Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 281
Filtrar
1.
Biochem Biophys Res Commun ; 321(4): 1024-31, 2004 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-15358131

RESUMEN

Growth differentiation factor 3 (GDF-3) is structurally a bone morphogenetic protein/growth differentiation factor subfamily member of the TGF-beta superfamily. GDF-3 exhibits highest level of expression in white fat tissue in mice and is greatly induced by high fat diet if fat metabolic pathway is blocked. To identify its biological function, GDF-3 was overexpressed in mice by adenovirus mediated gene transfer. Mice transduced with GDF-3 displayed profound weight gain when fed with high fat diet. The phenotypes included greatly expanded adipose tissue mass, increased body adiposity, highly hypertrophic adipocytes, hepatic steatosis, and elevated plasma leptin. GDF-3 stimulated peroxisome proliferator activated receptor expression in adipocytes, a master nuclear receptor that controls adipogenesis. However, GDF-3 was not involved in blood glucose homeostasis or insulin resistance, a condition associated with obesity. In contrast, similar phenotypes were not observed in GDF-3 mice fed with normal chow, indicating that GDF-3 is only active under high lipid load. Thus, GDF-3 is a new non-diabetic adipogenic factor tightly coupled with fat metabolism.


Asunto(s)
Tejido Adiposo/crecimiento & desarrollo , Grasas de la Dieta/administración & dosificación , Péptidos y Proteínas de Señalización Intercelular/fisiología , Células 3T3-L1 , Adipocitos/citología , Adipocitos/fisiología , Animales , Secuencia de Bases , Glucemia/metabolismo , Tamaño de la Célula/genética , Tamaño de la Célula/fisiología , Células Cultivadas , Citocinas/genética , Citocinas/fisiología , ADN/genética , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/patología , Expresión Génica , Factor 3 de Diferenciación de Crecimiento , Humanos , Insulina/sangre , Péptidos y Proteínas de Señalización Intercelular/genética , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/genética , Transducción Genética , Aumento de Peso/genética , Aumento de Peso/fisiología
2.
J Cell Biol ; 166(6): 901-12, 2004 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-15353548

RESUMEN

Actopaxin is an actin and paxillin binding protein that localizes to focal adhesions. It regulates cell spreading and is phosphorylated during mitosis. Herein, we identify a role for actopaxin phosphorylation in cell spreading and migration. Stable clones of U2OS cells expressing actopaxin wild-type (WT), nonphosphorylatable, and phosphomimetic mutants were developed to evaluate actopaxin function. All proteins targeted to focal adhesions, however the nonphosphorylatable mutant inhibited spreading whereas the phosphomimetic mutant cells spread more efficiently than WT cells. Endogenous and WT actopaxin, but not the nonphosphorylatable mutant, were phosphorylated in vivo during cell adhesion/spreading. Expression of the nonphosphorylatable actopaxin mutant significantly reduced cell migration, whereas expression of the phosphomimetic increased cell migration in scrape wound and Boyden chamber migration assays. In vitro kinase assays demonstrate that extracellular signal-regulated protein kinase phosphorylates actopaxin, and treatment of U2OS cells with the MEK1 inhibitor UO126 inhibited adhesion-induced phosphorylation of actopaxin and also inhibited cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteínas de Microfilamentos/metabolismo , Actinina , Secuencia de Aminoácidos , Butadienos/farmacología , Línea Celular Tumoral , Tamaño de la Célula/genética , Células Clonales , Inhibidores Enzimáticos/farmacología , Adhesiones Focales/metabolismo , Humanos , Cinética , Proteínas de Microfilamentos/química , Mutación , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína
3.
Cancer Res ; 64(16): 5659-63, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15313904

RESUMEN

We have established a primary pancreatic duct epithelial cell culture (PDEC) system to investigate the relationship between oncogenic activation of K-ras and pancreatic ductal tumorigenesis. We have found that the acute introduction of physiological levels of oncogenic K-ras (K-rasV12) into quiescent PDECs stimulates S-phase entry and induces a pronounced increase in cell size. Both effects are dependent on the functional integrity of the phosphatidylinositol 3'-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway. In addition, K-rasV12 promotes the loss of epithelial E-cadherin and the gain of mesenchymal N-cadherin in PDEC. Our observations indicate that the oncogenic activation of K-ras is sufficient to elicit mitogenic and morphogenic responses in pancreatic ductal cells and hence is likely to play an instructive role in the initiation of pancreatic ductal adenocarcinoma.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Transformación Celular Neoplásica/genética , Genes ras/genética , Conductos Pancreáticos/fisiología , Neoplasias Pancreáticas/genética , Animales , Carcinoma Ductal Pancreático/patología , Ciclo Celular/genética , División Celular/genética , Tamaño de la Célula/genética , Transformación Celular Neoplásica/patología , Células Cultivadas , Células Epiteliales/patología , Células Epiteliales/fisiología , Regulación de la Expresión Génica , Masculino , Mutación , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/patología , Fosfatidilinositol 3-Quinasas/fisiología , Ratas , Ratas Endogámicas F344
4.
Neuron ; 43(4): 475-85, 2004 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-15312647

RESUMEN

In Brn3b(-/-) mice, where 80% of retinal ganglion cells degenerate early in development, the remaining 20% include most or all ganglion cell types. Cells of the same type cover the retinal surface evenly but tile it incompletely, indicating that a regular mosaic and normal dendritic field size can be maintained in the absence of contact among homotypic cells. In Math5(-/-) mice, where only approximately 5% of ganglion cells are formed, the dendritic arbors of at least two types among the residual ganglion cells are indistinguishable from normal in shape and size, even though throughout development they are separated by millimeters from the nearest neighboring ganglion cell of the same type. It appears that the primary phenotype of retinal ganglion cells can develop without homotypic contact; dendritic repulsion may be an end-stage mechanism that fine-tunes the dendritic arbors for more efficient coverage of the retinal surface.


Asunto(s)
Comunicación Celular/fisiología , Dendritas/fisiología , Células Ganglionares de la Retina/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Comunicación Celular/genética , Recuento de Células/métodos , Tamaño de la Célula/genética , Tamaño de la Célula/fisiología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Dendritas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción Brn-3 , Factor de Transcripción Brn-3B , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
5.
Neuron ; 43(3): 359-72, 2004 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-15294144

RESUMEN

Genetic studies of neocortical area patterning are limited, because mice deficient for candidate regulatory genes die before areas emerge and have other complicating issues. To define roles for the homeodomain transcription factor EMX2, we engineered nestin-Emx2 transgenic mice that overexpress Emx2 in cortical progenitors coincident with expression of endogenous Emx2 and survive postnatally. Cortical size, lamination, thalamus, and thalamocortical pathfinding are normal in homozygous nestin-Emx2 mice. However, primary sensory and motor areas are disproportionately altered in size and shift rostrolaterally. Heterozygous transgenics have similar but smaller changes. Opposite changes are found in heterozygous Emx2 knockout mice. Fgf8 expression in the commissural plate of nestin-Emx2 mice is indistinguishable from wild-type, but Pax6 expression is downregulated in rostral cortical progenitors, suggesting that EMX2 repression of PAX6 specification of rostral identities contributes to reduced rostral areas. We conclude that EMX2 levels in cortical progenitors disproportionately specify sizes and positions of primary cortical areas.


Asunto(s)
Proteínas de Homeodominio/fisiología , Corteza Motora/metabolismo , Neocórtex/metabolismo , Corteza Somatosensorial/metabolismo , Células Madre/metabolismo , Animales , Tamaño de la Célula/genética , Proteínas del Ojo , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Corteza Motora/embriología , Neocórtex/embriología , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box , Ratas , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Corteza Somatosensorial/embriología , Factores de Transcripción
6.
Curr Biol ; 14(16): 1436-50, 2004 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-15324660

RESUMEN

BACKGROUND: 14-3-3 proteins are abundant and conserved polypeptides that mediate the cellular effects of basophilic protein kinases through their ability to bind specific peptide motifs phosphorylated on serine or threonine. RESULTS: We have used mass spectrometry to analyze proteins that associate with 14-3-3 isoforms in HEK293 cells. This identified 170 unique 14-3-3-associated proteins, which show only modest overlap with previous 14-3-3 binding partners isolated by affinity chromatography. To explore this large set of proteins, we developed a domain-based hierarchical clustering technique that distinguishes structurally and functionally related subsets of 14-3-3 target proteins. This analysis revealed a large group of 14-3-3 binding partners that regulate cytoskeletal architecture. Inhibition of 14-3-3 phosphoprotein recognition in vivo indicates the general importance of such interactions in cellular morphology and membrane dynamics. Using tandem proteomic and biochemical approaches, we identify a phospho-dependent 14-3-3 binding site on the A kinase anchoring protein (AKAP)-Lbc, a guanine nucleotide exchange factor (GEF) for the Rho GTPase. 14-3-3 binding to AKAP-Lbc, induced by PKA, suppresses Rho activation in vivo. CONCLUSION: 14-3-3 proteins can potentially engage around 0.6% of the human proteome. Domain-based clustering has identified specific subsets of 14-3-3 targets, including numerous proteins involved in the dynamic control of cell architecture. This notion has been validated by the broad inhibition of 14-3-3 phosphorylation-dependent binding in vivo and by the specific analysis of AKAP-Lbc, a RhoGEF that is controlled by its interaction with 14-3-3.


Asunto(s)
Citoesqueleto/fisiología , Estructura Terciaria de Proteína/fisiología , Proteínas/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Proteínas 14-3-3 , Actinas/fisiología , Animales , Diferenciación Celular/genética , Tamaño de la Célula/genética , Células Cultivadas , Análisis por Conglomerados , Biología Computacional , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citoesqueleto/genética , Cartilla de ADN , ADN Complementario/genética , Perros , Técnica del Anticuerpo Fluorescente , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Espectrometría de Masas , Ratones , Fosforilación , Pruebas de Precipitina , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Proteómica/métodos , Factores de Intercambio de Guanina Nucleótido Rho , Transfección
7.
Dev Cell ; 7(1): 85-93, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15239956

RESUMEN

Cells migrating through a tissue exert force via their cytoskeleton and are themselves subject to tension, but the effects of physical forces on cell behavior in vivo are poorly understood. Border cell migration during Drosophila oogenesis is a useful model for invasive cell movement. We report that this migration requires the activity of the transcriptional factor serum response factor (SRF) and its cofactor MAL-D and present evidence that nuclear accumulation of MAL-D is induced by cell stretching. Border cells that cannot migrate lack nuclear MAL-D but can accumulate it if they are pulled by other migrating cells. Like mammalian MAL, MAL-D also responds to activated Diaphanous, which affects actin dynamics. MAL-D/SRF activity is required to build a robust actin cytoskeleton in the migrating cells; mutant cells break apart when initiating migration. Thus, tension-induced MAL-D activity may provide a feedback mechanism for enhancing cytoskeletal strength during invasive migration.


Asunto(s)
Movimiento Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Oocitos/metabolismo , Oogénesis/genética , Factor de Respuesta Sérica/metabolismo , Citoesqueleto de Actina/metabolismo , Transporte Activo de Núcleo Celular/genética , Animales , Proteínas Portadoras/metabolismo , Tamaño de la Célula/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , ADN Complementario/análisis , ADN Complementario/genética , Drosophila/citología , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/genética , Proteínas de Drosophila/aislamiento & purificación , Retroalimentación/fisiología , Retroalimentación Fisiológica/fisiología , Forminas , Datos de Secuencia Molecular , Mutación/genética , Proteínas Nucleares/genética , Proteínas Nucleares/aislamiento & purificación , Oocitos/citología , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Factor de Respuesta Sérica/genética , Estrés Mecánico , Factores de Transcripción
8.
Mol Cell Neurosci ; 26(3): 450-7, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15234349

RESUMEN

Many cholinergic parasympathetic and enteric neurons require neurturin signaling through GDNF family receptor GFRalpha2 for target innervation. Since a distinct minority of sympathetic neurons are cholinergic, we examined whether GFRalpha2 is important for their development. We detected GFRalpha2 in neonatal sympathetic cholinergic neurons and neurturin mRNA in their target tissues, sweat glands in footpads, and periosteum. Lack of GFRalpha2 in mice did not affect the number of sympathetic cholinergic neurons, but their soma size was decreased in comparison to wild types. In adult and in 3-week-old GFRalpha2 knockout mice, the density of sympathetic cholinergic innervation was reduced by 50-70% in the sweat glands, and was completely absent in the periosteum. Sympathetic noradrenergic innervation of blood vessels in the footpads was unchanged. The density of sympathetic axons in sweat glands was unaffected at postnatal day P4 reflecting successful growth into the target area. Our results indicate that the cholinergic subpopulation of sympathetic neurons requires GFRalpha2 signaling for soma size and for growth or maintenance of target innervation. Thus, neurturin may be a general target-derived innervation factor for postganglionic cholinergic neurons in all parts of the autonomic nervous system.


Asunto(s)
Fibras Adrenérgicas/fisiología , Fibras Colinérgicas/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/deficiencia , Proteínas Tirosina Quinasas Receptoras/genética , Animales , Tamaño de la Célula/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Periostio/citología , Periostio/inervación , Periostio/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-ret , Proteínas Tirosina Quinasas Receptoras/fisiología , Glándulas Sudoríparas/citología , Glándulas Sudoríparas/inervación , Glándulas Sudoríparas/metabolismo
9.
J Chem Neuroanat ; 27(4): 237-46, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15261330

RESUMEN

In order to furnish a combined model of relevance to human inclusion-body myopathy and Alzheimer's disease, transgenic mice expressing human betaAPP-C99 in skeletal muscle and brain under the control of the cytomegalovirus/beta-actin promoter were produced (Tg13592). These transgenic mice develop Abeta deposits in muscles but not in brain. Cell metabolic activity was analyzed in brain regions and muscle by cytochrome oxidase (CO) histochemistry, the terminal enzyme of the electron transport chain. By comparison to age-matched controls of the C57BL/6 strain, CO activity was selectively increased in dark skeletal muscle fibers of Tg13592 mice. In addition, only increases in CO activity were obtained in those brain regions where a significant difference appeared. The CO activity of Tg13592 mice was elevated in several thalamic nuclei, including laterodorsal, ventromedial, and midline as well as submedial, intralaminar, and reticular. In contrast, the groups did not differ in most cortical regions, except for prefrontal, secondary motor, and auditory cortices, and in most brainstem regions, except for cerebellar (fastigial and interpositus) nuclei and related areas (red and lateral vestibular nuclei). No variation in cell density and surface area appeared in conjunction with these enzymatic alterations. The overproduction of betaAPP-C99 fragments in brain without (amyloidosis did not appear to affect the metabolic activity of structures particularly vulnerable in Alzheimer's disease.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Encéfalo/enzimología , Complejo IV de Transporte de Electrones/metabolismo , Metabolismo Energético/genética , Músculo Esquelético/enzimología , Fragmentos de Péptidos/biosíntesis , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/citología , Recuento de Células , Tamaño de la Célula/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Miositis por Cuerpos de Inclusión/genética , Miositis por Cuerpos de Inclusión/metabolismo , Neuronas/citología , Neuronas/enzimología , Fragmentos de Péptidos/genética , Tálamo/citología , Tálamo/enzimología , Regulación hacia Arriba/genética
10.
Exp Cell Res ; 298(2): 624-31, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15265708

RESUMEN

The coxsackie and adenovirus receptor (CAR) is a key factor in adenoviral cancer gene therapy. Reduced expression of CAR during progression of prostate and bladder cancer has been reported. In embryonic development and tissue differentiation, CAR is also differentially expressed. This study suggests a role of CAR expression in cell adhesion and cell motility of human cancer cells. Stable CAR-expressing clones from E-cadherin-deficient A2780 ovarian and CaSki cervical cancer cells with originally low and high CAR expression levels, respectively, were established. CAR reexpression in otherwise singularly growing A2780 parental cells resulted in formation of cell-cell contacts and aggregation in cell clusters. CAR overexpression in cell adhesion-forming CaSki cells did not result in morphological changes. Migration of the A2780 CAR clones was strongly reduced as characterized by using spread-off assays. Using migration chambers, formation of satellite colonies was reduced by 97% in CAR-expressing A2780 cell clones and by 23% in CAR-expressing CaSki cell clones. Parental A2780 and CaSki cells selected for high migratory ability by using migration chambers expressed endogenous CAR on lower levels associated with lower adenoviral transduction efficiency. Our data suggest CAR as a new inhibitory factor for cancer cell migration.


Asunto(s)
Movimiento Celular/genética , Metástasis de la Neoplasia/fisiopatología , Neoplasias/metabolismo , Receptores Virales/metabolismo , Adenoviridae/genética , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular/genética , Agregación Celular/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Tamaño de la Célula/genética , Células Clonales/citología , Células Clonales/metabolismo , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Vectores Genéticos/genética , Humanos , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/prevención & control , Neoplasias/genética , Receptores Virales/genética , Transducción Genética
11.
Cell ; 117(7): 899-913, 2004 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-15210111

RESUMEN

Cyclin-dependent kinase (CDK) activity initiates the eukaryotic cell division cycle by turning on a suite of gene expression in late G1 phase. In metazoans, CDK-dependent phosphorylation of the retinoblastoma tumor suppressor protein (Rb) alleviates repression of E2F and thereby activates G1/S transcription. However, in yeast, an analogous G1 phase target of CDK activity has remained elusive. Here we show that the cell size regulator Whi5 inhibits G1/S transcription and that this inhibition is relieved by CDK-mediated phosphorylation. Deletion of WHI5 bypasses the requirement for upstream activators of the G1/S transcription factors SBF/MBF and thereby accelerates the G1/S transition. Whi5 is recruited to G1/S promoter elements via its interaction with SBF/MBF in vivo and in vitro. In late G1 phase, CDK-dependent phosphorylation dissociates Whi5 from SBF and drives Whi5 out of the nucleus. Elimination of CDK activity at the end of mitosis allows Whi5 to reenter the nucleus to again repress G1/S transcription. These findings harmonize G1/S control in eukaryotes.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Anticuerpos Monoclonales/metabolismo , Núcleo Celular/metabolismo , Tamaño de la Célula/genética , Cromatina/metabolismo , Cruzamientos Genéticos , Epistasis Genética , Fase G1 , Eliminación de Gen , Regulación Fúngica de la Expresión Génica , Genes Fúngicos , Genes Reguladores , Modelos Biológicos , Fosforilación , Pruebas de Precipitina , Regiones Promotoras Genéticas , Proteínas/análisis , ARN/análisis , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Fase S , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Especificidad por Sustrato , Transcripción Genética
12.
J Leukoc Biol ; 76(3): 657-66, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15197230

RESUMEN

To test whether reduced immune responsiveness in early life may be related to the immaturity of neonatal antigen-presenting cells, we comparatively assessed the phenotypic and functional characteristics of dendritic epidermal leukocytes (DEL) and epidermal Langerhans cells (LC) in newborn (NB) and adult mice, respectively. We report that purified, 3-day-cultured DEL do not acquire the morphology and phenotype typical of LC and are significantly weaker stimulators of naive, allogeneic CD4+ and CD8+ T cells than LC. Freshly isolated DEL are twice as efficient as LC in the uptake of fluorescein isothiocyanate-conjugated tracers but are not able to present these to antigen-specific T cell hybridomas. To clarify the underlying cause, cytokine expression of NB and adult epidermal cells (EC) was examined. We found that DEL express considerable amounts of interleukin (IL)-10, that IL-10 in NB EC supernatants partially inhibits LC maturation, and that DEL-enriched EC from IL-10-/- mice induce stronger primary T cell responses compared with those from IL-10+/+ mice. We conclude that IL-10 is one of the factors preventing maturation and differentiation of DEL into immunocompetent LC in intrauterine life and is at least partly responsible for the poor immune responsiveness of neonates.


Asunto(s)
Comunicación Autocrina/inmunología , Diferenciación Celular/inmunología , Epidermis/inmunología , Interleucina-10/fisiología , Células de Langerhans/inmunología , Leucocitos/inmunología , Factores de Edad , Animales , Animales Recién Nacidos , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/genética , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Tamaño de la Célula/efectos de los fármacos , Tamaño de la Célula/genética , Tamaño de la Célula/inmunología , Células Cultivadas , Células Epidérmicas , Epidermis/crecimiento & desarrollo , Interleucina-10/genética , Interleucina-10/farmacología , Células de Langerhans/citología , Células de Langerhans/efectos de los fármacos , Leucocitos/citología , Leucocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/inmunología
13.
J Neuropathol Exp Neurol ; 63(5): 429-40, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15198122

RESUMEN

Down syndrome is the most frequent genetic cause of mental retardation, having an incidence of 1 in 700 live births. In the present study we used a transgenic mouse in vivo library consisting of 4 yeast artificial chromosome (YAC) transgenic mouse lines, each bearing a different fragment of the Down syndrome critical region 1 (DCR-1), implicated in brain abnormalities characterizing this pathology. The 152F7 fragment, in addition to genes also located on the other DCR-1 fragments, bears the DYRK1A gene, encoding for a serine-threonine kinase. The neurobehavioral analysis of these mouse lines showed that DYRK1A overexpressing 152F7 mice but not the other lines display learning impairment and hyperactivity during development. Additionally, 152F7 mice display increased brain weight and neuronal size. At a biochemical level we found DYRK1A overexpression associated with a development-dependent increase in phosphorylation of the transcription factor FKHR and with high levels of cyclin B1, suggesting for the first time in vivo a correlation between DYRK1A overexpression and cell cycle protein alteration. In addition, we found an altered phosphorylation of transcription factors of CREB family. Our findings support a role of DYRK1A overexpression in the neuronal abnormalities seen in Down syndrome and suggest that this pathology is linked to altered levels of proteins involved in the regulation of cell cycle.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Síndrome de Down/genética , Biblioteca Genómica , Proteínas Musculares/metabolismo , Malformaciones del Sistema Nervioso/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas , Animales , Proteínas de Ciclo Celular/genética , Tamaño de la Célula/genética , Aberraciones Cromosómicas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclina B/metabolismo , Ciclina B1 , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Síndrome de Down/metabolismo , Síndrome de Down/fisiopatología , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead , Humanos , Hipercinesia/genética , Hipercinesia/metabolismo , Hipercinesia/fisiopatología , Péptidos y Proteínas de Señalización Intracelular , Discapacidades para el Aprendizaje/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Mutación/genética , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/fisiopatología , Tamaño de los Órganos/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba/genética , Quinasas DyrK
14.
Eur J Neurosci ; 19(11): 2953-66, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15182302

RESUMEN

A synRas mouse model was used expressing constitutively activated Ha-Ras (Val12 mutation) in neurons to investigate the role of Ras-MAPkinase signalling for neuronal connectivity in adult brain. Expression of the transgene in the cortex of these mice starts after neuronal differentiation is completed and allows to directly investigate the effects of enhanced Ras activity in differentiated neurons. Activation of Ha-Ras induced an increase in soma size which was sensitive to MEK inhibitor in postnatal organotypic cultures. Adult cortical pyramidal neurons showed complex structural rearrangements associated with an increased size and ramification of dendritic arborization. Dendritic spine density was elevated and correlated with a twofold increase in number of synapses. In acute brain slices of the somatosensory and of the visual cortex, extracellular field potentials were recorded from layer II/III neurons. The input-output relation of synaptically evoked field potentials revealed a significantly higher basal excitability of the transgenic mice cortex compared to wild-type animals. In whole cell patch clamp preparations, the frequency of AMPA receptor-mediated spontaneous excitatory postsynaptic currents was increased while the ratio between NMDA and AMPA-receptor mediated signal amplitude was unchanged. A pronounced depression of paired pulse facilitation indicated that Ras contributes to changes at the presynaptic site. Furthermore, synRas mice showed an increased synaptic long-term potentiation, which was sensitive to blockers of NMDA-receptors and of MEK. We conclude that neuronal Ras is a common switch of plasticity in adult mammalian brain sculpturing neuronal architecture and synaptic connectivity in concert with tuning synaptic efficacy.


Asunto(s)
Corteza Cerebral/citología , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Células Piramidales/fisiología , Sinapsis/fisiología , Proteínas ras/metabolismo , 2-Amino-5-fosfonovalerato/farmacología , Animales , Animales Recién Nacidos , Axones/ultraestructura , Butadienos/farmacología , Recuento de Células/métodos , Tamaño de la Célula/genética , Tamaño de la Célula/fisiología , Células Cultivadas , Dendritas/ultraestructura , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Histocitoquímica/métodos , Inmunohistoquímica/métodos , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Ratones , Ratones Transgénicos/genética , Microscopía Electrónica/métodos , Nitrilos/farmacología , Técnicas de Placa-Clamp/métodos , Células Piramidales/ultraestructura , Quinoxalinas/farmacología , Ratas , Sinapsis/ultraestructura , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Sinaptofisina/metabolismo , Factores de Tiempo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología , Proteínas ras/genética
15.
Curr Opin Genet Dev ; 14(2): 210-7, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15196469

RESUMEN

Although the mechanisms of cell cycle control are well established, the factors controlling cell growth and target size are still poorly understood. Much evidence suggests that ribosome biogenesis, and in particular the synthesis of the rRNAs, plays a central role not only in permitting growth, but also in regulating it. In the past few years we have begun to penetrate the network linking rRNA gene transcription to growth.


Asunto(s)
Crecimiento/fisiología , ARN Ribosómico/fisiología , Tamaño de la Célula/genética , Tamaño de la Célula/fisiología , Silenciador del Gen/fisiología , ARN Polimerasa I/fisiología , ARN Polimerasa II/fisiología , Levaduras/genética , Levaduras/fisiología
16.
Glia ; 47(2): 180-8, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15185396

RESUMEN

Individuals with tuberous sclerosis complex (TSC) develop central nervous system abnormalities that may reflect astrocyte dysfunction. In an effort to model astrocyte dysfunction in TSC, we generated mice lacking Tsc1 (hamartin) expression in astrocytes and demonstrated that Tsc1-null astrocytes exhibit abnormalities in contact inhibition growth arrest. In this study, we demonstrate that hamartin-deficient astrocytes are also defective in cell size regulation. We show that the increase in Tsc1-null astrocyte size is associated with increased activation of the S6-kinase pathway. In keeping with recent reports that the hamartin/tuberin complex may regulate Rheb and downstream S6K activation, we demonstrate that expression of either Rheb or S6K in primary astrocytes results in increased S6 pathway activation, and that inhibition of Rheb activity in Tsc1-deficient astrocytes using either pharmacologic or genetic strategies markedly reduces S6 activation. Collectively, these observations suggest that TSC inactivation in astrocytes results in defective cell size regulation associated with dysregulated Rheb/mTOR/S6K pathway activity.


Asunto(s)
Astrocitos/metabolismo , Sistema Nervioso Central/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Neuropéptidos/metabolismo , Proteínas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Esclerosis Tuberosa/metabolismo , Animales , Astrocitos/patología , Diferenciación Celular/genética , Tamaño de la Célula/genética , Células Cultivadas , Sistema Nervioso Central/fisiopatología , Inhibición de Contacto/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Inhibidores Enzimáticos/farmacología , Ratones , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/genética , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/metabolismo , Proteínas/genética , Interferencia de ARN , Proteína Homóloga de Ras Enriquecida en el Cerebro , Transducción de Señal/genética , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR , Transfección , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/fisiopatología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor , Regulación hacia Arriba/genética
17.
J Comp Neurol ; 474(1): 136-48, 2004 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-15156583

RESUMEN

Neuronal apoptosis is a key component in the sculpting of tissues during embryonic and postnatal development and is driven largely by the action of caspases. In the mouse olfactory system, caspase-3 and -9 are expressed in olfactory receptor neurons (ORNs) of adult mice, and their selective retrograde activation drives ORN apoptosis following ablation of their target, the olfactory bulb (OB). Here, we show that both of these caspases are expressed at the earliest stages of ORN embryonic development, and their expression is concentrated in outgrowing ORN axons. The retention, in null mice for both caspases, of a population of ORNs that would normally undergo developmental apoptosis beginning at E13 of development, results in a permanently expanded population of ORNs. In turn, in some caspase-3 null mice, the ORN target organ, the OB, also develops abnormally, resulting in the formation of secondary, apparently functional, extracranial ectopic OBs.


Asunto(s)
Apoptosis/fisiología , Caspasas/fisiología , Vías Olfatorias/citología , Neuronas Receptoras Olfatorias/fisiología , Animales , Animales Recién Nacidos , Caspasa 3 , Caspasa 9 , Caspasas/genética , Recuento de Células/métodos , Tamaño de la Célula/genética , Embrión de Mamíferos , Genotipo , Inmunoquímica/métodos , Etiquetado Corte-Fin in Situ/métodos , Ratones , Ratones Noqueados , Vías Olfatorias/fisiología
18.
J Cell Biol ; 165(3): 433-45, 2004 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-15123739

RESUMEN

Biochemical experiments have shown that Smad6 and Smad ubiquitin regulatory factor 1 (Smurf1) block the signal transduction of bone morphogenetic proteins (BMPs). However, their in vivo functions are largely unknown. Here, we generated transgenic mice overexpressing Smad6 in chondrocytes. Smad6 transgenic mice showed postnatal dwarfism with osteopenia and inhibition of Smad1/5/8 phosphorylation in chondrocytes. Endochondral ossification during development in these mice was associated with almost normal chondrocyte proliferation, significantly delayed chondrocyte hypertrophy, and thin trabecular bone. The reduced population of hypertrophic chondrocytes after birth seemed to be related to impaired bone growth and formation. Organ culture of cartilage rudiments showed that chondrocyte hypertrophy induced by BMP2 was inhibited in cartilage prepared from Smad6 transgenic mice. We then generated transgenic mice overexpressing Smurf1 in chondrocytes. Abnormalities were undetectable in Smurf1 transgenic mice. Mating Smad6 and Smurf1 transgenic mice produced double-transgenic pups with more delayed endochondral ossification than Smad6 transgenic mice. These results provided evidence that Smurf1 supports Smad6 function in vivo.


Asunto(s)
Enfermedades Óseas Metabólicas/genética , Cartílago/metabolismo , Condrocitos/metabolismo , Proteínas de Unión al ADN/metabolismo , Enanismo/genética , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Animales Recién Nacidos , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Huesos/metabolismo , Huesos/patología , Huesos/fisiopatología , Cartílago/crecimiento & desarrollo , Cartílago/patología , Diferenciación Celular/genética , Tamaño de la Célula/efectos de los fármacos , Tamaño de la Célula/genética , Condrocitos/efectos de los fármacos , Condrocitos/patología , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Enanismo/metabolismo , Enanismo/patología , Feto , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Ratones Transgénicos , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Fosforilación/efectos de los fármacos , Proteínas Smad , Proteína Smad1 , Proteína smad6 , Transactivadores/genética , Ubiquitina-Proteína Ligasas/genética
19.
Brain Res Dev Brain Res ; 150(1): 41-5, 2004 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-15126036

RESUMEN

Immunohistochemistry for parvalbumin, a marker for primary proprioceptors, was performed on the dorsal root ganglion (DRG) of wildtype and knockout mice for Brn-3a at postnatal day 0 and embryonic day 18.5. The DRG contained many parvalbumin-immunoreactive (ir) neurons in wildtype (5.4%) and knockout mice (5.6%). Cell size analysis demonstrated that such neurons were mostly medium-sized to large in these mice. Therefore, it is unlikely that the survival of proprioceptors is dependent upon Brn-3a in the DRG. In the dorsal column and gray matter of the spinal cord of knockout mice, however, parvalbumin-ir nerve fibers were sparse compared to wildtype mice. The number of parvalbumin-ir varicosities around motoneurons decreased in the mutant. Thus, our data suggest that Brn-3a may play an important role in the central projection and terminal formation of DRG proprioceptors in the spinal cord.


Asunto(s)
Vías Aferentes/metabolismo , Proteínas de Unión al ADN/deficiencia , Ganglios Espinales/metabolismo , Mecanorreceptores/metabolismo , Neuronas Aferentes/metabolismo , Parvalbúminas/metabolismo , Factores de Transcripción/deficiencia , Vías Aferentes/citología , Animales , Diferenciación Celular/genética , Tamaño de la Célula/genética , Proteínas de Unión al ADN/genética , Ganglios Espinales/citología , Ganglios Espinales/embriología , Conos de Crecimiento/metabolismo , Conos de Crecimiento/ultraestructura , Inmunohistoquímica , Mecanorreceptores/citología , Ratones , Ratones Noqueados , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/ultraestructura , Neuronas Aferentes/citología , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Propiocepción/genética , Médula Espinal/citología , Médula Espinal/embriología , Médula Espinal/metabolismo , Factor de Transcripción Brn-3 , Factor de Transcripción Brn-3A , Factores de Transcripción/genética
20.
Development ; 131(11): 2631-40, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15128660

RESUMEN

The FGF receptor Heartless (HTL) is required for mesodermal cell migration in the Drosophila gastrula. We show that mesoderm cells undergo different phases of specific cell shape changes during mesoderm migration. During the migratory phase, the cells adhere to the basal surface of the ectoderm and exhibit extensive protrusive activity. HTL is required for the protrusive activity of the mesoderm cells. Moreover, the early phenotype of htl mutants suggests that HTL is required for the adhesion of mesoderm cells to the ectoderm. In a genetic screen we identified pebble (pbl) as a novel gene required for mesoderm migration. pbl encodes a guanyl nucleotide exchange factor (GEF) for RHO1 and is known as an essential regulator of cytokinesis. We show that the function of PBL in cell migration is independent of the function of PBL in cytokinesis. Although RHO1 acts as a substrate for PBL in cytokinesis, compromising RHO1 function in the mesoderm does not block cell migration. These data suggest that the function of PBL in cell migration might be mediated through a pathway distinct from RHO1. This idea is supported by allele-specific differences in the expressivity of the cytokinesis and cell migration phenotypes of different pbl mutants. We show that PBL is autonomously required in the mesoderm for cell migration. Like HTL, PBL is required for early cell shape changes during mesoderm migration. Expression of a constitutively active form of HTL is unable to rescue the early cellular defects in pbl mutants, suggesting that PBL is required for the ability of HTL to trigger these cell shape changes. These results provide evidence for a novel function of the Rho-GEF PBL in HTL-dependent mesodermal cell migration.


Asunto(s)
Movimiento Celular/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/citología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Mesodermo/citología , Proteínas Tirosina Quinasas/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , División Celular/fisiología , Tamaño de la Célula/genética , Drosophila/embriología , Drosophila/genética , Proteínas de Drosophila/genética , Embrión no Mamífero/citología , Gástrula/citología , Regulación del Desarrollo de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Mutación , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Proteínas Tirosina Quinasas/genética , Receptores de Factores de Crecimiento de Fibroblastos/genética , Factores de Transcripción/genética , Proteína 1 Relacionada con Twist , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA