Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 161: 105542, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34737043

RESUMO

BACKGROUND: Vitamin A (VitA), via its active metabolite retinoic acid (RA), is critical for the maintenance of memory function with advancing age. Although its role in Alzheimer's disease (AD) is not well understood, data suggest that impaired brain VitA signaling is associated with the accumulation of ß-amyloid peptides (Aß), and could thus contribute to the onset of AD. METHODS: We evaluated the protective action of a six-month-long dietary VitA-supplementation (20 IU/g), starting at 8 months of age, on the memory and the neuropathology of the 3xTg-AD mouse model of AD (n = 11-14/group; including 4-6 females and 7-8 males). We also measured protein levels of Retinoic Acid Receptor ß (RARß) and Retinoid X Receptor γ (RXRγ) in homogenates from the inferior parietal cortex of 60 participants of the Religious Orders study (ROS) divided in three groups: no cognitive impairment (NCI) (n = 20), mild cognitive impairment (MCI) (n = 20) and AD (n = 20). RESULTS: The VitA-enriched diet preserved spatial memory of 3xTg-AD mice in the Y maze. VitA-supplementation affected hippocampal RXR expression in an opposite way according to sex by tending to increase in males and decrease in females their mRNA expression. VitA-enriched diet also reduced the amount of hippocampal Aß40 and Aß42, as well as the phosphorylation of tau protein at sites Ser396/Ser404 (PHF-1) in males. VitA-supplementation had no effect on tau phosphorylation in females but worsened their hippocampal Aß load. However, the expression of Rxr-ß in the hippocampus was negatively correlated with the amount of both soluble and insoluble Aß in both males and females. Western immunoblotting in the human cortical samples of the ROS study did not reveal differences in RARß levels. However, it evidenced a switch from a 60-kDa-RXRγ to a 55-kDa-RXRγ in AD, correlating with ante mortem cognitive decline and the accumulation of neuritic plaques in the brain cortex. CONCLUSION: Our data suggest that (i) an altered expression of RXRs receptors is a contributor to ß-amyloid pathology in both humans and 3xTg-AD mice, (ii) a chronic exposure of 3xTg-AD mice to a VitA-enriched diet may be protective in males, but not in females.


Assuntos
Doença de Alzheimer , Vitamina A , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Dieta , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Receptores X de Retinoides/metabolismo , Proteínas tau/metabolismo
2.
Cell Death Dis ; 11(2): 101, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32029703

RESUMO

N-Methyl-D-aspartate (NMDA)-induced neuronal cell death is involved in a large spectrum of diseases affecting the brain and the retina such as Alzheimer's disease and diabetic retinopathy. Associated neurological impairments may result from the inhibition of neuronal plasticity by Nogo-A. The objective of the current study was to determine the contribution of Nogo-A to NMDA excitotoxicity in the mouse retina. We observed that Nogo-A is upregulated in the mouse vitreous during NMDA-induced inflammation. Intraocular injection of a function-blocking antibody specific to Nogo-A (11C7) was carried out 2 days after NMDA-induced injury. This treatment significantly enhanced visual function recovery in injured animals. Strikingly, the expression of potent pro-inflammatory molecules was downregulated by 11C7, among which TNFα was the most durably decreased cytokine in microglia/macrophages. Additional analyses suggest that TNFα downregulation may stem from cofilin inactivation in microglia/macrophages. 11C7 also limited gliosis presumably via P.Stat3 downregulation. Diabetic retinopathy was associated with increased levels of Nogo-A in the eyes of donors. In summary, our results reveal that Nogo-A-targeting antibody can stimulate visual recovery after retinal injury and that Nogo-A is a potent modulator of excitotoxicity-induced neuroinflammation. These data may be used to design treatments against inflammatory eye diseases.


Assuntos
Células Amácrinas/efeitos dos fármacos , Anti-Inflamatórios/farmacologia , Anticorpos Neutralizantes/farmacologia , Proteínas Nogo/antagonistas & inibidores , Células Ganglionares da Retina/efeitos dos fármacos , Retinite/prevenção & controle , Visão Ocular/efeitos dos fármacos , Idoso , Idoso de 80 Anos ou mais , Células Amácrinas/metabolismo , Células Amácrinas/patologia , Animais , Retinopatia Diabética/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , N-Metilaspartato , Plasticidade Neuronal/efeitos dos fármacos , Proteínas Nogo/metabolismo , Fosforilação , Recuperação de Função Fisiológica , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Retinite/induzido quimicamente , Retinite/metabolismo , Retinite/fisiopatologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
3.
Glia ; 66(10): 2079-2093, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30051920

RESUMO

Nogo-A is a potent glial-derived inhibitor of axon growth in the injured CNS and acts as a negative regulator of developmental angiogenesis by inhibiting vascular endothelial cell migration. However, its function in pathological angiogenesis has never been studied after ischemic injury in the CNS. Using the mouse model of oxygen-induced retinopathy (OIR) which yields defined zones of retinal ischemia, our goal was to investigate the role of Nogo-A in vascular regeneration. We demonstrate a marked upregulation of the Nogo-A receptor sphingosine 1-phosphate receptor 2 in blood vessels following OIR, while Nogo-A is abundantly expressed in surrounding glial cells. Acute inhibition of Nogo-A with function-blocking antibody 11C7 significantly improved vascular regeneration and consequently prevented pathological pre-retinal angiogenesis. Ultimately, inhibition of Nogo-A led to restoration of retinal function as determined by electrophysiological response of retinal cells to light stimulation. Our data suggest that anti-Nogo-A antibody may protect neuronal cells from ischemic damage by accelerating blood vessel repair in the CNS. Targeting Nogo-A by immunotherapy may improve CNS perfusion after vascular injuries.


Assuntos
Isquemia/metabolismo , Neovascularização Fisiológica/fisiologia , Proteínas Nogo/metabolismo , Regeneração/fisiologia , Doenças Retinianas/metabolismo , Vasos Retinianos/metabolismo , Indutores da Angiogênese/farmacologia , Animais , Modelos Animais de Doenças , Isquemia/tratamento farmacológico , Isquemia/patologia , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Proteínas Nogo/antagonistas & inibidores , Proteínas Nogo/imunologia , Receptores de Lisoesfingolipídeo/metabolismo , Regeneração/efeitos dos fármacos , Doenças Retinianas/tratamento farmacológico , Doenças Retinianas/patologia , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/patologia , Receptores de Esfingosina-1-Fosfato , Visão Ocular/efeitos dos fármacos , Visão Ocular/fisiologia
4.
Neural Plast ; 2017: 6818970, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29234527

RESUMO

The lack of axonal regeneration and neuronal cell death causes permanent neurological deficits in the injured CNS. Using the classical CNS injury model of optic nerve crush in mice, ciliary neurotrophic factor (CNTF) was found to stimulate retinal ganglion cell (RGC) survival and axonal growth, but in an incomplete fashion. The elucidation of molecular mechanisms impairing CNTF-induced axonal regeneration is paramount to promote visual recovery. In the present study, we sought to evaluate the contribution of sphingosine 1-phosphate receptor 1 (S1PR1) to the neuroprotective and regenerative effects of CNTF. The transduction of retinal cells with adeno-associated viruses (AAV) allowed to activate CNTF/signal transducer and activator of transcription 3 (Stat3) signaling and to modulate S1PR1 expression in RGCs. Our results showed that CNTF/Stat3 prevented injury-induced S1PR1 downregulation. Silencing S1PR1 in RGCs significantly enhanced CNTF-induced axonal growth in the injured optic nerve. In contrast, RGC survival was markedly decreased when S1PR1 was repressed with viral vectors. The level of phosphorylated Stat3 (P-Stat3), an intracellular mediator of CNTF, did not fluctuate after S1PR1 inhibition and CNTF stimulation. Collectively, these results suggest that S1PR1 acts as a major regulator of retinal neuron survival and restricts the RGC growth response induced by CNTF.


Assuntos
Axônios/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fator Neurotrófico Ciliar/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Axônios/metabolismo , Regulação para Baixo/efeitos dos fármacos , Camundongos , Regeneração Nervosa/fisiologia , Neuroproteção/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
5.
Neurosci Lett ; 627: 168-77, 2016 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-27276652

RESUMO

In the adult retina, we have previously shown that Nogo-A was highly expressed in Müller glia. However, the role of Nogo-A in the glial cell physiology is not clear. In this study, we investigated the possible influence that Nogo-A may exert on other polarized molecules in Müller cells, in particular inwardly rectifying potassium channel 4.1 (Kir4.1) and aquaporin 4 (AQP4) that respectively control potassium and water exchange in glial cells. Our results showed that adenovirus-mediated Nogo-A overexpression with AdNogo-A increased the immunofluorescent signal of Kir4.1 in rat Müller cell line 1 (rMC-1) cells but did not change its expression level by Western blotting. In vivo, AdNogo-A induced ectopic Kir4.1 immunoreactivity throughout the radial processes of Müller cells compared with AdLacZ control virus. Surprisingly, AdNogo-A did not modify the distribution of Dp71 and AQP4 that are common binding partners for Kir4.1 in the dystrophin-associated protein (DAP) complex anchored at the plasma membrane of Müller glia. Immunoprecipitation experiments revealed molecular interactions between Nogo-A and Kir4.1. In Nogo-A KO mouse retinae, the distribution of Kir4.1 was not different from that observed in Wild-Type (WT) animals. In addition, potassium conductance did not change in freshly dissociated Nogo-A KO Müller glia compared with WT cells. In summary, the increase of Nogo-A expression can selectively influence the distribution of Kir4.1 in glia but is not essential for Kir4.1-mediated potassium conductance at the plasma membrane in physiological conditions. Nogo-A-Kir4.1 interactions may, however, contribute to pathological processes taking place in the retina, for instance, after ischemia.


Assuntos
Aquaporina 4/metabolismo , Células Ependimogliais/metabolismo , Proteínas Nogo/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Adenoviridae/fisiologia , Animais , Células Cultivadas , Distrofina/metabolismo , Vetores Genéticos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Potássio/metabolismo , Ratos , Regulação para Cima
6.
J Neurochem ; 138(4): 571-86, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27309795

RESUMO

In this study, we used a classical optic nerve injury model to address the function of the sphingosine 1-phosphate (S1P)-S1P receptor (S1PR) axis in retinal ganglion cell (RGC) death and axonal growth. After lesion, the expression of S1PR1 was generally reduced in axotomized RGCs but persisted in αRGCs, a subpopulation of injury-resistant RGCs. Silencing S1PR1 with an adeno-associated virus serotype 2 (AAV2) containing a shRNA specific to S1PR1 (AAV2.shRNA-S1PR1) exacerbated the loss of RGCs induced by optic nerve crush; the rate of RGC survival was decreased by more than 24% in retinae infected with AAV2.shRNA-S1PR1 compared with AAV2.shRNA-scrambled or AAV2.GFP control treatments. In the superior and temporal regions of the retina, cell death rose by more than ~ 35% and ~ 50%, respectively, in comparison with control groups. In the optic nerve, S1PR1 silencing markedly reduced axonal sprouting after the lesion relative to control animals. Early after optic nerve crush, 67% of αRGCs stained for osteopontin were lost in retinae infected with AAV2.shRNA-S1PR1, whereas the number of intrinsically photosensitive RGCs expressing melanopsin, another injury-resistant RGC type, was not affected. Moreover, retinal infection with AAV2.shRNA-S1PR1 down-regulated mammalian target of rapamycin pathway activation in αRGCs. Together, our results reveal that S1PR1 contributes to survival and growth mechanisms in injured RGCs by regulating the mammalian target of rapamycin pathway. The role of sphingosine 1-phosphate receptor 1 (S1PR1) was studied in retinal ganglion cell survival and axonal growth after optic nerve injury. After axonal damage, S1PR1 expression was decreased in retinal neurons. Viral-mediated S1PR1 down-regulation enhanced injury-induced cell death and reduced spontaneous axonal growth. In injury-resistant retinal neurons, the activation of mTOR signalling was down-regulated by silencing S1PR1, suggesting an important role for S1PR1 in neuronal growth and survival mechanisms in vivo.


Assuntos
Traumatismos do Nervo Óptico/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Axônios/metabolismo , Sobrevivência Celular , Regulação para Baixo , Masculino , Camundongos Endogâmicos C57BL , Compressão Nervosa/métodos , Regeneração Nervosa/fisiologia , Neuritos/fisiologia , Nervo Óptico/patologia , Células Ganglionares da Retina/citologia , Receptores de Esfingosina-1-Fosfato
7.
Proc Natl Acad Sci U S A ; 110(21): E1943-52, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23625008

RESUMO

Nogo-A is an important axonal growth inhibitor in the adult and developing CNS. In vitro, Nogo-A has been shown to inhibit migration and cell spreading of neuronal and nonneuronal cell types. Here, we studied in vivo and in vitro effects of Nogo-A on vascular endothelial cells during angiogenesis of the early postnatal brain and retina in which Nogo-A is expressed by many types of neurons. Genetic ablation or virus-mediated knock down of Nogo-A or neutralization of Nogo-A with an antibody caused a marked increase in the blood vessel density in vivo. In culture, Nogo-A inhibited spreading, migration, and sprouting of primary brain microvascular endothelial cells (MVECs) in a dose-dependent manner and induced the retraction of MVEC lamellipodia and filopodia. Mechanistically, we show that only the Nogo-A-specific Delta 20 domain exerts inhibitory effects on MVECs, but the Nogo-66 fragment, an inhibitory domain common to Nogo-A, -B, and -C, does not. Furthermore, the action of Nogo-A Delta 20 on MVECs required the intracellular activation of the Ras homolog gene family, member A (Rho-A)-associated, coiled-coil containing protein kinase (ROCK)-Myosin II pathway. The inhibitory effects of early postnatal brain membranes or cultured neurons on MVECs were relieved significantly by anti-Nogo-A antibodies. These findings identify Nogo-A as an important negative regulator of developmental angiogenesis in the CNS. They may have important implications in CNS pathologies involving angiogenesis such as stroke, brain tumors, and retinopathies.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/crescimento & desenvolvimento , Células Endoteliais/metabolismo , Proteínas da Mielina/metabolismo , Neovascularização Fisiológica/fisiologia , Animais , Encéfalo/citologia , Células Cultivadas , Circulação Cerebrovascular/fisiologia , Células Endoteliais/citologia , Camundongos , Camundongos Knockout , Proteínas da Mielina/genética , Proteínas Nogo
8.
Neurobiol Dis ; 51: 202-13, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23194670

RESUMO

The optic nerve crush injury is a well-accepted model to study the mechanisms of axonal regeneration after trauma in the CNS. The infection of retinal ganglion cells (RGCs) with an adeno-associated virus serotype 2 - ciliary neurotrophic factor (AAV2.CNTF) was previously shown to stimulate axonal regeneration. However, the transfection of axotomized neurons themselves may not be optimal to promote full axonal regeneration in the visual system. Here, we show that the release of CNTF by glial cells is a very powerful stimulus for optic fiber regeneration and RGC survival after optic nerve crush. After 8 weeks, long-distance regeneration of severed optic axons was induced by CNTF until and beyond the optic chiasm. Regenerated axons stayed for at least 6 months in the damaged optic nerve. Strikingly, however, many regenerated axons showed one or several sharp U-turns along their course, suggesting that guidance cues are missing and that long-distance axonal regeneration is limited by the return of the growing axons toward the retina. Even more surprisingly, massive axonal sprouting was observed within the eye, forming a dense plexus of neurites at the inner surface of the retina. These results indicate that massive stimulation of the neuronal growth program can lead to aberrant growth; the absence of local regulatory and guidance factors in the adult, injured optic nerve may therefore represent a major, so far underestimated obstacle to successful axon regeneration.


Assuntos
Fator Neurotrófico Ciliar/genética , Técnicas de Transferência de Genes , Regeneração Nervosa/genética , Traumatismos do Nervo Óptico/genética , Animais , Axônios , Western Blotting , Sobrevivência Celular/genética , Fator Neurotrófico Ciliar/metabolismo , Imunofluorescência , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
J Neurosci ; 29(17): 5536-45, 2009 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-19403821

RESUMO

The central hypothesis of excitotoxicity is that excessive stimulation of neuronal NMDA-sensitive glutamate receptors is harmful to neurons and contributes to a variety of neurological disorders. Glial cells have been proposed to participate in excitotoxic neuronal loss, but their precise role is defined poorly. In this in vivo study, we show that NMDA induces profound nuclear factor kappaB (NF-kappaB) activation in Müller glia but not in retinal neurons. Intriguingly, NMDA-induced death of retinal neurons is effectively blocked by inhibitors of NF-kappaB activity. We demonstrate that tumor necrosis factor alpha (TNFalpha) protein produced in Müller glial cells via an NMDA-induced NF-kappaB-dependent pathway plays a crucial role in excitotoxic loss of retinal neurons. This cell loss occurs mainly through a TNFalpha-dependent increase in Ca(2+)-permeable AMPA receptors on susceptible neurons. Thus, our data reveal a novel non-cell-autonomous mechanism by which glial cells can profoundly exacerbate neuronal death following excitotoxic injury.


Assuntos
Agonistas de Aminoácidos Excitatórios/toxicidade , Neurônios Retinianos/patologia , Neurônios Retinianos/fisiologia , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , N-Metilaspartato/toxicidade , Neurônios Retinianos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/fisiologia
10.
Invest Ophthalmol Vis Sci ; 48(5): 2311-20, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17460296

RESUMO

PURPOSE: In a former study, it was demonstrated that the retina of juvenile Sprague-Dawley (SD) rat has a remarkable intrinsic resistance to light-induced retinopathy compared with the adult retina. The purpose of the present study was to investigate the cellular and molecular mechanisms underlying this endogenous resistance to light-induced damage. METHODS: Juvenile SD rats were exposed for 6 (from P14 to P20) or 14 (from P14 to P28) days to a bright, cyclic, luminous environment of 10,000 lux. Retinal histology was examined immediately after exposure to light or at 2 months of age, and photoreceptor cell death was quantified by measuring the thickness of the outer nuclear layer (ONL) and by TUNEL assays. Changes in protein levels and cellular localization of fibroblast growth factor (FGF)-2, ciliary neurotrophic factor (CNTF), and brain-derived neurotrophic factor (BDNF) were determined by Western blot analysis and retinal immunohistochemistry, respectively. RESULTS: The data demonstrate that although the rate of photoreceptor loss was different after 6 and 14 days of exposure to light, similar ONL thickness was reached at 2 months of age--that is, 4 to 5 weeks after exposure to light. A large number of TUNEL-positive photoreceptors was visualized immediately after 6 and 14 days of exposure to light, reflecting the intense cell death that was occurring in the ONL. Western blot analysis showed that exposure to light induced a strong upregulation of the neurotrophic factors FGF-2 and CNTF in juvenile retinas, whereas no change in BDNF protein expression was noted. Of interest, after exposure to light, endogenous FGF-2 and CNTF were selectively upregulated in Müller cells. CONCLUSIONS: The results show that endogenous expression of FGF-2 and CNTF by Müller glia may play a role in protecting the juvenile retina from light-induced damage.


Assuntos
Fator Neurotrófico Ciliar/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fármacos Neuroprotetores/metabolismo , Lesões Experimentais por Radiação/metabolismo , Retina/efeitos da radiação , Degeneração Retiniana/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Morte Celular , Citoproteção , Modelos Animais de Doenças , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Marcação In Situ das Extremidades Cortadas , Microscopia de Fluorescência , Gravidez , Lesões Experimentais por Radiação/patologia , Lesões Experimentais por Radiação/prevenção & controle , Ratos , Ratos Sprague-Dawley , Retina/metabolismo , Retina/patologia , Degeneração Retiniana/patologia , Degeneração Retiniana/prevenção & controle , Regulação para Cima
11.
Brain ; 129(Pt 4): 1014-26, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16418178

RESUMO

Trauma or disease in the CNS often leads to neuronal death and consequent loss of functional connections. The idea has been put forward that strategies aimed at repairing the injured CNS involve stimulation of both neuronal survival and axon regeneration. We tested this hypothesis in the adult rat retinocollicular system by combining two strategies: (i) exogenous administration of brain-derived neurotrophic factor (BDNF), a potent survival factor for damaged retinal ganglion cells (RGCs) and (ii) lens injury, which promotes robust growth of transected RGC axons. Our results demonstrate that BDNF and lens injury interact synergistically to promote neuronal survival: 71% of RGCs were alive at 2 weeks after optic nerve injury, a time when only approximately 10% of these neurons remain without treatment. Intravitreal injection of BDNF, however, led to regeneration failure following lens injury. The effect of BDNF could not be generalized to other growth factors, as ciliary neurotrophic factor did not cause a significant reduction of lens injury-induced regeneration. Growth arrest in optic nerves treated with BDNF and lens injury correlated with the formation of hypertrophic axonal swellings in the proximal optic nerve. These swellings were filled with numerous vesicular bodies, disorganized neurofilaments and degenerating organelles. Our results demonstrate that: (i) increased neuronal survival does not necessarily lead to enhanced axon regeneration and (ii) activation of survival and growth pathways may produce axonal dystrophy similar to that found in neurodegenerative disorders including glaucoma, Alzheimer's disease and multiple sclerosis. We propose that loss of axonal integrity may limit neuronal recovery in the injured, adult CNS.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Cristalino/lesões , Fármacos Neuroprotetores/farmacologia , Nervo Óptico/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Axônios/fisiologia , Sobrevivência Celular , Feminino , Hipertrofia/patologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Microscopia Eletrônica , Regeneração Nervosa/efeitos dos fármacos , Nervo Óptico/ultraestrutura , Traumatismos do Nervo Óptico/patologia , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/patologia
12.
Invest Ophthalmol Vis Sci ; 46(9): 3383-92, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16123443

RESUMO

PURPOSE: To test the hypothesis that adenovirus (Ad)-mediated gene delivery of brain-derived neurotrophic factor (BDNF) to Müller cells can protect photoreceptors from light-induced retinal degeneration. METHODS: Adult Sprague-Dawley rats received an intraocular injection of Ad.BDNF, control Ad containing the green fluorescent protein (GFP) gene, or BDNF recombinant protein. Animals were then exposed to 5, 10, or 16 days of constant light. The effect of Ad.BDNF on photoreceptor survival was examined histologically, by measuring the outer nuclear layer (ONL) thickness, and functionally, by measuring the electroretinographic (ERG) response. RESULTS: Ad.BDNF mediated sustained expression of bioactive neurotrophin by Müller cells that lasted for at least 30 days after viral vector administration. BDNF gene delivery to Müller glia markedly increased the survival and structural integrity of light-damaged photoreceptors. For example, after 10 days of exposure to light, the average percentage of ONL preservation in the superior central retina of eyes that received Ad.BDNF was 71%, compared with 46% in eyes that received a control Ad.GFP or 15% in contralateral eyes. Of importance, retinas exposed to Ad.BDNF had more photoreceptor nuclei than retinas that received a single intraocular injection of BDNF recombinant protein. The neuroprotective effect of Ad.BDNF was accompanied by preservation of the ERG response of the treated eyes. CONCLUSIONS: These data provide proof of the concept that BDNF gene transfer into Müller cells is an effective strategy for preserving structure and function of photoreceptors in retinal degeneration.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Regulação da Expressão Gênica/fisiologia , Neuroglia/metabolismo , Células Fotorreceptoras/efeitos da radiação , Lesões Experimentais por Radiação/prevenção & controle , Degeneração Retiniana/prevenção & controle , Adenoviridae/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sobrevivência Celular , Eletrorretinografia , Feminino , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas Imunoenzimáticas , Luz , Fatores de Crescimento Neural/biossíntese , Células Fotorreceptoras/fisiopatologia , Lesões Experimentais por Radiação/fisiopatologia , Ratos , Ratos Sprague-Dawley , Degeneração Retiniana/fisiopatologia
13.
Mol Ther ; 12(3): 402-12, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15975850

RESUMO

Glaucoma is the second leading cause of blindness in the world. Loss of vision in glaucomatous optic neuropathy is caused by the selective degeneration of retinal ganglion cells (RGCs). Ocular hypertension is a major risk factor in glaucoma, but visual field defects continue to progress in some patients despite the use of drugs that lower intraocular pressure. At present, there are no effective neuroprotective strategies for the treatment of this disease. The extracellular signal-regulated kinase (Erk) 1/2 pathway is an evolutionarily conserved mechanism used by several peptide factors to promote cell survival. Here we tested if selective activation of Erk1/2 protected RGCs in a rat model of experimental glaucoma. We used recombinant adeno-associated virus to transduce RGCs with genes encoding constitutively active or wild-type MEK1 (approved gene symbol MAP2K1), the upstream activator of Erk1/2. MEK1 gene transfer into RGCs markedly increased neuronal survival: 1366 +/- 70 RGCs/mm(2) (mean +/- SEM) were alive in the dorsal retina at 5 weeks after ocular hypertension surgery, a time when only 680 +/- 86 RGCs/mm(2) of these neurons remained in control eyes. We conclude that the Erk1/2 pathway plays a key role in the protection of RGCs from ocular hypertensive damage. This study identifies a novel gene therapy strategy in which selective activation of the Erk1/2 signaling pathway effectively slows cell death in glaucoma.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Glaucoma/metabolismo , Glaucoma/terapia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Western Blotting , Sobrevivência Celular , Dependovirus/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Vetores Genéticos , Humanos , Imuno-Histoquímica , Pressão Intraocular , MAP Quinase Quinase 1/metabolismo , Masculino , Microscopia de Fluorescência , Neurônios/metabolismo , Ratos , Retina/patologia , Fatores de Tempo
14.
J Neurochem ; 93(1): 72-83, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15773907

RESUMO

Neurotrophins play important roles in the response of adult neurons to injury. The intracellular signaling mechanisms used by neurotrophins to regulate survival and axon growth in the mature CNS in vivo are not well understood. The goal of this study was to define the role of the extracellular signal-regulated kinases 1/2 (Erk1/2) pathway in the survival and axon regeneration of adult rat retinal ganglion cells (RGCs), a prototypical central neuron population. We used recombinant adeno-associated virus (AAV) to selectively transduce RGCs with genes encoding constitutively active or wild-type mitogen-activated protein kinase kinase 1 (MEK1), the upstream activator of Erk1/2. In combination with anterograde and retrograde tracing techniques, we monitored neuronal survival and axon regeneration in vivo. MEK1 gene delivery led to robust and selective transgene expression in multiple RGC compartments including cell bodies, dendrites, axons and targets in the brain. Furthermore, MEK1 activation induced in vivo phosphorylation of Erk1/2 in RGC bodies and axons. Quantitative analysis of cell survival demonstrated that Erk1/2 activation promoted robust RGC neuroprotection after optic nerve injury. In contrast, stimulation of the Erk1/2 pathway was not sufficient to induce RGC axon growth beyond the lesion site. We conclude that the Erk1/2 pathway plays a key role in the survival of axotomized mammalian RGCs in vivo, and that activation of other signaling components is required for axon regeneration in the growth inhibitory CNS environment.


Assuntos
Sistema Nervoso Central/citologia , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Regeneração Nervosa/fisiologia , Neurônios/fisiologia , Traumatismos do Nervo Óptico/fisiopatologia , Animais , Axotomia/métodos , Western Blotting/métodos , Encéfalo/metabolismo , Encéfalo/virologia , Contagem de Células/métodos , Sobrevivência Celular/fisiologia , Toxina da Cólera/metabolismo , Dependovirus/fisiologia , Feminino , Regulação da Expressão Gênica/fisiologia , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde/metabolismo , Hemaglutininas/metabolismo , Neurônios/virologia , Nervo Óptico/metabolismo , Nervo Óptico/virologia , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/virologia , Ratos , Ratos Sprague-Dawley , Retina/metabolismo , Retina/virologia , Células Ganglionares da Retina/fisiologia , Células Ganglionares da Retina/virologia , Estilbamidinas/metabolismo , Fatores de Tempo
15.
J Comp Neurol ; 472(2): 208-20, 2004 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-15048688

RESUMO

The neuropeptide Antho-RFamide is extremely abundant in Renilla koellikeri (sea pansy), a representative of the cnidarians (octocorallians) considered to be closest to the stem ancestors of metazoans with nervous systems. Therefore, a knowledge of the distribution of Antho-RFamide-containing neurons in this species would contribute to our understanding of the early evolution of nervous systems. Using antisera raised against RFamide and FMRFamide, we detected immunostaining in numerous neurons throughout the nervous system of the sea pansy. The antisera revealed ectodermal nerve-nets on the upper and lower sides of the colony and on the oral side of tentacles, in the oral disk, and in the pharynx of feeding polyps. Neurons were immunostained also in the mesogleal nerve-net of feeding polyps and in the through-conducting mesogleal nerve-net of the colonial mass. Varying densities of stained neurons were observed in the different compartments of the endoderm: muscular walls of the feeding and water circulation polyps, mesenteric filaments and their derived follicles containing either ovocytes or spermatophores, in the endodermal channels connecting the different compartments of the colony, and in circular muscle of the peduncle. The distribution of immunostained neurons suggests that they play important roles in feeding, reproduction, neuromuscular transmission, and in neuro-neuronal transmission coordinating the different parts of the colony.


Assuntos
Antozoários/fisiologia , Neurônios/química , Neuropeptídeos/análise , Animais , Antozoários/anatomia & histologia , Antozoários/química , Comportamento Alimentar/fisiologia , Sistema Nervoso/química , Ácido Pirrolidonocarboxílico/análogos & derivados
16.
Gen Comp Endocrinol ; 129(1): 63-8, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12409097

RESUMO

In the sea pansy Renilla koellikeri, estradiol-17beta (E(2)) levels exhibited an annual pattern of secretion that correlated with the reproductive cycle, and displayed sex-specific and tissue-specific differences. The E(2) levels were low during the non-reproductive period extending from autumn to winter. A first rise of E(2) concentrations occurred in March when gonad maturation resumed, as indicated by an increase in lipid storage. This suggests that E(2) may influence the beginning of gonad maturation in the sea pansy. Estradiol-17beta returned to basal levels in April and May when lipids rose sharply. A second, more significant surge of E(2) levels occurred in June when spawning was initiated and it was more marked in female than in male colonies. This suggests that E(2) may participate in synchronising of oocyte maturation around spawning time to optimise the probability of fertilisation. The higher E(2) levels in somatic tissues (peduncle and polyps) than in eggs during the March peak revealed a non-reproductive origin for E(2) and the need for transport of E(2) through the gastrovascular cavity to reach reproductive tissues. Further analyses are required to determine the relative contribution of E(2) to reproductive events and its pathway of synthesis in this colonial anthozoan.


Assuntos
Antozoários/metabolismo , Estradiol/metabolismo , Reprodução/fisiologia , Animais , Estradiol/análise , Feminino , Masculino , Periodicidade , Estações do Ano , Distribuição por Sexo , Fatores Sexuais , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA