Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Mol Psychiatry ; 22(6): 910-919, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27698430

RESUMEN

Many antidepressants stimulate adult hippocampal neurogenesis, but the mechanisms by which they increase neurogenesis and modulate behavior are incompletely understood. Here we show that hippocampal bone morphogenetic protein (BMP) signaling is modulated by antidepressant treatment, and that the changes in BMP signaling mediate effects of antidepressant treatment on neural progenitor cell proliferation and behavior. Treatment with the selective serotonin reuptake inhibitor fluoxetine suppressed BMP signaling in the adult mouse hippocampus both by decreasing levels of BMP4 ligand and increasing production of the BMP inhibitor noggin. Increasing BMP signaling in the hippocampus via viral overexpression of BMP4 blocked the effects of fluoxetine on proliferation in the dentate gyrus and on depressive behavior. Conversely, inhibiting BMP signaling via viral overexpression of noggin in the hippocampus or infusion of noggin into the ventricles exerted antidepressant and anxiolytic activity along with an increase in hippocampal neurogenesis. Similarly, conditional genetic deletion of the type II BMP receptor in Ascl1-expressing cells promoted neurogenesis and reduced anxiety- and depression-like behaviors, suggesting that neural progenitor cells contribute to the effects of BMP signaling on affective behavior. These observations indicate that BMP signaling in the hippocampus regulates depressive behavior, and that decreasing BMP signaling may be required for the effects of some antidepressants. Thus BMP signaling is a new and powerful potential target for the treatment of depression.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/fisiología , Animales , Ansiolíticos/metabolismo , Ansiolíticos/farmacología , Antidepresivos/metabolismo , Antidepresivos/farmacología , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/fisiología , Proliferación Celular/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Depresión/tratamiento farmacológico , Trastorno Depresivo/tratamiento farmacológico , Fluoxetina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo
2.
J Intern Med ; 267(1): 71-88, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20059645

RESUMEN

Peptide nanostructures containing bioactive signals offer exciting novel therapies of broad potential impact in regenerative medicine. These nanostructures can be designed through self-assembly strategies and supramolecular chemistry, and have the potential to combine bioactivity for multiple targets with biocompatibility. It is also possible to multiplex their functions by using them to deliver proteins, nucleic acids, drugs and cells. In this review, we illustrate progress made in this new field by our group and others using peptide-based nanotechnology. Specifically, we highlight the use of self-assembling peptide amphiphiles towards applications in the regeneration of the central nervous system, vasculature and hard tissue along with the transplant of islets and the controlled release of nitric oxide to prevent neointimal hyperplasia. Also, we discuss other self-assembling oligopeptide technology and the progress made with these materials towards the development of potential therapies.


Asunto(s)
Nanotecnología/métodos , Péptidos/síntesis química , Medicina Regenerativa/métodos , Animales , Materiales Biocompatibles , Humanos , Nanoestructuras , Nanotecnología/tendencias , Unión Proteica , Medicina Regenerativa/tendencias
3.
J Neurosci Res ; 85(16): 3535-44, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17803219

RESUMEN

TWEAK/Fn14 signaling regulates progenitor cell proliferation, differentiation, and survival in multiple organ systems. This study examined the effects of TWEAK (tumor necrosis factor-like weak inducer of apoptosis) treatment on cultured mouse neural progenitor cells. The receptor for TWEAK is expressed by neural progenitor cells from the early embryonic stages through postnatal development. Although embryonic day 12 (E12) and postnatal day 1 (PN1) neural progenitor cells both express the receptor for TWEAK, TWEAK treatment of cultured E12 and PN1 progenitor cells resulted in age-dependent effects on proliferation and on neurite extension by neuronal progeny. TWEAK treatment did not alter proliferation of E12 neural progenitor cells but shifted PN1 progenitor cells toward cell-cycle phases G0 and G1 and reduced the rate at which they incorporated CldU. Conversely, the effects of TWEAK on axon elongation were more prominent in the earlier developmental stage. TWEAK induced extensive neurite outgrowth by the neuronal progeny of E12 but not PN1 progenitors. Treatment of the E12 progenitor cells with a TWEAK-neutralizing antibody repressed neurite extension, indicating that endogenous activation of this pathway may be required for neurite extension by the embryonic neuronal progeny. These studies indicate that TWEAK/Fn14 receptor activation exerts different effects on neural progenitor cells and their progeny depending on the developmental stage of the cells.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/embriología , Neuronas/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Células Madre/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocina TWEAK , Regulación del Desarrollo de la Expresión Génica/genética , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/metabolismo , Conos de Crecimiento/ultraestructura , Ratones , Receptores del Factor de Necrosis Tumoral/agonistas , Receptores del Factor de Necrosis Tumoral/genética , Células Madre/citología , Células Madre/efectos de los fármacos , Receptor de TWEAK , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/farmacología
4.
J Med Genet ; 42(12): 940-6, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15831595

RESUMEN

Neural tube defects (NTDs) are the second most common birth defects (1 in 1000 live births) in the world. Periconceptional maternal folate supplementation reduces NTD risk by 50-70%; however, studies of folate related and other developmental genes in humans have failed to definitively identify a major causal gene for NTD. The aetiology of NTDs remains unknown and both genetic and environmental factors are implicated. We present findings from a microsatellite based screen of 44 multiplex pedigrees ascertained through the NTD Collaborative Group. For the linkage analysis, we defined our phenotype narrowly by considering individuals with a lumbosacral level myelomeningocele as affected, then we expanded the phenotype to include all types of NTDs. Two point parametric analyses were performed using VITESSE and HOMOG. Multipoint parametric and nonparametric analyses were performed using ALLEGRO. Initial results identified chromosomes 7 and 10, both with maximum parametric multipoint lod scores (Mlod) >2.0. Chromosome 7 produced the highest score in the 24 cM interval between D7S3056 and D7S3051 (parametric Mlod 2.45; nonparametric Mlod 1.89). Further investigation demonstrated that results on chromosome 7 were being primarily driven by a single large pedigree (parametric Mlod 2.40). When this family was removed from analysis, chromosome 10 was the most interesting region, with a peak Mlod of 2.25 at D10S1731. Based on mouse human synteny, two candidate genes (Meox2, Twist1) were identified on chromosome 7. A review of public databases revealed three biologically plausible candidates (FGFR2, GFRA1, Pax2) on chromosome 10. The results from this screen provide valuable positional data for prioritisation of candidate gene assessment in future studies of NTDs.


Asunto(s)
Cromosomas Humanos Par 10 , Cromosomas Humanos Par 7 , Ligamiento Genético , Genoma Humano , Cresta Neural/patología , Defectos del Tubo Neural/genética , Salud de la Familia , Femenino , Marcadores Genéticos , Genotipo , Humanos , Masculino , Modelos Genéticos , Linaje , Mapeo Físico de Cromosoma
5.
Dev Biol ; 237(1): 212-21, 2001 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-11518517

RESUMEN

The differentiation, survival, and proliferation of developing sympathetic neuroblasts are all coordinately promoted by neurotrophins. In this study, we demonstrate that bone morphogenetic protein 4 (BMP4), a factor known to be necessary for the differentiation of sympathetic neurons (Schneider et al., 1999), conversely reduces both survival and proliferation of cultured E14 sympathetic neuroblasts. The anti-proliferative effects of BMP4 occur more rapidly than the pro-apoptotic actions and appear to involve different intracellular mechanisms. BMP4 treatment induces expression of the transcription factor Msx-2 and the cyclin-dependent kinase inhibitor p21(CIP1/WAF1) (p21). Treatment of cells with oligonucleotides antisense to either of these genes prevents cell death after BMP4 treatment but does not significantly alter the anti-proliferative effects. Thus Msx-2 and p21 are necessary for BMP4-mediated cell death but not for promotion of exit from cell cycle. Although treatment of cultured E14 sympathetic neuroblasts with neurotrophins alone did not alter cell numbers, BMP4-induced cell death was prevented by co-treatment with either neurotrophin-3 (NT-3) or nerve growth factor (NGF). This suggests that BMP4 may also induce dependence of the cells on neurotrophins for survival. Thus, sympathetic neuron numbers may be determined in part by factors that inhibit the proliferation and survival of neuroblasts and make them dependent upon exogenous factors for survival.


Asunto(s)
Apoptosis , Proteínas Morfogenéticas Óseas/farmacología , Ciclinas/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas Proto-Oncogénicas c-bcl-2 , Ganglio Cervical Superior/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 4 , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Proteínas de Homeodominio , Ratones , Mitosis/efectos de los fármacos , Factor de Crecimiento Nervioso/farmacología , Neurotrofina 3/farmacología , Oligonucleótidos Antisentido/farmacología , Proteínas Proto-Oncogénicas/fisiología , Ratas , Ratas Sprague-Dawley , Ganglio Cervical Superior/citología , Proteína X Asociada a bcl-2
6.
J Neurosci Res ; 61(6): 686-92, 2000 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-10972965

RESUMEN

The purpose of this study was to investigate the role of fas antigen, a member of the TNF receptor family, in cell death after focal cerebral ischemia. Focal ischemia was induced in the Sprague-Dawley rat. Evidence for apoptosis was determined by morphology as well as the presence of DNA fragmentation by the end labeling technique (TUNEL). Immunohistochemistry was performed to detect expression of both fas and fas ligand (fasL). In a separate set of experiments, two groups of mice were studied: lpr (that have a loss of function mutation for fas) and wild type. Infarct volume was measured at 24 hr as well as evidence for apoptosis. Twenty-four hours after ischemia, there was evidence for apoptosis based on morphological criteria as well as the TUNEL technique in the rat. Immunohistochemistry demonstrated increased expression of both fas and fasL in the ischemic region, with maximal staining occurring between 24-48 hr for both. Twenty-four hours after ischemia in the mice, there was evidence of apoptosis in both groups, however, the mutant mice (lpr) had significantly smaller infarcts as compared to the wild type. There was no difference in the cerebrovasculature of the two groups of mice. These data support the hypothesis that apoptosis plays a role in the pathophysiology of focal cerebral ischemia. Furthermore, these data suggest that fas-mediated apoptosis contributes to this process.


Asunto(s)
Apoptosis/fisiología , Ataque Isquémico Transitorio/fisiopatología , Receptor fas/genética , Receptor fas/metabolismo , Animales , Expresión Génica/fisiología , Etiquetado Corte-Fin in Situ , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Ataque Isquémico Transitorio/patología , Masculino , Ratones , Ratones Mutantes , Neuronas/química , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Receptor fas/análisis
7.
J Neurosci ; 20(11): 4198-205, 2000 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10818155

RESUMEN

Apoptosis may result either from positive induction by ligand binding to a plasma membrane receptor or from negative induction attributable to loss of a suppressor signal. For example, apoptosis of developing sympathetic neurons may be induced in culture either by exposure to leukemia inhibitory factor (LIF) or by deprivation of nerve growth factor. This study compared the cell death pathways activated in sympathetic neurons by these two different stimuli. Both types of cell death were developmentally regulated; both were maximal in the immediate postnatal period and disappeared over the next 2 weeks. Both types of cell death were reduced by genetic deletion of Bax or by virally mediated overexpression of Bcl-2. Similarly both were reduced by inhibition of caspase activity or by inhibition of Nedd-2 synthesis with antisense oligonucleotides. Finally, both involved activation of c-Jun N-terminal kinase (JNK) signaling. Nedd-2 expression by sympathetic neurons declined in parallel with the developmental loss of LIF-mediated cell death, suggesting that downregulation of the caspase during development may underlie the loss of cytokine-mediated apoptosis. Treatment of sympathetic neurons with an antibody that blocks the function of the low-affinity neurotrophin receptor (p75(LNTR)) prevented LIF-induced cell death. Similarly genetic deletion of p75(LNTR) prevented apoptosis after LIF treatment. These observations suggest that concurrent p75(LNTR) signaling is necessary for LIF-induced cell death and that cytokine-mediated cell death and growth factor deprivation appear to activate the same intracellular pathways involving JNK signaling.


Asunto(s)
Apoptosis/fisiología , Inhibidores de Crecimiento/fisiología , Interleucina-6 , Linfocinas/fisiología , Neuronas/fisiología , Receptor trkA/genética , Transducción de Señal/fisiología , Sistema Nervioso Simpático/fisiología , Transactivadores/fisiología , Proteínas Adaptadoras Transductoras de Señales , Adenoviridae/genética , Animales , Animales Recién Nacidos , Caspasa 2 , Caspasas/metabolismo , Células Cultivadas , Vectores Genéticos , Inmunohistoquímica , Factor Inhibidor de Leucemia , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Sprague-Dawley , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/fisiología , Sistema Nervioso Simpático/citología , Factores de Transcripción , Proteína X Asociada a bcl-2
8.
Brain Res Brain Res Rev ; 32(1): 57-71, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10751657

RESUMEN

Communication through gap junction channels provides a major signaling mechanism during early brain histogenesis, a developmental time during which neural progenitor cells are inexcitable and do not express ligand-gated channel responses to the major CNS neurotransmitters. Expression of different gap junction types during neurogenesis may therefore define intercellular pathways for transmission of developmentally relevant molecules. To better understand the molecular mechanism(s) by which growth and differentiation of neurons are modulated by gap junction channels, we have been examining the developmental effects of a specific set of cytokines on differentiation and gap junction expression in a conditionally immortalized mouse embryonic hippocampal neuronal progenitor cell line (MK31). When multipotent MK31 cells are in an uncommitted state, they uniformly express the neuroepithelial intermediate filament class VI marker, nestin, are strongly coupled by gap junctions composed of connexin43 (Cx43) and express connexin45 (Cx45) at the mRNA level. As these cells undergo neuronal lineage commitment and exit from cell cycle, they begin to express the early neurofilament marker, NF66, and coupling strength and expression of Cx43 begin to decline with concurrent expression of other connexin proteins, including Cx26, Cx33, Cx36, Cx40 and Cx45. Terminal neuronal differentiation is heralded by the expression of more advanced neurofilament proteins, increased morphologic maturation, the elaboration of inward currents and action potentials that possess mature physiological properties, and changing profiles of expression of connexin subtypes, including upregulation of Cx36 expression. These important developmental transitions are regulated by a complex network of cell cycle checkpoints. To begin to examine the precise roles of gap junction proteins in traversing these developmental checkpoints and in thus regulating neurogenesis, we have focused on individual members of two classes of genes involved in these seminal events: ID (inhibitor of differentiation)-1 and GAS (growth arrest-specific gene)5. When MK31 cells were maintained in an uncommitted state, levels of ID-1 mRNA were high and GAS5 transcripts were essentially undetectable. Application of cytokines that promote neuronal lineage commitment and cell cycle exit resulted in down-regulation of ID-1 and upregulation of GAS5 transcripts, whereas additional cytokine paradigms that promoted terminal neuronal differentiation resulted in the delayed down-regulation of GAS5 mRNA. Stable MK31 transfectants were generated for ID-1 and GAS5. In basal conditions, cellular proliferation was enhanced in the ID-1 transfectants and inhibited in the GAS5 transfectants when compared with control MK31 cells. When cytokine-mediated neurogenesis was examined in these transfected cell lines, constitutive expression of ID-1 inhibited and constitutive expression of GAS5 enhanced initial and terminal stages of neuronal differentiation, with evidence that terminal neuronal maturation in both transfectant lines was associated with decreased cellular viability, possibly due to the presence of conflicting cell cycle-associated developmental signals. These experimental reagents will prove to be valuable experimental tools to help define the functional interrelationships between changing profiles of connexin protein expression and cell cycle regulation during neuronal ontogeny in the mammalian brain. The present review summarizes the current state of research involving the temporal expression of such connexin types in differentiating hippocampal neurons and speculates on the possible role of these intercellular channels in the development and plasticity of the nervous system. In addition, we describe the functional properties and expression pattern of the newly discovered neuronal-specific gap junctional protein, Cx36, in the developing mouse fetal hippocampus and in the rat retina and brain.


Asunto(s)
Conexinas/genética , Uniones Comunicantes/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/crecimiento & desarrollo , Neuronas/fisiología , Sinapsis/fisiología , Animales , Uniones Comunicantes/química , Hipocampo/química , Hipocampo/citología , Neuronas/química , Sinapsis/química
9.
Methods ; 20(2): 129-39, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10671307

RESUMEN

A series of antipeptide antibodies designed to recognize specific sequences of the gap junction protein connexin 43 (Cx43) were developed and characterized immunochemically and immunohistologically. These antibodies bound to gap junctions and, on Western blots, to 43-kDa (often resolved as a doublet) and 41-kDa proteins in samples from heart, leptomeningeal cells, and brain. Relatively little of the 41-kDa protein was detectable in heart homogenates. Cultured rat leptomeningeal cells expressed high levels of the gap junction protein Cx43 and were used to analyze its turnover and phosphorylation. Pulse-chase experiments in leptomeningeal cells with [(35)S]methionine indicated that the 41-kDa form of connexin 43 was the first immunoprecipitable translation product. Radiolabel subsequently appeared in the lower band of the doublet at 43 kDa, followed by a shift into the higher band and turnover of the protein with a t(1/2) of 2.7 h. Pulse-chase labeling with [(32)P]P(i) indicated that phosphorylation of connexin 43 was limited to the 43-kDa protein, with a t(1/2) of 1.7 h. Treatment with alkaline phosphatase shifted the apparent molecular mass of the 43-kDa protein doublet such that it comigrated with the 41-kDa form. Hence, the 43-kDa protein observed on Western blots of both leptomeningeal cells and heart arises by phosphorylation of the 41 kDa precursor. Phosphorylation of serine residues accounts for most, if not all, of Cx43 phosphorylation in this system.


Asunto(s)
Encéfalo/metabolismo , Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Meninges/metabolismo , Miocardio/metabolismo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Anticuerpos , Encéfalo/citología , Conexina 43/química , Uniones Comunicantes/ultraestructura , Meninges/citología , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Miocardio/citología , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/inmunología , Fosforilación , Ratas
10.
Dev Neurosci ; 22(1-2): 74-85, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10657700

RESUMEN

Although multipotent progenitor cells capable of generating neurons, astrocytes and oligodendrocytes are present within the germinal zones of the brain throughout embryonic, postnatal and adult life, the different neural cell types are generated within discrete temporospatial developmental windows. This might suggest that multipotent progenitor cells encounter different signals during each developmental stage, thus accounting for separate waves of lineage commitment and cellular differentiation. This study demonstrates, however, that progenitor cell responses to the same class of signals, the bone morphogenetic proteins (BMPs), change during ontogeny, and that these same signals may thus initiate progenitor cell elaboration of several different lineages. BMPs promote cell death and inhibit the proliferation of early (embryonic day 13, E13) ventricular zone progenitor cells. At later embryonic (E16) stages of cerebral cortical development, BMPs exhibit a concentration-dependent dissociation of cellular actions, with either enhancement of neuronal and astroglial elaboration (at 1-10 ng/ml) or potentiation of cell death (at 100 ng/ml). Finally, during the period of perinatal cortical gliogenesis, BMPs enhance astroglial lineage elaboration. By contrast, oligodendroglial lineage elaboration is inhibited by the BMPs at all stages. Further, application of the BMP antagonist noggin to cultured progenitors promotes the generation of oligodendrocytes, indicating that endogenous BMP signaling can actively suppress oligodendrogliogenesis. These observations suggest that developmental changes in neural progenitor cell responsiveness to the BMPs may represent a novel mechanism for orchestrating context-specific cellular events such as lineage elaboration and cellular viability.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Corteza Cerebral/citología , Corteza Cerebral/embriología , Células Madre/efectos de los fármacos , Factor de Crecimiento Transformador beta , Animales , Astrocitos/citología , Proteína Morfogenética Ósea 2 , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/embriología , Desarrollo Embrionario y Fetal/fisiología , Humanos , Oligodendroglía/citología , Ratas , Ratas Sprague-Dawley , Células Madre/citología
11.
J Neurosci Res ; 59(3): 312-20, 2000 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-10679766

RESUMEN

Multipotent neural progenitor cells become progressively more biased towards a glial fate during development coincident with an increase in expression of the epidermal growth factor receptor (EGFR). To determine whether differences in lineage commitment of neural progenitor cells from different stages are causally related to expression of the EGFR and whether generation of glia is EGFR-dependent, we used an EGFR-specific tyrosine kinase inhibitor, PD158780, to block the activation of EGFR in progenitor cells. Treatment of cultured neonatal progenitor cells with PD158780 completely blocked EGF-induced proliferation of the cells but did not affect bFGF-induced proliferation. Nevertheless, treatment with the inhibitor failed to inhibit the generation of astroglia in the presence of either EGF or bFGF. Treatment with bone morphogenetic protein-2 (BMP2) enhanced astroglial differentiation and suppressed oligodendroglial (OL) differentiation. PD158780 treatment had no effect on the BMP2-induced astroglial differentiation or OL suppression. These observations suggest that the generation of astroglia is not dependent on EGFR activation. Because it was still possible that the progenitor cell responses reflected a prior history of EGFR signaling, rat forebrain cells were cultured in the presence of PD158780 from a time (E12.5) preceding expression of the EGFR. After time in culture, the E12.5 cells expressed EGFR by Western analysis both in the presence and in the absence of PD158780, but activation of EGFR kinase (receptor autophosphorylation) was undetectable in the presence of PD158780 and the cells did not proliferate in response to EGF. Nevertheless, astroglial differentiation was normal in PD158780-treated cells both in the absence and in the presence of BMPs or CNTF. Furthermore, the propensity towards glial differentiation increased with time in culture even in the absence of EGFR signaling. This suggests that the increased bias towards glial differentiation during development does not depend on EGFR signaling.


Asunto(s)
Receptores ErbB/fisiología , Prosencéfalo/citología , Prosencéfalo/embriología , Transducción de Señal/fisiología , Células Madre/citología , Animales , Animales Recién Nacidos , Astrocitos/citología , Proteínas Morfogenéticas Óseas/farmacología , División Celular/efectos de los fármacos , Línea Celular , Factor Neurotrófico Ciliar/farmacología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/fisiología , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Prosencéfalo/efectos de los fármacos , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo
12.
J Neurosci ; 19(22): 9848-55, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10559394

RESUMEN

The expression and functional properties of connexin36 (Cx36) were examined in two communication-deficient cell lines (N2A-neuroblastoma and PC-12 cells) transfected with Cx36 and in hippocampal neurons that express the connexin endogenously. Transfected cells expressed the expected 2.9 kb Cx36 transcript and Cx36 immunoreactivity, whereas nontransfected cells were devoid of Cx36. The relationship between steady-state junctional conductance (g(j)) and transjunctional voltage was well described by a two-state Boltzmann equation. The half-inactivation voltage (V(0)), the ratio of minimal to maximal g(j) (g(min)/g(max)), and the equivalent gating charge were +/- 75 mV, 0.55, and 1.75, respectively, indicating that Cx36 exhibits very low voltage sensitivity. Conductance of single Cx36 channels measured with patch pipettes containing 130 mM CsCl was 10-15 pS (n = 15 cell pairs); despite this low unitary conductance, Cx36 channels were permeable to the dye Lucifer yellow. Hippocampal neurons expressed Cx36 both in vivo and in culture. The electrophysiological properties of channels in cultured hippocampal neurons were similar to those of the channels expressed by the transfected cell lines, and the neuronal channels were similarly permeable to Lucifer yellow. The unique combination of weak voltage sensitivity, small unitary conductance, and permeation by anions as large as second messenger molecules endows Cx36 gap junction channels with properties well suited for mediating flexible electrical and biochemical interactions between neurons.


Asunto(s)
Encéfalo/fisiología , Conexinas/genética , Conexinas/fisiología , Proteínas del Ojo/genética , Proteínas del Ojo/fisiología , Uniones Comunicantes/fisiología , Neuronas/fisiología , Animales , Encéfalo/citología , Conexinas/análisis , Conductividad Eléctrica , Proteínas del Ojo/análisis , Hipocampo/fisiología , Neuroblastoma , Especificidad de Órganos , Células PC12 , ARN Mensajero/genética , Ratas , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Transfección , Células Tumorales Cultivadas , Proteína delta-6 de Union Comunicante
13.
Dev Biol ; 215(1): 118-29, 1999 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10525354

RESUMEN

Although Sonic Hedgehog (Shh) plays a critical role in brain development, its actions on neural progenitor cell proliferation and differentiation have not been clearly defined. Transcripts for the putative Shh-receptor genes patched (Ptc) and smoothened (Smo) are expressed by embryonic, postnatal, and adult progenitor cells, suggesting that Shh can act directly on these cells. The recombinant human amino-terminal fragment of Shh protein (Shh-N) alone did not support the survival of cultured progenitor cells, but treatment with Shh-N in the presence of bFGF increased progenitor cell proliferation. Furthermore, treatment of embryonic rat progenitor cells propagated either in primary culture or after mitogen expansion significantly increased the proportions of both beta-tubulin- (neuronal marker) and O4- (oligodendroglial marker) immunoreactive cells and reduced the proportion of nestin- (uncommitted neural progenitor cell marker) immunoreactive cells. By contrast Shh-N had no effect on the elaboration of GFAP- (astroglial marker) immunoreactive cells. Cotreatment with Shh-N and bone morphogenetic protein-2 (BMP2) inhibited the anti-proliferative, astroglial-inductive, and oligodendroglial-suppressive effects of BMP2. Our observations suggest that Shh-N selectively promotes the elaboration of both neuronal and oligodendroglial lineage species and inhibits the effects of BMP2 on progenitor cell proliferation and astroglial differentiation.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas/citología , Oligodendroglía/citología , Proteínas/farmacología , Transactivadores , Factor de Crecimiento Transformador beta , Envejecimiento , Animales , Proteína Morfogenética Ósea 2 , Encéfalo/citología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Desarrollo Embrionario y Fetal , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas Hedgehog , Humanos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Células Madre/citología , Células Madre/efectos de los fármacos
14.
J Neurosci Res ; 56(2): 131-45, 1999 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-10494102

RESUMEN

Multipotent progenitor cells have been identified within periventricular generative zones of the developing and adult brain. To determine whether the environmental responsiveness of these cells changes during development, progenitor cells were cultured from embryonic, postnatal, and adult rat brain in the presence of either basic fibroblast growth factor (bFGF) or epidermal growth factor (EGF). Embryonic cells cultured as intact progenitor neurospheres proliferated more robustly in response to bFGF than to EGF, whereas proliferation of postnatal and adult progenitor cells was enhanced more by EGF than bFGF. Progenitor cells generated in the presence of either bFGF or EGF had the capacity to generate neurons, astrocytes, and oligodendrocytes at all developmental stages. Most embryonic and neonatal bFGF-generated cells differentiated predominantly into neurons, whereas late stage embryonic and neonatal EGF-generated progenitors largely remained in an undifferentiated state. However, later postnatal and adult progenitor species, irrespective of whether they were generated in the presence of bFGF or EGF, gave rise preferentially to astrocytes. Treatment with bone morphogenetic protein (BMP)2 or BMP7 enhanced astroglial differentiation and suppressed oligodendroglial differentiation of both EGF- and bFGF-generated progenitor species, suggesting that the effects of the BMPs are not dependent on EGF receptor activation. Thus, while central nervous system (CNS) progenitor cells retain multipotent capacity and responsiveness to the BMPs throughout development, they exhibit significant changes in other cellular response properties, perhaps reflecting differences in the requirements for specific generative versus regenerative events.


Asunto(s)
Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Diferenciación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Madre/efectos de los fármacos , Animales , Astrocitos/citología , Proteína Morfogenética Ósea 2 , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/farmacología , Encéfalo/crecimiento & desarrollo , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Humanos , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley , Células Madre/fisiología , Factor de Crecimiento Transformador beta/farmacología
15.
J Neurosci ; 19(16): 7077-88, 1999 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-10436062

RESUMEN

Members of the bone morphogenetic protein (BMP) family have been implicated in multiple aspects of neural development in both the CNS and peripheral nervous system. BMP ligands and receptors, as well as the BMP antagonist noggin, are expressed in the developing cerebral cortex, making the BMPs likely candidates for regulating cortical development. To define the role of these factors in the developing cerebral cortex, we examined the effects of BMP2 and BMP4 on cortical cells in vitro. Cells were cultured from embryonic day 13 (E13) and E16 rat cerebral cortex in the absence or presence of different concentrations of fibroblast growth factor 2, a known regulator of cortical cell proliferation and differentiation. At E13, the BMPs promoted cell death and inhibited proliferation of cortical ventricular zone cells, resulting in the generation of fewer neurons and no glia. At E16, the effects of the BMPs were more complex. Concentrations of BMP2 in the range of 1-10 ng/ml promoted neuronal and astroglial differentiation and inhibited oligodendroglial differentiation, whereas 100 ng/ml BMP2 promoted cell death and inhibited proliferation. Addition of the BMP antagonist noggin promoted oligodendrogliogenesis in vitro, demonstrating that endogenous BMP signaling influences the differentiation of cortical cells in vitro. The distribution of BMP2 and noggin within the developing cortex suggests that local concentrations of ligands and antagonists define gradients of BMP signaling during corticogenesis. Together, these results support the hypothesis that the BMPs and their antagonist noggin co-regulate cortical cell fate and morphogenesis.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Corteza Cerebral/fisiología , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Recuento de Células , Diferenciación Celular/fisiología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Desarrollo Embrionario y Fetal/fisiología , Edad Gestacional , Neuronas/citología , Oligodendroglía/citología , Fenotipo , Ratas , Ratas Sprague-Dawley
16.
J Neurosci Res ; 57(4): 557-71, 1999 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-10440906

RESUMEN

Glutamate receptors guide the proliferation, migration, and differentiation of glial cells. Here, we characterize AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid) and NMDA receptor protein expression and function and mRNA expression in hippocampal glial cultures. By immunocytochemistry, GluR2 (the subunit that limits the Ca(2+) permeability of AMPA receptors) exhibited prominent labeling in hippocampal glial cultures. Double-labeling of GluR2 with GFAP and with A2B5 revealed GluR2 subunit expression on type-1 and type-2 astrocyte lineage cells. GluR1 subunit expression was more prominent in type-1 than in type-2 astrocytes. To characterize functional properties of glutamate receptors expressed in cultured hippocampal astrocytes, we performed whole-cell patch clamp recording. Application of L-glutamate, AMPA, and kainate, but not NMDA, to small, rounded cells (morphologically identified as type-2 astrocytes) elicited inward currents which were blocked by the AMPA/kainate antagonist 6-cyano-7-nitroquinoxaline-2, 3-dione (CNQX). Cyclothiazide potentiated AMPA- and kainate-elicited currents, indicative of AMPA-preferring receptors. Current voltage analysis indicated that type-2 astrocyte AMPA receptors were electrically linear, indicative of GluR2-containing, Ca(2+)-impermeable AMPA receptors. By Northern blot analysis, GluR1 mRNA was highest in astrocyte cultures from cerebellum and hippocampus and moderate in astrocyte cultures from neocortex and striatum. GluR3 mRNA was detectable in astrocyte cultures from cerebellum and neocortex. GluR2 and NR1 mRNA expression were not detected in astrocytes cultured from any brain region examined. In situ hybridization studies showed wide expression of GluR1 mRNA in cultured astrocytes; GluR2 and GluR3 mRNAs were near background levels. Thus, cultured type-2 astrocytes express functional AMPA receptors in a cell-specific and region-specific manner, consistent with their role in neuronal-glial communication.


Asunto(s)
Astrocitos/metabolismo , Hipocampo/metabolismo , Receptores AMPA/fisiología , Animales , Northern Blotting , Células Cultivadas , Hipocampo/citología , Inmunohistoquímica , Hibridación in Situ , Ácido Kaínico/farmacología , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores AMPA/biosíntesis , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
17.
Arch Neurol ; 56(7): 780-4, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10404978

RESUMEN

A few brief years ago, damage to the central nervous system was generally perceived to be irreparable, and loss of neurons was largely viewed as an irreversible process. However, major advances in the study of neural progenitor cells have altered these perceptions, and rational approaches to the repair of the damaged nervous system using transplanted progenitor cells now seem feasible. This review will discuss the basic biology of neural progenitor cells, the mechanisms regulating the generation of neurons and glia from these cells, and the techniques that are available for preparing such cells for transplantation into the nervous system. The potential uses for these cells in treating neurologic disease will then be reviewed, and the theoretical and technical problems that may be encountered will be discussed.


Asunto(s)
Regeneración Nerviosa/fisiología , Células Madre/fisiología , Encefalopatías/cirugía , Trasplante de Células/fisiología , Ingeniería Genética , Humanos , Neuroglía/fisiología , Neuronas/fisiología
18.
J Neurosci ; 18(23): 9800-11, 1998 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9822739

RESUMEN

We have previously isolated epidermal growth factor (EGF)-responsive multipotent progenitor cells from the early postnatal rodent cerebral cortex independent of generative zones. In this study we have examined the mechanisms regulating the generation of differentiated oligodendrocytes (OLs) from these multipotent cells. Although cultures of primary cortical OL progenitor cells propagated at clonal density spontaneously gave rise to differentiated OLs in defined medium, cultures of multipotent progenitors isolated from identical regions supported the elaboration of OL progenitors but not differentiated OLs. These observations indicate that the terminal maturation of OL progenitors derived from multipotent cells is dependent on signals present within the cellular environment. Application of cytokines such as basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF), or neurotrophin 3 (NT3) to clonal density cultures of cortical multipotent progenitors increased the proportion of OL progenitors but failed to support the generation of differentiated OLs. By contrast, application of factors that activate gp130/leukemia inhibitory factor beta (LIFbeta) heterodimeric receptors, such as ciliary neurotrophic factor (CNTF), activated signal transducers and activators of transcription-3 in these OL progenitor cells and promoted the generation of differentiated OLs. Clonal analysis also demonstrated that CNTF directly targets OL progenitors derived from the multipotent cells. These observations suggest that two distinct progenitor cell pathways contribute to the generation of differentiated OLs during postnatal cortical gliogenesis. Although oligodendroglial maturation of classical OL progenitor cells is driven by cell autonomous mechanisms, our findings demonstrate that the generation of differentiated OLs from cortical multipotent progenitor cells is dependent on environmental cues, including activation of gp130/LIFbeta receptors.


Asunto(s)
Antígenos CD/fisiología , Inhibidores de Crecimiento/fisiología , Interleucina-6 , Linfocinas/fisiología , Glicoproteínas de Membrana/fisiología , Oligodendroglía/citología , Transducción de Señal/fisiología , Células Madre/citología , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Núcleo Celular/química , Núcleo Celular/fisiología , Factor Neurotrófico Ciliar , Receptor gp130 de Citocinas , Dimerización , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor Inhibidor de Leucemia , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurotrofina 3 , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/fisiología , Prosencéfalo/citología , Ratas , Ratas Sprague-Dawley
19.
Invest Ophthalmol Vis Sci ; 39(11): 2132-9, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9761292

RESUMEN

PURPOSE: To determine whether the p53 protein plays a role in the selective vulnerability of the inner retina to transient ischemia. METHODS: Transient retinal ischemia was induced using a high intraocular pressure (HIOP) model in the Sprague-Dawley rat for 60 minutes. Histopathologic outcome was determined 7 days after ischemia. In addition, analysis for evidence for apoptosis (TdT-dUTP terminal nick-end label [TUNEL] staining) and p53 protein expression (immunohistochemistry) was performed at several points during the reperfusion period. In a separate set of experiments, wild-type mice and two groups of transgenic mice, one homozygous and the other heterozygous for the p53 null gene, were also subjected to HIOP for 60 minutes, and histopathology was performed 7 days later. RESULTS: At 7 days subsequent to 60 minutes of ischemia in the rat, there was marked thinning of the inner retinal layers. There were scattered TUNEL-positive cells within the inner retina, peaking at 24 to 48 hours and persisting for at least 7 days. p53 immunochemistry demonstrated elevated protein levels within the inner retina; this finding peaked at 24 to 48 hours but was no longer present at 4 days after ischemia. TUNEL staining of the inner retina of the mouse was most prominent 24 hours subsequent to ischemia but persisted at 48 hours. Seven days subsequent to 60 minutes of ischemia in the wild-type and transgenic mice, histopathologic evaluation demonstrated preservation of the retinal histoarchitecture in the heterozygous group compared with the wild-type or homozygous animals. CONCLUSIONS: These data further support the hypothesis that the delayed cell death that occurs after transient retinal ischemia is, in part, apoptotic. In addition, they suggest a role for the p53 protein in the selective vulnerability of the inner retina to transient ischemia. p53 protein may be a target for future therapeutic agents in the treatment of disorders of the retina where ischemia plays a pathogenetic role.


Asunto(s)
Daño por Reperfusión/metabolismo , Retina/metabolismo , Enfermedades de la Retina/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Animales , Apoptosis , ADN Nucleotidilexotransferasa , Nucleótidos de Desoxiuracil , Técnica del Anticuerpo Fluorescente Indirecta , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/etiología , Daño por Reperfusión/patología , Retina/ultraestructura , Enfermedades de la Retina/etiología , Enfermedades de la Retina/patología
20.
Neurology ; 51(3): 695-702, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9748012

RESUMEN

BACKGROUND: Preclinical studies have demonstrated that nerve growth factor may prevent or reverse peripheral neuropathy. We have therefore tested the effects of recombinant human nerve growth factor in patients with diabetic polyneuropathy. METHODS: A total of 250 patients with symptomatic diabetic polyneuropathy randomly received either placebo or one of two doses of recombinant human nerve growth factor for 6 months. Patients were assessed for symptoms and signs of polyneuropathy before and after treatment. RESULTS: Compared with placebo, recombinant human nerve growth factor led to significant improvement after 6 months of treatment, as measured by the sensory component of the neurologic examination, two quantitative sensory tests, and the impression of most subjects that their neuropathy had improved. Three prospectively identified multiple endpoint analyses indicated improvements in the nerve growth factor treatment groups over the placebo group in all three analyses (p = 0.032; p = 0.008; p = 0.005). Recombinant human nerve growth factor was well tolerated, with injection site discomfort reported as the most frequent adverse event. CONCLUSIONS: Recombinant human nerve growth factor appears to be safe and shows preliminary evidence of efficacy in patients with symptomatic diabetic polyneuropathy.


Asunto(s)
Neuropatías Diabéticas/tratamiento farmacológico , Factores de Crecimiento Nervioso/uso terapéutico , Adolescente , Adulto , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/efectos adversos , Conducción Nerviosa/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Sensación/efectos de los fármacos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA