Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 150: 105259, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33434618

RESUMO

Neuronal regeneration in the injured central nervous system is hampered by multiple extracellular proteins. These proteins exert their inhibitory action through interactions with receptors that are located in cholesterol rich compartments of the membrane termed lipid rafts. Here we show that cholesterol-synthesis inhibition prevents the association of the Neogenin receptor with lipid rafts. Furthermore, we show that cholesterol-synthesis inhibition enhances axonal growth both on inhibitory -myelin and -RGMa substrates. Following optic nerve injury, lowering cholesterol synthesis with both drugs and siRNA-strategies allows for robust axonal regeneration and promotes neuronal survival. Cholesterol inhibition also enhanced photoreceptor survival in a model of Retinitis Pigmentosa. Our data reveal that Lovastatin leads to several opposing effects on regenerating axons: cholesterol synthesis inhibition promotes regeneration whereas altered prenylation impairs regeneration. We also show that the lactone prodrug form of lovastatin has differing effects on regeneration when compared to the ring-open hydroxy-acid form. Thus the association of cell surface receptors with lipid rafts contributes to axonal regeneration inhibition, and blocking cholesterol synthesis provides a potential therapeutic approach to promote neuronal regeneration and survival in the diseased Central Nervous System. SIGNIFICANCE STATEMENT: Statins have been intensively used to treat high levels of cholesterol in humans. However, the effect of cholesterol inhibition in both the healthy and the diseased brain remains controversial. In particular, it is unclear whether cholesterol inhibition with statins can promote regeneration and survival following injuries. Here we show that late stage cholesterol inhibition promotes robust axonal regeneration following optic nerve injury. We identified distinct mechanisms of action for activated vs non-activated Lovastatin that may account for discrepancies found in the literature. We show that late stage cholesterol synthesis inhibition alters Neogenin association with lipid rafts, thereby i) neutralizing the inhibitory function of its ligand and ii) offering a novel opportunity to promote CNS regeneration and survival following injuries.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nervo Óptico/efeitos dos fármacos , Animais , Anticolesterolemiantes/farmacologia , Axônios/efeitos dos fármacos , Axônios/patologia , Sobrevivência Celular , Embrião de Galinha , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Bainha de Mielina , Neurônios/metabolismo , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Células Fotorreceptoras , Prenilação , Pró-Fármacos , Ratos , Retina , Retinose Pigmentar , Dicloridrato de trans-1,4-Bis(2-clorobenzaminometil)ciclo-hexano/farmacologia
2.
PLoS One ; 14(8): e0220056, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31369591

RESUMO

Abortive cell cycle (ACC) re-entry of apoptotic neurons is a recently characterized phenomenon that occurs after central nervous system (CNS) injury or over the course of CNS disease. Consequently, inhibiting cell cycle progression is neuroprotective in numerous CNS pathology models. Primary cilia are ubiquitous, centriole-based cellular organelles that prevent cell cycling, but their ability to modulate abortive cell cycle has not been described. Here, we show that neuronal cilia are ablated in-vitro and in-vivo following injury by hypoxia or optic nerve transection (ONT), respectively. Furthermore, forced cilia resorption sensitized neurons to these injuries and enhanced cell death. In contrast, pharmacological inhibition or shRNA knockdown of the proteins that disassemble the cilia increased neuron survival and decreased the phosphorylation of retinoblastoma (Rb), a master switch for cell cycle re-entry. Our findings show that the stabilization of neuronal primary cilia inhibits, at least transiently, apoptotic cell cycling, which has implications for future therapeutic strategies that halt or slow the progression of neurodegenerative diseases and acute CNS injuries.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo Celular , Sistema Nervoso Central/patologia , Cílios/fisiologia , Neurônios/patologia , Traumatismos do Nervo Óptico/patologia , Substâncias Protetoras/farmacologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/lesões , Cílios/efeitos dos fármacos , Etoposídeo/farmacologia , Feminino , Hipóxia , Neurônios/efeitos dos fármacos , Traumatismos do Nervo Óptico/tratamento farmacológico , Fosforilação , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia
3.
Invest Ophthalmol Vis Sci ; 60(2): 634-649, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30743263

RESUMO

Purpose: Integrin adherence to the extracellular matrix (ECM) is essential for retinal ganglion cell (RGC) survival: damage causes production and release of ECM degrading matrix metalloproteinases (MMPs) that disrupt integrin ligation, leading to RGC death. The interplay of MMPs, integrins, and focal adhesion kinase (FAK) was studied in RGCs after optic nerve injury. Methods: Optic nerve transection and optic nerve crush were used to study RGC survival and regeneration, respectively. Treatments were administered intravitreally or into the cut end of the optic nerve. RGC survival was assessed by fluorescence or confocal microscopy; cell counting, peptide levels, and localization were assessed by Western blot and immunohistochemistry. Results: MMP-9 was most strongly increased and localized to RGCs after injury. Pan-MMP, MMP-2/-9, and MMP-3 inhibition all significantly enhanced RGC survival and increased RGC axon regeneration. FAK activation was decreased at 4 days postaxotomy, when apoptosis begins. FAK inhibition reduced RGC survival and abrogated the neuroprotective effects of MMP inhibition, whereas FAK activation increased RGC survival despite MMP activation. Integrin ligation with CD29 antibody or glycine-arginine-glycine-aspatate-serine (GRGDS) peptide increased RGC survival after axotomy. Conclusions: ECM-integrin ligation promotes RGC survival and axon regeneration via FAK activation.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Integrina beta1/metabolismo , Metaloproteinases da Matriz/metabolismo , Traumatismos do Nervo Óptico/fisiopatologia , Regeneração/fisiologia , Células Ganglionares da Retina/fisiologia , Animais , Western Blotting , Contagem de Células , Sobrevivência Celular/fisiologia , Matriz Extracelular/enzimologia , Feminino , Imuno-Histoquímica , Inibidores de Metaloproteinases de Matriz/farmacologia , Microscopia Confocal , Microscopia de Fluorescência , Compressão Nervosa , Oligopeptídeos/farmacologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/citologia
4.
J Stroke Cerebrovasc Dis ; 27(4): 845-856, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29196198

RESUMO

BACKGROUND: A reliable model of ischemia-reperfusion is required to evaluate the efficacy and safety of neuroprotective therapies for stroke. We present a novel reproducible pterygopalatine-ophthalmic artery ligation model of ischemia-reperfusion injury in the retina. METHODS: Rats were subjected to ophthalmic artery/meningeal sheath ligation (OAML-standard method) or clamping of the pterygopalatine-ophthalmic artery (OAC-new method) for 30 minutes. Retinal ganglion cell (RGC) survival was assessed by prelabeling with FluoroGold (FG) (Santa Cruz Biotechnology, CA, USA) and RNA-binding protein with multiple splicing (RBPMS) at 14 days after ischemia, and all results were compared with a sham group (n = 7 in each group). RESULTS: RGC density in the normal-uninjured (FG-labeled) group was 2111 ± 38 cells/mm2 (mean ± standard error of mean) and that in the RBPMS-labeled group was 2142 ± 35 cells/mm2. The OAML procedure significantly reduced RGC density to 738 ± 23 cells/mm2 and 780 ± 41 cells/mm2 (P < .001) in the FG-labeled and RBPMS-labeled groups, respectively. Similarly, OAC reduced RGC survival to 782 ± 19 cells/mm2 and 813 ± 22 cells/mm2 (P < .001) in the FG-labeled and RBPMS-labeled groups, respectively. RGC survival was similar following OAC and OAML models, suggesting that both induce comparable levels of damage. However, RGC survival in the OAC model was found to have less dispersion than OAML-induced ischemia. CONCLUSIONS: These results suggest that the OAC procedure is a reliable reproduction of ischemia-reperfusion injury that mimics the effects of ophthalmic artery occlusion in humans and provides a useful research model for testing manipulations directed against pathways involved in RGC ischemic degeneration.


Assuntos
Procedimentos Neurocirúrgicos , Artéria Oftálmica/cirurgia , Traumatismo por Reperfusão/etiologia , Células Ganglionares da Retina/patologia , Animais , Biomarcadores/metabolismo , Sobrevivência Celular , Constrição , Modelos Animais de Doenças , Feminino , Ligadura , Artéria Oftálmica/fisiopatologia , Proteínas de Ligação a RNA/metabolismo , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Células Ganglionares da Retina/metabolismo , Fatores de Tempo
5.
Cell Rep ; 20(1): 99-111, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28683327

RESUMO

Developing strategies that promote axonal regeneration within the injured CNS is a major therapeutic challenge, as axonal outgrowth is potently inhibited by myelin and the glial scar. Although regeneration can be achieved using the genetic deletion of PTEN, a negative regulator of the mTOR pathway, this requires inactivation prior to nerve injury, thus precluding therapeutic application. Here, we show that, remarkably, fibroblast-derived exosomes (FD exosomes) enable neurite growth on CNS inhibitory proteins. Moreover, we demonstrate that, upon treatment with FD exosomes, Wnt10b is recruited toward lipid rafts and activates mTOR via GSK3ß and TSC2. Application of FD exosomes shortly after optic nerve injury promoted robust axonal regeneration, which was strongly reduced in Wnt10b-deleted animals. This work uncovers an intercellular signaling pathway whereby FD exosomes mobilize an autocrine Wnt10b-mTOR pathway, thereby awakening the intrinsic capacity of neurons for regeneration, an important step toward healing the injured CNS.


Assuntos
Comunicação Autócrina , Axônios/metabolismo , Exossomos/metabolismo , Regeneração Nervosa , Traumatismos do Nervo Óptico/metabolismo , Proteínas Wnt/metabolismo , Animais , Axônios/fisiologia , Células COS , Células Cultivadas , Chlorocebus aethiops , Fibroblastos/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Células HEK293 , Humanos , Microdomínios da Membrana/metabolismo , Camundongos , Nervo Óptico/metabolismo , Nervo Óptico/fisiologia , Células PC12 , Ratos , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo , Proteínas Wnt/genética
6.
Cell Rep ; 8(4): 1146-59, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25127134

RESUMO

Ideal strategies to ameliorate CNS damage should promote both neuronal survival and axon regeneration. The receptor Neogenin promotes neuronal apoptosis. Its ligand prevents death, but the resulting repulsive guidance molecule a (RGMa)-Neogenin interaction also inhibits axonal growth, countering any prosurvival benefits. Here, we explore strategies to inhibit Neogenin, thus simultaneously enhancing survival and regeneration. We show that bone morphogenetic protein (BMP) and RGMa-dependent recruitment of Neogenin into lipid rafts requires an interaction between RGMa and Neogenin subdomains. RGMa or Neogenin peptides that prevent this interaction, BMP inhibition by Noggin, or reduction of membrane cholesterol all block Neogenin raft localization, promote axon outgrowth, and prevent neuronal apoptosis. Blocking Neogenin raft association influences axonal pathfinding, enhances survival in the developing CNS, and promotes survival and regeneration in the injured adult optic nerve and spinal cord. Moreover, lowering cholesterol disrupts rafts and restores locomotor function after spinal cord injury. These data reveal a unified strategy to promote both survival and regeneration in the CNS.


Assuntos
Microdomínios da Membrana/fisiologia , Regeneração Nervosa , Animais , Axônios/fisiologia , Galinhas , Feminino , Cones de Crescimento/fisiologia , Macrolídeos , Proteínas de Membrana/metabolismo , Neurônios Motores/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Nervo Óptico/patologia , Nervo Óptico/fisiopatologia , Ratos Wistar , Células Ganglionares da Retina/fisiologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia
7.
PLoS One ; 9(7): e101349, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24983470

RESUMO

Retinal ganglion cells (RGCs) are neurons that relay visual signals from the retina to the brain. The RGC cell bodies reside in the retina and their fibers form the optic nerve. Full transection (axotomy) of the optic nerve is an extra-retinal injury model of RGC degeneration. Optic nerve transection permits time-kinetic studies of neurodegenerative mechanisms in neurons and resident glia of the retina, the early events of which are reported here. One day after injury, and before atrophy of RGC cell bodies was apparent, glia had increased levels of phospho-Akt, phospho-S6, and phospho-ERK1/2; however, these signals were not detected in injured RGCs. Three days after injury there were increased levels of phospho-Rb and cyclin A proteins detected in RGCs, whereas these signals were not detected in glia. DNA hyperploidy was also detected in RGCs, indicative of cell cycle re-entry by these post-mitotic neurons. These events culminated in RGC death, which is delayed by pharmacological inhibition of the MAPK/ERK pathway. Our data show that a remote injury to RGC axons rapidly conveys a signal that activates retinal glia, followed by RGC cell cycle re-entry, DNA hyperploidy, and neuronal death that is delayed by preventing glial MAPK/ERK activation. These results demonstrate that complex and variable neuro-glia interactions regulate healthy and injured states in the adult mammalian retina.


Assuntos
Ciclo Celular , Neuroglia/enzimologia , Traumatismos do Nervo Óptico/complicações , Degeneração Retiniana/etiologia , Células Ganglionares da Retina/metabolismo , Animais , Axotomia , Morte Celular , Feminino , Cinética , Nervo Óptico/fisiopatologia , Ratos , Ratos Wistar , Retina/enzimologia , Degeneração Retiniana/enzimologia , Degeneração Retiniana/metabolismo , Transdução de Sinais
8.
Invest Ophthalmol Vis Sci ; 55(3): 1919-29, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24526440

RESUMO

PURPOSE: BM88 is a cell-cycle exit and neuronal differentiation protein that has been used as a marker of surviving retinal ganglion cells (RGCs) after optic nerve injury. Thy1.1 has also been used as a marker for RGC loss, but after optic nerve crush (ONC) a decrease in Thy1.1 expression precedes the loss of RGCs. The purpose of this study was to determine if BM88 expression was correlated with RGC loss after ONC and optic nerve transection (ONT) injuries. METHODS: Rats were injected with Fluorogold (FG) into the superior colliculus to label RGCs and received ONC or ONT 7 days later. Eyes were collected 2 to 28 days after injury. Retinas were labeled with BM88 and intensity of the BM88 cell labeling was measured. RESULTS: In control retinas, 98.9% of RGCs were immunoreactive (-IR) for BM88. There was a significant downregulation of BM88 by 52% to 80% of RGCs 7 days after ONC or ONT. The staining intensity of the remaining labeled cells was reduced to 41% to 51% of the control after 28 days of optic nerve injury. However, early in the injury there was a significant increase in the staining intensity of BM88. CONCLUSIONS: Nearly all BM88-IR RGCs colocalized with FG-labeled RGCs in control retinas. However, both the number of BM88-IR RGCs and their intensity decreased gradually between 4 and 28 days, preceding the loss of FG-labeled cells. These findings indicate that BM88 is not a good marker of surviving RGCs but may indicate abnormal RGC functioning, which precedes cell death.


Assuntos
Regulação para Baixo , Proteínas de Membrana/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Traumatismos do Nervo Óptico/metabolismo , Células Ganglionares da Retina/patologia , Animais , Biomarcadores , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Traumatismos do Nervo Óptico/patologia , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/metabolismo
9.
Acta Pharmacol Sin ; 34(1): 91-103, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23202803

RESUMO

Retinal ischemia is a very useful model to study the impact of various cell death pathways, such as apoptosis and necrosis, in the ischemic retina. However, it is important to note that the retina is formed as an outpouching of the diencephalon and is part of the central nervous system. As such, the cell death pathways initiated in response to ischemic damage in the retina reflect those found in other areas of the central nervous system undergoing similar trauma. The retina is also more accessible than other areas of the central nervous system, thus making it a simpler model to work with and study. By utilizing the retinal model, we can greatly increase our knowledge of the cell death processes initiated by ischemia which lead to degeneration in the central nervous system. This paper examines work that has been done so far to characterize various aspects of cell death in the retinal ischemia model, such as various pathways which are activated, and the role neurotrophic factors, and discusses how these are relevant to the treatment of ischemic damage in both the retina and the greater central nervous system.


Assuntos
Sistema Nervoso Central/fisiopatologia , Isquemia/fisiopatologia , Retina/fisiopatologia , Doenças Retinianas/fisiopatologia , Vasos Retinianos/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Morte Celular , Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Humanos , Isquemia/metabolismo , Fatores de Crescimento Neural/metabolismo , Retina/metabolismo , Doenças Retinianas/metabolismo , Vasos Retinianos/metabolismo , Acidente Vascular Cerebral/metabolismo
10.
Invest Ophthalmol Vis Sci ; 53(7): 3973-89, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22531700

RESUMO

PURPOSE: Retinal ganglion cell (RGC) degeneration is an important cause of visual impairment and can be modeled by optic nerve transection, which causes the death of 90% of RGCs within 14 days postaxotomy. We performed a proteomic study to identify and quantify proteins in the rat retina after optic nerve transection. Our goal was to isolate potential targets for therapeutic intervention to prevent RGC degeneration. METHODS: iTRAQ proteomics was used to analyze adult rat retinas at 1, 3, 4, 7, 14, and 21 days postaxotomy. Hepatoma-derived growth factor (HDGF), a target identified by iTRAQ, was delivered by intraocular injections. Wortmannin or PD98059 were coadministered with HDGF to determine if the protective effects of HDGF are dependent on PI3 kinase or MAP kinase activity, respectively. RESULTS: At a false-discovery rate of 5%, 216 proteins were identified by iTRAQ proteomics, 71 of which showed changes in expression (<0.7× or >1.3×) at one time point after injury: 52 proteins had expression peaks, whereas 19 showed downward expression spikes. Levels of GAPDH did not change after axotomy. Among these differentially expressed proteins was HDGF. HDGF delivery significantly increased RGC survival compared with control treatments, and increased Akt phosphorylation in the retina at 24 hours after intraocular injection. RGC rescue by HDGF was dependent on both MAP kinase and PI3 kinase activity in the retina. CONCLUSIONS: We have identified numerous proteins that are differentially regulated at key time points after axotomy, and how the temporal profiles of their expression parallel RGC death. Using these data, we showed that HDGF is a potent neuroprotective factor for injured adult RGCs.


Assuntos
Proteínas do Olho/análise , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Fármacos Neuroprotetores/farmacologia , Traumatismos do Nervo Óptico/metabolismo , Nervo Óptico/metabolismo , Degeneração Retiniana/prevenção & controle , Células Ganglionares da Retina/metabolismo , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/complicações , Traumatismos do Nervo Óptico/patologia , Ratos , Ratos Sprague-Dawley , Degeneração Retiniana/etiologia , Degeneração Retiniana/metabolismo , Células Ganglionares da Retina/patologia , Transdução de Sinais
11.
J Neurotrauma ; 28(9): 1863-79, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21902538

RESUMO

Recent studies have identified anti-apoptotic functions for vascular endothelial growth factor (VEGF) in the central nervous system (CNS). However, VEGF therapy has been hampered by a tendency to promote vascular permeability, edema, and inflammation. Recently, engineered zinc finger proteins (ZFPs) that upregulate multiple forms of VEGF in their natural biological ratios, have been developed to overcome these negative side effects. We used retinal trauma and ischemia models, and a cortical pial strip ischemia model to determine if VEGF upregulating ZFPs are neuroprotective in the adult CNS. Optic nerve transection and ophthalmic artery ligation lead to the apoptotic degeneration of retinal ganglion cells (RGCs) and are, respectively, two highly reproducible models for CNS trauma or ischemia. Adeno-associated vectors (AAV) vectors encoding VEGF-ZFPs (AAV-VEGF-ZFP) significantly increased RGC survival by ∼twofold at 14 days after optic nerve transection or ophthalmic artery ligation. Furthermore, AAV-VEGF-ZFP enhanced recovery of the pupillary light reflex. RECA-1 immunostaining demonstrated no appreciable differences between retinas treated with AAV-VEGF-ZFP and controls, suggesting that AAV-VEGF-ZFP treatment did not affect retinal vasculature. Following pial strip of the forelimb motor cortex, brains treated with an adenovirus encoding VEGF ZFPs (AdV-ZFP) showed higher neuronal survival, accelerated wound contraction, and reduced lesion volume between 1 and 6 weeks after injury. Behavioral testing using the cylinder test for vertical exploration showed that AdV-VEGF-ZFP treatment enhanced contralateral forelimb function within the first 2 weeks after injury. Our results indicate that VEGF ZFP therapy is neuroprotective following traumatic injury or stroke in the adult mammalian CNS.


Assuntos
Lesões Encefálicas/terapia , Terapia Genética/métodos , Acidente Vascular Cerebral/terapia , Regulação para Cima/genética , Fator A de Crescimento do Endotélio Vascular/genética , Dedos de Zinco/genética , Animais , Comportamento Animal/fisiologia , Lesões Encefálicas/genética , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/terapia , Engenharia de Proteínas , Ratos , Recuperação de Função Fisiológica/genética , Acidente Vascular Cerebral/genética , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
J Neurosci ; 31(29): 10494-505, 2011 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-21775595

RESUMO

To promote functional recovery after CNS injuries, it is crucial to develop strategies that enhance both neuronal survival and regeneration. Here, we report that caspase-6 is upregulated in injured retinal ganglion cells and that its inhibition promotes both survival and regeneration in these adult CNS neurons. Treatment of rat retinal whole mounts with Z-VEID-FMK, a selective inhibitor of caspase-6, enhanced ganglion cell survival. Moreover, retinal explants treated with this drug extended neurites on myelin. We also show that caspase-6 inhibition resulted in improved ganglion cell survival and robust axonal regeneration following optic nerve injury in adult rats. The effects of Z-VEID-FMK were similar to other caspase inhibitory peptides including Z-LEHD-FMK and Z-VAD-FMK. In searching for downstream effectors for caspase-6, we identified caspase-8, whose expression pattern resembled that of caspase-6 in the injured eye. We then showed that caspase-8 is activated downstream of caspase-6 in the injured adult retina. Furthermore, we investigated the role of caspase-8 in RGC apoptosis and regenerative failure both in vitro and in vivo. We observed that caspase-8 inhibition by Z-IETD-FMK promoted survival and regeneration to an extent similar to that obtained with caspase-6 inhibition. Our results indicate that caspase-6 and caspase-8 are components of a cellular pathway that prevents neuronal survival and regeneration in the adult mammalian CNS.


Assuntos
Apoptose/fisiologia , Caspase 6/metabolismo , Caspase 8/metabolismo , Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/enzimologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Axônios/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Toxina da Cólera , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Proteína GAP-43/metabolismo , Injeções Intraoculares/métodos , Masculino , Bainha de Mielina/metabolismo , Regeneração Nervosa/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Degeneração Retrógrada/prevenção & controle , Estilbamidinas
13.
J Proteome Res ; 10(8): 3344-62, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21627321

RESUMO

Retinal ganglion cells (RGCs) are central nervous system (CNS) neurons that transmit visual information from the retina to the brain. Apoptotic RGC degeneration causes visual impairment that can be modeled by optic nerve crush. Neuronal apoptosis is also a salient feature of CNS trauma, ischemia (stroke), and diseases of the CNS such as Alzheimer's, Parkinson's, multiple sclerosis, and amyotrophic lateral sclerosis. Optic nerve crush induces the apoptotic cell death of ∼ 70% of RGCs within the first 14 days after injury. This model is particularly attractive for studying adult neuron apoptosis because the time-course of RGC death is well established and axon regeneration within the myelinated optic nerve can be concurrently evaluated. Here, we performed a large scale iTRAQ proteomic study to identify and quantify proteins of the rat retina at 1, 3, 4, 7, 14, and 21 days after optic nerve crush. In total, 337 proteins were identified, and 110 were differentially regulated after injury. Of these, 58 proteins were upregulated (>1.3 ×), 46 were downregulated (<0.7 ×), and 6 showed both positive and negative regulation over 21 days, relative to normal retinas. Among the differentially expressed proteins, Thymosin-ß4 showed an early upregulation at 3 days, the time-point that immediately precedes the induction of RGC apoptosis after injury. We examined the effect of exogenous Thymosin-ß4 administration on RGC death after optic nerve injury. Intraocular injections of Thymosin-ß4 significantly increased RGC survival by ∼ 3-fold compared to controls and enhanced axon regeneration after crush, demonstrating therapeutic potential for CNS insults. Overall, our study identified numerous proteins that are differentially regulated at key time-points after optic nerve crush, and how the temporal profiles of their expression parallel RGC death. This data will aid in the future development of novel therapeutics to promote neuronal survival and regeneration in the adult CNS.


Assuntos
Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/patologia , Espectrometria de Massas em Tandem/métodos , Animais , Apoptose , Cromatografia Líquida de Alta Pressão , Proteínas do Olho/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Proteômica , Ratos , Células Ganglionares da Retina/metabolismo
14.
J Vis Exp ; (51)2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21610673

RESUMO

Retinal ganglion cells (RGCs) are CNS neurons that output visual information from the retina to the brain, via the optic nerve. The optic nerve can be accessed within the orbit of the eye and completely transected (axotomized), cutting the axons of the entire RGC population. Optic nerve transection is a reproducible model of apoptotic neuronal cell death in the adult CNS (1-4). This model is particularly attractive because the vitreous chamber of the eye acts as a capsule for drug delivery to the retina, permitting experimental manipulations via intraocular injections. The diffusion of chemicals through the vitreous fluid ensures that they act upon the entire RGC population. Moreover, RGCs can be selectively transfected by applying short interfering RNAs (siRNAs), plasmids, or viral vectors to the cut end of the optic nerve (5-7) or injecting vectors into their target, the superior colliculus (8). This allows researchers to study apoptotic mechanisms in the desired neuronal population without confounding effects on other bystander neurons or surrounding glia. An additional benefit is the ease and accuracy with which cell survival can be quantified after injury. The retina is a flat, layered tissue and RGCs are localized in the innermost layer, the ganglion cell layer. The survival of RGCs can be tracked over time by applying a fluorescent tracer (3% Fluorogold) to the cut end of the optic nerve at the time of axotomy, or by injecting the tracer into the superior colliculus (RGC target) one week prior to axotomy. The tracer is retrogradely transported, labeling the entire RGC population. Because the ganglion cell layer is a monolayer (one cell thick), RGC densities can be quantified in flat-mounted tissue, without the need for stereology. Optic nerve transection leads to the apoptotic death of 90% of injured RGCs within 14 days postaxotomy (9-11). RGC apoptosis has a characteristic time-course whereby cell death is delayed 3-4 days postaxotomy, after which the cells rapidly degenerate. This provides a time window for experimental manipulations directed against pathways involved in apoptosis.


Assuntos
Apoptose/fisiologia , Sistema Nervoso Central/citologia , Neurônios/citologia , Procedimentos Neurocirúrgicos/métodos , Nervo Óptico/citologia , Nervo Óptico/cirurgia , Células Ganglionares da Retina/citologia , Animais , Ratos
15.
J Vis Exp ; (51)2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21610674

RESUMO

Retinal ganglion cells (RGCs) are CNS neurons that output visual information from the retina to the brain, via the optic nerve. The optic nerve can be accessed within the orbit of the eye and completely transected (axotomized), cutting the axons of the entire RGC population. Optic nerve transection is a reproducible model of apoptotic neuronal cell death in the adult CNS (1-4). This model is particularly attractive because the vitreous chamber of the eye acts as a capsule for drug delivery to the retina, permitting experimental manipulations via intraocular injections. The diffusion of chemicals through the vitreous fluid ensures that they act upon the entire RGC population. Viral vectors, plasmids or short interfering RNAs (siRNAs) can also be delivered to the vitreous chamber in order to infect or transfect retinal cells (5-12). The high tropism of Adeno-Associated Virus (AAV) vectors is beneficial to target RGCs, with an infection rate approaching 90% of cells near the injection site (6, 7, 13-15). Moreover, RGCs can be selectively transfected by applying siRNAs, plasmids, or viral vectors to the cut end of the optic nerve (16-19) or injecting vectors into their target the superior colliculus (10). This allows researchers to study apoptotic mechanisms in the injured neuronal population without confounding effects on other bystander neurons or surrounding glia. RGC apoptosis has a characteristic time-course whereby cell death is delayed 3-4 days postaxotomy, after which the cells rapidly degenerate. This provides a window for experimental manipulations directed against pathways involved in apoptosis. Manipulations that directly target RGCs from the transected optic nerve stump are performed at the time of axotomy, immediately after cutting the nerve. In contrast, when substances are delivered via an intraocular route, they can be injected prior to surgery or within the first 3 days after surgery, preceding the initiation of apoptosis in axotomized RGCs. In the present article, we demonstrate several methods for experimental manipulations after optic nerve transection.


Assuntos
Sistema Nervoso Central/cirurgia , Procedimentos Neurocirúrgicos/métodos , Nervo Óptico/cirurgia , Células Ganglionares da Retina/citologia , Animais , Ratos
16.
Channels (Austin) ; 4(5): 337-46, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20699649

RESUMO

Retinal ganglion cell (RGC) degeneration is an important cause of visual impairment, and results in part from microglia-mediated inflammation. Numerous experimental studies have focused on identifying drug targets to rescue these neurons. We recently showed that K(V)1.1 and K(V)1.3 channels are expressed in adult rat RGCs and that siRNA-mediated knockdown of either channel reduces RGC death after optic nerve transection. Earlier we found that K(V)1.3 channels also contribute to microglial activation and neurotoxicity; raising the possibility that these channels contribute to neurodegeneration through direct roles in RGCs and through inflammatory mechanisms. Here, RGC survival was increased by combined siRNA-mediated knockdown of K(V)1.1 and K(V)1.3 in RGCs, but survival was much greater when knockdown of either channel was combined with intraocular injection of a K(V)1.3 channel blocker (agitoxin-2 or margatoxin). After axotomy, increased expression of several inflammation-related molecules preceded RGC loss and, consistent with a dual mechanism, their expression was differentially affected when channel knockdown in RGCs was combined with K(V)1.3 blocker injection. K(V)1.3 blockers reduced activation of retinal microglia and their tight apposition along RGC axon fascicles after axotomy, but did not prevent their migration from the inner plexiform to the damaged ganglion cell layer. Expression of several growth factors increased after axotomy; and again, there were differences following blocker injection compared with RGC-selective channel knockdown. These results provide evidence that K(V)1.3 channels play important roles in apoptotic degeneration of adult RGCs through cell-autonomous mechanisms mediated by channels in the neurons, and nonautonomous mechanisms mediated by microglia and inflammation.


Assuntos
Inflamação/tratamento farmacológico , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Apoptose , Axotomia , Canal de Potássio Kv1.3/antagonistas & inibidores , Degeneração Neural , Ratos , Células Ganglionares da Retina/patologia , Venenos de Escorpião/farmacologia
17.
J Neurosci ; 27(1): 234-44, 2007 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-17202491

RESUMO

Brain damage and disease involve activation of microglia and production of potentially neurotoxic molecules, but there are no treatments that effectively target their harmful properties. We present evidence that the small-conductance Ca2+/calmodulin-activated K+ channel KCNN4/ KCa3.1/SK4/IK1 is highly expressed in rat microglia and is a potential therapeutic target for acute brain damage. Using a Transwell cell-culture system that allows separate treatment of the microglia or neurons, we show that activated microglia killed neurons, and this was markedly reduced by treating only the microglia with a selective inhibitor of KCa3.1 channels, triarylmethane-34 (TRAM-34). To assess the role of KCa3.1 channels in microglia activation and key signaling pathways involved, we exploited several fluorescence plate-reader-based assays. KCa3.1 channels contributed to microglia activation, inducible nitric oxide synthase upregulation, production of nitric oxide and peroxynitrite, and to consequent neurotoxicity, protein tyrosine nitration, and caspase 3 activation in the target neurons. Microglia activation involved the signaling pathways p38 mitogen-activated protein kinase (MAPK) and nuclear factor kappaB (NF-kappaB), which are important for upregulation of numerous proinflammatory molecules, and the KCa3.1 channels were functionally linked to activation of p38 MAPK but not NF-kappaB. These in vitro findings translated into in vivo neuroprotection, because we found that degeneration of retinal ganglion cells after optic nerve transection was reduced by intraocular injection of TRAM-34. This study provides evidence that KCa3.1 channels constitute a therapeutic target in the CNS and that inhibiting this K+ channel might benefit acute and chronic neurodegenerative disorders that are caused by or exacerbated by inflammation.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Microglia/metabolismo , Microglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico/metabolismo , Animais , Células Cultivadas , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Ratos , Ratos Wistar
18.
J Neurobiol ; 59(1): 162-80, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15007834

RESUMO

Both attractive and repellent cues are required to guide developing axons to their targets in the central nervous system. Critical guidance molecules in the developing brain include the semaphorins, netrins, slits, and ephrins. Current research indicates that many of these molecules and their receptors are expressed in the adult central nervous system (CNS), and that injury can alter the levels of these ligands/receptors. Recent studies have begun the process of elucidating the functions of these receptors in adult mammals, and the effects that they have on the regeneration of adult neurons. This review addresses our current knowledge with respect to the response of adult CNS neurons to axonal injury, interventions for enhancing the survival and regeneration of injured neurons, and the expression of developmental axon guidance cues in the injured mature CNS, with specific focus on the retino-tectal projection.


Assuntos
Axônios/fisiologia , Sinais (Psicologia) , Regeneração Nervosa/fisiologia , Neurônios/fisiologia , Animais , Apoptose/fisiologia , Lesões Encefálicas/fisiopatologia , Lesões Encefálicas/terapia , Sistema Nervoso Central/citologia , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/fisiologia , Efrinas/metabolismo , Lateralidade Funcional/fisiologia , Modelos Neurológicos , Proteínas da Mielina/metabolismo , Fatores de Crescimento Neural/metabolismo , Proteínas Nogo , Nervos Periféricos/transplante , Células Ganglionares da Retina/fisiologia , Semaforinas/metabolismo , Transplantes
19.
Neuroscience ; 110(3): 555-67, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11906793

RESUMO

In the present study we localized glial cell line-derived neurotrophic factor (GDNF), and the high affinity receptor for GDNF (GFRalpha-1) in the rat retina. We also examined the effects of neurturin on the survival of axotomized retinal ganglion cells (RGCs) and compared neurturin-mediated RGC rescue to GDNF and brain-derived neurotrophic factor (BDNF) neuroprotection. We administered combined injections of neurturin with BDNF or GDNF in order to determine if these factors rescue RGCs by different mechanisms. GDNF immunoreactivity was localized to RGCs, photoreceptors, and retinal pigment epithelial cells. GFRalpha-1 immunoreactivity was localized to RGCs, Müller cells, and photoreceptors. RGC densities in control retinas decreased from the original value of 2481+/-121 (RGCs/mm(2)+/-S.D.) to 347+/-100 at 14 days post-axotomy. Neurturin treatment significantly increased RGC survival after axotomy (745+/-94) similar to GDNF (868+/-110). BDNF treatment resulted in higher RGC survival (1109+/-156) than either neurturin or GDNF. Combined administration of neurturin with BDNF had additive effects on the survival of axotomized RGCs (1962+/-282), similar to combined administration of GDNF and BDNF (1825+/-269). Combined administration of neurturin and GDNF (1265+/-178) had an enhanced effect on RGC survival. These results suggest that neurturin, GDNF, and BDNF act independently to rescue injured RGCs. Our results also suggest that RGCs and retinal Müller cells may be responsive to GDNF because they both express GFRalpha-1. The present findings have implications for the rescue of injured retinal ganglion cells, as well as other CNS neurons that are responsive to neurturin, GDNF, and BDNF, including midbrain dopaminergic neurons and motor neurons.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Drosophila , Degeneração Neural/tratamento farmacológico , Fatores de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/farmacologia , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Axotomia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Interações Medicamentosas/fisiologia , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Imuno-Histoquímica , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neurturina , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ret , Ratos , Receptores Proteína Tirosina Quinases/metabolismo , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...