Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Ther ; 26(10): 2379-2396, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30217731

RESUMO

Dysregulation of the complement system is implicated in neurodegeneration, including human and animal glaucoma. Optic nerve and retinal damage in glaucoma is preceded by local complement upregulation and activation, but whether targeting this early innate immune response could have therapeutic benefit remains undefined. Because complement signals through three pathways that intersect at complement C3 activation, here we targeted this step to restore complement balance in the glaucomatous retina and to determine its contribution to degeneration onset and/or progression. To achieve this, we combined adeno-associated virus retinal gene therapy with the targeted C3 inhibitor CR2-Crry. We show that intravitreal injection of AAV2.CR2-Crry produced sustained Crry overexpression in the retina and reduced deposition of the activation product complement C3d on retinal ganglion cells and the inner retina of DBA/2J mice. This resulted in neuroprotection of retinal ganglion cell axons and somata despite continued intraocular pressure elevation, suggesting a direct restriction of neurodegeneration onset and progression and significant delay to terminal disease stages. Our study uncovers a damaging effect of complement C3 or downstream complement activation in glaucoma, and it establishes AAV2.CR2-Crry as a viable therapeutic strategy to target pathogenic C3-mediated complement activation in the glaucomatous retina.


Assuntos
Complemento C3/genética , Glaucoma/terapia , Degeneração Neural/terapia , Proteínas Recombinantes de Fusão/genética , Animais , Complemento C3/antagonistas & inibidores , Dependovirus/genética , Modelos Animais de Doenças , Progressão da Doença , Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Glaucoma/genética , Glaucoma/patologia , Humanos , Pressão Intraocular/efeitos dos fármacos , Injeções Intravítreas , Camundongos , Degeneração Neural/genética , Degeneração Neural/patologia , Proteínas Recombinantes de Fusão/administração & dosagem , Retina/efeitos dos fármacos , Retina/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia
2.
J Neurochem ; 142(2): 323-337, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28498493

RESUMO

Glaucoma is a leading cause of blindness, characterized by retinal ganglion cell (RGC) loss and optic nerve (ON) damage. Cumulative evidence suggests glial cell involvement in the degeneration of the ON and RGCs. We analyzed the contribution of microglial reactivity to early axoglial alterations of the ON in an induced model of ocular hypertension. For this purpose, vehicle or chondroitin sulfate (CS) were weekly injected into the eye anterior chamber from Wistar rats for different intervals. The amount of Brn3a(+) RGC significantly decreased in CS-injected eyes for 10 and 15 (but not 6) weeks. A reduction in anterograde transport of ß-subunit cholera toxin was observed in the superior colliculus and the lateral geniculate nucleus contralateral to CS-injected eyes for 6 and 15 weeks. A disruption of cholera toxin ß-subunit transport was observed at the proximal myelinated ON. A significant decrease in phosphorylated neurofilament heavy chain immunoreactivity, an increase in ionized calcium-binding adaptor molecule 1(+), ED1(+) (microglial markers), and glial fibrillary acidic protein (astrocytes) (+) area, and decreased luxol fast blue staining were observed in the ON at 6 and 15 weeks of ocular hypertension. Microglial reactivity involvement was examined through a daily treatment with minocycline (30 mg/kg, i.p.) for 2 weeks, after 4 weeks of ocular hypertension. Minocycline prevented the increase in ionized calcium-binding adaptor molecule 1(+), ED-1(+), and glial fibrillary acidic protein(+) area, the decrease in phosphorylated neurofilament heavy-chain immunoreactivity and luxol fast blue staining, and the deficit in anterograde transport induced by 6 weeks of ocular hypertension. Thus, targeting microglial reactivity might prevent early axoglial alterations in the glaucomatous ON. Cover Image for this issue: doi: 10.1111/jnc.13807.


Assuntos
Glaucoma/tratamento farmacológico , Nervo Óptico/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Modelos Animais de Doenças , Corpos Geniculados/efeitos dos fármacos , Glaucoma/metabolismo , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Minociclina/administração & dosagem , Minociclina/farmacologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Nervo Óptico/metabolismo , Ratos Wistar , Retina/efeitos dos fármacos , Retina/metabolismo
3.
Exp Eye Res ; 150: 34-43, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26851485

RESUMO

Within the white matter, axonal loss by neurodegeneration is coupled to glial cell changes in gene expression, structure and function commonly termed gliosis. Recently, we described the highly variable expansion of gliosis alebosco@neuro.utah.edu in degenerative optic nerves from the DBA/2J mouse model of chronic, age-related glaucoma. Here, to estimate and compare the levels of axonal loss with the expansion of glial coverage and axonal degeneration in DBA/2J nerves, we combined semiautomatic axon counts with threshold-based segmentation of total glial/scar areas and degenerative axonal profiles in plastic cross-sections. In nerves ranging from mild to severe degeneration, we found that the progression of axonal dropout is coupled to an increase of gliotic area. We detected a strong correlation between axon loss and the aggregate coverage by glial cells and scar, whereas axon loss did not correlate with the small fraction of degenerating profiles. Nerves with low to medium levels of axon loss displayed moderate glial reactivity, consisting of hypertrophic astrocytes, activated microglia and normal distribution of oligodendrocytes, with minimal reorganization of the tissue architecture. In contrast, nerves with extensive axonal loss showed prevalent rearrangement of the nerve, with loss of axon fascicle territories and enlarged or almost continuous gliotic and scar domains, containing reactive astrocytes, oligodendrocytes and activated microglia. These findings support the value of optic nerve gliotic expansion as a quantitative estimate of optic neuropathy that correlates with axon loss, applicable to grade the severity of optic nerve damage in mouse chronic glaucoma.


Assuntos
Glaucoma/patologia , Gliose/complicações , Neuroglia/patologia , Doenças do Nervo Óptico/patologia , Nervo Óptico/patologia , Células Ganglionares da Retina/patologia , Animais , Astrócitos/patologia , Axônios/patologia , Doença Crônica , Modelos Animais de Doenças , Feminino , Glaucoma/complicações , Gliose/patologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Microscopia Confocal , Doenças do Nervo Óptico/etiologia , Fotomicrografia
4.
bioRxiv ; 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38979153

RESUMO

Microglia, the parenchymal macrophage of the central nervous system serve crucial remodeling functions throughout development. Microglia are transcriptionally heterogenous, suggesting that distinct microglial states confer discrete roles. Currently, little is known about how dynamic these states are, the cues that promote them, or how they impact microglial function. In the developing retina, we previously found a significant proportion of microglia express CD11c (Integrin αX, complement receptor 4, Itgax) which has also been reported in other developmental and disease contexts. Here, we sought to understand the regulation and function of CD11c+ microglia. We found that CD11c+ microglia track with prominent waves of neuronal apoptosis in postnatal retina. Using genetic fate mapping, we provide evidence that microglia transition out of the CD11c state to return to homeostasis. We show that CD11c+ microglia have elevated lysosomal content and contribute to the clearance of apoptotic neurons, and found that acquisition of CD11c expression is, in part, dependent upon the TAM receptor Axl. Using selective ablation, we found CD11c+ microglia are not uniquely critical for phagocytic clearance of apoptotic cells. Together, our data suggest CD11c+ microglia are a transient state induced by developmental apoptosis rather than a specialized subset mediating phagocytic elimination.

5.
Elife ; 112022 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-35481836

RESUMO

Microglia serve critical remodeling roles that shape the developing nervous system, responding to the changing neural environment with phagocytosis or soluble factor secretion. Recent single-cell sequencing (scRNAseq) studies have revealed the context-dependent diversity in microglial properties and gene expression, but the cues promoting this diversity are not well defined. Here, we ask how interactions with apoptotic neurons shape microglial state, including lysosomal and lipid metabolism gene expression and dependence on Colony-stimulating factor 1 receptor (CSF1R) for survival. Using early postnatal mouse retina, a CNS region undergoing significant developmental remodeling, we performed scRNAseq on microglia from mice that are wild-type, lack neuronal apoptosis (Bax KO), or are treated with CSF1R inhibitor (PLX3397). We find that interactions with apoptotic neurons drive multiple microglial remodeling states, subsets of which are resistant to CSF1R inhibition. We find that TAM receptor Mer and complement receptor 3 are required for clearance of apoptotic neurons, but that Mer does not drive expression of remodeling genes. We show TAM receptor Axl is negligible for phagocytosis or remodeling gene expression but is consequential for microglial survival in the absence of CSF1R signaling. Thus, interactions with apoptotic neurons shift microglia toward distinct remodeling states and through Axl, alter microglial dependence on survival pathway, CSF1R.


Assuntos
Microglia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos , Animais , Apoptose , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Fagocitose , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Transdução de Sinais
6.
Front Cell Dev Biol ; 8: 581136, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33043015

RESUMO

Retinal development follows a conserved neurogenic program in vertebrates to orchestrate the generation of specific cell types from multipotent progenitors in sequential but overlapping waves. In this program, retinal ganglion cells (RGCs) are the first cell type generated. RGCs are the final output neurons of the retina and are essential for vision and circadian rhythm. Key molecular steps have been defined in multiple vertebrate species to regulate competence, specification, and terminal differentiation of this cell type. This involves neuronal-specific transcription factor networks, regulators of chromatin dynamics and miRNAs. In mammals, RGCs and their optic nerve axons undergo neurodegeneration and loss in glaucoma and other optic neuropathies, resulting in irreversible vision loss. The incapacity of RGCs and axons to regenerate reinforces the need for the design of efficient RGC replacement strategies. Here we describe the essential molecular pathways for the differentiation of RGCs in vertebrates, as well as experimental manipulations that extend the competence window for generation of this early cell type from late progenitors. We discuss recent advances in regeneration of retinal neurons in vivo in both mouse and zebrafish and discuss possible strategies and barriers to achieving RGC regeneration as a therapeutic approach for vision restoration in blinding diseases such as glaucoma.

7.
Cell Rep ; 27(7): 2002-2013.e5, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31091440

RESUMO

Microglia have important remodeling functions in neurodevelopment, aging, and disease, with evidence for molecular diversity. However, the signaling pathways and environmental cues that drive diverse states of microglia are incompletely understood. We profiled microglia of a discrete developing CNS region, the murine retina. We found distinct transcriptional signatures for retinal microglia across development and peak postnatal density of a population that resembles aging and disease-associated microglia (DAM) and CD11c+ microglia of developing white matter. While TREM2 signaling modulates the expression of select genes, the DAM-related signature is significantly reduced in retinas lacking Bax, a proapoptotic factor required for neuronal death. Furthermore, we found postnatal retinal microglia highly expressing CD11c are resistant to loss or inhibition of colony stimulating factor 1 receptor (CSF1R), while most microglia can be eliminated in Bax knockout retina. Thus, developmental apoptosis promotes a microglia gene signature linked to CSF1R independence that shares features with microglia in developing white matter and in disease.


Assuntos
Apoptose , Diferenciação Celular , Microglia/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Retina/metabolismo , Doenças Retinianas/metabolismo , Transdução de Sinais , Transcriptoma , Animais , Camundongos , Camundongos Knockout , Microglia/patologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Retina/patologia , Doenças Retinianas/genética , Doenças Retinianas/patologia
8.
Invest Ophthalmol Vis Sci ; 49(4): 1437-46, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18385061

RESUMO

PURPOSE: In the context of the retinal ganglion cell (RGC) axon degeneration in the optic nerve that occurs in glaucoma, microglia become activated, then phagocytic, and redistribute in the optic nerve head. The authors investigated the potential contribution of retinal microglia activation to glaucoma progression in the DBA/2J chronic mouse glaucoma model. METHODS: The authors treated 6-week-old DBA/2J mice for 25 weeks with minocycline, a tetracycline derivative known to reduce microglia activation and to improve neuronal survival in other models of neurodegenerative disease. They quantified RGC numbers and characterized microglia activation, gliosis, and both axonal integrity and retrograde tracer transport by RGCs in mice systemically treated with minocycline or vehicle only. RESULTS: Minocycline reduced microglial activation and improved RGC axonal transport and integrity, yet it had no effect on the characteristic age-related ocular changes that lead to chronically elevated pressure and did not alter Müller or astrocyte gliosis. Specifically, minocycline increased the fraction of microglia with resting ramified morphology and reduced levels of Iba1 mRNA and protein, a microglia-specific calcium ligand linked to activation. The reduction in microglial activation was coupled to significant improvement in RGC axonal transport, as measured by neuronal retrograde tracing from the superior colliculus. Finally, minocycline treatment significantly decoupled RGC axon loss from increased intraocular pressure. CONCLUSIONS: These observations suggest that in glaucoma, retina and optic nerve head microglia activation may be a factor in the early decline in function of the optic nerve and its subsequent degeneration.


Assuntos
Modelos Animais de Doenças , Glaucoma/prevenção & controle , Microglia/efeitos dos fármacos , Minociclina/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Doenças do Nervo Óptico/metabolismo , Retina/citologia , Animais , Transporte Axonal/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/genética , Sobrevivência Celular , Glaucoma/metabolismo , Glaucoma/patologia , Gliose/metabolismo , Injeções Intraperitoneais , Pressão Intraocular , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos DBA , Proteínas dos Microfilamentos , Microglia/metabolismo , Doenças do Nervo Óptico/patologia , RNA Mensageiro/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Front Neurosci ; 10: 526, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27932942

RESUMO

Neurodegeneration in glaucoma results in decline and loss of retinal ganglion cells (RGCs), and is associated with activation of myeloid cells such as microglia and macrophages. The chemokine fractalkine (FKN or Cx3cl1) mediates communication from neurons to myeloid cells. Signaling through its receptor Cx3cr1 has been implicated in multiple neurodegenerative diseases, but the effects on neuronal pathology are variable. Since it is unknown how FKN-mediated crosstalk influences RGC degeneration in glaucoma, we assessed this in a chronic mouse model, DBA/2J. We analyzed a DBA/2J substrain deficient in Cx3cr1, and compared compartmentalized RGC degeneration and myeloid cell responses to those in standard DBA/2J mice. We found that loss of FKN signaling exacerbates axon transport dysfunction, an early event in neurodegeneration, with a significant increase in RGCs with somal accumulation of the axonal protein phosphorylated neurofilament, and reduced retinal expression of genes involved in axon transport, Kif1b, and Atp8a2. There was no change in the loss of Brn3-positive RGCs, and no difference in the extent of damage to the proximal optic nerve, suggesting that the loss of fractalkine signaling primarily affects axon transport. Since Cx3cr1 is specifically expressed in myeloid cells, we assessed changes in retinal microglial number and activation, changes in gene expression, and the extent of macrophage infiltration. We found that loss of fractalkine signaling led to innate immune changes within the retina, including increased infiltration of peripheral macrophages and upregulated nitric oxide synthase-2 (Nos-2) expression in myeloid cells, which contributes to the production of NO and can promote axon transport deficits. In contrast, resident retinal microglia appeared unchanged either in number, morphology, or expression of the myeloid activation marker ionized calcium binding adaptor molecule 1 (Iba1). There was also no significant increase in the proinflammatory gene interleukin 1 beta (Il1ß). We conclude that loss of fractalkine signaling causes a selective worsening of axon transport dysfunction in RGCs, which is linked to enhanced Nos-2 expression in myeloid cells. Our findings suggest that distinct mechanisms may contribute to different aspects of RGC decline in glaucoma, with axonal transport selectively altered after loss of Cx3cr1 in microglia and/or macrophages.

10.
J Neurosci ; 23(16): 6413-22, 2003 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-12878681

RESUMO

During development of the retina, programmed cell death helps to establish the final size and distribution of various cell classes in distinct layers of the tissue. Here we show that dying cells in the developing ganglion and inner nuclear layers are clustered spatially and that gap junction inhibitors decrease the clustering of dying cells. To confirm the role of gap junctions in cell death, we induced targeted cell death via intracellular cytochrome c (Cc) and examined the induced cells and their neighbors for apoptotic morphology or caspase-3 cleavage. These studies indicate that bystander killing extends to coupled cells. Quantitative studies of bystander killing were performed by scrape-loading retinas with Cc in the presence of rhodamine dextran (RD; to identify Cc-loaded cells) and by counting pyknotic cells in cryosections. Although only 1.5% of control scrape-loaded cells (RD alone) showed apoptotic morphology, 97% of Cc scrape-loaded cells were pyknotic. Moreover, bystander killing extended to neighboring cells, not labeled with RD, and was reduced significantly by the gap junction inhibitors octanol and carbenoxolone. We hypothesize that dying cells in the retina generate a gap junction-permeant apoptotic signal that mediates bystander killing. This novel finding of naturally occurring bystander cell death may have important implications in the histogenesis and pathology of the nervous system.


Assuntos
Efeito Espectador/fisiologia , Junções Comunicantes/metabolismo , Retina/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Carbenoxolona/farmacologia , Contagem de Células , Morte Celular/fisiologia , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Endogâmicos , Ratos Long-Evans , Retina/citologia , Retina/efeitos dos fármacos
11.
Invest Ophthalmol Vis Sci ; 46(10): 3869-75, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16186376

RESUMO

PURPOSE: In adult retina, aquaporin-4 (AQP4) and inwardly rectifying K(+) (Kir4.1) channels localize to astrocyte and Müller cell membranes facing vascular and vitreous compartments, optimizing clearance of extracellular K(+) and water from the synaptic layers. However, it is unknown whether these channels are expressed at early developmental stages, before gliogenesis or angiogenesis take place in the neural retina. This study was conducted to determine the presence of AQP4 and Kir4.1 proteins in the developing mouse retina. METHODS: Simultaneous AQP4 and Kir4.1 immunodetection was performed in postnatal mice 1, 9, 15, and 30 days of age. Confocal microscopy was used to identify the cellular distribution of AQP4 and Kir4.1 proteins, as well as their coexpression with the cell-selective immunomarkers Prox-1, calbindin, and neurofilament. RESULTS: AQP4 and Kir4.1 proteins were coexpressed in calbindin- and Prox1-expressing retinal neurons at birth. These neurons were identified as horizontal cells based on their position and morphology. By P15, when vision starts, AQP4 and Kir4.1 localization coordinately switched from horizontal cells to Müller glial cells. CONCLUSIONS: The findings showed that AQP4 and Kir4.1 protein expression is confined to differentiating horizontal cells before its expression in Müller cells. The finding of AQP4 in neurons is novel, since AQP4 expression within the central nervous system is restricted to glia. Also, the results demonstrated that AQP4 is a horizontal cell-specific immunomarker in neonatal retina. The transitory coexpression of AQP4 and Kir4.1 proteins by differentiating horizontal interneurons suggests that these cells mediate K(+) and water transcellular uptake until the initiation of phototransduction, when glial cells assume these functions.


Assuntos
Aquaporina 4/metabolismo , Proteínas do Olho/metabolismo , Neuroglia/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Retina/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Calbindinas , Técnica Indireta de Fluorescência para Anticorpo , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Proteínas de Neurofilamentos/metabolismo , Retina/citologia , Retina/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo , Proteínas Supressoras de Tumor , Equilíbrio Hidroeletrolítico
12.
J Vis Exp ; (99): e52731, 2015 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-25992962

RESUMO

Microglia, which are CNS-resident neuroimmune cells, transform their morphology and size in response to CNS damage, switching to an activated state with distinct functions and gene expression profiles. The roles of microglial activation in health, injury and disease remain incompletely understood due to their dynamic and complex regulation in response to changes in their microenvironment. Thus, it is critical to non-invasively monitor and analyze changes in microglial activation over time in the intact organism. In vivo studies of microglial activation have been delayed by technical limitations to tracking microglial behavior without altering the CNS environment. This has been particularly challenging during chronic neurodegeneration, where long-term changes must be tracked. The retina, a CNS organ amenable to non-invasive live imaging, offers a powerful system to visualize and characterize the dynamics of microglia activation during chronic disorders. This protocol outlines methods for long-term, in vivo imaging of retinal microglia, using confocal ophthalmoscopy (cSLO) and CX3CR1(GFP/+) reporter mice, to visualize microglia with cellular resolution. Also, we describe methods to quantify monthly changes in cell activation and density in large cell subsets (200-300 cells per retina). We confirm the use of somal area as a useful metric for live tracking of microglial activation in the retina by applying automated threshold-based morphometric analysis of in vivo images. We use these live image acquisition and analyses strategies to monitor the dynamic changes in microglial activation and microgliosis during early stages of retinal neurodegeneration in a mouse model of chronic glaucoma. This approach should be useful to investigate the contributions of microglia to neuronal and axonal decline in chronic CNS disorders that affect the retina and optic nerve.


Assuntos
Glaucoma/patologia , Microglia/patologia , Retina/patologia , Fatores Etários , Animais , Axônios/patologia , Receptor 1 de Quimiocina CX3C , Diagnóstico por Imagem/métodos , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/química , Camundongos , Camundongos Endogâmicos DBA , Oftalmoscopia/métodos , Nervo Óptico/patologia , Receptores de Quimiocinas/análise , Receptores de Quimiocinas/química
13.
Dis Model Mech ; 8(5): 443-55, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25755083

RESUMO

Microglia serve key homeostatic roles, and respond to neuronal perturbation and decline with a high spatiotemporal resolution. The course of all chronic CNS pathologies is thus paralleled by local microgliosis and microglia activation, which begin at early stages of the disease. However, the possibility of using live monitoring of microglia during early disease progression to predict the severity of neurodegeneration has not been explored. Because the retina allows live tracking of fluorescent microglia in their intact niche, here we investigated their early changes in relation to later optic nerve neurodegeneration. To achieve this, we used the DBA/2J mouse model of inherited glaucoma, which develops progressive retinal ganglion cell degeneration of variable severity during aging, and represents a useful model to study pathogenic mechanisms of retinal ganglion cell decline that are similar to those in human glaucoma. We imaged CX3CR1(+/GFP) microglial cells in vivo at ages ranging from 1 to 5 months by confocal scanning laser ophthalmoscopy (cSLO) and quantified cell density and morphological activation. We detected early microgliosis at the optic nerve head (ONH), where axonopathy first manifests, and could track attenuation of this microgliosis induced by minocycline. We also observed heterogeneous and dynamic patterns of early microglia activation in the retina. When the same animals were aged and analyzed for the severity of optic nerve pathology at 10 months of age, we found a strong correlation with the levels of ONH microgliosis at 3 to 4 months. Our findings indicate that live imaging and monitoring the time course and levels of early retinal microgliosis and microglia activation in glaucoma could serve as indicators of future neurodegeneration severity.


Assuntos
Glaucoma/patologia , Microglia/patologia , Degeneração Neural/patologia , Animais , Receptor 1 de Quimiocina CX3C , Modelos Animais de Doenças , Feminino , Gliose/patologia , Lasers , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Oftalmoscopia , Disco Óptico/patologia , Receptores de Quimiocinas/metabolismo , Retina/patologia
14.
Brain Res Dev Brain Res ; 139(2): 325-30, 2002 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12480149

RESUMO

Developing amacrine cells in the vertebrate retina undergo naturally-occurring cell death which is accentuated by the early removal of retinal ganglion cells. We show that providing BDNF or decreasing endogenous BDNF via competitive binding with soluble TrkB receptors in a whole-retina culture assay modulates the frequency of dying cells in the amacrine cell layer. Ganglion cells synthesize BDNF, and amacrine cells express TrkB receptors, suggesting a likely signaling mechanism.


Assuntos
Células Amácrinas/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Morte Celular/fisiologia , Diferenciação Celular/fisiologia , Receptor trkB/metabolismo , Retina/crescimento & desenvolvimento , Células Ganglionares da Retina/metabolismo , Envelhecimento/metabolismo , Células Amácrinas/citologia , Células Amácrinas/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Contagem de Células , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Endogâmicos , Receptor trkB/efeitos dos fármacos , Retina/citologia , Retina/metabolismo , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
15.
PLoS One ; 7(8): e43602, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22952717

RESUMO

Glaucoma is a neurodegenerative disease that results in the progressive decline and ultimate death of retinal ganglion cells (RGCs). While multiple risk factors are associated with glaucoma, the mechanisms leading to onset and progression of the disease remain unknown. Molecular analysis in various glaucoma models has revealed involvement of non-neuronal cell populations, including astrocytes, Mueller glia and microglia, at early stages of glaucoma. High-dose irradiation was reported to have a significant long-term protective effect in the DBA/2J (D2) mouse model of glaucoma, although the cellular and molecular basis for this effect remains unclear. In particular, the acute effects of irradiation on specific cell populations, including non-neuronal cells, in the D2 retina and nerve have not been assessed. Here we report that irradiation induces transient reduction in proliferating microglia within the optic nerve head and glial lamina within the first week post-irradiation. This was accompanied by reduced microglial activation, with no effect on astrocyte gliosis in those regions. At later stages we confirm that early high-dose irradiation of the mouse head results in improvement of axonal structural integrity and anterograde transport function, without reduction of intraocular pressure. Thus reduced microglial activation induced by irradiation at early stages is associated with reduced optic nerve and retinal neurodegeneration in the D2 mouse model of glaucoma.


Assuntos
Glaucoma/patologia , Microglia/patologia , Microglia/efeitos da radiação , Animais , Axônios/patologia , Axônios/efeitos da radiação , Proliferação de Células/efeitos da radiação , Doença Crônica , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Feminino , Gliose/patologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Nervo Óptico/patologia , Nervo Óptico/efeitos da radiação , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/efeitos da radiação , Fatores de Tempo
16.
J Comp Neurol ; 519(4): 599-620, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21246546

RESUMO

Changes in microglial cell activation and distribution are associated with neuronal decline in the central nervous system (CNS), particularly under pathological conditions. Activated microglia converge on the initial site of axonal degeneration in human glaucoma, yet their part in its pathophysiology remains unresolved. To begin with, it is unknown whether microglia activation precedes or is a late consequence of retinal ganglion cell (RGC) neurodegeneration. Here we address this critical element in DBA/2J (D2) mice, an established model of chronic inherited glaucoma, using as a control the congenic substrain DBA/2J Gpnmb(+/SjJ) (D2G), which is not affected by glaucoma. We analyzed the spatial distribution and timecourse of microglial changes in the retina, as well as within the proximal optic nerve prior to and throughout ages when neurodegeneration has been reported. Exclusively in D2 mice, we detected early microglia clustering in the inner central retina and unmyelinated optic nerve regions, with microglia activation peaking by 3 months of age. Between 5 and 8 months of age, activated microglia persisted and concentrated in the optic disc, but also localized to the retinal periphery. Collectively, our findings suggest microglia activation is an early alteration in the retina and optic nerve in D2 glaucoma, potentially contributing to disease onset or progression. Ultimately, detection of microglial activation may have value in early disease diagnosis, while modulation of microglial responses may alter disease progression.


Assuntos
Glaucoma/fisiopatologia , Microglia/fisiologia , Animais , Receptor 1 de Quimiocina CX3C , Modelos Animais de Doenças , Progressão da Doença , Humanos , Pressão Intraocular , Camundongos , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Microglia/citologia , Nervo Óptico/citologia , Nervo Óptico/crescimento & desenvolvimento , Nervo Óptico/fisiopatologia , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Retina/citologia , Retina/crescimento & desenvolvimento , Retina/fisiopatologia
17.
J Biol Chem ; 282(13): 9789-9796, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17284442

RESUMO

Scaffolding of membrane proteins is a common strategy for forming complexes of proteins, including some connexins, within membrane microdomains. Here we describe studies indicating that Cx32 interacts with a PDZ-containing scaffolding protein, Dlgh1 (Discs Large homolog 1). Initial screens of liver lysates using antibody arrays indicated an interaction between Cx32 and Dlgh1 that was confirmed using coimmunoprecipitation studies. Yeast two-hybrid complementation determined that the Cx32 bound via interaction with the SH3/Hook domain of Dlgh1. Confocal microscopy of liver sections revealed that Cx32 and Dlgh1 could colocalize in hepatocyte membranes in wild type mice. Examination of levels and localization of Dlgh1 in livers from Cx32 null mice indicate that, in the absence of Cx32, Dlgh1 was decreased, and the remainder was translocated from the hepatocyte membrane to the nucleus with some remaining in cytoplasmic compartments. This translocation was confirmed by Western blots comparing Dlgh1 levels in nuclear extracts from wild type and Cx32 null murine livers. Using SKHep cells stably transfected with Cx32 under the control of a tet-off promoter, we found that acute removal of Cx32 led to a decrease of membrane-localized Dlgh1 and an increase in the nuclear localization of this tumor suppressor protein. Together, these results suggest that loss of Cx32 alters the levels, localization, and interactions of the tumor suppressor protein Dlgh1, events known in other systems to alter cell cycle and increase tumorigenicity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Conexinas/fisiologia , Junções Comunicantes/fisiologia , Proteínas de Membrana/fisiologia , Domínios de Homologia de src/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Conexinas/deficiência , Conexinas/metabolismo , Proteína 1 Homóloga a Discs-Large , Junções Comunicantes/genética , Guanilato Quinases , Proteínas de Membrana/metabolismo , Camundongos , Transporte Proteico/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Domínios de Homologia de src/genética , Proteína beta-1 de Junções Comunicantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA