Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Med Sci ; 19(1): 164-174, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34975310

RESUMEN

Over the past two decades, the development of targeted immunotherapeutics for relapsing-remitting multiple sclerosis has been successfully orchestrated through the efficacious modulation of neuroinflammatory outcomes demonstrated in the experimental autoimmune encephalomyelitis (EAE) model. In this model, the focus of developing immunomodulatory therapeutics has been demonstrated through their effectiveness in modifying the pro-inflammatory Th1 and Th17-dependent neuropathological outcomes of demyelination, oligodendrocytopathy and axonal dystrophy. However, recent successful preclinical and clinical trials have advocated for the significance of B cell-dependent immunopathogenic responses and has led to the development of novel biologicals that target specific B cell phenotypes. In this context, a new molecule, B-cell activating factor (BAFF), has emerged as a positive regulator of B cell survival and differentiation functioning through various signaling pathways and potentiating the activity of various receptor complexes through pleiotropic means. One possible cognate receptor for BAFF includes the Nogo receptor (NgR) and its homologs, previously established as potent inhibitors of axonal regeneration during central nervous system (CNS) injury and disease. In this review we provide current evidence for BAFF-dependent signaling through the NgR multimeric complex, elucidating their association within the CNS compartment and underlying the importance of these potential pathogenic molecular regulators as possible therapeutic targets to limit relapse rates and potentially MS progression.


Asunto(s)
Factor Activador de Células B/fisiología , Linfocitos B/fisiología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Animales , Autoinmunidad , Factor Activador de Células B/metabolismo , Factor Activador de Células B/uso terapéutico , Humanos , Agentes Inmunomoduladores/uso terapéutico , Esclerosis Múltiple/terapia , Proteínas Nogo/fisiología , Transducción de Señal
2.
J Mol Neurosci ; 69(3): 360-370, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31286407

RESUMEN

Intracerebral hemorrhage (ICH) leads to widespread pathological lesions in the brain, especially impacting neuronal survival and axonal regeneration. This study aimed to elucidate whether the Nogo-A (a myelin-related protein)/paired immunoglobulin-like receptor B (Pir-B)/tropomyosin receptor kinase B (TrkB) pathway could exert a regulatory effect in ICH. An ICH model was first established in Sprague Dawley rats, followed by different administrations of vehicle, k252a, or NSC 87877. The Morris water maze test was performed to observe ICH-induced cognitive dysfunction in rats. Rats in the ICH + NSC 87877 group showed better cognitive performance compared with those injected with vehicle or k252a. Neurobehavioral scores were identical. By harvesting brain tissues at different time points after ICH, we detected the expression levels of Nogo-A and PirB with western blot and immunofluorescence and found that they were markedly upregulated at 48 h after ICH. TUNEL and Fluoro-Jade B staining showed that NSC 87877 treatment attenuated ICH-induced apoptosis and neuronal death, whereas k252a treatment aggravated these pathological changes. The expression levels of growth-associated protein 43 (GAP43) and neurofilament 200 (NF200) were higher in the ICH + NSC 87877 group compared with the ICH + vehicle group, but were lower in the ICH + k252a group. Finally, we confirmed the protective role of p-TrkB/TrkB in ICH by western blot. To sum up, our study identified the inhibitory role of the Nogo-A/PirB/TrkB pathway in ICH; however, p-TrkB/TrkB may serve as a potential target for secondary brain injury post-ICH.


Asunto(s)
Hemorragia Cerebral/fisiopatología , Proyección Neuronal/fisiología , Neuronas/fisiología , Proteínas Nogo/fisiología , Receptor trkB/fisiología , Receptores Inmunológicos/fisiología , Transducción de Señal , Animales , Apoptosis , Encéfalo/patología , Carbazoles/toxicidad , Muerte Celular , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/patología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/prevención & control , Alcaloides Indólicos/toxicidad , Masculino , Aprendizaje por Laberinto , Actividad Motora , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Proteínas Nogo/biosíntesis , Quinolinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores Inmunológicos/biosíntesis , Regeneración , Regulación hacia Arriba
3.
J Cereb Blood Flow Metab ; 37(2): 614-631, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27927704

RESUMEN

Recently, we discovered a new role for the well-known axonal growth inhibitory molecule Nogo-A as a negative regulator of angiogenesis in the developing central nervous system. However, how Nogo-A affected the three-dimensional (3D) central nervous system (CNS) vascular network architecture remained unknown. Here, using vascular corrosion casting, hierarchical, synchrotron radiation µCT-based network imaging and computer-aided network analysis, we found that genetic ablation of Nogo-A significantly increased the three-dimensional vascular volume fraction in the postnatal day 10 (P10) mouse brain. More detailed analysis of the cerebral cortex revealed that this effect was mainly due to an increased number of capillaries and capillary branchpoints. Interestingly, other vascular parameters such as vessel diameter, -length, -tortuosity, and -volume were comparable between both genotypes for non-capillary vessels and capillaries. Taken together, our three-dimensional data showing more vessel segments and branchpoints at unchanged vessel morphology suggest that stimulated angiogenesis upon Nogo-A gene deletion results in the insertion of complete capillary micro-networks and not just single vessels into existing vascular networks. These findings significantly enhance our understanding of how angiogenesis, vascular remodeling, and three-dimensional vessel network architecture are regulated during central nervous system development. Nogo-A may therefore be a potential novel target for angiogenesis-dependent central nervous system pathologies such as brain tumors or stroke.


Asunto(s)
Sistema Nervioso Central/crecimiento & desarrollo , Neovascularización Fisiológica , Proteínas Nogo/fisiología , Animales , Sistema Nervioso Central/irrigación sanguínea , Corteza Cerebral/irrigación sanguínea , Eliminación de Gen , Imagenología Tridimensional/métodos , Ratones , Proteínas Nogo/genética
4.
Genetics ; 205(1): 295-302, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27821431

RESUMEN

Nogo-A is a membrane-bound protein that functions to inhibit neuronal migration, adhesion, and neurite outgrowth during development. In the mature nervous system, Nogo-A stabilizes neuronal wiring to inhibit neuronal plasticity and regeneration after injury. Here, we show that RET-1, the sole Nogo-A homolog in Caenorhabditis elegans, is required to control developmental wiring of a specific subset of neurons. In ret-1 deletion mutant animals, specific ventral nerve cord axons are misguided where they fail to respect the ventral midline boundary. We found that ret-1 is expressed in multiple neurons during development, and, through mosaic analysis, showed that ret-1 controls axon guidance in a cell-autonomous manner. Finally, as in mammals, ret-1 regulates ephrin expression, and dysregulation of the ephrin ligand VAB-2 is partially responsible for the ret-1 mutant axonal defects. Together, our data present a previously unidentified function for RET-1 in the nervous system of C. elegans.


Asunto(s)
Caenorhabditis elegans/crecimiento & desarrollo , Neuronas/fisiología , Proteínas Nogo/fisiología , Animales , Axones/metabolismo , Axones/fisiología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Efrinas/metabolismo , Proteínas de la Membrana/genética , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Neuronas Motoras/fisiología , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Proteínas Nogo/biosíntesis , Proteínas Nogo/genética , Proteínas Nogo/metabolismo
5.
Sci Rep ; 6: 39586, 2016 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-28000762

RESUMEN

Mitral cells are major projection neurons of the olfactory bulb (OB) that form an axonal bundle known as the lateral olfactory tract (LOT). After axonal bundle formation, collateral branches sprout from primary axons of the LOT. Recently, we identified LOT usher substance (LOTUS) as an endogenous Nogo receptor-1 (NgR1) antagonist and demonstrated that LOTUS contributes to the formation of the LOT axonal bundle. Immunoblots revealed that the expression level of Nogo-A in the OB developmentally increased during axonal collateral formation. Next, we found that the axonal collateral branches were increased in cultured OB neurons from LOTUS-knockout (KO) mice, whereas they were decreased in cultured OB neurons from NgR1-KO mice. Knockdown of Nogo-A in cultured OB neurons reduced the number of axonal collateral branches, suggesting that endogenous Nogo-A induces axonal branching. Finally, the collateral branches of the LOT were increased in LOTUS-KO mice, whereas those in NgR1-KO mice were decreased. Moreover, the abnormal increase of axonal branching observed in LOTUS-KO mice was rescued in the double mutant of LOTUS- and NgR1-KO mice. These findings suggest that Nogo-A and NgR1 interactions may contribute to axonal branching in LOT development.


Asunto(s)
Axones/fisiología , Proteínas Nogo/fisiología , Bulbo Olfatorio/embriología , Bulbo Olfatorio/fisiología , Transducción de Señal , Animales , Proteínas de Unión al Calcio/fisiología , Células Cultivadas , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Neuronas/fisiología , Receptor Nogo 1/fisiología , Prosencéfalo/fisiología
7.
Oncol Rep ; 35(6): 3395-402, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27109183

RESUMEN

Nogo or reticulon-4 (RTN4), also known as neurite outgrowth inhibitor, is a member of the reticulon family of genes. Nogo-A, one of the three isoforms, is enriched in the central nervous system (CNS). The extracellular domain of Nogo-A, Nogo-66, has neurite growth inhibitory activity that is specific for neurons and is mediated by the Nogo receptor. However, most of its functions are not known yet. We investigated whether Nogo-A modulates the migration and invasion of a glioblastoma cell line, as well as the factors that have an effect on Nogo-A. The expression of Nogo-A was evaluated using western blotting and immunohistochemistry in human brain tumor specimens. U87MG cells were transfected with a sense-Nogo-A cDNA construct (U87-Nogo-A cells expressing Nogo-A) and an empty vector (U87MG-E cells not expressing Nogo-A). The migration and invasion abilities of these cells were investigated using simple scratch and Matrigel invasion assays. Morphologic and cytoskeletal changes were documented by confocal microscopy. The proliferation rate was estimated using doubling time assay. The effects of Nogo-A on Rho activity and phosphorylated cofilin were determined by a Rho activity assay and western blotting. Among primary brain tumors, Nogo-A expression was found in a higher percentage of oligodendrogliomas (90.0%) compared with the percentage in the glioblastomas (68.4%). In addition, the percentage in mixed gliomas was 42.9%, while it was not expressed in pituitary adenomas or schwannomas. The migration and invasion abilities of the U87-Nogo-A cells were decreased compared with the control. In the U87-Nogo-A cell line, Rho activity and phosphorylated cofilin expression were also decreased and morphology became more flat in comparison with the U87MG-E cell line. Nogo-A may inhibit the migration and invasion of human malignant glioma cells via the downregulation of RhoA-cofilin signaling.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Proteínas Nogo/fisiología , Factores Despolimerizantes de la Actina/metabolismo , Actinas/análisis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Invasividad Neoplásica , Proteínas Nogo/análisis , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...