absolutely required for reproductive competence in vertebrate animals. Several lines of evidence strongly suggest that Fgf8 is critical for normal development of the GnRH system, therefore, the GT1-7 cells provided us with an additional endpoint, Gnrh gene expression and promoter activity, to assess potential downstream consequences of FGF8-induced modulation of FGF receptor levels. Results from this study suggest that the autoregulation of its cognate receptor represents a common downstream effect of FGF8. Further, we show that Fgfr1 and Fgfr3 are differentially regulated within the same cell type, implicating these two receptors in different biological roles. Moreover, Fgfr1 and Fgfr3 are differentially regulated among different cell types, suggesting such autoregulation occurs in a cell type-specific fashion. Lastly, we demonstrate that FGF8b decreases Gnrh promoter activity and gene expression, possibly reflecting a downstream consequence of altered FGF receptor populations. Together, our data bring forth the possibility that, in addition to the FGF synexpression group, autoregulation of FGFR expression by FGF8 represents a mechanism by which FGF8 could fine-tune its regulatory actions.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/fisiología , Regulación de la Expresión Génica , Neuronas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Animales , Química Encefálica , Línea Celular , Linaje de la Célula , Fibroblastos/citología , Hormona Liberadora de Gonadotropina , Hipocampo/citología , Hipotálamo/citología , Ratones
12.
Ann Endocrinol (Paris) ; 71 Suppl 1: S33-41, 2010 Sep.
Artículo en Francés | MEDLINE | ID: mdl-21237329

RESUMEN

Hypogonadotropic hypogonadism (HH) is defined by the absence of sex steroid synthesis associated with the lack of appropriate gonadotrophin secretion. This leads to a variable degree of impuberism, often diagnosed during childhood or adolescence. Genetics of HH involve many genes. However, molecular defects have been identified in only 30 % of patients. Kallmann syndrome (KS) is defined by the association of HH and anosmia. Six genes are involved in KS (KAL1, FGFR1, FGF8, PROK2, PROKR2 and CHD7). However, genetics of KS is complex, because of the variability of the phenotype for a similar molecular defect. Otherwise, heterozygous anomalies are frequently described. Identification in the same patient of several mutations in some of these genes (digenism) could account for this variability. Autosomal recessive transmission is frequently observed in familial cases of HH without anosmia. Molecular alterations have been identified for several neuropeptides or their corresponding receptors, which are involved in the physiology of the gonadotropic axis : GNRHR, KISS1R/GPR54, neurokinin B (TAC3), TACR3 and GNRH1 (and PROK2, PROKR2 and CHD7). Anomalies of leptin or its receptor are also involved in HH cases. A new negative regulating element has been recently identified in humans : RFRP3, which is ortholog of the avian GnIH (gonadotrophin inhibitory hormone). Recent progress about these neuropeptides leads to a new model of comprehension of the gonadotropic axis physiology, from a linear model to a network model, which regulates the central element of regulation of the gonadotropic axis, represented by the GnRH neurons.


Asunto(s)
Gónadas/metabolismo , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Hipófisis/metabolismo , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Proteínas de la Matriz Extracelular/genética , Femenino , Factor 8 de Crecimiento de Fibroblastos/genética , Hormonas Gastrointestinales/genética , Humanos , Hipogonadismo/genética , Síndrome de Kallmann/genética , Síndrome de Kallmann/metabolismo , Leptina/genética , Masculino , Proteínas del Tejido Nervioso/genética , Neuropéptidos/genética , Trastornos del Olfato/genética , Hormonas Inhibidoras de la Liberación de Hormona Hipofisaria/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Leptina/genética , Receptores de Péptidos/genética
13.
J Neurosci ; 29(42): 13389-400, 2009 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-19846726

RESUMEN

Habenular nuclei play a key role in the control of motor and cognitive behavior, processing emotion, motivation, and reward values in the brain. Thus, analysis of the molecular and cellular mechanisms underlying the development and evolution of this region will contribute to a better understanding of brain function. The Fgf8 gene is expressed in the dorsal midline of the diencephalon, close to the area in which the habenular region will develop. Given that Fgf8 is an important morphogenetic signal, we decided to investigate the role of Fgf8 signaling in diencephalic development. To this end, we analyzed the effects of altered Fgf8 expression in the mouse embryo, using molecular and cellular markers. Decreasing Fgf8 activity in the diencephalon was found to be associated with dosage-dependent alterations in the epithalamus: the habenular region and pineal gland are reduced or lacking in Fgf8 hypomorphic mice. Actually, our findings indicate that Fgf8 may be the master gene for these diencephalic domains, acting as an inductive and morphogenetic regulator. Therefore, the emergence of the habenular region in vertebrates could be understood in terms of a phylogenetic territorial addition caused by de novo expression of Fgf8 in the diencephalic alar plate. This region specializes to permit the development of adaptive control of the motor function in the vertebrate brain.


Asunto(s)
Tipificación del Cuerpo/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neuronas/fisiología , Tálamo , Factores de Edad , Animales , Apoptosis/genética , Encéfalo/anatomía & histología , Encéfalo/embriología , Encéfalo/metabolismo , Movimiento Celular/genética , Proliferación Celular , Embrión de Pollo , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo , Embrión de Mamíferos/fisiología , Factor 8 de Crecimiento de Fibroblastos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/genética , Tálamo/citología , Tálamo/embriología , Tálamo/fisiología , Proteína Wnt1/genética
14.
Semin Cell Dev Biol ; 20(6): 719-25, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19596327

RESUMEN

Correct patterning of the developing brain is crucial importance for accurate wiring and function. Although the adult brain contains many complex structures, it begins with a simple structure-the neural tube. As it develops, the neural tube is divided into several regions, including the telencephalon, diencephalon, midbrain, and hindbrain. In each of these regions, signaling molecules are secreted from discrete zones, which establish positional information and regulate regional growth. There are many mechanistic questions that remain to be resolved about the action of these growth and differentiation factors. The cellular factors mediating patterning in response to these factors are largely unknown. Furthermore, identical differentiation factors are expressed in different regions of the brain and yet control significantly different patterning mechanisms, and the factors that control region-specific responses to these factors are mostly obscure. Furthermore, differentiation factors also show dramatically different expression patterns in different vertebrate species that may underlie changes in brain structure, but the mechanisms by which these changes in gene expression occur poorly understood. To address these issues, we discuss the role of Fgf8, which controls anterior/posterior patterning in different regions of the developing brain. We also discuss how modifications of Fgf8 expression in the diencephalon controlled by retrotransposons can change the shape and function of the brain in various species.


Asunto(s)
Tipificación del Cuerpo , Diencéfalo/embriología , Factor 8 de Crecimiento de Fibroblastos/fisiología , Telencéfalo/embriología , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular/fisiología , Diencéfalo/citología , Factor 8 de Crecimiento de Fibroblastos/genética , Humanos , Mesencéfalo/citología , Mesencéfalo/embriología , Ratones , Rombencéfalo/citología , Rombencéfalo/embriología , Transducción de Señal/fisiología , Telencéfalo/citología , Tálamo/citología , Tálamo/embriología , Factores de Transcripción/metabolismo
15.
Development ; 135(17): 2873-81, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18653561

RESUMEN

The vertebrate thalamus contains multiple sensory nuclei and serves as a relay station to receive sensory information and project to corresponding cortical areas. During development, the progenitor region of the diencephalon is divided into three parts, p1, p2 (presumptive thalamus) and p3, along its longitudinal axis. Besides the local expression of signaling molecules such as sonic hedgehog (Shh), Wnt proteins and Fgf8, the patterning mechanisms of the thalamic nuclei are largely unknown. Using mouse in utero electroporation to overexpress or inhibit endogenous Fgf8 at the diencephalic p2/p3 border, we revealed that it affected gene expression only in the p2 region without altering overall diencephalic size or the expression of other signaling molecules. We demonstrated that two distinctive populations in p2, which can be distinguished by Ngn2 and Mash1 in early embryonic diencephalon, are controlled by Fgf8 activity in complementary manner. Furthermore, we found that FGF activity shifts thalamic sensory nuclei on the A/P axis in postnatal brain. Moreover, gene expression analysis demonstrated that FGF signaling shifts prethalamic nuclei in complementary manner to the thalamic shift. These findings suggest conserved roles of FGF signaling in patterning along the A/P axis in CNS, and reveal mechanisms of nucleogenesis in the developing thalamus.


Asunto(s)
Tipificación del Cuerpo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Tálamo/embriología , Animales , Biomarcadores/metabolismo , Electroporación , Desarrollo Embrionario , Factor 8 de Crecimiento de Fibroblastos/genética , Proteínas de Unión al GTP/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cuerpos Geniculados/citología , Cuerpos Geniculados/embriología , Proteínas Hedgehog/metabolismo , Ratones , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Tálamo/citología , Proteínas Wnt/metabolismo
17.
Endocrinology ; 149(10): 4997-5003, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18566132

RESUMEN

GnRH neurons are essential for the onset and maintenance of reproduction. Mutations in both fibroblast growth factor receptor (Fgfr1) and Fgf8 have been shown to cause Kallmann syndrome, a disease characterized by hypogonadotropic hypogonadism and anosmia, indicating that FGF signaling is indispensable for the formation of a functional GnRH system. Presently it is unclear which stage of GnRH neuronal development is most impacted by FGF signaling deficiency. GnRH neurons express both FGFR1 and -3; thus, it is also unclear whether FGFR1 or FGFR3 contributes directly to GnRH system development. In this study, we examined the developing GnRH system in mice deficient in FGF8, FGFR1, or FGFR3 to elucidate the individual contribution of these FGF signaling components. Our results show that the early emergence of GnRH neurons from the embryonic olfactory placode requires FGF8 signaling, which is mediated through FGFR1, not FGFR3. These data provide compelling evidence that the developing GnRH system is exquisitely sensitive to reduced levels of FGF signaling. Furthermore, Kallmann syndrome stemming from FGF signaling deficiency may be due primarily to defects in early GnRH neuronal development prior to their migration into the forebrain.


Asunto(s)
Factor 8 de Crecimiento de Fibroblastos/metabolismo , Hormona Liberadora de Gonadotropina/fisiología , Vías Olfatorias/embriología , Vías Olfatorias/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Movimiento Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Hipotálamo/citología , Hipotálamo/embriología , Hipotálamo/fisiología , Proteínas de Filamentos Intermediarios/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Vías Olfatorias/citología , Periferinas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Transactivadores/metabolismo , Órgano Vomeronasal/citología , Órgano Vomeronasal/embriología , Órgano Vomeronasal/fisiología
18.
J Neurosci Res ; 85(9): 1884-93, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17471553

RESUMEN

Methods for identification and in vitro expansion of ventral mesencephalic dopaminergic precursor cells are of interest in the search for transplantable neurons for cell therapy in Parkinson's disease (PD). We investigated the potential use of fibroblast growth factor 2 (FGF2) and fibroblast growth factor 8 (FGF8) for expansion of such dopaminergic precursor cells, and fetal antigen-1 (FA1), a secreted neuronal protein of unknown function, as a non-invasive dopaminergic marker. Tissue from embryonic day (ED) 12 rat ventral mesencephalon was dissociated mechanically and cultured for 4 days in the presence of FGF2, FGF8, or without mitogens (control). After mitogen withdrawal and addition of 0.5% bovine serum, cells were differentiated for 6 days. Before differentiation, significantly more cells incorporated BrdU in cultures exposed to FGF2 (19-fold; P < 0.001) and FGF8 (3-fold; P < 0.05) compared to controls. After differentiation, biochemical analyses showed significantly more dopamine and FA1 in conditioned medium from both FGF2 and FGF8 expanded cultures than in controls. Correspondingly, numbers of tyrosine hydroxylase (TH)- and FA1-immunoreactive cells had increased 16-fold (P < 0.001) and 2.1-fold (P < 0.001), respectively in the FGF2 group and 10-fold (P < 0.001) and 1.8-fold (P < 0.05), respectively in the FGF8 group. In conclusion, the present procedure allows efficient expansion and differentiation of dopaminergic precursor cells and provides novel evidence of FGF8 as a mitogen for these cells. Furthermore, FA1 was identified as a potential supplementary non-invasive marker of cultured dopaminergic neurons.


Asunto(s)
Dopamina/fisiología , Mesencéfalo/citología , Mitógenos/farmacología , Células Madre/fisiología , Animales , Antimetabolitos , Biomarcadores/análisis , Bromodesoxiuridina , Recuento de Células , Diferenciación Celular/fisiología , Células Cultivadas , Cromatografía Líquida de Alta Presión , Medios de Cultivo Condicionados , Dopamina/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Embarazo , Ratas , Serotonina/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Ácido gamma-Aminobutírico/metabolismo
19.
Bone ; 40(2): 360-73, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17085092

RESUMEN

Marrow-isolated adult multilineage inducible (MIAMI) cells were differentiated in vitro to neuronal cells in a neurotrophin-dependent fashion. After induction, the cells revealed electrophysiological features similar to those observed in mature neurons. Primary early passage human MIAMI cells without any type of co-cultures with other cell types were used. The developmental program involved a multi-step process requiring the concerted action of brain-derived neurotrophic factor, nerve growth factor and depended on neurotrophin-3, after basic fibroblast growth factor withdrawal. MIAMI-derived neuron-like cells sequentially expressed the neuronal markers, developed a complex neurite outgrowth and arborization, and acquired electrophysiological characteristics similar to those observed in mature neurons. The young and old MIAMI-derived neuronal cells developed both inward and outward currents upon depolarization, similar to those observed in normal neurons. These results represent the earliest evidence that neurotrophin-3 can direct the differentiation of non-neural stem cells from human adult bone marrow stroma to neuron-like cells in vitro. Supplementing the aforementioned multi-step process with sonic hedgehog, fibroblast growth factor 8, and retinoic acid increased the expression of molecules involved in dopaminergic differentiation and of tyrosine hydroxylase, the rate limiting enzyme of dopamine synthesis. MIAMI cells from young and old individuals represent autologous human cell populations for the treatment of disorders of the skeletal and nervous systems and for applications in cell therapy and reparative medicine approaches.


Asunto(s)
Células de la Médula Ósea/fisiología , Dopamina/metabolismo , Neuronas/fisiología , Neurotrofina 3/fisiología , Células del Estroma/fisiología , Adolescente , Adulto , Células Madre Adultas/citología , Células Madre Adultas/fisiología , Factores de Edad , Anciano , Células de la Médula Ósea/citología , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Femenino , Factor 8 de Crecimiento de Fibroblastos/farmacología , Proteínas Hedgehog/farmacología , Humanos , Masculino , Persona de Mediana Edad , Neuritas/fisiología , Neuronas/citología , Neuronas/metabolismo , Neurotrofina 3/farmacología , Proteínas Recombinantes/farmacología , Células del Estroma/citología , Tretinoina/farmacología , Tirosina 3-Monooxigenasa/metabolismo
20.
Nat Cell Biol ; 8(3): 271-7, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16489341

RESUMEN

Somitogenesis requires bilateral rhythmic segmentation of paraxial mesoderm along the antero-posterior axis. The location of somite segmentation depends on opposing signalling gradients of retinoic acid (generated by retinaldehyde dehydrogenase-2; Raldh2) anteriorly and fibroblast growth factor (FGF; generated by Fgf8) posteriorly. Retinoic-acid-deficient embryos exhibit somite left-right asymmetry, but it remains unclear how retinoic acid mediates left-right patterning. Here, we demonstrate that retinoic-acid signalling is uniform across the left-right axis and occurs in node ectoderm but not node mesoderm. In Raldh2(-/-) mouse embryos, ectodermal Fgf8 expression encroaches anteriorly into node ectoderm and neural plate, but its expression in presomitic mesoderm is initially unchanged. The late stages of somitogenesis were rescued in Raldh2(-/-) mouse embryos when the maternal diet was supplemented with retinoic acid until only the 6-somite stage, demonstrating that retinoic acid is only needed during node stages. A retinoic-acid-reporter transgene marking the action of maternal retinoic acid in rescued Raldh2(-/-) embryos revealed that the targets of retinoic-acid signalling during somitogenesis are the node ectoderm and the posterior neural plate, not the presomitic mesoderm. Our findings suggest that antagonism of Fgf8 expression by retinoic acid occurs in the ectoderm and that failure of this mechanism generates excessive FGF8 signalling to adjacent mesoderm, resulting initially in smaller somites and then left-right asymmetry.


Asunto(s)
Tipificación del Cuerpo , Ectodermo/metabolismo , Somitos/metabolismo , Tretinoina/fisiología , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal , Tretinoina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA