Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
J Neuroinflammation ; 15(1): 210, 2018 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-30029608

RESUMEN

BACKGROUND: Alzheimer's disease is characterized by progressive accumulation of ß-amyloid (Aß)-containing amyloid plaques, and microglia play a critical role in internalization and degradation of Aß. Our previous research confirmed that Nogo-66 binding to Nogo receptors (NgR) expressed on microglia inhibits cell adhesion and migration in vitro. METHODS: The adhesion and migration of microglia isolated from WT and APP/PS1 mice from different ages were measured by adhesion assays and transwells. After NEP1-40 (a competitive antagonist of Nogo/NgR pathway) was intracerebroventricularly administered via mini-osmotic pumps for 2 months in APP/PS1 transgenic mice, microglial recruitment toward Aß deposits and CD36 expression were determined. RESULTS: In this paper, we found that aging led to a reduction of microglia adhesion and migration to fAß1-42 in WT and APP/PS1 mice. The adhesion and migration of microglia to fAß1-42 were downregulated by the Nogo, which was mediated by NgR, and the increased inhibitory effects of the Nogo could be observed in aged mice. Moreover, Rho GTPases contributed to the effects of the Nogo on adhesion and migration of microglia to fAß1-42 by regulating cytoskeleton arrangement. Furthermore, blocking the Nogo/NgR pathway enhanced recruitment of microglia toward Aß deposits and expression of CD36 in APP/PS1 mice. CONCLUSION: Taken together, Nogo/NgR pathway could take part in Aß pathology in AD by modulating microglial adhesion and migration to Aß and the Nogo/NgR pathway might be an important target for treating AD.


Asunto(s)
Envejecimiento , Péptidos beta-Amiloides/farmacología , Adhesión Celular/efectos de los fármacos , Microglía/efectos de los fármacos , Proteínas Nogo/metabolismo , Receptores Nogo/metabolismo , Fragmentos de Péptidos/farmacología , Envejecimiento/efectos de los fármacos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/patología , Adhesión Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de la Mielina/farmacología , Presenilina-1/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
2.
J Neuroinflammation ; 13(1): 56, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26939570

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is characterized by extracellular ß-amyloid (Aß) plaques, neurofibrillary tangles (NFTs), and microglia-dominated neuroinflammation. The Nogo/NgR signal pathway is involved in AD pathological features, but the detailed mechanism needs further investigation. Our previous studies have confirmed that the activation of NgR on microglia by Nogo promotes the expression of proinflammatory cytokines and inhibits cell adhesion and migration behaviors. In the present study, we investigated the effects of Nogo/NgR signaling pathway on the pathological features of AD and possible mechanisms. METHODS: After NEP1-40 (a competitive antagonist of Nogo/NgR pathway) was intracerebroventricularly administered via mini-osmotic pumps for 2 months in amyloid precursor protein (APP)/PS1 transgenic mice, plaque load, tau phosphorylation, and inflammatory responses were determined. After primary mouse neurons were exposed to the conditioned medium from BV-2 microglia stimulated by Nogo, the production of Aß and phosphorylation of tau was quantified by ELISA and western blot. RESULTS: Inhibition of the Nogo/NgR signaling pathway ameliorated pathological features including amyloid plaques and phosphorylated levels of tau in APP/PS1 mice. In addition, after treatment with the conditioned medium from BV-2 microglia stimulated by Nogo, Aß production and tau phosphorylation in cultured neurons were increased. The conditioned medium also increased the expression of APP, its amyloidogenic processing, and the activity of GSK3ß in neurons. The conditioned medium was also proinflammatory medium, and the blockage of the Nogo/NgR pathway improved the neuroinflammatory environment in APP/PS1 mice. CONCLUSIONS: Taken together, the neuroinflammation mediated by Nogo/NgR pathway in microglia could directly take part in the pathological process of AD by influencing the amyloidogenesis and tau phosphorylation. These results contribute to a better understanding of AD pathogenesis and could offer a new therapeutic option for delaying the progression of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Microglía/efectos de los fármacos , Proteínas de la Mielina/farmacología , Proteínas Nogo/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Placa Amiloide/prevención & control , Transducción de Señal/efectos de los fármacos , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Medios de Cultivo Condicionados , Inflamación/patología , Inflamación/prevención & control , Ratones , Ratones Endogámicos C57BL , Fosforilación
3.
J Neurochem ; 132(1): 70-84, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25314656

RESUMEN

Axonal regeneration after injury to the CNS is hampered by myelin-derived inhibitors, such as Nogo-A. Natural products, such as green tea, which are neuroprotective and safe for long-term therapy, would complement ongoing various pharmacological approaches. In this study, using nerve growth factor-differentiated neuronal-like Neuroscreen-1 cells, we show that extremely low concentrations of unfractionated green tea polyphenol mixture (GTPP) and its active ingredient, epigallocatechin-3-gallate (EGCG), prevent both the neurite outgrowth-inhibiting activity and growth cone-collapsing activity of Nogo-66 (C-terminal domain of Nogo-A). Furthermore, a synergistic interaction was observed among GTPP constituents. This preventive effect was dependent on 67-kDa laminin receptor (67LR) to which EGCG binds with high affinity. The antioxidants N-acetylcysteine and cell-permeable catalase abolished this preventive effect of GTPP and EGCG, suggesting the involvement of sublethal levels of H2 O2 in this process. Accordingly, exogenous sublethal concentrations of H2 O2 , added as a bolus dose (5 µM) or more effectively through a steady-state generation (1-2 µM), mimicked GTPP in counteracting the action of Nogo-66. Exogenous H2 O2 mediated this action by bypassing the requirement of 67LR. Taken together, these results show for the first time that GTPP and EGCG, acting through 67LR and elevating intracellular sublethal levels of H2 O2 , inhibit the antineuritogenic action of Nogo-A. Currently, several agents are being evaluated for overcoming axonal growth inhibitors to promote functional recovery after stroke and spinal cord injury. Epigallocatechin-3-gallate (EGCG), present in green tea polyphenol mixture (GTPP), prevents antineuritogenic activity of Nogo-A, a myelin-derived axonal growth inhibitor. The preventive action of EGCG involves the cell-surface-associated 67-kDa laminin receptor and H2 O2 . GTPP may complement ongoing efforts to treat neuronal injuries.>


Asunto(s)
Peróxido de Hidrógeno/farmacología , Proteínas de la Mielina/antagonistas & inhibidores , Proteínas de la Mielina/farmacología , Neuritas/efectos de los fármacos , Oxidantes/farmacología , Polifenoles/farmacología , Receptores de Laminina/efectos de los fármacos , Té/química , Animales , Células Cultivadas , Conos de Crecimiento/efectos de los fármacos , Ratones , Proteínas Nogo , Polifenoles/química , Seudópodos/efectos de los fármacos
4.
Stem Cells ; 32(6): 1636-48, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24449409

RESUMEN

Neural stem cells (NSCs) and neural progenitors (NPs) in the mammalian neocortex give rise to the main cell types of the nervous system. The biological behavior of these NSCs and NPs is regulated by extracellular niche derived autocrine-paracrine signaling factors on a developmental timeline. Our previous reports [Plos One 2010;5:e15341; J Neurochem 2011;117:565-578] have shown that chondroitin sulfate proteoglycan and ApolipoproteinE are autocrine-paracrine survival factors for NSCs. NogoA, a myelin related protein, is expressed in the cortical ventricular zones where NSCs reside. However, the functional role of Nogo signaling proteins in NSC behavior is not completely understood. In this study, we show that NogoA receptors, NogoR1 and PirB, are expressed in the ventricular zone where NSCs reside between E10.5 and 14.5 but not at E15.5. Nogo ligands stimulate NSC survival and proliferation in a dosage-dependent manner in vitro. NogoR1 and PirB are low and high affinity Nogo receptors, respectively and are responsible for the effects of Nogo ligands on NSC behavior. Inhibition of autocrine-paracrine Nogo signaling blocks NSC survival and proliferation. In NSCs, NogoR1 functions through Rho whereas PirB uses Shp1/2 signaling pathways to control NSC behavior. Taken together, this work suggests that Nogo signaling is an important pathway for survival of NSCs.


Asunto(s)
Proteínas de la Mielina/metabolismo , Células-Madre Neurales/citología , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Apolipoproteínas E/metabolismo , Comunicación Autocrina/efectos de los fármacos , Recuento de Células , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula , Supervivencia Celular/efectos de los fármacos , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Embrión de Mamíferos/citología , Femenino , Proteínas Ligadas a GPI/deficiencia , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Proteínas de la Mielina/deficiencia , Proteínas de la Mielina/farmacología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Proteínas Nogo , Receptor Nogo 1 , Comunicación Paracrina/efectos de los fármacos , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores Inmunológicos/deficiencia , Transducción de Señal/efectos de los fármacos , Esferoides Celulares/citología , Esferoides Celulares/metabolismo
5.
J Biol Chem ; 287(21): 17206-17213, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22493449

RESUMEN

It has become increasingly clear that only antibodies recognizing conformation-dependent epitopes of myelin oligodendrocyte glycoprotein (MOG) have a demyelinating potential in the animal model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Nevertheless, for the induction of EAE, most studies to date have used MOG peptides or bacterially expressed MOG, neither of which contain the tertiary structure of the native antigen. Non-refolded recombinant human MOG does not induce EAE in DA rats. Therefore, we refolded this protein in order to assess the influence of MOG conformation on its pathogenicity in DA rats. DA rats immunized with refolded human MOG developed severe acute EAE. As expected, rats immunized with the refolded protein had a higher amount of conformational MOG antibodies present in serum. But in addition, a striking effect of MOG refolding on the generation of T-cell responses was found. Indeed, T-cell responses against the encephalitogenic MOG 91-108 epitope were greatly enhanced after refolding. Therefore, we conclude that refolding of MOG increases its pathogenicity both by generating conformation-dependent MOG antibodies and by enhancing its processing or/and presentation on MHC molecules. These data are important in regard to investigations of the pathogenic potential of many (auto)antigens.


Asunto(s)
Autoanticuerpos/inmunología , Linfocitos B/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Proteínas de la Mielina/inmunología , Pliegue de Proteína , Linfocitos T/inmunología , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Autoanticuerpos/farmacología , Autoantígenos/inmunología , Linfocitos B/patología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Proteínas de la Mielina/química , Proteínas de la Mielina/farmacología , Glicoproteína Mielina-Oligodendrócito , Estructura Terciaria de Proteína , Ratas , Linfocitos T/patología
6.
J Neurosci ; 31(16): 5977-88, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21508223

RESUMEN

Stroke is the leading cause of disability in much of the world, with few treatment options available. Following unilateral stroke in rats, inosine, a naturally occurring purine nucleoside, stimulates the growth of projections from the undamaged hemisphere into denervated areas of the spinal cord and improves skilled use of the impaired forelimb. Inosine augments neurons' intrinsic growth potential by activating Mst3b, a component of the signal transduction pathway through which trophic factors regulate axon outgrowth. The present study investigated whether inosine would complement the effects of treatments that promote plasticity through other mechanisms. Following unilateral stroke in the rat forelimb motor area, inosine combined with NEP1-40, a Nogo receptor antagonist, doubled the number of axon branches extending from neurons in the intact hemisphere into the denervated side of the spinal cord compared with either treatment alone, and restored rats' level of skilled reaching using the impaired forepaw to preoperative levels. Similar functional improvements were seen when inosine was combined with environmental enrichment (EE). The latter effect was associated with changes in gene expression in layer 5 pyramidal neurons of the undamaged cortex well beyond those seen with inosine or EE alone. Inosine is now in clinical trials for other indications, making it an attractive candidate for the treatment of stroke patients.


Asunto(s)
Miembro Anterior/efectos de los fármacos , Inosina/uso terapéutico , Proteínas de la Mielina/uso terapéutico , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Axones/efectos de los fármacos , Axones/fisiología , Ambiente , Miembro Anterior/fisiopatología , Inosina/farmacología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Movimiento/efectos de los fármacos , Movimiento/fisiología , Proteínas de la Mielina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Fragmentos de Péptidos/farmacología , Ratas , Recuperación de la Función/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiopatología , Accidente Cerebrovascular/fisiopatología
7.
J Biol Chem ; 286(3): 1876-83, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21087927

RESUMEN

Myelin components that inhibit axonal regeneration are believed to contribute significantly to the lack of axonal regeneration noted in the adult central nervous system. Three proteins found in myelin, Nogo, myelin-associated glycoprotein, and oligodendrocyte-myelin glycoprotein, inhibit neurite outgrowth in vitro. All of these proteins interact with the same receptors, namely, the Nogo receptor (NgR) and paired immunoglobulin-like receptor B (PIR-B). As per previous reports, corticospinal tract (CST) regeneration is not enhanced in NgR-knock-out mice after spinal cord injury. Therefore, we assessed CST regeneration in PIR-B-knock-out mice. We found that hindlimb motor function, as assessed using the Basso mouse scale, footprint test, inclined plane test, and beam walking test, did not differ between the PIR-B-knock-out and wild-type mice after dorsal hemisection of the spinal cord. Further, tracing of the CST fibers after injury did not reveal enhanced axonal regeneration or sprouting in the CST of the PIR-B-knock-out mice. Systemic administration of NEP1-40, a NgR antagonist, to PIR-B knock-out mice did not enhance the regenerative response. These results indicate that PIR-B knock-out is not sufficient to induce extensive axonal regeneration after spinal cord injury.


Asunto(s)
Actividad Motora , Neuritas/metabolismo , Receptores Inmunológicos/metabolismo , Recuperación de la Función , Regeneración , Traumatismos de la Médula Espinal/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Noqueados , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Proteínas de la Mielina/farmacología , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Neuritas/patología , Proteínas Nogo , Fragmentos de Péptidos/farmacología , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/genética , Traumatismos de la Médula Espinal/patología
8.
J Neurochem ; 120(5): 721-31, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22145612

RESUMEN

Nogo-66 is a 66-amino-acid-residue extracellular domain of Nogo-A, which plays a key role in inhibition neurite outgrowth of central nervous system through binding to the Nogo-66 receptor (NgR) expressed on the neuron. Recent studies have confirmed that NgR is also expressed on the surface of macrophages/microglia in multiple sclerosis, but its biological effects remain unknown. In the present study, our results demonstrated that Nogo-66 triggered microglia anti-adhesion and inhibited their migration in vitro, which was mediated by NgR. We also assessed the roles of small GTP (glycosyl phosphatidylinositol)-binding proteins of the Rho family as the downstream signal transducers on the microglia adhesion and mobility induced by Nogo-66. The results showed that Nogo-66 activated RhoA and reduced the activity of Cdc42 in the meanwhile, which further triggered the anti-adhesion and migration inhibition effects to microglia. Nogo-66 inhibited microglia polarization and membrane protrusion formation, thus might eventually contribute to the decreasing capability of cell mobility. Taken together, the Nogo-66/NgR pathway may modulate neuroinflammation via mediating microglia adhesion and migration in addition to its role in neurons. Better understanding the relationship between Nogo-66/NgR and neuroinflammation may help targeting NgR for treating central nervous system diseases related with inflammation.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Movimiento Celular/fisiología , Microglía/efectos de los fármacos , Proteínas de la Mielina/farmacología , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Adhesión Celular/fisiología , Movimiento Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Proteínas Ligadas a GPI/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Microglía/fisiología , Proteínas de la Mielina/inmunología , Proteínas de la Mielina/metabolismo , Proteínas Nogo , Receptor Nogo 1 , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rho/genética
9.
Proc Natl Acad Sci U S A ; 106(41): 17511-6, 2009 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-19805174

RESUMEN

Blood vessel formation during ischemia and wound healing requires coordination of the inflammatory response with genes that regulate blood vessel assembly. Here we show that the reticulon family member 4B, aka Nogo-B, is upregulated in response to ischemia and is necessary for blood flow recovery secondary to ischemia and wound healing. Mice lacking Nogo-B exhibit reduced arteriogenesis and angiogenesis that are linked to a decrease in macrophage infiltration and inflammatory gene expression in vivo. Bone marrow-derived macrophages isolated from Nogo knock-out mice have reduced spreading and chemotaxis due to impaired Rac activation. Bone marrow reconstitution experiments show that Nogo in myeloid cells is necessary to promote macrophage homing and functional recovery after limb ischemia. Thus, endogenous Nogo coordinates macrophage-mediated inflammation with arteriogenesis, wound healing, and blood flow control.


Asunto(s)
Macrófagos/fisiología , Proteínas de la Mielina/farmacología , Proteínas de la Mielina/fisiología , Animales , Velocidad del Flujo Sanguíneo/fisiología , Movimiento Celular/fisiología , Inflamación/prevención & control , Isquemia/fisiopatología , Isquemia/prevención & control , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/fisiología , Proteínas de la Mielina/deficiencia , Proteínas de la Mielina/genética , Proteínas Nogo , Flujo Sanguíneo Regional/fisiología , Regulación hacia Arriba , Cicatrización de Heridas/fisiología
10.
J Biol Chem ; 285(9): 6425-33, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20018888

RESUMEN

The N-terminal domain of NogoA, called amino-Nogo, inhibits axonal outgrowth and cell spreading via a largely unknown mechanism. In the present study, we show that amino-Nogo decreases Rac1 activity and inhibits fibroblast spreading. 12-O-Tetradecanoylphorbol-13-acetate-type tumor promoters, such as phorbol 12-myristate 13-acetate (PMA) and teleocidin, increase Rac1 activity and overcome the amino-Nogo-induced inhibition of cell spreading. The stimulating effect of tumor promoters on cell spreading requires activation of protein kinase D and the subsequent activation of Akt1. Furthermore, we identified Akt1 as a new signaling component of the amino-Nogo pathway. Akt1 phosphorylation is decreased by amino-Nogo. Activation of Akt1 with a cell-permeable peptide, TAT-TCL1, blocks the amino-Nogo inhibition. Finally, we provide evidence that these signaling pathways operate in neurons in addition to fibroblasts. Our results suggest that activation of protein kinase D and Akt1 are approaches to promote axonal regeneration after injury.


Asunto(s)
Proteínas de la Mielina/farmacología , Acetato de Tetradecanoilforbol/farmacología , Células 3T3 , Animales , Carcinógenos , Adhesión Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Fibroblastos/metabolismo , Inhibidores de Crecimiento , Humanos , Ratones , Regeneración Nerviosa , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Neuronas/metabolismo , Proteínas Nogo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo
11.
J Neurochem ; 116(5): 900-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21214567

RESUMEN

In addition to supporting rapid nerve conduction, myelination nurtures and stabilizes axons and protects them from acute toxic insults. One myelin molecule that protects and sustains axons is myelin-associated glycoprotein (MAG). MAG is expressed on the innermost wrap of myelin, apposed to the axon surface, where it interacts with axonal receptors that reside in lateral membrane domains including gangliosides, the glycosylphosphatidylinositol-anchored Nogo receptors, and ß1-integrin. We report here that MAG protection extends beyond the axon to the neurons from which those axons emanate, protecting them from excitotoxicity. Compared to wild type mice, Mag-null mice displayed markedly increased seizure activity in response to intraperitoneal injection of kainic acid, an excitotoxic glutamate receptor agonist. Mag-null mice also had larger lesion volumes in response to intrastriatal injection of the excitotoxin NMDA. Prior injection of a soluble form of MAG partially protected Mag-null mice from NMDA-induced lesions. Hippocampal neurons plated on proteins extracted from wild-type rat or mouse myelin were resistant to kainic acid-induced excitotoxicity, whereas neurons plated on proteins from Mag-null myelin were not. Protection was reversed by anti-MAG antibody and replicated by addition of soluble MAG. MAG-mediated protection from excitotoxicity was dependent on Nogo receptors and ß1-integrin. We conclude that MAG engages membrane-domain resident neuronal receptors to protect neurons from excitotoxicity, and that soluble MAG mitigates excitotoxic damage in vivo.


Asunto(s)
Agonistas de Aminoácidos Excitadores/toxicidad , Ácido Kaínico/toxicidad , N-Metilaspartato/toxicidad , Receptores de Superficie Celular/uso terapéutico , Convulsiones/prevención & control , Animales , Anticuerpos/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inducido químicamente , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/patología , Susceptibilidad a Enfermedades/terapia , Inhibidores Enzimáticos/farmacología , Hipocampo/citología , Humanos , Técnicas In Vitro , Cadenas beta de Integrinas/inmunología , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Mielina/farmacología , Glicoproteína Asociada a Mielina , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fosfoinositido Fosfolipasa C/farmacología , Receptores de Superficie Celular/deficiencia , Convulsiones/inducido químicamente , Convulsiones/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tubulina (Proteína)/metabolismo
12.
Neurochem Res ; 36(6): 1129-34, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21461775

RESUMEN

Communication between astrocytes via the gap junction is crucial for maintaining homeostasis of the extra-neuronal microenvironment of the central nervous system. Dysfunction of astrocytic gap junctions is involved in many brain disorders. Our previous studies demonstrated a novel co-localization of Nogo-66 receptor at glial gap junctions in rat cerebellum and posterior pituitary. The present study was aimed at exploring whether Nogo-66 can modulate glial gap junctions in vitro. We confirmed the co-localization of Nogo-66 receptor with Cx43 in cultured astrocytes, and stimulated astrocytes with myelin extracts, or Nogo-66-Fc conditioned medium. Finally, we expressed and purified a functionally effective GST-Nogo-66 peptide. Lucifer yellow transfer assay was adopted to measure the gap junction permeability. The results showed that the spreading of Lucifer yellow was inhibited significantly by all three treatments as compared with their corresponding controls. Therefore, this study shows a novel inhibitory effect of Nogo-66 on the permeability of astrocytic gap junctions, suggesting a presumable role of Nogo-66 receptor in modulating the glial gap junction.


Asunto(s)
Astrocitos/efectos de los fármacos , Colorantes/química , Uniones Comunicantes/efectos de los fármacos , Proteínas de la Mielina/farmacología , Animales , Astrocitos/fisiología , Medios de Cultivo Condicionados , Uniones Comunicantes/fisiología , Inmunohistoquímica , Proteínas de la Mielina/química , Proteínas Nogo , Ratas
13.
Cereb Cortex ; 20(10): 2380-90, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20093372

RESUMEN

Although Nogo-A has been intensively studied for its inhibitory effect on axonal regeneration in the adult central nervous system, little is known about its function during brain development. In the embryonic mouse cortex, Nogo-A is expressed by radial precursor/glial cells and by tangentially migrating as well as postmigratory neurons. We studied radially migrating neuroblasts in wild-type and Nogo-A knockout (KO) mouse embryos. In vitro analysis showed that Nogo-A and its receptor components NgR, Lingo-1, TROY, and p75 are expressed in cells emigrating from embryonic forebrain-derived neurospheres. Live imaging revealed an increased cell motility when Nogo-A was knocked out or blocked with antibodies. Antibodies blocking NgR or Lingo-1 showed the same motility-enhancing effect supporting a direct role of surface Nogo-A on migration. Bromodeoxyuridine (BrdU) labeling of embryonic day (E)15.5 embryos demonstrated that Nogo-A influences the radial migration of neuronal precursors. At E17.5, the normal transient accumulation of radially migrating precursors within the subventricular zone was not detectable in the Nogo-A KO mouse cortex. At E19, migration to the upper cortical layers was disturbed. These findings suggest that Nogo-A and its receptor complex play a role in the interplay of adhesive and repulsive cell interactions in radial migration during cortical development.


Asunto(s)
Movimiento Celular/genética , Corteza Cerebral/citología , Corteza Cerebral/embriología , Proteínas de la Mielina/fisiología , Células-Madre Neurales/fisiología , Animales , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Adhesión Celular/genética , Diferenciación Celular/genética , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Dominio Doblecortina , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de la Mielina/deficiencia , Proteínas de la Mielina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/efectos de los fármacos , Neuropéptidos/metabolismo , Proteínas Nogo , Receptores de Péptidos/metabolismo , Estadísticas no Paramétricas , Tubulina (Proteína)/metabolismo
14.
J Neurosci ; 29(48): 15266-76, 2009 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19955379

RESUMEN

Rho GTPases are thought to mediate the action of several axonal growth inhibitors in the adult brain and spinal cord. RhoA has been targeted pharmacologically in both humans and animals to promote neurite outgrowth and functional recovery following CNS trauma. However, rat spinal cord injury studies suggest a complicated and partial benefit of inhibiting Rho or its downstream effector, Rho-associated kinase (ROCKII). This limited benefit may reflect inhibition of other kinases, poor access, or a minimal role of ROCKII in vivo. Therefore, we studied ROCKII mutant mice to probe this pathway genetically. ROCKII(-/-) dorsal root ganglion neurons are less sensitive to inhibition by Nogo protein or by chondroitin sulfate proteoglycan in vitro. We examined adult ROCKII(-/-) mice in two injury paradigms, cervical multilevel dorsal rhizotomy and midthoracic dorsal spinal cord hemisection. After dorsal root crush injury, the ROCKII(-/-) mice recovered use of the affected forepaw more quickly than did controls. Moreover, multiple classes of sensory axons regenerated across the dorsal root entry zone into the spinal cord of mice lacking ROCKII. After the spinal cord injury, ROCKII(-/-) mice showed enhanced local growth of raphespinal axons in the caudal spinal cord and corticospinal axons into the lesion site. Improved functional recovery was not observed by Basso Mouse Scale score following dorsal hemisection, likely due to developmental defects in the nervous system. Together, these findings demonstrate that the ROCKII gene product limits axonal growth after CNS trauma.


Asunto(s)
Axones/patología , Axones/fisiología , Traumatismos de la Médula Espinal/patología , Quinasas Asociadas a rho/fisiología , Amidas/farmacología , Análisis de Varianza , Animales , Axones/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Región CA1 Hipocampal/citología , Células Cultivadas , Toxina del Cólera/metabolismo , Inhibidores Enzimáticos/farmacología , Ganglios Espinales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Neuropatía Mediana/etiología , Neuropatía Mediana/patología , Neuropatía Mediana/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Mielina/farmacología , Regeneración Nerviosa/fisiología , Neuronas/clasificación , Neuronas/efectos de los fármacos , Neuronas/patología , Proteínas Nogo , Piridinas/farmacología , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Rizotomía/métodos , Traumatismos de la Médula Espinal/fisiopatología , Factores de Tiempo , Versicanos/farmacología , Quinasas Asociadas a rho/deficiencia
15.
J Neurochem ; 113(6): 1644-58, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20374426

RESUMEN

Lesioned axons do not regenerate in the adult mammalian CNS, owing to the over-expression of inhibitory molecules such as myelin-derived proteins or chondroitin sulphate proteoglycans. In order to overcome axon inhibition, strategies based on extrinsic and intrinsic treatments have been developed. For myelin-associated inhibition, blockage with NEP1-40, receptor bodies or IN-1 antibodies has been used. In addition, endogenous blockage of cell signalling mechanisms induced by myelin-associated proteins is a potential tool for overcoming axon inhibitory signals. We examined the participation of glycogen synthase kinase 3beta (GSK3beta) and extracellular-related kinase (ERK) 1/2 in axon regeneration failure in lesioned cortical neurons. We also investigated whether pharmacological blockage of GSK3beta and ERK1/2 activities facilitates regeneration after myelin-directed inhibition in two models: (i) cerebellar granule cells and (ii) lesioned entorhino-hippocampal pathway in slice cultures, and whether the regenerative effects are mediated by Nogo Receptor 1 (NgR1). We demonstrate that, in contrast to ERK1/2 inhibition, the pharmacological treatment of GSK3beta inhibition strongly facilitated regrowth of cerebellar granule neurons over myelin independently of NgR1. Finally, these regenerative effects were corroborated in the lesioned entorhino-hippocampal pathway in NgR1-/- mutant mice. These results provide new findings for the development of new assays and strategies to enhance axon regeneration in injured cortical connections.


Asunto(s)
Corteza Cerebral/citología , Glucógeno Sintasa Quinasa 3/metabolismo , Neuritas/fisiología , Neuronas/citología , Aminofenoles/farmacología , Animales , Animales Recién Nacidos , Axotomía/métodos , Células Cultivadas , Técnicas de Cocultivo/métodos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Proteínas Ligadas a GPI , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/citología , Humanos , Indoles/farmacología , Lisina/análogos & derivados , Lisina/metabolismo , Maleimidas/farmacología , Ratones , Ratones Noqueados , Mutación/genética , Proteínas de la Mielina/metabolismo , Proteínas de la Mielina/farmacología , Neuritas/efectos de los fármacos , Proteínas Nogo , Receptor Nogo 1 , Embarazo , Ratas , Receptores de Superficie Celular , Receptores de Péptidos/deficiencia , Factores de Tiempo , Versicanos/metabolismo
16.
Mol Ther ; 17(7): 1173-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19293775

RESUMEN

Artemin is a neurotrophic factor of the glial cell line-derived neurotrophic factor (GDNF) family of ligands that acts through the GDNF family receptor alpha3 (GFRalpha3)/ret receptor found predominantly on sensory and sympathetic neurons. In order to explore the potential utility of artemin to improve functional outcome after spinal cord injury (SCI), we constructed a nonreplicating herpes simplex virus (HSV)-based vector to express artemin (QHArt). We found that QHArt efficiently transfects spinal cord neurons to produce artemin. Transgene-mediated artemin supported the extension of neurites by primary dorsal root ganglion neurons in culture, and allowed those cells to overcome myelin inhibition of neurite extension through activation of protein kinase A (PKA) to phosphorylate cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) and increase expression of arginase I. Intraspinal injection of QHArt immediately after thoracic spinal cord dorsal over hemisection produced a statistically significant improvement in motor recovery over the course of four weeks measured by locomotor rating score.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/genética , Proteínas de la Mielina/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Simplexvirus/genética , Traumatismos de la Médula Espinal/terapia , Animales , Arginasa/metabolismo , Axones/fisiología , Western Blotting , Carbazoles/farmacología , Línea Celular , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Ganglios Espinales , Inmunoquímica , Neuritas/fisiología , Neuronas/metabolismo , Fosforilación/inmunología , Pirroles/farmacología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/citología
17.
Zhonghua Yan Ke Za Zhi ; 46(6): 503-7, 2010 Jun.
Artículo en Zh | MEDLINE | ID: mdl-21055194

RESUMEN

OBJECTIVE: To study the effect of protein Nogo-66 on the expression of CD11b and MHC-II in retinal microglia of chronic ocular hypertension SD rats and the effects of protein Nogo-66 on the immunogenicity of injured retinal tissues. METHODS: It was a control experimental study. Chronic ocular hypertension rat model was established by laser photocoagulation on the anterior chamber angle and superficial vein of the sclera. One ml of Nogo-66 (0.01%) in PBS was injected subcutaneously on the day of laser treatment and 0.005% Nogo-66 PBS solution was injected into the vitreous 7 days and 1 month latter. PBS without Nogo-66 was injected in the control group. The expression of cell surface antigen CD11b and MHC-II were detected by immunohistochemistry 1 month and 1 day after the establishment of hypertension model. The difference of average IOP among groups was analyzed by variance analysis. The difference of expression of CD11b and MHC-II between the experimental and control groups was analyzed by t-test. RESULTS: The intraocular pressure (IOP) of experimental groups rised from the seventh day after model-building and the highest IOP was (24.16 ± 2.70) mm Hg (1 mm Hg = 0.133 kPa) 1 month later while that in the control groups was (15.93 ± 3.28) mm Hg. The difference between them was statistically significant (F = 2.10, P < 0.05). Expression of CD11b was (1.78 ± 0.63)% and MHC-II was (3.92 ± 1.03)% in Nogo-66 with hypertension groups, these results was significantly lower than those in Nogo-66 with normal intraocular pressure groups in which the expression of CD11b was (8.15 ± 1.97)% (t = 2.35, P < 0.05) and MHC-II was (11.45 ± 1.97)% (t = 2.14, P < 0.05). CONCLUSIONS: Protein Nogo-66 activated the cell surface antigen CD11b in nerve fiber layer of retina and induced antigen presenting molecules (MHC-II). This indicates that Nogo has the center immunogenicity and this protein could activate antigen-presenting cells to present injury antigen.


Asunto(s)
Microglía/metabolismo , Proteínas de la Mielina/farmacología , Hipertensión Ocular/metabolismo , Animales , Antígeno CD11b/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Masculino , Proteínas Nogo , Ratas , Ratas Sprague-Dawley , Retina/citología , Retina/metabolismo
18.
J Cell Biol ; 116(4): 1063-70, 1992 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1370837

RESUMEN

To gain insights into the functional role of the molecular association between neural adhesion molecules and extracellular matrix constituents, soluble forms of the myelin-associated glycoprotein (MAG) and the neural cell adhesion molecule (N-CAM), representing most of the extracellular domains of the molecules, were investigated in their ability to modify fibrillogenesis of collagen type I. MAG and N-CAM retarded the rate of fibril formation, as measured by changes in turbidity, and increased the diameter of the fibrils formed, but did not change the banding pattern when compared to collagen type I in the absence of adhesion molecules. Scatchard plot analysis of the binding of MAG and N-CAM to the fibril-forming collagen types I, II, III, and V suggest one binding site for N-CAM and two binding sites for MAG. Binding of MAG, but not of N-CAM, to collagen type I was decreased during fibril formation, probably due to a reduced accessibility of one binding site for MAG during fibrillogenesis. These results indicate that the neural adhesion molecules can influence the configuration of extracellular matrix constituents, thus, implicating them in the modulation of cell-substrate interactions.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Colágeno/metabolismo , Proteínas de la Mielina/farmacología , Animales , Sitios de Unión , Moléculas de Adhesión Celular Neuronal/aislamiento & purificación , Moléculas de Adhesión Celular Neuronal/metabolismo , Heparina/farmacología , Ratones , Proteínas de la Mielina/aislamiento & purificación , Proteínas de la Mielina/metabolismo , Glicoproteína Asociada a Mielina , Ensayo de Unión Radioligante , Solubilidad
19.
Mol Biol Cell ; 17(5): 2278-86, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16510522

RESUMEN

The neural cell adhesion molecule (NCAM) plays a pivotal role in the development of the nervous system, promoting neuronal differentiation via homophilic (NCAM-NCAM) as well as heterophilic (NCAM-fibroblast growth factor receptor [FGFR]) interactions. NCAM-induced intracellular signaling has been shown to affect and be dependent on the cytoplasmic Ca2+ concentration ([Ca2+]i). However, the molecular basis of this remains unclear. In this study, we determined [Ca2+]i regulating mechanisms involved in intracellular signaling induced by NCAM. To mimic the effect of homophilic NCAM interaction on [Ca2+]i in vitro, we used a peptide derived from a homophilic binding site of NCAM, termed P2, which triggers signaling cascades similar to those activated by NCAM-NCAM interaction. We found that P2 increased [Ca2+]i in primary hippocampal neurons. This effect depended on two signaling pathways. The first pathway was associated with activation of FGFR, phospholipase Cgamma, and production of diacylglycerol, and the second pathway involved Src-family kinases. Moreover, NCAM-mediated Ca2+ entry required activation of nonselective cation and T-type voltage-gated Ca2+ channels. These channels, together with the Src-family kinases, were also involved in neuritogenesis induced by physiological, homophilic NCAM interactions. Thus, unanticipated mechanisms of Ca2+ homeostasis are shown to be activated by NCAM and to contribute to neuronal differentiation.


Asunto(s)
Canales de Calcio Tipo T/efectos de los fármacos , Calcio/metabolismo , Canales Iónicos/efectos de los fármacos , Proteínas de la Mielina/farmacología , Moléculas de Adhesión de Célula Nerviosa/farmacología , Animales , Sitios de Unión , Canales de Calcio Tipo T/metabolismo , Diferenciación Celular , Citoplasma/metabolismo , Diglicéridos/metabolismo , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Homeostasis , Canales Iónicos/metabolismo , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosfolipasa C gamma/metabolismo , Ratas , Ratas Wistar , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo
20.
Neuron ; 15(6): 1375-81, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8845160

RESUMEN

The MAG-deficient mouse was used to test whether MAG acts as a significant inhibitor of axonal regeneration in the adult mammalian CNS, as suggested by cell culture experiments. Cell spreading, neurite elongation, or growth cone collapse of different cell types in vitro was not significantly different when myelin preparations or optic nerve cryosections from either MAG-deficient or wild-type mice were used as a substrate. More importantly, the extent of axonal regrowth in lesioned optic nerve and corticospinal tract in vivo was similarly poor in MAG-deficient and wild-type mice. However, axonal regrowth increased significantly and to a similar extent in both genotypes after application of the IN-1 antibody directed against the neurite growth inhibitors NI-35 and NI-250. These observations do not support the view that MAG is a significant inhibitor of axonal regeneration in the adult CNS.


Asunto(s)
Axones/fisiología , Sistema Nervioso Central/fisiología , Glicoproteína Asociada a Mielina/fisiología , Regeneración Nerviosa , Inhibición Neural/fisiología , Células 3T3 , Animales , Cerebelo/citología , Ganglios Espinales/citología , Inhibidores de Crecimiento/antagonistas & inhibidores , Ratones , Ratones Mutantes , Proteínas de la Mielina/farmacología , Glicoproteína Asociada a Mielina/deficiencia , Glicoproteína Asociada a Mielina/farmacología , Neuritas/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Nervio Óptico/citología , Células PC12 , Tractos Piramidales/citología , Ratas , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA