Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Biomater Sci ; 6(7): 1723-1734, 2018 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-29845137

RESUMO

Injury-activated endogenous neural stem cells (NSCs) in the spinal cord have promising therapeutic applications for rebuilding the neuronal relays after spinal cord injury (SCI) because of their lack of immune-rejection following exogenous cell transplantation. However, these NSCs rarely differentiate into neurons and the damaged axonal regenerative ability is drastically reduced due to the adverse SCI microenvironment. Cetuximab, an EGFR signaling antagonist, has demonstrated the ability of promoting NSC differentiation into neurons. Taxol, in addition to stabilizing microtubules, has shown potential for enhancing axonal regeneration and reducing scar formation after SCI. In this study, we further verified the combined therapeutic effects of Cetuximab and Taxol on inhibition of scar deposition and promotion of neuronal differentiation, axonal outgrowth and functional recovery in a rat severe SCI model. A linear orderly collagen scaffold modified with Cetuximab and Taxol was grafted into the SCI site after the complete removal of 4 mm of spinal tissue. The results showed that the combined functional scaffold implantation significantly increased neural regeneration to reconnect the neural network. Moreover, scaffold transplantation decreases the deposition of varied scar-related inhibitors within the lesion center, further reflecting the need for a combination dedicated to increasing motor function following SCI. Collagen scaffold based-combined therapy provides a potential strategy for improving functional restoration of the injured spinal cord.


Assuntos
Cetuximab/farmacologia , Proteínas da Mielina/farmacologia , Fármacos Neuroprotetores/farmacologia , Paclitaxel/farmacologia , Traumatismos da Medula Espinal/tratamento farmacológico , Alicerces Teciduais , Animais , Diferenciação Celular/efeitos dos fármacos , Cicatriz/prevenção & controle , Colágeno/química , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Proteínas da Mielina/isolamento & purificação , Regeneração Nervosa/efeitos dos fármacos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Telencéfalo/química
2.
Int J Mol Med ; 39(2): 437-445, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28075461

RESUMO

Interleukin-6 (IL)-6 was originally discovered as a factor that contributes to the secondary pathological and inflammatory response in the central nervous system (CNS) following injury. However, accumulating evidence suggests that IL-6 is also involved in functional and structural recovery following CNS injury by promoting axonal sprou-ting. This suggests a potential dual role of IL-6 in CNS injury. However, the definitive function of IL-6 in neural injury and the corresponding underlying mechanisms are still topics of controversy. The present study was carried out to examine the potential function of IL-6 in resistance to neurite growth­inhibitory effects via regulation of the expression of growth associated protein-43 (GAP-43), myelin-associated neurite outgrowth inhibitor (Nogo-A) and its receptor (NgR). Rat dorsal root ganglion (DRG) neurons cultured in an inhibitory microenvironment mimicking injured CNS were used to investigate the effects of IL-6 on the outgrowth of neuronal processes. Additionally, IL-6 was subarachnoidally injected into rats to establish a spinal cord injury (SCI) model, and the neurobehavioral manifestations and neural morphology were subsequently evaluated to determine the effect of IL-6 on neural regeneration. Finally, the potential molecular mechanisms of IL-6-mediated rege-neration and functional recovery following CNS injury are discussed. The results of the present study demonstrated that the in vitro administration of IL-6 enhanced the neurite outgrowth of DRG neurons in a dose-dependent manner via resisting the inhibitory function of myelin proteins. All doses of the IL-6 subarachnoid injection improved the Basso, Beattie and Bresnahan scores following SCI, with a large number of axonal sproutings observed at the spinal lesion site, and several sprouting fibers being elongated and bypassing the lesion and entered the caudal spinal cord. Additionally, a significantly increased density area of diaminobenzidine-labeled neural fiber was observed in rats that received a subarachnoid injection of IL-6, and the rats exhibited increased expression of GAP-43 and decreased expression of Nogo-A. In conclusion, the results of the present study indicated that IL-6 interferes with the inhibitory functions of myelin proteins by upregulating the expression of GAP-43 and simultaneously downregulating the expression of Nogo-A and NgR to promote axonal sprouting and functional recovery following SCI.


Assuntos
Axônios/metabolismo , Interleucina-6/metabolismo , Regeneração Nervosa , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/reabilitação , Animais , Axônios/efeitos dos fármacos , Biomarcadores , Modelos Animais de Doenças , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Gânglios Espinais , Expressão Gênica , Interleucina-6/farmacologia , Masculino , Proteínas da Mielina/metabolismo , Proteínas da Mielina/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Proteínas Nogo/metabolismo , Receptor Nogo 1/genética , Receptor Nogo 1/metabolismo , Tratos Piramidais/efeitos dos fármacos , Tratos Piramidais/metabolismo , Ratos , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/genética
3.
J Neurochem ; 132(1): 70-84, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25314656

RESUMO

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.>


Assuntos
Peróxido de Hidrogênio/farmacologia , Proteínas da Mielina/antagonistas & inibidores , Proteínas da Mielina/farmacologia , Neuritos/efeitos dos fármacos , Oxidantes/farmacologia , Polifenóis/farmacologia , Receptores de Laminina/efeitos dos fármacos , Chá/química , Animais , Células Cultivadas , Cones de Crescimento/efeitos dos fármacos , Camundongos , Proteínas Nogo , Polifenóis/química , Pseudópodes/efeitos dos fármacos
4.
Neuropsychopharmacology ; 40(4): 974-86, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25308353

RESUMO

Anxiety disorder is related to the pathophysiology of psychiatric diseases, including major depression, substance abuse, and schizophrenia. The amygdala is important for manifestation and modulation of anxiety. However, relatively little is known regarding the mechanisms that control the amygdala inhibitory activity that is involved in anxiety. We found that almost all ErbB4, which is the only autonomous receptor of neuregulin 1 (NRG1) in the basolateral amygdala (BLA), was expressed in GABAergic neurons. Endogenous NRG1-ErbB4 signaling pathway in the BLA could modulate anxiety-like behaviors and GABA release, whereas it had no effect on glutamatergic transmission. The administration of NRG1 into the BLA of high-anxiety mice alleviated their anxiety and enhanced GABAergic neurotransmission. Moreover, exogenous NRG1 also produced an anxiolytic effect in the stressed mice. Together, these observations indicated that NRG1-ErbB4 signaling is critical to maintaining GABAergic activity in the amygdala and thus to modulating anxiety-like behaviors. Because NRG1 and ErbB4 are susceptibility genes of schizophrenia, our findings might also help to explain the potential mechanism of emotional abnormality in schizophrenia.


Assuntos
Tonsila do Cerebelo/metabolismo , Ansiedade/patologia , Ansiedade/fisiopatologia , Regulação da Expressão Gênica/fisiologia , Proteínas da Mielina/metabolismo , Receptor ErbB-4/metabolismo , Receptores de Superfície Celular/metabolismo , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Comportamento Alimentar/efeitos dos fármacos , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Proteínas Ligadas por GPI/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Técnicas In Vitro , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas da Mielina/genética , Proteínas da Mielina/farmacologia , Receptor Nogo 1 , Pirimidinas/farmacologia , Quinazolinas/farmacologia , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Receptor ErbB-4/genética , Receptor ErbB-4/farmacologia , Receptores de Superfície Celular/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tirfostinas/farmacologia , Ácido gama-Aminobutírico/metabolismo
5.
Mol Brain ; 7: 86, 2014 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-25406759

RESUMO

BACKGROUND: CNS axon regeneration inhibitors such as Nogo and CSPGs (Chondroitin Sulfate Proteoglycans) are major extrinsic factors limiting outgrowth of severed nerve fibers. However, knowledge on intracellular signaling cascades and gene expression programs activated by these inhibitors in neurons is sparse. Herein we studied intracellular signaling cascades activated by total myelin, Nogo and CSPGs in primary mouse CNS neurons. RESULTS: Total myelin, Nogo and CSPGs stimulated gene expression activity of the serum response factor (SRF), a central gene regulator of immediate early (IEG) and actin cytoskeletal gene transcription. As demonstrated by pharmacological interference, SRF-mediated IEG activation by myelin, Nogo or CSPGs depended on MAP kinase, to a lesser extent on Rho-GTPase but not on PKA signaling. Stimulation of neurons with all three axon growth inhibitors activated the MAP kinase ERK. In addition to ERK activation, myelin activated the IEG c-Fos, an important checkpoint of neuronal survival vs. apoptosis. Employing Srf deficient neurons revealed that myelin-induced IEG activation requires SRF. This suggests an SRF function in mediating neuronal signaling evoked by axon regeneration associated inhibitors. Besides being a signaling target of axon growth inhibitors, we show that constitutively-active SRF-VP16 can be employed to circumvent neurite growth inhibition imposed by myelin, Nogo and CSPGs. CONCLUSION: In sum, our data demonstrate that axon regeneration inhibitors such as Nogo trigger gene expression programs including an SRF-dependent IEG response via MAP kinases and Rho-GTPases.


Assuntos
Axônios/fisiologia , Sistema Nervoso Central/fisiologia , Genes Precoces , Sistema de Sinalização das MAP Quinases/genética , Regeneração Nervosa , Fator de Resposta Sérica/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Axônios/efeitos dos fármacos , Proteoglicanas de Sulfatos de Condroitina/farmacologia , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas da Mielina/farmacologia , Bainha de Mielina/metabolismo , Regeneração Nervosa/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Proteínas Nogo , Proteínas Proto-Oncogênicas c-fos/metabolismo
6.
Stem Cells ; 32(6): 1636-48, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24449409

RESUMO

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.


Assuntos
Proteínas da Mielina/metabolismo , Células-Tronco Neurais/citologia , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais , Apolipoproteínas E/metabolismo , Comunicação Autócrina/efeitos dos fármacos , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Tamanho Celular , Sobrevivência Celular/efeitos dos fármacos , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Embrião de Mamíferos/citologia , Feminino , Proteínas Ligadas por GPI/deficiência , Proteínas Ligadas por GPI/metabolismo , Células HEK293 , Humanos , Proteínas da Mielina/deficiência , Proteínas da Mielina/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Proteínas Nogo , Receptor Nogo 1 , Comunicação Parácrina/efeitos dos fármacos , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Receptores de Superfície Celular/deficiência , Receptores Imunológicos/deficiência , Transdução de Sinais/efeitos dos fármacos , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo
7.
Spine J ; 14(8): 1722-33, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24462452

RESUMO

BACKGROUND CONTEXT: Transplantation of bone marrow cells into spinal cord lesions promotes functional recovery in animal models, and recent clinical trials suggest possible recovery also in humans. The mechanisms responsible for these improvements are still unclear. PURPOSE: To characterize spinal cord motor neurite interactions with human bone marrow stromal cells (MSCs) in an in vitro model of spinal cord injury (SCI). STUDY DESIGN/SETTING: Previously, we have reported that human MSCs promote the growth of extending sensory neurites from dorsal root ganglia (DRG), in the presence of some of the molecules present in the glial scar, which are attributed with inhibiting axonal regeneration after SCI. We have adapted and optimized this system replacing the DRG with a spinal cord culture to produce a central nervous system (CNS) model, which is more relevant to the SCI situation. METHODS: We have developed and characterized a novel spinal cord culture system. Human MSCs were cocultured with spinal motor neurites in substrate choice assays containing glial scar-associated inhibitors of nerve growth. In separate experiments, MSC-conditioned media were analyzed and added to spinal motor neurites in substrate choice assays. RESULTS: As has been reported previously with DRG, substrate-bound neurocan and Nogo-A repelled spinal neuronal adhesion and neurite outgrowth, but these inhibitory effects were abrogated in MSC/spinal cord cocultures. However, unlike DRG, spinal neuronal bodies and neurites showed no inhibition to substrates of myelin-associated glycoprotein. In addition, the MSC secretome contained numerous neurotrophic factors that stimulated spinal neurite outgrowth, but these were not sufficient stimuli to promote spinal neurite extension over inhibitory concentrations of neurocan or Nogo-A. CONCLUSIONS: These findings provide novel insight into how MSC transplantation may promote regeneration and functional recovery in animal models of SCI and in the clinic, especially in the chronic situation in which glial scars (and associated neural inhibitors) are well established. In addition, we have confirmed that this CNS model predominantly comprises motor neurons via immunocytochemical characterization. We hope that this model may be used in future research to test various other potential interventions for spinal injury or disease states.


Assuntos
Células-Tronco Mesenquimais/citologia , Neurônios Motores/citologia , Neuritos/fisiologia , Adulto , Animais , Embrião de Galinha , Técnicas de Cocultura , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/fisiologia , Proteínas da Mielina/farmacologia , Neuritos/efeitos dos fármacos , Neurocam/farmacologia , Proteínas Nogo , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia
8.
Mol Med Rep ; 8(3): 883-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23842701

RESUMO

Hypoxic ischemic encephalopathy is a serious condition due to inadequate oxygen supply to the brain. Regeneration of neural cells is a critical process for repairing the damaged brain. Nogo has been identified as an inhibitor of neurite outgrowth that is specific to the brain. In the present study, the Nogo-A receptor (NgR) antagonist NEP1-40 was used to study the effects of inhibition of NgR on the regeneration of neural cells and the related Wnt signaling pathway in newborn rats. The investigation focused on the transcription factors regulated in the Wnt signaling pathway during the repair process, together with the proliferation of neural cells. The results indicated that c-Jun and c-Myc were the main transcription factors involved in the Wnt signaling pathway, while neural cell proliferation in the subventricular zone was increased during this process.


Assuntos
Proteínas da Mielina/antagonistas & inibidores , Proteínas da Mielina/farmacologia , Fragmentos de Peptídeos/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Proliferação de Células/efeitos dos fármacos , Dinoprosta/análogos & derivados , Dinoprosta/análise , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Proteínas da Mielina/metabolismo , Proteínas da Mielina/uso terapêutico , Regeneração Nervosa/efeitos dos fármacos , Neurônios/citologia , Neurônios/metabolismo , Proteínas Nogo , Fragmentos de Peptídeos/uso terapêutico , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ratos , Ratos Wistar , Regulação para Cima/efeitos dos fármacos
9.
Cell Transplant ; 21(6): 1177-97, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22236767

RESUMO

Adult central mammalian axons show minimal regeneration after spinal cord injury due to loss of oligodendrocytes, demyelination of surviving axons, absence of growth-promoting molecules, and inhibitors of axonal outgrowth. In the present study, we attempted to address these impediments to regeneration by using a combinatory strategy to enhance cell survival and regeneration after complete spinal cord transection (SCT) in adult rats. The strategy comprised: 1) adult rat brain-derived neural stem/progenitor cells (NSPCs) preseeded on laminin-coated chitosan channels; 2) extramedullary chitosan channels to promote axonal regrowth and reduce the barrier caused by scarring; 3) local delivery of a novel rat soluble Nogo-66 receptor protein [NgR(310)ecto-Fc, referred to as NgR] to block the inhibitory effect of myelin-based inhibitors; and 4) local delivery of basic fibroblast growth factor, epidermal growth factor, and platelet-derived growth factor to enhance survival and promote differentiation of transplanted cells. Compared with our previous studies where brain-derived NSPCs preseeded in extramedullary chitosan channels were implanted in the same SCT model but without growth factors and NgR, the present channel-growth factor combination produced greater numbers of surviving NSPCs after SCT. Also, the growth factors promoted preferential differentiation of NSPCs toward oligodendrocytes, while NgR significantly decreased astrocytic differentiation of NSPCs. NgR alone or in combination with NSPCs significantly enhanced the total number of myelinated fibers in the bridge and increased the area of the bridging tissue between the cord stumps. The combination of NgR, growth factors, and NSPCs had synergistic effect on bridge formation. However, only a small number of descending corticospinal tract axons grew into the central portions of the bridges as shown by anterograde tracing of the corticospinal tract with BDA. The majority of the regenerated axons in the channels originated from local host neurons adjacent to the tissue bridges. In conclusion, we showed that growth factors increased survival of transplanted NSPCs whereas NgR enhanced axonal regeneration, but the combination did not have additive effects on functional recovery or regeneration.


Assuntos
Axônios/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas da Mielina/farmacologia , Células-Tronco Neurais/citologia , Traumatismos da Medula Espinal/terapia , Animais , Diferenciação Celular , Quitosana/química , Fator de Crescimento Epidérmico/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Proteínas Ligadas por GPI/farmacologia , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Regeneração Nervosa/fisiologia , Células-Tronco Neurais/transplante , Receptor Nogo 1 , Oligodendroglia/citologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia
10.
Br J Pharmacol ; 163(7): 1507-19, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21449980

RESUMO

BACKGROUND AND PURPOSE: Cannabis extracts and several cannabinoids have been shown to exert broad anti-inflammatory activities in experimental models of inflammatory CNS degenerative diseases. Clinical use of many cannabinoids is limited by their psychotropic effects. However, phytocannabinoids like cannabidiol (CBD), devoid of psychoactive activity, are, potentially, safe and effective alternatives for alleviating neuroinflammation and neurodegeneration. EXPERIMENTAL APPROACH: We used experimental autoimmune encephalomyelitis (EAE) induced by myelin oligodendrocyte glycoprotein (MOG) in C57BL/6 mice, as a model of multiple sclerosis. Using immunocytochemistry and cell proliferation assays we evaluated the effects of CBD on microglial activation in MOG-immunized animals and on MOG-specific T-cell proliferation. KEY RESULTS: Treatment with CBD during disease onset ameliorated the severity of the clinical signs of EAE. This effect of CBD was accompanied by diminished axonal damage and inflammation as well as microglial activation and T-cell recruitment in the spinal cord of MOG-injected mice. Moreover, CBD inhibited MOG-induced T-cell proliferation in vitro at both low and high concentrations of the myelin antigen. This effect was not mediated via the known cannabinoid CB(1) and CB(2) receptors. CONCLUSIONS AND IMPLICATIONS: CBD, a non-psychoactive cannabinoid, ameliorates clinical signs of EAE in mice, immunized against MOG. Suppression of microglial activity and T-cell proliferation by CBD appeared to contribute to these beneficial effects.


Assuntos
Canabidiol/farmacologia , Microglia/efeitos dos fármacos , Esclerose Múltipla/tratamento farmacológico , Medula Espinal/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Animais , Lesão Axonal Difusa/tratamento farmacológico , Modelos Animais de Doenças , Progressão da Doença , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/tratamento farmacológico , Feminino , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Proteínas da Mielina/farmacologia , Glicoproteína Mielina-Oligodendrócito , Degeneração Neural/tratamento farmacológico , Linfócitos T/imunologia
11.
J Neurochem ; 116(5): 900-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21214567

RESUMO

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.


Assuntos
Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Caínico/toxicidade , N-Metilaspartato/toxicidade , Receptores de Superfície Celular/uso terapêutico , Convulsões/prevenção & controle , Animais , Anticorpos/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Suscetibilidade a Doenças/induzido quimicamente , Suscetibilidade a Doenças/metabolismo , Suscetibilidade a Doenças/patologia , Suscetibilidade a Doenças/terapia , Inibidores Enzimáticos/farmacologia , Hipocampo/citologia , Humanos , Técnicas In Vitro , Cadeias beta de Integrinas/imunologia , Imageamento por Ressonância Magnética/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas da Mielina/farmacologia , Glicoproteína Associada a Mielina , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Fosfoinositídeo Fosfolipase C/farmacologia , Receptores de Superfície Celular/deficiência , Convulsões/induzido quimicamente , Convulsões/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tubulina (Proteína)/metabolismo
12.
Cell Death Differ ; 17(9): 1392-408, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20094059

RESUMO

Neuronal outgrowth is guided by both extrinsic and intrinsic factors, involving transcriptional regulation. The acetylation of histones and transcription factors, which facilitates promoter accessibility, ultimately promotes transcription, and depends on the balance between histone deacetylases (HDACs) and histone acetyltransferases (HATs) activities. However, a critical function for specific acetylation modifying enzymes in neuronal outgrowth has yet to be investigated. To address this issue, we have used an epigenetic approach to facilitate gene expression in neurons, by using specific HDAC inhibitors. Neurons treated with a combination of HDAC and transcription inhibitors display an acetylation and transcription-dependent increase in outgrowth and a reduction in growth cone collapse on both 'permissive' (poly-D-lysine, PDL) and 'non-permissive' substrates (myelin and chondroitin sulphate proteoglycans (CSPGs)). Next, we specifically show that the expression of the histone acetyltransferases CBP/p300 and P/CAF is repressed in neurons by inhibitory substrates, whereas it is triggered by HDAC inhibition on both permissive and inhibitory conditions. Gene silencing and gain of function experiments show that CBP/p300 and P/CAF are key players in neuronal outgrowth, acetylate histone H3 at K9-14 and the transcription factor p53, thereby initiating a pro-neuronal outgrowth transcriptional program. These findings contribute to the growing understanding of transcriptional regulation in neuronal outgrowth and may lay the molecular groundwork for the promotion of axonal regeneration after injury.


Assuntos
Crescimento Celular/efeitos dos fármacos , Cones de Crescimento/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Neurônios/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Animais , Células Cultivadas , Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Proteoglicanas de Sulfatos de Condroitina/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Inativação Gênica , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 2/antagonistas & inibidores , Histona Desacetilases/metabolismo , Histonas/metabolismo , Modelos Neurológicos , Proteínas da Mielina/farmacologia , Neuritos/efeitos dos fármacos , Neurônios/citologia , Neurônios/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ratos , Ratos Endogâmicos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/genética , Fatores de Transcrição de p300-CBP/genética
13.
J Biol Chem ; 285(9): 6425-33, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-20018888

RESUMO

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.


Assuntos
Proteínas da Mielina/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Células 3T3 , Animais , Carcinógenos , Adesão Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Fibroblastos/metabolismo , Inibidores do Crescimento , Humanos , Camundongos , Regeneração Nervosa , Neuritos/efeitos dos fármacos , Neuritos/ultraestrutura , Neurônios/metabolismo , Proteínas Nogo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo
14.
Proc Natl Acad Sci U S A ; 106(41): 17511-6, 2009 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-19805174

RESUMO

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.


Assuntos
Macrófagos/fisiologia , Proteínas da Mielina/farmacologia , Proteínas da Mielina/fisiologia , Animais , Velocidade do Fluxo Sanguíneo/fisiologia , Movimento Celular/fisiologia , Inflamação/prevenção & controle , Isquemia/fisiopatologia , Isquemia/prevenção & controle , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/fisiologia , Proteínas da Mielina/deficiência , Proteínas da Mielina/genética , Proteínas Nogo , Fluxo Sanguíneo Regional/fisiologia , Regulação para Cima , Cicatrização/fisiologia
15.
Neurosci Lett ; 460(1): 41-6, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19446604

RESUMO

Myelin contains many axonal outgrowth inhibitory components which contribute to regeneration failure after neuronal injury in the mammalian central nervous system (CNS). In an attempt to develop small molecular agents to promote axonal outgrowth, we screened a compound library purified from traditional Chinese herbs, and found a small molecular compound polygalasaponin G (PS-G), extracted from Polygala japonica, which has a potent neurotrophic activity on PC12 cells and cultured cortical neurons. We reported, to our knowledge for the first time, that PS-G could promote neurite outgrowth of neurons cultured on the myelin substrates and inhibit the activation of RhoA. Thus, our results could represent a therapeutic approach to improve axon regeneration after CNS injuries.


Assuntos
Bainha de Mielina/fisiologia , Neuritos/efeitos dos fármacos , Neurônios/citologia , Polygala/química , Saponinas/farmacologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Cerebelo/citologia , Cerebelo/fisiologia , Relação Dose-Resposta a Droga , Marcação In Situ das Extremidades Cortadas/métodos , Proteínas da Mielina/farmacologia , Fator de Crescimento Neural/farmacologia , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Proteínas Nogo , Células PC12 , Ratos , Saponinas/química , Proteína rhoA de Ligação ao GTP/metabolismo
16.
Mol Ther ; 17(7): 1173-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19293775

RESUMO

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.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/genética , Proteínas da Mielina/farmacologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Simplexvirus/genética , Traumatismos da Medula Espinal/terapia , Animais , Arginase/metabolismo , Axônios/fisiologia , Western Blotting , Carbazóis/farmacologia , Linhagem Celular , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Gânglios Espinais , Imunoquímica , Neuritos/fisiologia , Neurônios/metabolismo , Fosforilação/imunologia , Pirróis/farmacologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/citologia
17.
J Neuroimmunol ; 200(1-2): 27-40, 2008 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-18657868

RESUMO

A role for mast cells (MC) in the pathogenesis of multiple sclerosis (MS) has been suggested, based on the analysis of human lesions and on an animal model of the disease (EAE). What role MC play in the development of MS is not well understood. We hypothesized that the link connecting MC with demyelinating diseases may be represented by their interaction with myelin. Here we show that myelin can activate mast cells. This process could be a key event in the mast cell function required for inducing EAE in mice and possibly in MS in man.


Assuntos
Mastócitos/fisiologia , Bainha de Mielina/metabolismo , Receptores Depuradores/fisiologia , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Células Cultivadas , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Citometria de Fluxo , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Masculino , Mastócitos/efeitos dos fármacos , Camundongos , Microscopia Eletrônica/métodos , Proteínas da Mielina/farmacologia , Neutrófilos/metabolismo , Neutrófilos/ultraestrutura , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Ratos , Ratos Wistar , Superóxidos/metabolismo , Fatores de Tempo , Aderências Teciduais/metabolismo
18.
J Cell Sci ; 120(Pt 11): 1877-87, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17488779

RESUMO

The migration of olfactory ensheathing cells (OECs) is essential for pioneering the olfactory nerve pathway during development and for promoting axonal regeneration when implanted into the injured central nervous system (CNS). In the present study, recombinant Nogo-66 enhanced the adhesion of OECs and inhibited their migration. Using immunocytochemistry and western blot, we showed that the Nogo-66 receptor (NgR) was expressed on OECs. When NgR was released from the cell surface with phosphatidylinositol-specific phospholipase C or neutralized by NgR antibody, the effect of Nogo-66 on OEC adhesion and migration was markedly attenuated. Nogo-66 was found to promote the formation of focal adhesion in OECs and inhibited their membrane protrusion through the activation of RhoA. Furthermore, the co-culture migration assay demonstrated that OEC motility was significantly restricted by Nogo-A expressed on Cos7 cell membranes or oligodendrocytes. Moreover, treatment with anti-NgR antibody facilitated migration of implanted OECs in a spinal cord hemisection injury model. Taken together, we demonstrate, for the first time, that Nogo, a myelin-associated inhibitor of axon regeneration in the CNS, enhances the adhesion and inhibits the migration of OECs via NgR regulation of RhoA.


Assuntos
Movimento Celular/efeitos dos fármacos , Proteínas da Mielina/farmacologia , Condutos Olfatórios/citologia , Condutos Olfatórios/efeitos dos fármacos , Animais , Adesão Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Proteínas Ligadas por GPI , Proteínas da Mielina/metabolismo , Proteínas Nogo , Receptor Nogo 1 , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Células PC12 , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular , Receptores de Peptídeos/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
19.
FASEB J ; 20(11): 1939-41, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16849393

RESUMO

After binding, central nervous system (CNS) myelin-derived axon growth inhibitory ligands, the Nogo-66 receptor (NgR), complexes with LINGO-1 and either the low-affinity neurotrophin receptor (p75(NTR)) or TROY to initiate growth cone collapse via a Rho-A inhibitory signaling pathway and/or Ca(2+)-dependent activation of epidermal growth factor receptor (EGFR) through an unknown signaling pathway. We have shown that axon growth through CNS myelin is disinhibited after neurotrophic factor administration by 1) initiating intramembranous proteolysis (RIP) of p75(NTR), leading to cleavage of the extracellular (p75(ECD)) and intracellular domains (p75(ICD)) by alpha- and gamma-secretase, respectively, thereby paralyzing inhibitory signaling; 2) shedding of soluble NgR(ECD), which acts as a competitive antagonist to NgR for binding of inhibitory ligands; and 3) antagonizing NgR/p75(NTR) clustering by competitive p75(ECD)/NgR interaction. Here, we report that TNF-alpha converting enzyme (TACE) (a disintegrin and metalloproteinase 17, ADAM17) induces disinhibition of FGF2-stimulated neurite outgrowth of dorsal root ganglion neurons (DRGN) cultured in the presence of a predetermined concentration of inhibitory CNS myelin-derived ligands. After addition of TACE (which has alpha-secretase activity) to mitotically arrested adult rat mixed DRG cultures, we demonstrate 1) NgR(ECD) shedding; 2) release of p75(ECD) and p75(ICD) by RIP of p75(NTR); 3) blockade of Rho-A activation; 4) reduced EGFR phosphorylation; and 5) increased FGF2-stimulated DRGN neurite outgrowth and branching in the presence of CNS myelin-derived inhibitory ligands. Thus, TACE-induced cleavage of NgR and RIP of p75(NTR) abrogates axon growth inhibitory signaling, thereby disinhibiting CNS axon/neurite growth.


Assuntos
Proteínas ADAM/metabolismo , Gânglios Espinais/fisiologia , Proteínas da Mielina/farmacologia , Neuritos/fisiologia , Receptor de Fator de Crescimento Neural/fisiologia , Receptores de Superfície Celular/fisiologia , Proteínas ADAM/farmacologia , Proteína ADAM17 , Animais , Células Cultivadas , Sistema Nervoso Central/fisiologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteínas Ligadas por GPI , Gânglios Espinais/citologia , Imuno-Histoquímica , Proteínas da Mielina/fisiologia , Neuritos/efeitos dos fármacos , Neuritos/ultraestrutura , Receptor Nogo 1 , Ratos , Ratos Sprague-Dawley
20.
J Neurochem ; 96(6): 1616-25, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16441511

RESUMO

CNS myelin inhibits axon growth due to the expression of several growth-inhibitory proteins, including myelin-associated glycoprotein, oligodendrocyte myelin glycoprotein and Nogo. Myelin-associated inhibitory proteins activate rho GTPase in responsive neurons. Rho kinase (ROCK) has been implicated as a critical rho effector in this pathway due to the ability of the pharmacological inhibitor Y-27632 to circumvent myelin-dependent inhibition. Y-27632, however, inhibits the activity of additional kinases. Using three independent approaches, we provide direct evidence that ROCKII is activated in response to the myelin-associated inhibitor Nogo. We demonstrate that Nogo treatment enhances ROCKII translocation to the cellular membrane in PC12 cells and enhances ROCKII kinase activity towards an in vitro substrate. In addition, Nogo treatment enhances phosphorylation of myosin light chain II, a known ROCK substrate. Further, we demonstrate that primary dorsal root ganglia neurons can be rendered insensitive to the inhibitory effects of myelin via infection with dominant negative ROCK. Together these data provide direct evidence for a rho-ROCK-myosin light chain-II signaling cascade in response to myelin-associated inhibitors.


Assuntos
Inibidores do Crescimento/metabolismo , Proteínas da Mielina/metabolismo , Sistema Nervoso/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Amidas/farmacologia , Animais , Miosinas Cardíacas/metabolismo , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Gânglios Espinais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas da Mielina/farmacologia , Cadeias Leves de Miosina/metabolismo , Regeneração Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Proteínas Nogo , Células PC12 , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Piridinas/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA