Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38363718

RESUMO

ABSTRACT: Medical provider's ability to detect, diagnose, and treat sport-related concussion (SRC) has greatly improved in recent years. Though more is known about the biomechanical forces involved in concussion, it is still uncertain whether there are preventative measures athletes can take to prevent a SRC from occurring. The objective of this review was to determine if either neck size or neck strength is related to a decreased risk of sustaining a SRC. A literature review was conducted on Google Scholar and Ovid MEDLINE for pertinent articles. Findings indicate that isometric neck strength, but not neck size, has been shown to be a predictor for SRC prevention. Formal neck strengthening programs are feasible and lead to decreased SRC risk. Additionally, there may be greater opportunity to increase neck strength in amateur athletes compared to professional. In conclusion, cervical strengthening programs have been shown to be feasible and beneficial for athletes to decrease their risk of sustaining a SRC, though the optimal duration, intensity, and frequency has yet to be determined.

2.
Adv Exp Med Biol ; 1185: 33-37, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31884585

RESUMO

The complement system, commonly associated with innate immune responses to invading pathogens, has been found in the CNS to exert a host of noncanonical functions influential during development and disease. In the retina, local complement expression and activation have been detected in response to injury, and polymorphisms in complement genes have also been linked to the genetic risk for retinal disease. While knowledge regarding the functions, effects, and mechanisms underlying complement in the retina is incomplete, complement expression and activation have been intriguingly linked to both increases and decreases in retinal degeneration in separate contexts and model systems. Here we review the evidence for the varying adaptive and maladaptive contributions of complement and comment on the implications for therapeutic strategies at complement modulation in retinal pathologies.


Assuntos
Ativação do Complemento , Retina/fisiopatologia , Degeneração Retiniana/fisiopatologia , Proteínas do Sistema Complemento , Humanos , Imunidade Inata
3.
J Exp Med ; 216(8): 1925-1943, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31209071

RESUMO

Complement activation has been implicated as contributing to neurodegeneration in retinal and brain pathologies, but its role in retinitis pigmentosa (RP), an inherited and largely incurable photoreceptor degenerative disease, is unclear. We found that multiple complement components were markedly up-regulated in retinas with human RP and the rd10 mouse model, coinciding spatiotemporally with photoreceptor degeneration, with increased C3 expression and activation localizing to activated retinal microglia. Genetic ablation of C3 accelerated structural and functional photoreceptor degeneration and altered retinal inflammatory gene expression. These phenotypes were recapitulated by genetic deletion of CR3, a microglia-expressed receptor for the C3 activation product iC3b, implicating C3-CR3 signaling as a regulator of microglia-photoreceptor interactions. Deficiency of C3 or CR3 decreased microglial phagocytosis of apoptotic photoreceptors and increased microglial neurotoxicity to photoreceptors, demonstrating a novel adaptive role for complement-mediated microglial clearance of apoptotic photoreceptors in RP. These homeostatic neuroinflammatory mechanisms are relevant to the design and interpretation of immunomodulatory therapeutic approaches to retinal degenerative disease.


Assuntos
Ativação do Complemento/imunologia , Complemento C3/metabolismo , Antígeno de Macrófago 1/metabolismo , Microglia/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Retinose Pigmentar/imunologia , Animais , Apoptose/genética , Complemento C3/genética , Modelos Animais de Doenças , Feminino , Humanos , Antígeno de Macrófago 1/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fagocitose/genética , Células Fotorreceptoras de Vertebrados/imunologia , RNA Mensageiro/genética , Retina/patologia , Transdução de Sinais/imunologia
4.
Elife ; 82019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30666961

RESUMO

Constitutive TGFß signaling is important in maintaining retinal neurons and blood vessels and is a factor contributing to the risk for age-related macular degeneration (AMD), a retinal disease involving neurodegeneration and microglial activation. How TGFß signaling to microglia influences pathological retinal neuroinflammation is unclear. We discovered that ablation of the TGFß receptor, TGFBR2, in retinal microglia of adult mice induced abnormal microglial numbers, distribution, morphology, and activation status, and promoted a pathological microglial gene expression profile. TGFBR2-deficient retinal microglia induced secondary gliotic changes in Müller cells, neuronal apoptosis, and decreased light-evoked retinal function reflecting abnormal synaptic transmission. While retinal vasculature was unaffected, TGFBR2-deficient microglia demonstrated exaggerated responses to laser-induced injury that was associated with increased choroidal neovascularization, a hallmark of advanced exudative AMD. These findings demonstrate that deficiencies in TGFß-mediated microglial regulation can drive neuroinflammatory contributions to AMD-related neurodegeneration and neovascularization, highlighting TGFß signaling as a potential therapeutic target.


Assuntos
Neovascularização de Coroide/metabolismo , Microglia/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Retina/metabolismo , Degeneração Retiniana/metabolismo , Animais , Apoptose/genética , Neovascularização de Coroide/genética , Células Ependimogliais/metabolismo , Degeneração Macular/genética , Degeneração Macular/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Degeneração Retiniana/genética , Transdução de Sinais/genética , Transmissão Sináptica/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
5.
Annu Rev Vis Sci ; 4: 45-77, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29852094

RESUMO

Microglia, the primary resident immune cell type, constitute a key population of glia in the retina. Recent evidence indicates that microglia play significant functional roles in the retina at different life stages. During development, retinal microglia regulate neuronal survival by exerting trophic influences and influencing programmed cell death. During adulthood, ramified microglia in the plexiform layers interact closely with synapses to maintain synaptic structure and function that underlie the retina's electrophysiological response to light. Under pathological conditions, retinal microglia participate in potentiating neurodegeneration in diseases such as glaucoma, retinitis pigmentosa, and age-related neurodegeneration by producing proinflammatory neurotoxic cytokines and removing living neurons via phagocytosis. Modulation of pathogenic microglial activation states and effector mechanisms has been linked to neuroprotection in animal models of retinal diseases. These findings have led to the design of early proof-of-concept clinical trials with microglial modulation as a therapeutic strategy.


Assuntos
Microglia/fisiologia , Retina/embriologia , Retina/fisiologia , Doenças Retinianas/patologia , Animais , Humanos , Microglia/patologia , Terapia de Alvo Molecular/métodos , Doenças Retinianas/terapia , Neurônios Retinianos/fisiologia
6.
Exp Eye Res ; 161: 71-81, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28603016

RESUMO

Oxidative injuries, such as those related to reactive oxygen species (ROS), have been implicated in various retinal and optic nerve disorders. Many ROS detection methods have been developed. Although widely utilized, many of these methods are useful only in post mortem tissues, or require relatively expensive equipment, or involve intraocular injection. In the present study, we demonstrated and characterized a chemiluminescent probe L-012 as a noninvasive, in vivo ROS detection agent in the mouse retina. Using optic nerve crush (ONC) and retinal ischemia/reperfusion (I/R) as injury models, we show that L-012 produced intensive luminescent signals specifically in the injured eyes. Histological examination showed that L-012 administration was safe to the retina. Additionally, compounds that reduce tissue superoxide levels, apocynin and TEMPOL, decreased injury-induced L-012 chemiluminescence. The decrease in L-012 signals correlated with their protective effects against retinal I/R-induced morphological and functional changes in the retina. Together, these data demonstrate the feasibility of a fast, simple, reproducible, and non-invasive detection method to monitor in vivo ROS in the retina. Furthermore, the results also show that reduction of ROS is a potential therapeutic approach for protection from these retinal injuries.


Assuntos
Traumatismos do Nervo Óptico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Neurônios Retinianos/metabolismo , Animais , Modelos Animais de Doenças , Eletrorretinografia , Feminino , Substâncias Luminescentes/metabolismo , Luminol/análogos & derivados , Luminol/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Compressão Nervosa , Estresse Oxidativo , Reprodutibilidade dos Testes
7.
Mol Neurodegener ; 11: 30, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-27098079

RESUMO

BACKGROUND: The c-Jun N-terminal kinase (JNK) signaling pathway plays an important role in neuronal pathophysiology. Using JNK inhibitors, we examined involvement of the JNK pathway in cultured rat retinal ganglion cell (RGC) death and in mouse retinal ischemia/reperfusion (I/R) injury of the visual axis. The in vitro effects of JNK inhibitors were evaluated in cultured adult rat retinal cells enriched in RGCs. Retinal I/R was induced in C57BL/6J mice through elevation of intraocular pressure to 120 mmHg for 60 min followed by reperfusion. SP600125 was administered intraperitoneally once daily for 28 days. Phosphorylation of JNK and c-Jun in the retina was examined by immunoblotting and immunohistochemistry. The thickness of retinal layers and cell numbers in the ganglion cell layer (GCL) were examined using H&E stained retinal cross sections and spectral domain optical coherence tomography (SD-OCT). Retinal function was measured by scotopic flash electroretinography (ERG). Volumetric measurement of the superior colliculus (SC) as well as VGLUT2 and PSD95 expression were studied. RESULTS: JNK inhibitors SP600125 and TAT-JNK-III, dose-dependently and significantly (p < 0.05) protected against glutamate excitotoxicity and trophic factor withdrawal induced RGC death in culture. In the I/R model, phosphorylation of JNK (pJNK) in the retina was significantly (p < 0.05) increased after injury. I/R injury significantly (p < 0.05) decreased the thickness of retinal layers, including the whole retina, inner plexiform layer, and inner nuclear layer and cell numbers in the GCL. Administration of SP600125 for 28 days protected against all these degenerative morphological changes (p < 0.05). In addition, SP600125 significantly (p < 0.05) protected against I/R-induced reduction in scotopic ERG b-wave amplitude at 3, 7, 14, 21 and 28 days after injury. SP600125 also protected against the I/R-induced losses in volume and levels of synaptic markers in the SC. Moreover, the protective effects of SP600125 in the retina and SC were also detected even with only 7 days (Days 1-7 after I/R) of SP600125 treatment. CONCLUSIONS: Our results demonstrate the important role the JNK pathway plays in retinal degeneration in both in vitro and in vivo models and suggest that JNK inhibitors may be a useful therapeutic strategy for neuroprotection of RGCs in the retina.


Assuntos
Antracenos/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Retina/metabolismo , Doenças Retinianas/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Pressão Intraocular/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo
8.
Mol Neurodegener ; 11: 24, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-27008854

RESUMO

BACKGROUND: C1q represents the initiating protein of the classical complement cascade, however recent findings indicate pathway independent roles such as developmental pruning of retinal ganglion cell (RGC) axons. Furthermore, chronic neuroinflammation, including increased expression of C1q and activation of microglia and astrocytes, appears to be a common finding among many neurodegenerative disease models. Here we compare the effects of a retinal ischemia/reperfusion (I/R) injury on glial activation and neurodegeneration in wild type (WT) and C1qa-deficient mice in the retina and superior colliculus (SC). Retinal I/R was induced in mice through elevation of intraocular pressure to 120 mmHg for 60 min followed by reperfusion. Glial cell activation and population changes were assessed using immunofluorescence. Neuroprotection was determined using histological measurements of retinal layer thickness, RGC counts, and visual function by flash electroretinography (ERG). RESULTS: Retinal I/R injury significantly upregulated C1q expression in the retina as early as 72 h and within 7 days in the superficial SC, and was sustained as long as 28 days. Accompanying increased C1q expression was activation of microglia and astrocytes as well as a significantly increased glial population density observed in the retina and SC. Microglial activation and changes in density were completely ablated in C1qa-deficient mice, interestingly however there was no effect on astrocytes. Furthermore, loss of C1qa significantly rescued I/R-induced loss of RGCs and protected against retinal layer thinning in comparison to WT mice. ERG assessment revealed early preservation of b-wave amplitude deficits from retinal I/R injury due to C1qa-deficiency that was lost by day 28. CONCLUSIONS: Our results for the first time demonstrate the spatiotemporal changes in the neuroinflammatory response following retinal I/R injury at both local and distal sites of injury. In addition, we have shown a role for C1q as a primary mediator of microglial activation and pathological damage. This suggests developmental mechanisms of C1q may be re-engaged during injury response, modulation of which may be beneficial for neuroprotection.


Assuntos
Glicoproteínas de Membrana/metabolismo , Microglia/metabolismo , Neuroglia/metabolismo , Receptores de Complemento/metabolismo , Traumatismo por Reperfusão/metabolismo , Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Axônios/metabolismo , Modelos Animais de Doenças , Feminino , Pressão Intraocular/fisiologia , Masculino , Camundongos Transgênicos , Células Ganglionares da Retina/patologia
9.
Invest Ophthalmol Vis Sci ; 53(11): 7043-51, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-22956608

RESUMO

PURPOSE: We previously discovered elevated levels of secreted frizzled-related protein 1 (sFRP1), the Wnt signaling pathway inhibitor, in the glaucomatous trabecular meshwork (GTM), and found that key canonical Wnt signaling pathway genes are expressed in the trabecular meshwork (TM). The purpose of our study was to determine whether a functional canonical Wnt signaling pathway exists in the human TM (HTM). METHODS: Western immunoblotting and/or immunofluorescent microscopy were used to study ß-catenin translocation as well as the actin cytoskeleton in transformed and primary HTM cells. A TCF/LEF luciferase assay was used to study functional canonical Wnt signaling, which was confirmed further by WNT3a-induced expression of a pathway target gene, AXIN2, via quantitative PCR. Intravitreal injection of an Ad5 adenovirus expressing Dickkopf-related protein-1 (DKK1) was used to study the in vivo effect of canonical Wnt signaling on IOP in mice. RESULTS: WNT3a induced ß-catenin translocation in the HTM, which was blocked by co-treatment with sFRP1. Similarly, WNT3a enhanced luciferase levels in TCF/LEF luciferase assays, which also were blocked by sFRP1. Furthermore, AXIN2 expression was elevated significantly by WNT3a. However, neither WNT3a nor sFRP1 affected actin cytoskeleton organization, which theoretically could be regulated by noncanonical Wnt signaling in HTM cells. Exogenous DKK1, a specific inhibitor for the canonical Wnt signaling pathway, or sFRP1 elevated mouse IOP to equivalent levels. CONCLUSIONS: There is a canonical Wnt signaling pathway in the TM, and this canonical Wnt pathway, but not the noncanonical Wnt signaling pathway, regulates IOP.


Assuntos
Malha Trabecular/metabolismo , Via de Sinalização Wnt/fisiologia , Actinas/metabolismo , Adenoviridae/genética , Proteína Axina/genética , Western Blotting , Células Cultivadas , Densitometria , Eletroforese em Gel de Poliacrilamida , Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Pressão Intraocular/fisiologia , Injeções Intravítreas , Microscopia de Fluorescência , Transporte Proteico/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Malha Trabecular/efeitos dos fármacos , Proteína Wnt3A/farmacologia , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...