Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Dev Neurosci ; 41: 98-104, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25660516

RESUMEN

The TGF-ß ligand superfamily members activin A and BMP control important aspects of embryonic neuronal development and differentiation. Both are known to bind to activin receptor subtypes IIA (ActRIIA) and IIB, while in the avian ciliary ganglion (CG), so far only ActRIIA-expression has been described. We show that the expression of ACVR2B, coding for the ActRIIB, is tightly regulated during CG development and the knockdown of ACVR2B expression leads to a deregulation in the execution of neuronal apoptosis and therefore affects ontogenetic programmed cell death in vivo. While the differentiation of choroid neurons was impeded in the knockdown, pointing toward a reduction in activin A-mediated neural differentiation signaling, naturally occurring neuronal cell death in the CG was not prevented by follistatin treatment. Systemic injections of the BMP antagonist noggin, on the other hand, reduced the number of apoptotic neurons to a similar extent as ACVR2B knockdown. We therefore propose a novel pathway in the regulation of CG neuron ontogenetic programmed cell death, which could be mediated by BMP and signals via the ActRIIB.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Ganglios Parasimpáticos/citología , Ganglios Parasimpáticos/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas/metabolismo , Transducción de Señal/fisiología , Receptores de Activinas Tipo II/genética , Factores de Edad , Animales , Proteínas Aviares/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bromodesoxiuridina/metabolismo , Proteínas Portadoras/farmacología , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Embrión de Pollo , Productos del Gen gag/metabolismo , Técnicas de Transferencia de Gen , Análisis por Micromatrices , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Colorantes de Rosanilina/metabolismo , Somatostatina/metabolismo
3.
Neuroscience ; 150(3): 647-57, 2007 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-17997227

RESUMEN

For neurotrophins and also for members of the transforming growth factor beta (TGF-beta) family an activity-dependent regulation of synthesis and release has been proposed. Together with the observation that the secretion of neurotransmitters is initiated by neurotrophic factors, it is reasonable to assume that they might act as retrograde modulators enhancing the efficacy and stabilization of synapses. In the present study, we have tested this hypothesis and studied the release and regulation of TGF-beta in vitro using mouse primary hippocampal neurons at embryonic day E16.5 as model. We show that neuronal activity regulates TGF-beta release and TGF-beta expression in vitro. Treatment of the cultures with KCl, 3-veratroylveracevine (veratridine), glutamate or carbamylcholine chloride (carbachol) increased the levels of secreted TGF-beta, as assessed by the MLEC/plasminogen activator inhibitor (PAI)-luciferase-assay, whereas TGF-beta release stimulated by KCl or veratridine was reduced in the presence of tetrodotoxin or 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA). In addition, application of glutamate significantly upregulated expression of TGF-beta2 and TGF-beta3 in the culture. Notably, KCl stimulation caused Smad (composite term from SMA (C. elegans) and MAD=mothers against dpp (Drosophila)) translocation into the nucleus and upregulated TGF-beta inducible early gene (Tieg1) expression, demonstrating that activity-dependent released TGF-beta may exert autocrine actions and thereby activate the TGF-beta-dependent signaling pathway. Together, these results suggest an activity-dependent release and gene transcription of TGF-beta from mouse hippocampal neurons in vitro as well as subsequent autocrine functions of the released TGF-beta within the hippocampal network.


Asunto(s)
Hipocampo/citología , Neuronas/fisiología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Comunicación Autocrina/fisiología , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Femenino , Técnicas In Vitro , Ratones , Neuronas/citología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Embarazo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta3/genética , Factor de Crecimiento Transformador beta3/metabolismo
4.
J Neural Transm (Vienna) ; 114(7): 867-75, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17308981

RESUMEN

TGF-beta-induced apoptosis is essential for embryonic development and mainteanance of adult tissues. Impairment of the apoptotic pathway, regulated by TGF-beta, plays a center role in tumorigenesis and manifestations of different diseases. TIEG2/KLF11 is a recently identified human TGF-beta-inducible zinc finger protein belonging to the family of Sp1/KLF-like transcription factors. In human and murine tissues it has been shown that TIEG1 and TIEG2 induce apoptosis and inhibit cell growth. Since TGF-beta and Tieg1 are able to induce apoptosis in the oligodendroglial cell line OLI-neu, we analysed the ability of TIEG2 to mimic the effects observed after treatment with TGF-beta and overexpression of Tieg1. Herein we report that TIEG2 induces Caspase3-dependent apoptosis in murine OLI-neu cells. Furthermore, we could demonstrate that TIEG2 decreases the levels of the anti-apoptotic protein Bcl-X(L) and inhibits transcription driven by the Bcl-X(L) promoter. These data suggest that TIEG2 serves as a downstream mediator of TGF-beta, bridging TGF-beta-dependent signaling to the intracellular pathway of apoptosis.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Regulación hacia Abajo/genética , Regulación de la Expresión Génica/fisiología , Oligodendroglía/metabolismo , Proteínas Represoras/fisiología , Proteína bcl-X/antagonistas & inhibidores , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Muerte Celular/genética , Línea Celular , Humanos , Ratones , Oligodendroglía/patología , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Transcripción Genética , Proteína bcl-X/biosíntesis , Proteína bcl-X/genética
5.
Eur J Neurosci ; 22(1): 28-38, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16029193

RESUMEN

Transforming growth factor (TGF)-beta and insulin display opposite effects in regulating programmed cell death during vertebrate retina development; the former induces apoptosis while the latter prevents it. In the present study we investigated coordinated actions of TGF-beta and insulin in an organotypic culture system of early postnatal mouse retina. Addition of exogenous TGF-beta resulted in a significant increase in cell death whereas exogenous insulin attenuated apoptosis and was capable of blocking TGF-beta-induced apoptosis. This effect appeared to be modulated via insulin-induced transcriptional down-regulation of TGF-beta receptor II levels. The analysis of downstream signalling molecules also revealed opposite effects of both factors; insulin provided survival signalling by increasing the level of anti-apoptotic Bcl-2 protein expression and phosphorylation and down-regulating caspase 3 activity whereas pro-apoptotic TGF-beta signalling reduced Bcl-2 mRNA levels and Bcl-2 phosphorylation and induced the expression of TGF-induced immediate-early gene (TIEG), a Krüppel-like zinc-finger transcription factor, mimicking TGF-beta activity.


Asunto(s)
Apoptosis/fisiología , Insulina/metabolismo , Neuronas/metabolismo , Retina/crecimiento & desarrollo , Retina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasas/genética , Caspasas/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Interacciones Farmacológicas/fisiología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Insulina/farmacología , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Organogénesis/efectos de los fármacos , Organogénesis/fisiología , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Retina/efectos de los fármacos , Proteínas Smad , Transactivadores/efectos de los fármacos , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
6.
Prog Neurobiol ; 73(3): 151-77, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15236834

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder. The etiology of PD is likely due to combinations of environmental and genetic factors. In addition to the loss of neurons, including dopaminergic neurons in the substantia nigra pars compacta, a further morphologic hallmark of PD is the presence of Lewy bodies and Lewy neurites. The formation of these proteinaceous inclusions involves interaction of several proteins, including alpha-synuclein, synphilin-1, parkin and UCH-L1. Animal models allow to get insight into the mechanisms of several symptoms of PD, allow investigating new therapeutic strategies and, in addition, provide an indispensable tool for basic research. In animals PD does not arise spontaneously, thus, characteristic and specific functional changes have to be mimicked by application of neurotoxic agents or by genetic manipulations. In this review we will focus on genes and gene loci involved in PD, the functions of proteins involved in the formation of cytoplasmatic inclusions, their interactions, and their possible role in PD. In addition, we will review the current animal models of PD.


Asunto(s)
Proteínas Portadoras/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurotoxinas/metabolismo , Enfermedad de Parkinson/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Muerte Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Humanos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Sinucleínas , alfa-Sinucleína
7.
J Cell Biol ; 159(1): 157-67, 2002 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-12370242

RESUMEN

We have previously shown that the neurotrophic effect of glial cell line-derived neurotrophic factor (GDNF) in vitro and in vivo requires the presence of transforming growth factor (TGF)beta. Using primary neurons (chick E8 ciliary) we show that the combination of GDNF plus TGFbeta promotes survival, whereas the single factors do not. This cooperative effect is inhibited by blocking the extracellular signal-regulated kinase (ERK)/MAPK pathway, but not by interfering with the PI3 kinase signaling cascade. Although there is no functional GDNF signaling in the absence of TGFbeta, pretreatment with TGFbeta confers GDNF responsiveness to the cells. This is not due to upregulation of GDNF receptors mRNA and protein, but to TGFbeta-induced recruitment of the glycosyl-phosphatidylinositol-anchored GDNF receptor (GFR)alpha1 to the plasma membrane. This is supported by the fact that GDNF in the presence of a soluble GFRalpha1 can promote survival in the absence of TGFbeta. Our data suggest that TGFbeta is involved in GFRalpha1 membrane translocation, thereby permitting GDNF signaling and neurotrophic effects.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de Drosophila , Factores de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Brefeldino A/farmacología , Supervivencia Celular , Células Cultivadas , Embrión de Pollo , Factor Neurotrófico Derivado de la Línea Celular Glial , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Microdominios de Membrana/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-ret , Proteínas Tirosina Quinasas Receptoras/genética , Factor de Crecimiento Transformador beta/metabolismo
8.
J Neural Transm (Vienna) ; 109(3): 267-77, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11956950

RESUMEN

Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is a member of the EGF-family of ligands signaling via the EGF-receptor tyrosine kinase. In the present study we show that HB-EGF which is expressed in close proximity of developing mesencephalic dopaminergic neurons promotes the survival of TH-positive neurons in vitro. The survival promoting effect of HB-EGF is mediated via astroglial cells and utilizes the MAPK as well as the Akt-signaling pathway. Most notably endogenous HB-EGF significantly contributes to the survival of TH-+ neurons in control cultures, suggesting a relevant developmental role of HB-EGF for dopaminergic neurons. These findings indicate that HB-EGF may be an important molecule for developing dopaminergic neurons of the ventral midbrain.


Asunto(s)
Astrocitos/metabolismo , Supervivencia Celular/fisiología , Dopamina/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas , Transducción de Señal/fisiología , Sustancia Negra/embriología , Animales , Anticuerpos/farmacología , Astrocitos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Receptores ErbB/efectos de los fármacos , Receptores ErbB/metabolismo , Femenino , Feto , Factor de Crecimiento Similar a EGF de Unión a Heparina , Péptidos y Proteínas de Señalización Intercelular , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Enfermedad de Parkinson/fisiopatología , Embarazo , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo
9.
Mech Dev ; 109(2): 295-302, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11731241

RESUMEN

Adrenal chromaffin cells and sympathetic neurons are related, but phenotypically distinct derivatives of the neural crest. Molecular cues that determine the chromaffin cell phenotype have not yet been identified; in contrast to a widely held belief, glucocorticoid signaling is apparently not relevant (Development 126 (1999) 2935). Transforming growth factor-betas (TGF-betas) regulate various aspects of embryonic development and are expressed in the environment of sympathoadrenal (SA) progenitor cells. We have previously shown that neutralization of endogenous TGF-beta from E4 to E8 in the quail embryo significantly increases numbers of adrenal tyrosine hydroxylase-positive cells. Whether endogenous TGF-beta may also be involved in influencing phenotypic development of adrenal chromaffin cells and their SA progenitors has not been analyzed. We now demonstrate that neutralization of endogenous TGF-beta1, -beta2 and -beta3 with a pan-anti-TGF-beta antibody in quail embryos during distinct time windows does not alter phenotypic development of chromaffin cells. In situ hybridizations revealed unaltered expression of neurofilament (NF-160), synaptotagmin I and neurexin I in adrenal glands. Likewise, the NF-associated antigen 3A10, and polyphosphorylated NF epitopes (RT 97) were unaltered. Most importantly, the typical ultrastructure of adrenal chromaffin cells including their large chromaffin secretory granules, a hallmark of the neuroendocrine phenotype, which distinguishes them from sympathetic neurons, was not affected. We therefore conclude that neutralization of endogenous TGF-beta influences chromaffin cell proliferation, but does not interfere with the development of the typical chromaffin cell phenotype.


Asunto(s)
Glándulas Suprarrenales/citología , Glándulas Suprarrenales/embriología , Proteínas de Unión al Calcio , Células Cromafines/citología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Linaje de la Célula , ADN Complementario/metabolismo , Glicoproteínas , Inmunohistoquímica , Hibridación in Situ , Glicoproteínas de Membrana/metabolismo , Microscopía Electrónica , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuropéptidos , Fenotipo , Codorniz , Células Madre , Sinaptotagmina I , Sinaptotagminas , Factores de Tiempo
10.
Development ; 128(11): 1933-42, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11493517

RESUMEN

Programmed cell death (PCD) is a key phenomenon in the regulation of cell number in multicellular organisms. We have shown that reduction of endogenous transforming growth factor beta (TGF-beta) prevents apoptotic PCD of neurons in the developing peripheral and central nervous system, suggesting that TGF-beta is an important mediator of ontogenetic neuron death. Previous studies suggested that there are other pro-apoptotic molecules, nerve growth factor (NGF) and brain-derived neurotrophic factor, that induce cell death in the nervous system. In the developing chick retina, NGF induces PCD by activation of the p75 receptor. We have studied the role of TGF-beta and its putative interdependence with NGF-mediated PCD in the chick retina. We found that TGF-beta is present in the developing chick retina during the period of PCD and is essentially required to regulate PCD of retinal cells. TGF-beta 2, TGF-beta 3 and the ligand-binding TGF-beta receptor can be detected immunocytochemically in the central retina, a region where apoptosis is most prominent during the early period of PCD. Application of a TGF-beta-neutralizing antibody to chick embryos in ovo resulted in a decrease in the number of TUNEL-positive cells and a reduction of free nucleosome levels. In terms of magnitude, reduction of PCD caused by the neutralization of endogenous TGF-beta was equivalent to that seen after anti-NGF application. Neutralization of both factors did not result in a further decrease in apoptosis, indicating that NGF and TGF-beta may act on the same cell population. Furthermore, neutralization of TGF-beta did not affect the expression of NGF or the p75-receptor. Our results suggest that TGF-beta and NGF are both required to regulate cell death in the chick retina in vivo.


Asunto(s)
Apoptosis , Retina/citología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Recuento de Células , División Celular , Embrión de Pollo , Microglía/citología , Microglía/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/análisis , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Retina/embriología
11.
Eur J Biochem ; 267(24): 6982-8, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11106407

RESUMEN

Transforming growth factors-beta (TGF-beta) are multifunctional molecules with profound biological effects in many developmental processes including regulation of cell proliferation, differentiation, cell adhesion, skeletal development, haematopoiesis, inflammatory responses, and wound healing. To learn about the role of TGF-beta in vivo, phenotypes of targeted mutations of molecules within the TGF-beta signalling pathway, TGF-beta1, -beta2, -beta3, TGF-beta receptor (TbetaR-II) and the signalling molecules SMAD2, SMAD3 and SMAD4, are discussed in this review. The three individual TGF-beta mutants show distinct and only partially overlapping phenotypes. In mice, targeted disruption of the TGF-beta1 gene results in diffuse and lethal inflammation about 3 weeks after birth, suggesting a prominent role of TGF-beta in the regulation of immune cell proliferation and extravasation into tissues. However, just half of the TGF-beta1 (-/-) conceptuses actually reach partuition due to defective haematopoiesis and endothelial differentiation. Targeted disruption of both TGF-beta2 and TGF-beta3 genes results in perinatal lethality. TGF-beta2 null mice exhibit a broad range of developmental defects, including cardiac, lung, craniofacial, limb, eye, ear and urogenital defects, whereas TGF-beta3 gene ablation results exclusively in defective palatogenesis and delayed pulmonary development. The TbetaR-II null phenotype closely resembles that of TGF-beta1 (-/-) conceptuses, which die in utero by E10.5. Loss of SMAD2 or SMAD4 results in related phenotypes: the mutants fail to form an organized egg cylinder, lack mesoderm required for gastrulation and die prior to E8.5. Together, gene ablation within the TGF-beta signalling pathway supports the notion of a prominent role of TGF-beta during development.


Asunto(s)
Desarrollo Embrionario y Fetal/fisiología , Homeostasis/genética , Mutación , Factor de Crecimiento Transformador beta/genética , Animales , Proteínas de Unión al ADN/genética , Marcación de Gen , Ratones , Proteína Smad2 , Transactivadores/genética
12.
J Neurochem ; 75(6): 2227-40, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11080174

RESUMEN

Transforming growth factor-betas (TGF-betas) are among the most widespread and versatile cytokines. Here, we first provide a brief overview of their molecular biology, biochemistry, and signaling. We then review distribution and functions of the three mammalian TGF-beta isoforms, beta1, beta2, and beta3, and their receptors in the developing and adult nervous system. Roles of TGF-betas in the regulation of radial glia, astroglia, oligodendroglia, and microglia are addressed. Finally, we review the current state of knowledge concerning the roles of TGF-betas in controlling neuronal performances, including the regulation of proliferation of neuronal precursors, survival/death decisions, and neuronal differentiation.


Asunto(s)
Sistema Nervioso/crecimiento & desarrollo , Sistema Nervioso/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/química , Factor de Crecimiento Transformador beta/metabolismo , Animales , Muerte Celular , Diferenciación Celular , División Celular , Supervivencia Celular , Humanos , Morfogénesis , Familia de Multigenes , Sistema Nervioso/embriología , Neuroglía/metabolismo , Neuronas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/genética
13.
Nat Neurosci ; 3(11): 1085-90, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11036264

RESUMEN

We show that following immunoneutralization of endogenous transforming growth factors beta (TGF-beta) in the chick embryo, ontogenetic neuron death of ciliary, dorsal root and spinal motor neurons was largely prevented, and neuron losses following limb bud ablation were greatly reduced. Likewise, preventing TGF-beta signaling by treatment with a TbetaR-II fusion protein during the period of ontogenetic cell death in the ciliary ganglion rescued all neurons that normally die. TUNEL staining revealed decreased numbers of apoptotic cells following antibody treatment. Exogenous TGF-beta rescued the TGF-beta-deprived phenotype. We conclude that TGF-beta is critical in regulating ontogenetic neuron death as well as cell death following neuronal target deprivation.


Asunto(s)
Apoptosis/fisiología , Cuerpo Ciliar/fisiología , Ganglios Espinales/fisiología , Neuronas Motoras/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Anticuerpos/farmacología , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Supervivencia Celular/fisiología , Embrión de Pollo , Cuerpo Ciliar/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Inmunoglobulina G/farmacología , Neuronas Motoras/efectos de los fármacos , Factor de Crecimiento Transformador beta/efectos de los fármacos
14.
Exp Neurol ; 164(2): 303-13, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10915569

RESUMEN

Chromaffin cells have been recognized for their ability to transform into sympathetic ganglion-like cells in response to nerve growth factor (NGF) or to stimulation of other neurotrophic factors. Transforming growth factor beta (TGFbeta) family members have been shown to potentiate the effect of different trophic factors. The aim of this study was to investigate if TGFbeta may influence NGF-induced neuronal transformation and regulation of NGF, TGFbeta1, and their receptors in the adult rat chromaffin tissue after grafting. Intraocular transplantation of adult chromaffin tissue was employed and grafts were treated with TGFbeta1 and/or NGF. Graft survival time was 18 days after which the grafts were processed for TGFbeta luciferase detection assay, NGF enzyme immunoassay, or in situ hybridization. In grafts stimulated with NGF, increased levels of TGFbeta1 and TGFbeta1 mRNA were detected. When grafts instead were treated with TGFbeta1, enhanced levels of NGF protein were found. Furthermore, a positive mRNA signal corresponding to the transforming growth factor II receptor (TbetaRII) was found in the chromaffin cells of the normal adrenal medulla as well as after grafting. No increase of TbetaRII mRNA levels was detected after transplantation or after TGFbeta1 treatment. Instead a reduction of TbetaRII mRNA expression was noted after NGF treatment. NGF stimulation of grafts increased the message for NGF receptors p75 and trkA in the chromaffin transplants. Grafts processed for evaluations of neurite outgrowth were allowed to survive for 28 days and were injected weekly with NGF and/or TGFbeta1. NGF treatment resulted in a robust innervation of the host irides. TGFbeta1 had no additive effect on nerve fiber formation when combined with NGF. Combined treatment of NGF and anti-TGFbeta1 resulted in a significantly larger area of reinnervation. In conclusion, it was found that NGF and TGFbeta1 may regulate the expression of each other's protein in adult chromaffin grafts. Furthermore, TbetaRII mRNA was present in the adult rat chromaffin cells and became downregulated as a result of NGF stimulation. Although no synergistic effects of TGFbeta1 were found on NGF-induced neurite outgrowth, it was found that TGFbeta1 and NGF signaling are closely linked in the chromaffin cells of the adrenal medulla.


Asunto(s)
Médula Suprarrenal/metabolismo , Células Cromafines/metabolismo , Células Cromafines/trasplante , Factor de Crecimiento Nervioso/biosíntesis , Factor de Crecimiento Transformador beta/biosíntesis , Médula Suprarrenal/citología , Médula Suprarrenal/efectos de los fármacos , Animales , Cámara Anterior/citología , Cámara Anterior/cirugía , Supervivencia Celular/efectos de los fármacos , Células Cromafines/citología , Células Cromafines/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Vías de Administración de Medicamentos , Femenino , Supervivencia de Injerto , Hibridación in Situ , Inyecciones , Factor de Crecimiento Nervioso/administración & dosificación , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Proteínas Serina-Treonina Quinasas , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptor trkA/genética , Receptor trkA/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/administración & dosificación , Factor de Crecimiento Transformador beta/genética
15.
Exp Neurol ; 163(1): 55-62, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10785444

RESUMEN

Free radical formation is considered to be a major cause of dopaminergic (DAergic) cell death in the substantia nigra leading to Parkinson's disease (PD). In this study we employed several radical donors including iron and sodium nitroprusside to induce toxic effects on DAergic neurons cultured from the embryonic rat midbrain floor. Overall cell survival was assessed by assaying LDH, and DAergic neuron survival was monitored by counting tyrosine hydroxylase-positive cells. Our data suggest that the DAergic neuron population is about fourfold more susceptible to free-radical-mediated damage than the total population of midbrain neurons. Application of the neurotrophic factors GDNF and NT-4, for which DAergic neurons have specific receptors, prior to toxin administration protected these neurons from toxin-mediated death, which, fully or in part, occurs under the signs of apoptosis. These findings underscore the importance of GDNF and NT-4 in designing future therapeutical concepts for PD.


Asunto(s)
Hierro/toxicidad , Factores de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Óxido Nítrico/toxicidad , Animales , Apoptosis , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Radicales Libres/antagonistas & inhibidores , Radicales Libres/toxicidad , Factor Neurotrófico Derivado de la Línea Celular Glial , Mesencéfalo/citología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/embriología , Mesencéfalo/enzimología , Neuronas/citología , Neuronas/enzimología , Neurotrofina 3/farmacología , Nitroprusiato/toxicidad , Ratas , Ratas Wistar , Tirosina 3-Monooxigenasa/metabolismo
16.
Cytokine Growth Factor Rev ; 11(1-2): 97-102, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10708957

RESUMEN

This article summarizes and interprets recent data from our laboratories suggesting that transforming growth factor-ss (TGF-ss1, -ss2, -ss3) is essentially required, in vitro and in vivo, for the neurotrophic signaling of glial cell line-derived neurotrophic factor (GDNF). TGF-ss, which is synthesized by and released from neurons, also synergizes with neurotrophins and members of the neurokine and fibroblast growth factor families by increasing their efficacies. However, when applied to purified neuron populations without other factors being added, TGF-ss does not promote survival or differentiation. Together, these data suggest that neither TGF-ss nor GDNF fulfil essential criteria of a typical neurotrophic factor, as e.g. nerve growth factor (NGF). Moreover, the neurotrophic activity of NGF and other classic neurotrophic factors is apparently based, to a significant extent, on their co-operativity with TGF-ss. Mechanisms, by which TGF-ss generates neurotrophic effects and synergizes with other cytokines are beginning to emerge. Recruitment and/or stabilization of receptors and cross-talks at different levels of signal transduction are likely to be implied in generating the neurotrophic potential of the TGF-ss/cytokine synergisms. Together, these data outline a novel role of TGF-ss in a key event of nervous system development, ontogenetic neuron death. Conceptually more important, however, may be the broadening of the neurotrophic factor concept, which now has to imply the possibility that two cytokines, each being ineffective by itself, become neurotrophically active when acting in concert.


Asunto(s)
Citocinas/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Factor Neurotrófico Ciliar/metabolismo , Ganglios Sensoriales/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial , Humanos , Proteínas del Tejido Nervioso/metabolismo
17.
J Neurosci Res ; 59(3): 379-83, 2000 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-10679774

RESUMEN

Chromaffin cells and sympathetic neurons are derivatives of the sympathoadrenal cell lineage of the neural crest. Although they are similar with respect to many cell biological aspects, chromaffin cells, in contrast to sympathetic neurons, continue to synthesize DNA and proliferate through their whole life span. Large numbers of neural and hormonal signals have been implicated in the regulation of chromaffin cell proliferation based on in vitro studies. We have previously shown that chromaffin cells synthesize and release transforming growth factor-beta (TGF-beta) and that exogenously applied TGF-beta suppresses chromaffin cell proliferation in vitro. We show now that TGF-beta is also a physiologically relevant factor in the control of cell division in developing chromaffin cells. We have neutralized endogenous TGF-beta in quail embryos using a monoclonal antibody recognizing all three TGF-beta isoforms, TGF-beta1, -beta2, and -beta3. Embryos deprived of TGF-beta show a prominent increase in numbers of tyrosine hydroxylase (TH)-immunoreactive adrenal chromaffin cells and TH-positive cells incorporating 5'-bromo-2'deoxyuridine. This is the first documentation of the physiological significance of a factor that has been suggested to play a role in the regulation of chromaffin cell mitosis based on in vitro experiments.


Asunto(s)
Glándulas Suprarrenales/citología , Glándulas Suprarrenales/metabolismo , Células Cromafines/citología , Células Cromafines/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Glándulas Suprarrenales/embriología , Glándulas Suprarrenales/enzimología , Animales , Recuento de Células , División Celular/fisiología , Embrión de Pollo , Células Cromafines/enzimología , Oxidación-Reducción , Codorniz/embriología , Tirosina 3-Monooxigenasa/metabolismo
18.
J Neurosci ; 20(23): 8597-603, 2000 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11102463

RESUMEN

Transforming growth factor-betas (TGF-betas) constitute an expanding family of multifunctional cytokines with prominent roles in development, cell proliferation, differentiation, and repair. We have cloned, expressed, and raised antibodies against a distant member of the TGF-betas, growth/differentiation factor-15 (GDF-15). GDF-15 is identical to macrophage inhibitory cytokine-1 (MIC-1). GDF-15/MIC-1 mRNA and protein are widely distributed in the developing and adult CNS and peripheral nervous systems, including choroid plexus and CSF. GDF-15/MIC-1 is a potent survival promoting and protective factor for cultured and iron-intoxicated dopaminergic (DAergic) neurons cultured from the embryonic rat midbrain floor. The trophic effect of GDF-15/MIC-1 was not accompanied by an increase in cell proliferation and astroglial maturation, suggesting that GDF-15/MIC-1 probably acts directly on neurons. GDF-15/MIC-1 also protects 6-hydroxydopamine (6-OHDA)-lesioned nigrostriatal DAergic neurons in vivo. Unilateral injections of GDF-15/MIC-1 into the medial forebrain bundle just above the substantia nigra (SN) and into the left ventricle (20 microgram each) immediately before a 6-OHDA injection (8 microgram) prevented 6-OHDA-induced rotational behavior and significantly reduced losses of DAergic neurons in the SN. This protection was evident for at least 1 month. Administration of 5 microgram of GDF-15/MIC-1 in the same paradigm also provided significant neuroprotection. GDF-15/MIC-1 also promoted the serotonergic phenotype of cultured raphe neurons but did not support survival of rat motoneurons. Thus, GDF-15/MIC-1 is a novel neurotrophic factor with prominent effects on DAergic and serotonergic neurons. GDF-15/MIC-1 may therefore have a potential for the treatment of Parkinson's disease and disorders of the serotonergic system.


Asunto(s)
Citocinas/metabolismo , Dopamina/metabolismo , Mesencéfalo/metabolismo , Neuronas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Clonación Molecular , Cuerpo Estriado/efectos de los fármacos , Citocinas/genética , Citocinas/farmacología , Femenino , Factor 15 de Diferenciación de Crecimiento , Humanos , Hierro/farmacología , Cinesis/efectos de los fármacos , Mesencéfalo/citología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/embriología , Neuronas Motoras/citología , Neuronas Motoras/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Especificidad de Órganos , Oxidopamina/antagonistas & inhibidores , Núcleos del Rafe/citología , Núcleos del Rafe/efectos de los fármacos , Núcleos del Rafe/embriología , Núcleos del Rafe/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Serotonina/metabolismo , Médula Espinal , Sustancia Negra/efectos de los fármacos , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología
19.
J Neural Transm Suppl ; (60): 273-6, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11205146

RESUMEN

We have cloned, expressed, and raised antibodies against a novel member of the TGF-beta superfamily, growth/differentiation factor-15 (GDF-15). The predicted protein is identical to macrophage inhibitory cytokine-1 (MIC-1), which was discovered simultaneously. GDF-15 is a more distant member of the TGF-beta superfamily and does not belong to one of the known TGF-beta subfamilies. In the CNS, GDF-15/MIC-1 mRNA is abundantly expressed by the choroid plexus. In addition we have preliminary evidence that GDF-15/MIC-1 is a potent trophic factor for selected classes of neurons in vitro and in vivo. Thus, GDF-15 is a novel neurotrophic factor with prospects for the treatment of disorders of the CNS.


Asunto(s)
Citocinas , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Líquido Cefalorraquídeo/metabolismo , Factor 15 de Diferenciación de Crecimiento , Humanos , Estructura Terciaria de Proteína/fisiología , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido
20.
Eur J Clin Invest ; 30 Suppl 3: 87-90, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11281376

RESUMEN

Based on recent evidence from in vitro and gene knockout/insertion studies, this short review summarizes the molecular scenario underlying the development of adrenal chromaffin cells and their preganglionic innervation. During migration of neural crest cells from the dorsal surface of the neural tube to their destinations in the sympathetic primordia and adrenal glands, precursors of the so-called sympathoadrenal (SA) cell lineage are exposed to signals from the notochord and ventral neural tube probably including the protein, Sonic hedgehog. These, and signals in the region of the dorsal aorta (members of the family of bone morphogentic proteins), where SA progenitor cells subsequently assemble, are essential for the induction of the adrenergic phenotype. SA progenitor cells subsequently differentiate into paravertebral and prevertebral sympathetic neurones, intra- and extra-adrenal chromaffin cells and intermediate SIF (small intensely fluorescent) cells. Based on in vitro studies with isolated SA and chromaffin progenitor cells, glucocortiocids have been claimed as essential for suppressing neuronal commitment and for channelling SA cells towards the chromaffin phenotype. However, mice deficient for a functional glucocorticoid receptor possess the full complement of adrenal chromaffin cells at birth, suggesting that signals other than glucocorticoid hormones may be important in triggering chromaffin cell differentiation. The cholinergic neurones that are preganglionic to adrenal chromaffin cells have their cell bodies located in the intermediolateral column (IML) of the spinal cord. For their normal development, these neurones require signals from the adrenal medulla, which include neurotrophin-4, a major neurotrophic factor of adrenal chromaffin cells. Taken together, these data provide a more complete picture of molecular signalling in the development of one of the most important neuroendocrine tissues in vertebrates.


Asunto(s)
Fibras Autónomas Preganglionares/fisiología , Células Cromafines/fisiología , Células Madre/fisiología , Sistema Nervioso Simpático/fisiología , Animales , Glucocorticoides/fisiología , Humanos , Cresta Neural/citología , Cresta Neural/fisiología , Médula Espinal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA