Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
EBioMedicine ; 104: 105170, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38823088

RESUMEN

BACKGROUND: Ebola virus disease (EVD) survivors experience ocular sequelae including retinal lesions, cataracts, and vision loss. While monoclonal antibodies targeting the Ebola virus glycoprotein (EBOV-GP) have shown promise in improving prognosis, their effectiveness in mitigating ocular sequelae remains uncertain. METHODS: We developed and characterized a BSL-2-compatible immunocompetent mouse model to evaluate therapeutics targeting EBOV-GP by inoculating neonatal mice with vesicular stomatitis virus expressing EBOV-GP (VSV-EBOV). To examine the impact of anti-EBOV-GP antibody treatment on acute retinitis and ocular sequelae, VSV-EBOV-infected mice were treated with polyclonal antibodies or monoclonal antibody preparations with antibody-dependent cellular cytotoxicity (ADCC-mAb) or neutralizing activity (NEUT-mAb). FINDINGS: Treatment with all anti-EBOV-GP antibodies tested dramatically reduced viremia and improved survival. Further, all treatments reduced the incidence of cataracts. However, NEUT-mAb alone or in combination with ADCC-mAb reduced viral load in the eyes, downregulated the ocular immune and inflammatory responses, and minimized retinal damage more effectively. INTERPRETATION: Anti-EBOV-GP antibodies can improve survival among EVD patients, but improved therapeutics are needed to reduce life altering sequelae. This animal model offers a new platform to examine the acute and long-term effect of the virus in the eye and the relative impact of therapeutic candidates targeting EBOV-GP. Results indicate that even antibodies that improve systemic viral clearance and survival can differ in their capacity to reduce acute ocular inflammation, and long-term retinal pathology and corneal degeneration. FUNDING: This study was partly supported by Postgraduate Research Fellowship Awards from ORISE through an interagency agreement between the US DOE and the US FDA.


Asunto(s)
Anticuerpos Antivirales , Modelos Animales de Enfermedad , Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Ratones , Ebolavirus/inmunología , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/virología , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/farmacología , Humanos , Carga Viral , Glicoproteínas/inmunología , Glicoproteínas/metabolismo , Proteínas del Envoltorio Viral/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos
2.
Prog Retin Eye Res ; 95: 101147, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36402656

RESUMEN

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.


Asunto(s)
Retina , Retinosquisis , Masculino , Humanos , Estructura Molecular , Retina/metabolismo , Retinosquisis/genética , Retinosquisis/metabolismo , Mutación , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Proteínas del Ojo/genética
3.
Invest Ophthalmol Vis Sci ; 63(11): 8, 2022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-36227606

RESUMEN

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.


Asunto(s)
Quistes , Retinosquisis , Animales , Ratones , Quistes/metabolismo , Quistes/patología , Electrorretinografía , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Retina/metabolismo , Retina/patología , Células Bipolares de la Retina/metabolismo , Retinosquisis/genética , Retinosquisis/metabolismo
4.
Hum Gene Ther ; 32(13-14): 667-681, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33019822

RESUMEN

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.


Asunto(s)
Retinosquisis , Animales , Electrorretinografía , Proteínas del Ojo/genética , Redes Reguladoras de Genes , Terapia Genética , Vectores Genéticos/genética , Ratones , Microglía , Retina , Retinosquisis/genética , Retinosquisis/terapia
5.
PLoS Genet ; 16(12): e1009259, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33362196

RESUMEN

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.


Asunto(s)
Autofagia , Endocitosis , Factores de Intercambio de Guanina Nucleótido/genética , Neurogénesis , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/metabolismo , Animales , Femenino , Factores de Intercambio de Guanina Nucleótido/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Células Fotorreceptoras/citología , Degeneración Retiniana/genética , Transcriptoma
6.
Sci Adv ; 4(3): eaap8492, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29750189

RESUMEN

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.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Microglía/metabolismo , Retina/citología , Envejecimiento/fisiología , Animales , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular , Proliferación Celular , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microglía/ultraestructura , Transducción de Señal
7.
Sci Rep ; 7(1): 8433, 2017 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-28814744

RESUMEN

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.


Asunto(s)
Proliferación Celular , Células Epiteliales/metabolismo , Monocitos/metabolismo , Células Mieloides/metabolismo , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Homeostasis , Yodatos/toxicidad , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microscopía Confocal , Receptores CCR2/genética , Receptores CCR2/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología
8.
Hum Mol Genet ; 26(12): 2218-2230, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28369466

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Membrana/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Vesículas Cubiertas por Clatrina/metabolismo , Proteínas del Ojo/genética , Fototransducción , Proteínas de la Membrana , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Noqueados , Mutación , Células Fotorreceptoras de Vertebrados/metabolismo , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Proteínas Qa-SNARE/metabolismo , Degeneración Retiniana/metabolismo , Retinitis Pigmentosa/genética , Proteínas SNARE/metabolismo
9.
Brain Struct Funct ; 222(6): 2759-2771, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28213784

RESUMEN

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.


Asunto(s)
Envejecimiento , Forma de la Célula , Macaca mulatta , Microglía/citología , Retina/citología , Factores de Edad , Animales , Biomarcadores/análisis , Proteínas del Ojo/análisis , Femenino , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional , Inmunohistoquímica , Microglía/química , Microscopía Confocal , Retina/química
10.
Exp Eye Res ; 159: 132-146, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27865785

RESUMEN

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.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , ADN/genética , Mutación , Células Fotorreceptoras de Vertebrados/metabolismo , Animales , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , ADN Metiltransferasa 3A , Análisis Mutacional de ADN , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Mutantes , Microscopía Electrónica , Modelos Animales , Células Fotorreceptoras de Vertebrados/ultraestructura , Reacción en Cadena en Tiempo Real de la Polimerasa , Neuronas Retinianas/metabolismo , Neuronas Retinianas/ultraestructura , ADN Metiltransferasa 3B
11.
Invest Ophthalmol Vis Sci ; 57(9): OCT105-11, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27409460

RESUMEN

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.


Asunto(s)
Aumento de la Imagen/métodos , Luz/efectos adversos , Enfermedades de la Retina/diagnóstico , Segmento Externo de las Células Fotorreceptoras Retinianas/patología , Tomografía de Coherencia Óptica/métodos , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades de la Retina/etiología , Células Ganglionares de la Retina/patología , Segmento Externo de las Células Fotorreceptoras Retinianas/efectos de la radiación
12.
Glia ; 64(9): 1479-91, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27314452

RESUMEN

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.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Microglía/metabolismo , Fagocitosis/fisiología , Células Fotorreceptoras/metabolismo , Retinitis Pigmentosa/metabolismo , Animales , Modelos Animales de Enfermedad , Macrófagos/metabolismo , Ratones Transgénicos , Fármacos Neuroprotectores/farmacología , Receptores de Quimiocina/metabolismo , Retina/metabolismo , Transducción de Señal/efectos de los fármacos
13.
J Neurosci ; 36(9): 2827-42, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26937019

RESUMEN

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.


Asunto(s)
Microglía/fisiología , Neuronas/fisiología , Retina/citología , Sinapsis/fisiología , Animales , Muerte Celular/genética , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Proteínas del Ojo/metabolismo , Femenino , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Nistagmo Optoquinético/genética , ARN no Traducido/genética , ARN no Traducido/metabolismo , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Sinapsis/genética , Trastornos de la Visión/genética , Trastornos de la Visión/patología , Trastornos de la Visión/fisiopatología , Vías Visuales/fisiología
14.
EMBO Mol Med ; 7(9): 1179-97, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26139610

RESUMEN

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.


Asunto(s)
Microglía/fisiología , Fagocitosis , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/congénito , Retinitis Pigmentosa/patología , Animales , Muerte Celular , Movimiento Celular , Modelos Animales de Enfermedad , Integrina alfaVbeta3/antagonistas & inhibidores , Ratones , Imagen Óptica , Péptidos Cíclicos/metabolismo
15.
Hum Mol Genet ; 24(13): 3775-91, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25859007

RESUMEN

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.


Asunto(s)
Cilios/metabolismo , Hidrocefalia/genética , Enfermedades Renales Quísticas/genética , Proteínas Nucleares/genética , Animales , Antígenos de Neoplasias , Proteínas de Ciclo Celular , Cilios/genética , Proteínas del Citoesqueleto , Modelos Animales de Enfermedad , Femenino , Humanos , Hidrocefalia/metabolismo , Enfermedades Renales Quísticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/metabolismo , Especificidad de Órganos
16.
Sci Rep ; 5: 9144, 2015 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-25775051

RESUMEN

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.


Asunto(s)
Cetocolesteroles/metabolismo , Degeneración Macular/etiología , Degeneración Macular/patología , Microglía/metabolismo , Neovascularización Patológica , Retina/metabolismo , Retina/patología , Animales , Receptor 1 de Quimiocinas CX3C , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factores Quimiotácticos/metabolismo , Factores Quimiotácticos/farmacología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Cetocolesteroles/farmacología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/inmunología , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Retina/efectos de los fármacos
17.
Invest Ophthalmol Vis Sci ; 55(9): 6031-40, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25159211

RESUMEN

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.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Microglía/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Degeneración Retiniana/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Atrofia/metabolismo , Atrofia/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Modelos Animales de Enfermedad , Fondo de Ojo , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Receptores de Hidrocarburo de Aril/inmunología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/patología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis/metabolismo , Retinitis/patología , Tomografía de Coherencia Óptica , Transcriptoma
18.
Hum Mol Genet ; 23(21): 5827-37, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899048

RESUMEN

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.


Asunto(s)
Estudios de Asociación Genética , Variación Genética , Degeneración Macular/genética , Proteínas de Microfilamentos/genética , Adulto , Anciano , Secuencia de Aminoácidos , Lámina Basal de la Coroides/metabolismo , Análisis Mutacional de ADN , Exoma , Matriz Extracelular/metabolismo , Fibrilina-2 , Fibrilinas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Degeneración Macular/diagnóstico , Masculino , Metaanálisis como Asunto , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Linaje , Conformación Proteica , Estabilidad Proteica , Retina/metabolismo , Retina/patología , Alineación de Secuencia
19.
Nat Commun ; 5: 4207, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24947469

RESUMEN

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.


Asunto(s)
Centriolos/metabolismo , Cilios/patología , Proteínas/genética , Alelos , Animales , Transporte Biológico , Centrosoma/ultraestructura , Cilios/genética , Citoplasma/metabolismo , Proteínas del Citoesqueleto , Fibroblastos/metabolismo , Citometría de Flujo , Proteínas Hedgehog/metabolismo , Macaca mulatta , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Microtúbulos/metabolismo , Mutación , Fenotipo , Proteínas/fisiología , Transducción de Señal , Transgenes
20.
Annu Rev Genomics Hum Genet ; 15: 151-71, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24773320

RESUMEN

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.


Asunto(s)
Factor H de Complemento/genética , Predisposición Genética a la Enfermedad , Degeneración Macular/genética , Polimorfismo de Nucleótido Simple/genética , Estudio de Asociación del Genoma Completo , Haplotipos , Humanos , Degeneración Macular/patología , Mutación , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...