Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Prog Retin Eye Res ; 95: 101147, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36402656

RESUMO

Retinoschisin (RS1) is a secreted protein that is essential for maintaining integrity of the retina. Numerous mutations in RS1 cause X-linked retinoschisis (XLRS), a progressive degeneration of the retina that leads to vision loss in young males. A key manifestation of XLRS is the formation of cavities (cysts) in the retina and separation of the layers (schisis), disrupting synaptic transmission. There are currently no approved treatments for patients with XLRS. Strategies using adeno-associated viral (AAV) vectors to deliver functional copies of RS1 as a form of gene augmentation therapy, are under clinical evaluation. To improve therapeutic strategies for treating XLRS, it is critical to better understand the secretion of RS1 and its molecular function. Immunofluorescence and immunoelectron microscopy show that RS1 is located on the surfaces of the photoreceptor inner segments and bipolar cells. Sequence homology indicates a discoidin domain fold, similar to many other proteins with demonstrated adhesion functions. Recent structural studies revealed the tertiary structure of RS1 as two back-to-back octameric rings, each cross-linked by disulfides. The observation of higher order structures in vitro suggests the formation of an adhesive matrix spanning the distance between cells (∼100 nm). Several studies indicated that RS1 readily binds to other proteins such as the sodium-potassium ATPase (NaK-ATPase) and extracellular matrix proteins. Alternatively, RS1 may influence fluid regulation via interaction with membrane proteins such as the NaK-ATPase, largely inferred from the use of carbonic anhydrase inhibitors to shrink the typical intra-retinal cysts in XLRS. We discuss these models in light of RS1 structure and address the difficulty in understanding the function of RS1.


Assuntos
Retina , Retinosquise , Masculino , Humanos , Estrutura Molecular , Retina/metabolismo , Retinosquise/genética , Retinosquise/metabolismo , Mutação , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Proteínas do Olho/genética
2.
Invest Ophthalmol Vis Sci ; 63(11): 8, 2022 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-36227606

RESUMO

Purpose: Loss of retinoschisin (RS1) function underlies X-linked retinoschisis (XLRS) pathology. In the retina, both photoreceptor inner segments and bipolar cells express RS1. However, the loss of RS1 function causes schisis primarily in the inner retina. To understand these cell type-specific phenotypes, we decoupled RS1 effects in bipolar cells from that in photoreceptors. Methods: Bipolar cell transgene RS1 expression was achieved using two inner retina-specific promoters: (1) a minimal promoter engineered from glutamate receptor, metabotropic glutamate receptor 6 gene (mini-mGluR6/ Grm6) and (2) MiniPromoter (Ple155). Adeno-associated virus vectors encoding RS1 gene under either the mini-mGluR6 or Ple-155 promoter were delivered to the XLRS mouse retina through intravitreal or subretinal injection on postnatal day 14. Retinal structure and function were assessed 5 weeks later: immunohistochemistry for morphological characterization, optical coherence tomography and electroretinography (ERG) for structural and functional evaluation. Results: Immunohistochemical analysis of RS1expression showed that expression with the MiniPromoter (Ple155) was heavily enriched in bipolar cells. Despite variations in vector penetrance and gene transfer efficiency across the injected retinas, those retinal areas with robust bipolar cell RS1 expression showed tightly packed bipolar cells with fewer cavities and marked improvement in inner retinal structure and synaptic function as judged by optical coherence tomography and electroretinography, respectively. Conclusions: These results demonstrate that RS1 gene expression primarily in bipolar cells of the XLRS mouse retina, independent of photoreceptor expression, can ameliorate retinoschisis structural pathology and provide further evidence of RS1 role in cell adhesion.


Assuntos
Cistos , Retinosquise , Animais , Camundongos , Cistos/metabolismo , Cistos/patologia , Eletrorretinografia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Retina/metabolismo , Retina/patologia , Células Bipolares da Retina/metabolismo , Retinosquise/genética , Retinosquise/metabolismo
3.
Hum Gene Ther ; 32(13-14): 667-681, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33019822

RESUMO

To understand RS1 gene interaction networks in the X-linked retinoschisis (XLRS) mouse retina (Rs1-/y), we analyzed the transcriptome by RNA sequencing before and after in vivo expression of exogenous retinoschisin (RS1) gene delivered by AAV8. RS1 is a secreted cell adhesion protein that is critical for maintaining structural lamination and synaptic integrity of the neural retina. RS1 loss-of-function mutations cause XLRS disease in young boys and men, with splitting ("schisis") of retinal layers and synaptic dysfunction that cause progressive vision loss with age. Analysis of differential gene expression profiles and pathway enrichment analysis of Rs1-KO (Rs1-/y) retina identified cell surface receptor signaling and positive regulation of cell adhesion as potential RS1 gene interaction networks. Most importantly, it also showed massive dysregulation of immune response genes at early age, with characteristics of a microglia-driven proinflammatory state. Delivery of AAV8-RS1 primed the Rs1-KO retina toward structural and functional recovery. The disease transcriptome transitioned toward a recovery phase with upregulation of genes implicated in wound healing, anatomical structure (camera type eye) development, metabolic pathways, and collagen IV networks that provide mechanical stability to basement membrane. AAV8-RS1 expression also attenuated the microglia gene signatures to low levels toward immune quiescence. This study is among the first to identify RS1 gene interaction networks that underlie retinal structural and functional recovery after RS1 gene therapy. Significantly, it also shows that providing wild-type RS1 gene function caused the retina immune status to transition from a degenerative inflammatory phenotype toward immune quiescence, even though the transgene is not directly linked to microglia function. This study indicates that inhibition of microglial proinflammatory responses is an integral part of therapeutic rescue in XLRS gene therapy, and gene therapy might realize its full potential if delivered before microglia activation and photoreceptor cell death. Clinical Trials. gov Identifier NTC 02317887.


Assuntos
Retinosquise , Animais , Eletrorretinografia , Proteínas do Olho/genética , Redes Reguladoras de Genes , Terapia Genética , Vetores Genéticos/genética , Camundongos , Microglia , Retina , Retinosquise/genética , Retinosquise/terapia
4.
PLoS Genet ; 16(12): e1009259, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33362196

RESUMO

Rab-GTPases and associated effectors mediate cargo transport through the endomembrane system of eukaryotic cells, regulating key processes such as membrane turnover, signal transduction, protein recycling and degradation. Using developmental transcriptome data, we identified Rabgef1 (encoding the protein RabGEF1 or Rabex-5) as the only gene associated with Rab GTPases that exhibited strong concordance with retinal photoreceptor differentiation. Loss of Rabgef1 in mice (Rabgef1-/-) resulted in defects specifically of photoreceptor morphology and almost complete loss of both rod and cone function as early as eye opening; however, aberrant outer segment formation could only partly account for visual function deficits. RabGEF1 protein in retinal photoreceptors interacts with Rabaptin-5, and RabGEF1 absence leads to reduction of early endosomes consistent with studies in other mammalian cells and tissues. Electron microscopy analyses reveal abnormal accumulation of macromolecular aggregates in autophagosome-like vacuoles and enhanced immunostaining for LC3A/B and p62 in Rabgef1-/- photoreceptors, consistent with compromised autophagy. Transcriptome analysis of the developing Rabgef1-/- retina reveals altered expression of 2469 genes related to multiple pathways including phototransduction, mitochondria, oxidative stress and endocytosis, suggesting an early trajectory of photoreceptor cell death. Our results implicate an essential role of the RabGEF1-modulated endocytic and autophagic pathways in photoreceptor differentiation and homeostasis. We propose that RabGEF1 and associated components are potential candidates for syndromic traits that include a retinopathy phenotype.


Assuntos
Autofagia , Endocitose , Fatores de Troca do Nucleotídeo Guanina/genética , Neurogênese , Células Fotorreceptoras/metabolismo , Degeneração Retiniana/metabolismo , Animais , Feminino , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células Fotorreceptoras/citologia , Degeneração Retiniana/genética , Transcriptoma
5.
Sci Adv ; 4(3): eaap8492, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29750189

RESUMO

Microglia have been discovered to undergo repopulation following ablation. However, the functionality of repopulated microglia and the mechanisms regulating microglia repopulation are unknown. We examined microglial homeostasis in the adult mouse retina, a specialized neural compartment containing regular arrays of microglia in discrete synaptic laminae that can be directly visualized. Using in vivo imaging and cell-fate mapping techniques, we discovered that repopulation originated from residual microglia proliferating in the central inner retina that subsequently spread by centrifugal migration to fully recapitulate pre-existing microglial distributions and morphologies. Repopulating cells fully restored microglial functions including constitutive "surveying" process movements, behavioral and physiological responses to retinal injury, and maintenance of synaptic structure and function. Microglial repopulation was regulated by CX3CL1-CX3CR1 signaling, slowing in CX3CR1 deficiency and accelerating with exogenous CX3CL1 administration. Microglial homeostasis following perturbation can fully recover microglial organization and function under the regulation of chemokine signaling between neurons and microglia.


Assuntos
Receptor 1 de Quimiocina CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Microglia/metabolismo , Retina/citologia , Envelhecimento/fisiologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Movimento Celular , Proliferação de Células , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Microglia/ultraestrutura , Transdução de Sinais
6.
Sci Rep ; 7(1): 8433, 2017 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-28814744

RESUMO

Age-related macular degeneration (AMD), a leading contributor of vision loss, currently lacks comprehensive treatment. While AMD histopathology involves retinal pigment epithelium (RPE) injury associated with immune cell infiltration, the nature of immune cell responses to RPE injury remains undefined. We induced RPE injury pharmacologically and genetically in transgenic mouse models in which microglia and systemic monocytes were separately tagged, enabling a spatial and temporal dissection of the relative contributions of microglia vs. monocytes to post-injury changes. We found that myeloid cell responses to RPE injury occur in stages: (1) an early mobilization of endogenous microglia from the inner retina to the RPE layer, followed by (2) subsequent monocyte infiltration from the retinal vasculature into the inner retina that replenishes the local myeloid cell population in a CCR2-regulated manner. These altered distributions of myeloid cells post-injury were long-lived, with recruited monocytes acquiring the distribution, markers, and morphologies of neighboring endogenous microglia in a durable manner. These findings indicate the role played by infiltrating monocytes in maintaining myeloid cell homeostasis in the retina following AMD-relevant RPE injury and provide a foundation for understanding and therapeutically modulating immune aspects in retinal disease.


Assuntos
Proliferação de Células , Células Epiteliais/metabolismo , Monócitos/metabolismo , Células Mieloides/metabolismo , Retina/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Animais , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Homeostase , Iodatos/toxicidade , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Microscopia Confocal , Receptores CCR2/genética , Receptores CCR2/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/patologia
7.
Hum Mol Genet ; 26(12): 2218-2230, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28369466

RESUMO

In retinal photoreceptors, vectorial transport of cargo is critical for transduction of visual signals, and defects in intracellular trafficking can lead to photoreceptor degeneration and vision impairment. Molecular signatures associated with routing of transport vesicles in photoreceptors are poorly understood. We previously reported the identification of a novel rod photoreceptor specific isoform of Receptor Expression Enhancing Protein (REEP) 6, which belongs to a family of proteins involved in intracellular transport of receptors to the plasma membrane. Here we show that loss of REEP6 in mice (Reep6-/-) results in progressive retinal degeneration. Rod photoreceptor dysfunction is observed in Reep6-/- mice as early as one month of age and associated with aberrant accumulation of vacuole-like structures at the apical inner segment and reduction in selected rod phototransduction proteins. We demonstrate that REEP6 is detected in a subset of Clathrin-coated vesicles and interacts with the t-SNARE, Syntaxin3. In concordance with the rod degeneration phenotype in Reep6-/- mice, whole exome sequencing identified homozygous REEP6-E75K mutation in two retinitis pigmentosa families of different ethnicities. Our studies suggest a critical function of REEP6 in trafficking of cargo via a subset of Clathrin-coated vesicles to selected membrane sites in retinal rod photoreceptors.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Animais , Vesículas Revestidas por Clatrina/metabolismo , Proteínas do Olho/genética , Transdução de Sinal Luminoso , Proteínas de Membrana , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Mutação , Células Fotorreceptoras de Vertebrados/metabolismo , Isoformas de Proteínas/metabolismo , Transporte Proteico , Proteínas Qa-SNARE/metabolismo , Degeneração Retiniana/metabolismo , Retinose Pigmentar/genética , Proteínas SNARE/metabolismo
8.
Brain Struct Funct ; 222(6): 2759-2771, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28213784

RESUMO

Microglia, the principal resident immune cell in the retina, play constitutive roles in immune surveillance and synapse maintenance, and are also associated with retinal disease, including those occurring in the macula. Perspectives on retinal microglia function have derived largely from rodent models and how these relate to the macula-bearing primate retina is unclear. In this study, we examined microglial distribution and cellular morphology in the adult rhesus macaque retina, and performed comparative characterizations in three retinal locations along the center-to-periphery axis (parafoveal, macular, and the peripheral retina). We found that microglia density peaked in the parafoveal retina and decreased in the peripheral retina. Individual microglial morphology reflected macular specialization, with macular microglia demonstrating the largest and most complex dendritic arbors relative to other retinal locations. Comparing retinal microglia between young and middle-aged animals, microglial density increased in the macular, but not in the peripheral retina with age, while microglial morphology across all locations remained relatively unchanged. Our findings indicate that microglial distribution and morphology demonstrate regional specialization in the retina, correlating with gradients of other retinal cell types. As microglia are innate immune cells implicated in age-related macular diseases, age-related microglial changes may be related to the increased vulnerability of the aged macula to immune-related neurodegeneration.


Assuntos
Envelhecimento , Forma Celular , Macaca mulatta , Microglia/citologia , Retina/citologia , Fatores Etários , Animais , Biomarcadores/análise , Proteínas do Olho/análise , Feminino , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional , Imuno-Histoquímica , Microglia/química , Microscopia Confocal , Retina/química
9.
Exp Eye Res ; 159: 132-146, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27865785

RESUMO

Characterizing the role of epigenetic regulation in the mammalian retina is critical for understanding fundamental mechanisms of retinal development and disease. DNA methylation, an epigenetic modifier of genomic DNA, plays an important role in modulating networks of tissue and cell-specific gene expression. However, the impact of DNA methylation on retinal development and homeostasis of retinal neurons remains unclear. Here, we have created a tissue-specific DNA methyltransferase (Dnmt) triple mutant mouse in an effort to characterize the impact of DNA methylation on retinal development and homeostasis. An Rx-Cre transgene was used to drive targeted mutation of all three murine Dnmt genes in the mouse retina encoding major DNA methylation enzymes DNMT1, DNMT3A and DNMT3B. The triple mutant mice represent a hypomorph model since Dnmt1 catalytic activity was still present and excision of Dnmt3a and Dnmt3b had only about 90% efficiency. Mutation of all three Dnmts resulted in global genomic hypomethylation and dramatic reorganization of the photoreceptor and synaptic layers within retina. Transcriptome and proteomic analyses demonstrated enrichment of dysregulated phototransduction and synaptic genes. The 5 mC signal in triple mutant retina was confined to the central heterochromatin but reduced in the peripheral heterochromatin region of photoreceptor nuclei. In addition, we found a reduction of the 5 mC signal in ganglion cell nuclei. Collectively, this data suggests cooperation of all three Dnmts in the formation and homeostasis of photoreceptors and other retinal neurons within the mammalian retina, and highlight the relevance of epigenetic regulation to sensory retinal disorders and vision loss.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , DNA/genética , Mutação , Células Fotorreceptoras de Vertebrados/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Análise Mutacional de DNA , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Microscopia Eletrônica , Modelos Animais , Células Fotorreceptoras de Vertebrados/ultraestrutura , Reação em Cadeia da Polimerase em Tempo Real , Neurônios Retinianos/metabolismo , Neurônios Retinianos/ultraestrutura , DNA Metiltransferase 3B
10.
Invest Ophthalmol Vis Sci ; 57(9): OCT105-11, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27409460

RESUMO

PURPOSE: We examined if light induces changes in the retinal structure that can be observed using optical coherence tomography (OCT). METHODS: Normal C57BL/6J mice (age 3-6 months) adapted to either room light (15 minutes to ∼5 hours, 50-500 lux) or darkness (overnight) were imaged using a Bioptigen UHR-OCT system. Confocal histologic images were obtained from mice killed under light- or dark-adapted conditions. RESULTS: The OCT image of eyes adapted to room light exhibited significant increases (6.1 ± 0.8 µm, n = 13) in total retina thickness compared to the same eyes after overnight dark adaptation. These light-adapted retinal thickness changes occurred mainly in the outer retina, with the development of a hyporeflective band between the RPE and photoreceptor-tip layers. Histologic analysis revealed a light-evoked elongation between the outer limiting membrane and Bruch's membrane from 45.8 ± 1.7 µm in the dark (n = 5) to 52.1 ± 3.7 µm (n = 5) in the light. Light-adapted retinas showed an increase of actin staining in RPE apical microvilli at the same location as the hyporeflective band observed in OCT images. Elongation of the outer retina could be detected even with brief light exposures, increasing 2.1 ± 0.3 µm after 15 minutes (n = 9), and 4.1 ± 1.0 µm after 2 hours (n = 6). Conversely, dark-adaptation caused outer retinal shortening of 1.4 ± 0.4 µm (n = 7) and 3.0 ± 0.5 µm (n = 8) after 15 minutes and 2 hours, respectively. CONCLUSIONS: Light-adaption induces an increase in the thickness of the outer retina and the appearance of a hyporeflective band in the OCT image. This is consistent with previous reports of light-induced fluid accumulation in the subretinal space.


Assuntos
Aumento da Imagem/métodos , Luz/efeitos adversos , Doenças Retinianas/diagnóstico , Segmento Externo das Células Fotorreceptoras da Retina/patologia , Tomografia de Coerência Óptica/métodos , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Retinianas/etiologia , Células Ganglionares da Retina/patologia , Segmento Externo das Células Fotorreceptoras da Retina/efeitos da radiação
11.
Glia ; 64(9): 1479-91, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27314452

RESUMO

Retinitis pigmentosa (RP), a disease characterized by the progressive degeneration of mutation-bearing photoreceptors, is a significant cause of incurable blindness in the young worldwide. Recent studies have found that activated retinal microglia contribute to photoreceptor demise via phagocytosis and proinflammatory factor production, however mechanisms regulating these contributions are not well-defined. In this study, we investigate the role of CX3CR1, a microglia-specific receptor, in regulating microglia-mediated degeneration using the well-established rd10 mouse model of RP. We found that in CX3CR1-deficient (CX3CR1(GFP/GFP) ) rd10 mice microglial infiltration into the photoreceptor layer was significantly augmented and associated with accelerated photoreceptor apoptosis and atrophy compared with CX3CR1-sufficient (CX3CR1(GFP/+) ) rd10 littermates. CX3CR1-deficient microglia demonstrated increased phagocytosis as evidenced by (1) having increased numbers of phagosomes in vivo, (2) an increased rate of phagocytosis of fluorescent beads and photoreceptor cellular debris in vitro, and (3) increased photoreceptor phagocytosis dynamics on live cell imaging in retinal explants, indicating that CX3CR1 signaling in microglia regulates the phagocytic clearance of at-risk photoreceptors. We also found that CX3CR1 deficiency in retinal microglia was associated with increased expression of inflammatory cytokines and microglial activation markers. Significantly, increasing CX3CL1-CX3CR1 signaling in the rd10 retina via exogenous intravitreal delivery of recombinant CX3CL1 was effective in (1) decreasing microglial infiltration, phagocytosis and activation, and (2) improving structural and functional features of photoreceptor degeneration. These results indicate that CX3CL1-CX3CR1 signaling is a molecular mechanism capable of modulating microglial-mediated degeneration and represents a potential molecular target in therapeutic approaches to RP. GLIA 2016;64:1479-1491.


Assuntos
Receptor 1 de Quimiocina CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Microglia/metabolismo , Fagocitose/fisiologia , Células Fotorreceptoras/metabolismo , Retinose Pigmentar/metabolismo , Animais , Modelos Animais de Doenças , Macrófagos/metabolismo , Camundongos Transgênicos , Fármacos Neuroprotetores/farmacologia , Receptores de Quimiocinas/metabolismo , Retina/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
J Neurosci ; 36(9): 2827-42, 2016 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-26937019

RESUMO

Microglia, the principal resident immune cell of the CNS, exert significant influence on neurons during development and in pathological situations. However, if and how microglia contribute to normal neuronal function in the mature uninjured CNS is not well understood. We used the model of the adult mouse retina, a part of the CNS amenable to structural and functional analysis, to investigate the constitutive role of microglia by depleting microglia from the retina in a sustained manner using genetic methods. We discovered that microglia are not acutely required for the maintenance of adult retinal architecture, the survival of retinal neurons, or the laminar organization of their dendritic and axonal compartments. However, sustained microglial depletion results in the degeneration of photoreceptor synapses in the outer plexiform layer, leading to a progressive functional deterioration in retinal light responses. Our results demonstrate that microglia are constitutively required for the maintenance of synaptic structure in the adult retina and for synaptic transmission underlying normal visual function. Our findings on constitutive microglial function are relevant in understanding microglial contributions to pathology and in the consideration of therapeutic interventions that reduce or perturb constitutive microglial function. SIGNIFICANCE STATEMENT: Microglia, the principal resident immune cell population in the CNS, has been implicated in diseases in the brain and retina. However, how they contribute to the everyday function of the CNS is unclear. Using the model of the adult mouse retina, we examined the constitutive role of microglia by depleting microglia from the retina. We found that in the absence of microglia, retinal neurons did not undergo overt cell death or become structurally disorganized in their processes. However, connections between neurons called synapses begin to break down, leading to a decreased ability of the retina to transmit light responses. Our results indicate that retinal microglia contribute constitutively to the maintenance of synapses underlying healthy vision.


Assuntos
Microglia/fisiologia , Neurônios/fisiologia , Retina/citologia , Sinapses/fisiologia , Animais , Morte Celular/genética , Modelos Animais de Doenças , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Proteínas do Olho/metabolismo , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Nistagmo Optocinético/genética , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Sinapses/genética , Transtornos da Visão/genética , Transtornos da Visão/patologia , Transtornos da Visão/fisiopatologia , Vias Visuais/fisiologia
13.
EMBO Mol Med ; 7(9): 1179-97, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26139610

RESUMO

Retinitis pigmentosa, caused predominantly by mutations in photoreceptor genes, currently lacks comprehensive treatment. We discover that retinal microglia contribute non-cell autonomously to rod photoreceptor degeneration by primary phagocytosis of living rods. Using rd10 mice, we found that the initiation of rod degeneration is accompanied by early infiltration of microglia, upregulation of phagocytic molecules in microglia, and presentation of "eat-me" signals on mutated rods. On live-cell imaging, infiltrating microglia interact dynamically with photoreceptors via motile processes and engage in rapid phagocytic engulfment of non-apoptotic rods. Microglial contribution to rod demise is evidenced by morphological and functional amelioration of photoreceptor degeneration following genetic ablation of retinal microglia. Molecular inhibition of microglial phagocytosis using the vitronectin receptor antagonist cRGD also improved morphological and functional parameters of degeneration. Our findings highlight primary microglial phagocytosis as a contributing mechanism underlying cell death in retinitis pigmentosa and implicate microglia as a potential cellular target for therapy.


Assuntos
Microglia/fisiologia , Fagocitose , Células Fotorreceptoras Retinianas Bastonetes/patologia , Retinose Pigmentar/congênito , Retinose Pigmentar/patologia , Animais , Morte Celular , Movimento Celular , Modelos Animais de Doenças , Integrina alfaVbeta3/antagonistas & inibidores , Camundongos , Imagem Óptica , Peptídeos Cíclicos/metabolismo
14.
Hum Mol Genet ; 24(13): 3775-91, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25859007

RESUMO

Distinct mutations in the centrosomal-cilia protein CEP290 lead to diverse clinical findings in syndromic ciliopathies. We show that CEP290 localizes to the transition zone in ciliated cells, precisely to the region of Y-linkers between central microtubules and plasma membrane. To create models of CEP290-associated ciliopathy syndromes, we generated Cep290(ko/ko) and Cep290(gt/gt) mice that produce no or a truncated CEP290 protein, respectively. Cep290(ko/ko) mice exhibit early vision loss and die from hydrocephalus. Retinal photoreceptors in Cep290(ko/ko) mice lack connecting cilia, and ciliated ventricular ependyma fails to mature. The minority of Cep290(ko/ko) mice that escape hydrocephalus demonstrate progressive kidney pathology. Cep290(gt/gt) mice die at mid-gestation, and the occasional Cep290(gt/gt) mouse that survives shows hydrocephalus and severely cystic kidneys. Partial loss of CEP290-interacting ciliopathy protein MKKS mitigates lethality and renal pathology in Cep290(gt/gt) mice. Our studies demonstrate domain-specific functions of CEP290 and provide novel therapeutic paradigms for ciliopathies.


Assuntos
Cílios/metabolismo , Hidrocefalia/genética , Doenças Renais Císticas/genética , Proteínas Nucleares/genética , Animais , Antígenos de Neoplasias , Proteínas de Ciclo Celular , Cílios/genética , Proteínas do Citoesqueleto , Modelos Animais de Doenças , Feminino , Humanos , Hidrocefalia/metabolismo , Doenças Renais Císticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/metabolismo , Especificidade de Órgãos
15.
Sci Rep ; 5: 9144, 2015 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-25775051

RESUMO

Age-related macular degeneration (AMD) has been associated with both accumulation of lipid and lipid oxidative products, as well as increased neuroinflammatory changes and microglial activation in the outer retina. However, the relationships between these factors are incompletely understood. 7-Ketocholesterol (7KCh) is a cholesterol oxidation product localized to the outer retina with prominent pro-inflammatory effects. To explore the potential relationship between 7KCh and microglial activation, we localized 7KCh and microglia to the outer retina of aged mice and investigated 7KCh effects on retinal microglia in both in vitro and in vivo systems. We found that retinal microglia demonstrated a prominent chemotropism to 7KCh and readily internalized 7KCh. Sublethal concentrations of 7KCh resulted in microglial activation and polarization to a pro-inflammatory M1 state via NLRP3 inflammasome activation. Microglia exposed to 7KCh reduced expression of neurotrophic growth factors but increased expression of angiogenic factors, transitioning to a more neurotoxic and pro-angiogenic phenotype. Finally, subretinal transplantation of 7KCh-exposed microglia promoted choroidal neovascularization (CNV) relative to control microglia in a Matrigel-CNV model. The interaction of retinal microglia with 7KCh in the aged retina may thus underlie how outer retinal lipid accumulation in intermediate AMD results in neuroinflammation that ultimately drives progression towards advanced AMD.


Assuntos
Cetocolesteróis/metabolismo , Degeneração Macular/etiologia , Degeneração Macular/patologia , Microglia/metabolismo , Neovascularização Patológica , Retina/metabolismo , Retina/patologia , Animais , Receptor 1 de Quimiocina CX3C , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fatores Quimiotáticos/metabolismo , Fatores Quimiotáticos/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Cetocolesteróis/farmacologia , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/imunologia , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Retina/efeitos dos fármacos
16.
Invest Ophthalmol Vis Sci ; 55(9): 6031-40, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25159211

RESUMO

PURPOSE: The aryl hydrocarbon receptor (AHR) is a ligand-activated nuclear receptor that regulates cellular response to environmental signals, including UV and blue wavelength light. This study was undertaken to elucidate AHR function in retinal homeostasis. METHODS: RNA-seq data sets were examined for Ahr expression in the mouse retina and rod photoreceptors. The Ahr(-/-) mice were evaluated by fundus imaging, optical coherence tomography, histology, immunohistochemistry, and ERG. For light damage experiments, adult mice were exposed to 14,000 to 15,000 lux of diffuse white light for 2 hours. RESULTS: In mouse retina, Ahr transcripts were upregulated during development, with continued increase in aging rod photoreceptors. Fundus examination of 3-month-old Ahr(-/-) mice revealed subretinal autofluorescent spots, which increased in number with age and following acute light exposure. Ahr(-/-) retina also showed subretinal microglia accumulation that correlated with autofluorescence changes, RPE abnormalities, and reactivity against immunoglobulin, complement factor H, and glial fibrillary acidic protein. Functionally, Ahr(-/-) mice displayed reduced ERG c-wave amplitudes. CONCLUSIONS: The Ahr(-/-) mice exhibited subretinal accumulation of microglia and focal RPE atrophy, phenotypes observed in AMD. Together with a recently published report on another Ahr(-/-) mouse model, our study suggests that AHR has a protective role in the retina as an environmental stress sensor. As such, its altered function may contribute to human AMD progression and provide a target for pharmacological intervention.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Microglia/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Degeneração Retiniana/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Animais , Atrofia/metabolismo , Atrofia/patologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Modelos Animais de Doenças , Fundo de Olho , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Receptores de Hidrocarboneto Arílico/imunologia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Epitélio Pigmentado da Retina/patologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Retinite/metabolismo , Retinite/patologia , Tomografia de Coerência Óptica , Transcriptoma
17.
Nat Commun ; 5: 4207, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24947469

RESUMO

The primary cilium originates from the mother centriole and participates in critical functions during organogenesis. Defects in cilia biogenesis or function lead to pleiotropic phenotypes. Mutations in centrosome-cilia gene CC2D2A result in Meckel and Joubert syndromes. Here we generate a Cc2d2a(-/-) mouse that recapitulates features of Meckel syndrome including embryonic lethality and multiorgan defects. Cilia are absent in Cc2d2a(-/-) embryonic node and other somatic tissues; disruption of cilia-dependent Shh signalling appears to underlie exencephaly in mutant embryos. The Cc2d2a(-/-) mouse embryonic fibroblasts (MEFs) lack cilia, although mother centrioles and pericentriolar proteins are detected. Odf2, associated with subdistal appendages, is absent and ninein is reduced in mutant MEFs. In Cc2d2a(-/-) MEFs, subdistal appendages are lacking or abnormal by transmission electron microscopy. Consistent with this, CC2D2A localizes to subdistal appendages by immuno-EM in wild-type cells. We conclude that CC2D2A is essential for the assembly of subdistal appendages, which anchor cytoplasmic microtubules and prime the mother centriole for axoneme biogenesis.


Assuntos
Centríolos/metabolismo , Cílios/patologia , Proteínas/genética , Alelos , Animais , Transporte Biológico , Centrossomo/ultraestrutura , Cílios/genética , Citoplasma/metabolismo , Proteínas do Citoesqueleto , Fibroblastos/metabolismo , Citometria de Fluxo , Proteínas Hedgehog/metabolismo , Macaca mulatta , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Microtúbulos/metabolismo , Mutação , Fenótipo , Proteínas/fisiologia , Transdução de Sinais , Transgenes
18.
Hum Mol Genet ; 23(21): 5827-37, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24899048

RESUMO

Neurodegenerative diseases affecting the macula constitute a major cause of incurable vision loss and exhibit considerable clinical and genetic heterogeneity, from early-onset monogenic disease to multifactorial late-onset age-related macular degeneration (AMD). As part of our continued efforts to define genetic causes of macular degeneration, we performed whole exome sequencing in four individuals of a two-generation family with autosomal dominant maculopathy and identified a rare variant p.Glu1144Lys in Fibrillin 2 (FBN2), a glycoprotein of the elastin-rich extracellular matrix (ECM). Sanger sequencing validated the segregation of this variant in the complete pedigree, including two additional affected and one unaffected individual. Sequencing of 192 maculopathy patients revealed additional rare variants, predicted to disrupt FBN2 function. We then undertook additional studies to explore the relationship of FBN2 to macular disease. We show that FBN2 localizes to Bruch's membrane and its expression appears to be reduced in aging and AMD eyes, prompting us to examine its relationship with AMD. We detect suggestive association of a common FBN2 non-synonymous variant, rs154001 (p.Val965Ile) with AMD in 10 337 cases and 11 174 controls (OR = 1.10; P-value = 3.79 × 10(-5)). Thus, it appears that rare and common variants in a single gene--FBN2--can contribute to Mendelian and complex forms of macular degeneration. Our studies provide genetic evidence for a key role of elastin microfibers and Bruch's membrane in maintaining blood-retina homeostasis and establish the importance of studying orphan diseases for understanding more common clinical phenotypes.


Assuntos
Estudos de Associação Genética , Variação Genética , Degeneração Macular/genética , Proteínas dos Microfilamentos/genética , Adulto , Idoso , Sequência de Aminoácidos , Lâmina Basilar da Corioide/metabolismo , Análise Mutacional de DNA , Exoma , Matriz Extracelular/metabolismo , Fibrilina-2 , Fibrilinas , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Degeneração Macular/diagnóstico , Masculino , Metanálise como Assunto , Proteínas dos Microfilamentos/metabolismo , Pessoa de Meia-Idade , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Linhagem , Conformação Proteica , Estabilidade Proteica , Retina/metabolismo , Retina/patologia , Alinhamento de Sequência
19.
Annu Rev Genomics Hum Genet ; 15: 151-71, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24773320

RESUMO

Genetic and genomic studies have enhanced our understanding of complex neurodegenerative diseases that exert a devastating impact on individuals and society. One such disease, age-related macular degeneration (AMD), is a major cause of progressive and debilitating visual impairment. Since the pioneering discovery in 2005 of complement factor H (CFH) as a major AMD susceptibility gene, extensive investigations have confirmed 19 additional genetic risk loci, and more are anticipated. In addition to common variants identified by now-conventional genome-wide association studies, targeted genomic sequencing and exome-chip analyses are uncovering rare variant alleles of high impact. Here, we provide a critical review of the ongoing genetic studies and of common and rare risk variants at a total of 20 susceptibility loci, which together explain 40-60% of the disease heritability but provide limited power for diagnostic testing of disease risk. Identification of these susceptibility loci has begun to untangle the complex biological pathways underlying AMD pathophysiology, pointing to new testable paradigms for treatment.


Assuntos
Fator H do Complemento/genética , Predisposição Genética para Doença , Degeneração Macular/genética , Polimorfismo de Nucleotídeo Único/genética , Estudo de Associação Genômica Ampla , Haplótipos , Humanos , Degeneração Macular/patologia , Mutação , Fatores de Risco
20.
J Neurosci ; 34(10): 3793-806, 2014 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24599476

RESUMO

Chronic retinal inflammation in the form of activated microglia and macrophages are implicated in the etiology of neurodegenerative diseases of the retina, including age-related macular degeneration, diabetic retinopathy, and glaucoma. However, molecular biomarkers and targeted therapies for immune cell activation in these disorders are currently lacking. To address this, we investigated the involvement and role of translocator protein (TSPO), a biomarker of microglial and astrocyte gliosis in brain degeneration, in the context of retinal inflammation. Here, we find that TSPO is acutely and specifically upregulated in retinal microglia in separate mouse models of retinal inflammation and injury. Concomitantly, its endogenous ligand, diazepam-binding inhibitor (DBI), is upregulated in the macroglia of the mouse retina such as astrocytes and Müller cells. In addition, we discover that TSPO-mediated signaling in microglia via DBI-derived ligands negatively regulates features of microglial activation, including reactive oxygen species production, TNF-α expression and secretion, and microglial proliferation. The inducibility and effects of DBI-TSPO signaling in the retina reveal a mechanism of coordinated macroglia-microglia interactions, the function of which is to limit the magnitude of inflammatory responses after their initiation, facilitating a return to baseline quiescence. Our results indicate that TSPO is a promising molecular marker for imaging inflammatory cell activation in the retina and highlight DBI-TSPO signaling as a potential target for immodulatory therapies.


Assuntos
Comunicação Celular/fisiologia , Microglia/metabolismo , Receptores de GABA/fisiologia , Retina/metabolismo , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Astrócitos/metabolismo , Células Cultivadas , Feminino , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Receptores de GABA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA