Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 37: 441-468, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34351785

RESUMEN

Visual opsin genes expressed in the rod and cone photoreceptor cells of the retina are core components of the visual sensory system of vertebrates. Here, we provide an overview of the dynamic evolution of visual opsin genes in the most species-rich group of vertebrates, teleost fishes. The examination of the rich genomic resources now available for this group reveals that fish genomes contain more copies of visual opsin genes than are present in the genomes of amphibians, reptiles, birds, and mammals. The expansion of opsin genes in fishes is due primarily to a combination of ancestral and lineage-specific gene duplications. Following their duplication, the visual opsin genes of fishes repeatedly diversified at the same key spectral-tuning sites, generating arrays of visual pigments sensitive to the ultraviolet to red spectrum of light. Species-specific opsin gene repertoires correlate strongly with underwater light habitats, ecology, and color-based sexual selection.


Asunto(s)
Opsinas , Opsinas de Bastones , Animales , Peces/genética , Mamíferos , Opsinas/genética , Filogenia , Pigmentos Retinianos/genética , Opsinas de Bastones/genética , Vertebrados/genética
2.
J Transl Med ; 22(1): 245, 2024 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-38448948

RESUMEN

BACKGROUND: In diabetic retinopathy (DR), hypoxia-inducible factor (HIF-1α) induces oxidative stress by upregulating glycolysis. This process leads to neurodegeneration, particularly photoreceptor cell damage, which further contributes to retinal microvascular deterioration. Further, the regulation of Wnt-inhibitory factor 1 (WIF1), a secreted Wnt signaling antagonist, has not been fully characterized in neurodegenerative eye diseases. We aimed to explore the impact of WIF1 on photoreceptor function within the context of DR. METHOD: Twelve-week-old C57BL/KsJ-db/db mice were intravitreally injected with WIF1 overexpression lentivirus. After 4 weeks, optical coherence tomography (OCT), transmission electron microscopy (TEM), H&E staining, and electroretinography (ERG) were used to assess the retinal tissue and function. The potential mechanism of action of WIF1 in photoreceptor cells was explored using single-cell RNA sequencing. Under high-glucose conditions, 661 W cells were used as an in vitro DR model. WIF1-mediated signaling pathway components were assessed using quantitative real-time PCR, immunostaining, and western blotting. RESULT: Typical diabetic manifestations were observed in db/db mice. Notably, the expression of WIF1 was decreased at the mRNA and protein levels. These pathological manifestations and visual function improved after WIF1 overexpression in db/db mice. TEM demonstrated that WIF1 restored damaged mitochondria, the Golgi apparatus, and photoreceptor outer segments. Moreover, ERG indicated the recovery of a-wave potential amplitude. Single-cell RNA sequencing and in vitro experiments suggested that WIF1 overexpression prevented the expression of glycolytic enzymes and lactate production by inhibiting the canonical Wnt signaling pathway, HIF-1α, and Glut1, thereby reducing retinal and cellular reactive oxygen species levels and maintaining 661 W cell viability. CONCLUSIONS: WIF1 exerts an inhibitory effect on the Wnt/ß-catenin-HIF-1α-Glut1 glycolytic pathway, thereby alleviating oxidative stress levels and mitigating pathological structural characteristics in retinal photoreceptor cells. This mechanism helps preserve the function of photoreceptor cells in DR and indicates that WIF1 holds promise as a potential therapeutic candidate for DR and other neurodegenerative ocular disorders.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Animales , Ratones , Transportador de Glucosa de Tipo 1 , Ratones Endogámicos C57BL , Células Fotorreceptoras , Retina
3.
FASEB J ; 37(5): e22885, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37000492

RESUMEN

Recent studies have reported the promising value of differential gene expression analysis and weighted gene coexpression network analysis (WGCNA) for identifying disease biomarkers. Based on this method, this study intends to characterize the hub genes and pathways related to retinal photoreceptor cell (PRC) injury in the context of retinitis pigmentosa (RP). A total of 53 coexpression modules were identified by WGCNA, among which lightpink4, darkolivegreen, tan4, blue2, skyblue2, and navajowhite2 ranked at the top. By analyzing the RP microarrays retrieved from the GEO database, 338 differentially expressed genes (DEGs) were identified in the RP samples. Forty-five candidate genes were selected from these DEGs by intersection with the genes in the coexpression modules. These intersection genes were subjected to GO and KEGG analyses. Furthermore, the genes and pathways involved in PRC damage were identified based on analyses utilizing GeneCards and STRING tools. Transcription factor 7-like 1 (TCF7L1, also called TCF3) was suggested to participate in the RP-associated PRC damage through the Wnt signaling pathway. It was validated in a blue light-irradiated cell model that TCF7L1 overexpression boosted PRC viability and repressed apoptosis. Inhibition of the Wnt signaling pathway also contributed to protective effects. Together, the data mentioned above supported the conclusion that either elevation of TCF7L1 or blockade of the Wnt signaling pathway could prevent RP progression by protecting PRCs from damage.


Asunto(s)
Redes Reguladoras de Genes , Retinitis Pigmentosa , Humanos , Células Fotorreceptoras de Vertebrados , Análisis por Micromatrices , Bases de Datos Genéticas , Retinitis Pigmentosa/genética , Perfilación de la Expresión Génica/métodos , Proteína 1 Similar al Factor de Transcripción 7
4.
J Neurochem ; 167(4): 538-555, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37840219

RESUMEN

GPR75 is an orphan G protein-coupled receptor for which there is currently limited information and its function in physiology and disease is only recently beginning to emerge. This orphan receptor is expressed in the retina but its function in the eye is unknown. The earliest studies on GPR75 were conducted in the retina, where the receptor was first identified and cloned and mutations in the receptor were identified as a possible contributor to retinal degenerative disease. Despite these sporadic reports, the function of GPR75 in the retina and in retinal disease has yet to be explored. To assess whether GPR75 has a functional role in the retina, the retina of Gpr75 knockout mice was characterized. Knockout mice displayed a mild progressive retinal degeneration, which was accompanied by oxidative stress. The degeneration was because of the loss of both M-cone and S-cone photoreceptor cells. Housing mice under constant dark conditions reduced oxidative stress but did not prevent cone photoreceptor cell loss, indicating that oxidative stress is not a primary cause of the observed retinal degeneration. Studies here demonstrate an important role for GPR75 in maintaining the health of cone photoreceptor cells and that Gpr75 knockout mice can be used as a model to study cone photoreceptor cell loss.


Asunto(s)
Células Fotorreceptoras Retinianas Conos , Degeneración Retiniana , Ratones , Animales , Degeneración Retiniana/genética , Ratones Noqueados , Retina , Ratones Endogámicos C57BL
5.
Cell Mol Life Sci ; 79(9): 493, 2022 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-36001156

RESUMEN

Opsins are universal photoreceptive proteins in animals. Vertebrate rhodopsin in ciliary photoreceptor cells photo-converts to a metastable active state to regulate cyclic nucleotide signaling. This active state cannot photo-convert back to the dark state, and thus vertebrate rhodopsin is categorized as a mono-stable opsin. By contrast, mollusk and arthropod rhodopsins in rhabdomeric photoreceptor cells photo-convert to a stable active state to stimulate IP3/calcium signaling. This active state can photo-convert back to the dark state, and thus these rhodopsins are categorized as bistable opsins. Moreover, the negatively charged counterion position crucial for the visible light sensitivity is different between vertebrate rhodopsin (Glu113) and mollusk and arthropod rhodopsins (Glu181). This can be explained by an evolutionary scenario where vertebrate rhodopsin newly acquired Glu113 as a counterion, which is thought to have led to higher signaling efficiency of vertebrate rhodopsin. However, the detailed evolutionary steps which led to the higher efficiency in vertebrate rhodopsin still remain unknown. Here, we analyzed the xenopsin group, which is phylogenetically distinct from vertebrate rhodopsin and functions in protostome ciliary cells. Xenopsins are blue-sensitive bistable opsins that regulate cAMP signaling. We found that a bistable xenopsin of Leptochiton asellus had Glu113 as a counterion but did not exhibit elevated signaling efficiency. Therefore, our results show that vertebrate rhodopsin and L. asellus xenopsin regulate cyclic nucleotide signaling in ciliary cells and displaced the counterion position from Glu181 to Glu113 via convergent evolution, whereas subsequently only vertebrate rhodopsin elevated its signaling efficiency by acquiring the mono-stable property.


Asunto(s)
Opsinas , Rodopsina , Animales , Nucleótidos Cíclicos/metabolismo , Opsinas/genética , Opsinas/metabolismo , Células Fotorreceptoras/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Vertebrados
6.
Int J Mol Sci ; 24(14)2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-37511262

RESUMEN

Establishing apicobasal polarity, involving intricate interactions among polarity regulators, is key for epithelial cell function. Though phosphatase of regenerating liver (PRL) proteins are implicated in diverse biological processes, including cancer, their developmental role remains unclear. In this study, we explore the role of Drosophila PRL (dPRL) in photoreceptor cell development. We reveal that dPRL, requiring a C-terminal prenylation motif, is highly enriched in the apical membrane of developing photoreceptor cells. Moreover, dPRL knockdown during retinal development results in adult Drosophila retinal degeneration, caused by hid-induced apoptosis. dPRL depletion also mislocalizes cell adhesion and polarity proteins like Armadillo, Crumbs, and DaPKC and relocates the basolateral protein, alpha subunit of Na+/K+-ATPase, to the presumed apical membrane. Importantly, this polarity disruption is not secondary to apoptosis, as suppressing hid expression does not rescue the polarity defect in dPRL-depleted photoreceptor cells. These findings underscore dPRL's crucial role in photoreceptor cell polarity and emphasize PRL's importance in establishing epithelial polarity and maintaining cell survival during retinal development, offering new insights into PRL's role in normal epithelium.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Hígado/metabolismo , Polaridad Celular/genética
7.
J Physiol ; 599(17): 4085-4100, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34252195

RESUMEN

KEY POINTS: Although cone and rod photoreceptor cells in the retina have a type of cannabinoid receptor called a CB1 receptor, little is known about how cannabinoids, the active component in marijuana, affect retinal function. Studies have shown that a circadian (24-h) clock in the retina uses dopamine receptors, which are also on photoreceptors, to regulate gap junctions (a type of cell-to-cell communication) between rods and cones, so that they are functional (open) at night but closed in the day. We show that CB1 receptors have opposite effects on rod-cone gap junctions in day and night, decreasing communication in the day when dopamine receptors are active and increasing communication when dopamine receptors are inactive. CB1 and dopamine receptors thus work together to enhance the day/night difference in rod-cone gap junction communication. The increased rod-cone communication at night due to cannabinoid CB1 receptors may help improve night vision. ABSTRACT: Cannabinoid CB1 receptors and dopamine D4 receptors in the brain form receptor complexes that interact but the physiological function of these interactions in intact tissue remains unclear. In vertebrate retina, rods and cones, which are connected by gap junctions, express both CB1 and D4 receptors. Because the retinal circadian clock uses cone D4 receptors to decrease rod-cone gap junction coupling in the day and to increase it at night, we studied whether an interaction between cone CB1 and D4 receptors increases the day/night difference in rod-cone coupling compared to D4 receptors acting alone. Using electrical recording and injections of Neurobiotin tracer into individual cones in intact goldfish retinas, we found that SR141716A (a CB1 receptor antagonist) application alone in the day increased both the extent of rod-cone tracer coupling and rod input to cones, which reaches cones via open gap junctions. Conversely, SR141716A application alone at night or SR141716A application in the day following 30-min spiperone (a D4 receptor antagonist) application decreased both rod-cone tracer coupling and rod input to cones. These results show that endogenous activation of cone CB1 receptors decreases rod-cone coupling in the day when D4 receptors are activated but increases it at night when D4 receptors are not activated. Therefore, the D4 receptor-dependent day/night switch in the effects of CB1 receptor activation results in an enhancement of the day/night difference in rod-cone coupling. This synergistic interaction increases detection of very dim large objects at night and fine spatial details in the day.


Asunto(s)
Cannabinoides , Carpa Dorada , Animales , Cannabinoides/farmacología , Uniones Comunicantes , Receptores de Dopamina D4 , Retina , Células Fotorreceptoras Retinianas Conos
8.
Pflugers Arch ; 473(9): 1469-1491, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33779813

RESUMEN

Light-evoked voltage responses of rod and cone photoreceptor cells in the vertebrate retina must be converted to a train of synaptic vesicle release events for transmission to downstream neurons. This review discusses the processes, proteins, and structures that shape this critical early step in vision, focusing on studies from salamander retina with comparisons to other experimental animals. Many mechanisms are conserved across species. In cones, glutamate release is confined to ribbon release sites although rods are also capable of release at non-ribbon sites. The role of non-ribbon release in rods remains unclear. Release from synaptic ribbons in rods and cones involves at least three vesicle pools: a readily releasable pool (RRP) matching the number of membrane-associated vesicles along the ribbon base, a ribbon reserve pool matching the number of additional vesicles on the ribbon, and an enormous cytoplasmic reserve. Vesicle release increases in parallel with Ca2+ channel activity. While the opening of only a few Ca2+ channels beneath each ribbon can trigger fusion of a single vesicle, sustained release rates in darkness are governed by the rate at which the RRP can be replenished. The number of vacant release sites, their functional status, and the rate of vesicle delivery in turn govern replenishment. Along with an overview of the mechanisms of exocytosis and endocytosis, we consider specific properties of ribbon-associated proteins and pose a number of remaining questions about this first synapse in the visual system.


Asunto(s)
Canales de Calcio/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Animales , Humanos , Estimulación Luminosa/métodos , Retina/metabolismo , Retina/ultraestructura , Células Fotorreceptoras Retinianas Conos/ultraestructura , Células Fotorreceptoras Retinianas Bastones/ultraestructura , Sinapsis/ultraestructura
9.
Pflugers Arch ; 473(9): 1361-1376, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33591421

RESUMEN

Rhodopsin is the light receptor in rod photoreceptor cells that initiates scotopic vision. Studies on the light receptor span well over a century, yet questions about the organization of rhodopsin within the photoreceptor cell membrane still persist and a consensus view on the topic is still elusive. Rhodopsin has been intensely studied for quite some time, and there is a wealth of information to draw from to formulate an organizational picture of the receptor in native membranes. Early experimental evidence in apparent support for a monomeric arrangement of rhodopsin in rod photoreceptor cell membranes is contrasted and reconciled with more recent visual evidence in support of a supramolecular organization of rhodopsin. What is known so far about the determinants of forming a supramolecular structure and possible functional roles for such an organization are also discussed. Many details are still missing on the structural and functional properties of the supramolecular organization of rhodopsin in rod photoreceptor cell membranes. The emerging picture presented here can serve as a springboard towards a more in-depth understanding of the topic.


Asunto(s)
Membrana Celular/química , Células Fotorreceptoras Retinianas Bastones/química , Rodopsina/química , Animales , Membrana Celular/metabolismo , Humanos , Multimerización de Proteína/fisiología , Estructura Secundaria de Proteína , Células Fotorreceptoras Retinianas Bastones/metabolismo , Rodopsina/metabolismo
10.
Proc Natl Acad Sci U S A ; 115(27): E6264-E6273, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29915052

RESUMEN

Retinal detachment (RD) is a sight-threatening complication common in many highly prevalent retinal disorders. RD rapidly leads to photoreceptor cell death beginning within 12 h following detachment. In patients with sustained RD, progressive visual decline due to photoreceptor cell death is common, leading to significant and permanent loss of vision. Microglia are the resident immune cells of the central nervous system, including the retina, and function in the homeostatic maintenance of the neuro-retinal microenvironment. It is known that microglia become activated and change their morphology in retinal diseases. However, the function of activated microglia in RD is incompletely understood, in part because of the lack of microglia-specific markers. Here, using the newly identified microglia marker P2ry12 and microglial depletion strategies, we demonstrate that retinal microglia are rapidly activated in response to RD and migrate into the injured area within 24 h post-RD, where they closely associate with infiltrating macrophages, a population distinct from microglia. Once in the injured photoreceptor layer, activated microglia can be observed to contain autofluorescence within their cell bodies, suggesting they function to phagocytose injured or dying photoreceptors. Depletion of retinal microglia results in increased disease severity and inhibition of macrophage infiltration, suggesting that microglia are involved in regulating neuroinflammation in the retina. Our work identifies that microglia mediate photoreceptor survival in RD and suggests that this effect may be due to microglial regulation of immune cells and photoreceptor phagocytosis.


Asunto(s)
Macrófagos/inmunología , Microglía/inmunología , Células Fotorreceptoras de Vertebrados/inmunología , Receptores Purinérgicos P2Y12/inmunología , Desprendimiento de Retina/inmunología , Animales , Muerte Celular/genética , Muerte Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Macrófagos/patología , Ratones , Ratones Transgénicos , Microglía/patología , Células Fotorreceptoras de Vertebrados/patología , Receptores Purinérgicos P2Y12/genética , Desprendimiento de Retina/genética , Desprendimiento de Retina/patología
11.
J Neurosci ; 39(34): 6798-6810, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31285302

RESUMEN

A major cause of human blindness is the death of rod photoreceptors. As rods degenerate, synaptic structures between rod and rod bipolar cells disappear and the rod bipolar cells extend their dendrites and occasionally make aberrant contacts. Such changes are broadly observed in blinding disorders caused by photoreceptor cell death and are thought to occur in response to deafferentation. How the remodeled retinal circuit affects visual processing following rod rescue is not known. To address this question, we generated male and female transgenic mice wherein a disrupted cGMP-gated channel (CNG) gene can be repaired at the endogenous locus and at different stages of degeneration by tamoxifen-inducible cre-mediated recombination. In normal rods, light-induced closure of CNG channels leads to hyperpolarization of the cell, reducing neurotransmitter release at the synapse. Similarly, rods lacking CNG channels exhibit a resting membrane potential that was ~10 mV hyperpolarized compared to WT rods, indicating diminished glutamate release. Retinas from these mice undergo stereotypic retinal remodeling as a consequence of rod malfunction and degeneration. Upon tamoxifen-induced expression of CNG channels, rods recovered their structure and exhibited normal light responses. Moreover, we show that the adult mouse retina displays a surprising degree of plasticity upon activation of rod input. Wayward bipolar cell dendrites establish contact with rods to support normal synaptic transmission, which is propagated to the retinal ganglion cells. These findings demonstrate remarkable plasticity extending beyond the developmental period and support efforts to repair or replace defective rods in patients blinded by rod degeneration.SIGNIFICANCE STATEMENT Current strategies for treatment of neurodegenerative disorders are focused on the repair of the primary affected cell type. However, the defective neurons function within a complex neural circuitry, which also becomes degraded during disease. It is not known whether rescued neurons and the remodeled circuit will establish communication to regain normal function. We show that the adult mammalian neural retina exhibits a surprising degree of plasticity following rescue of rod photoreceptors. The wayward dendrites of rod bipolar cells re-establish contact with rods to support normal synaptic transmission, which is propagated to the retinal ganglion cells. These findings support efforts to repair or replace defective rods in patients blinded by rod cell loss.


Asunto(s)
Retina/patología , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones , Transducción de Señal/fisiología , Sinapsis/fisiología , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos/fisiología , Electrorretinografía , Humanos , Ratones , Ratones Transgénicos , Plasticidad Neuronal/fisiología , Estimulación Luminosa , Células Fotorreceptoras de Vertebrados/fisiología , Células Bipolares de la Retina/fisiología , Degeneración Retiniana/inducido químicamente , Transmisión Sináptica , Tamoxifeno
12.
J Cell Mol Med ; 24(6): 3346-3358, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32003125

RESUMEN

Our study aimed to evaluate the protective role and mechanisms of bone marrow mesenchymal stem cells (BMSCs) in hypoxic photoreceptors and experimental retinal detachment. The cellular morphology, viability, apoptosis and autophagy of hypoxic 661w cells and cells cocultured with BMSCs were analysed. In retinal detachment model, BMSCs were intraocularly transplanted, and then, the retinal morphology, outer nuclear layer (ONL) thickness and rhodopsin expression were studied as well as apoptosis and autophagy of the retinal cells. The hypoxia-induced apoptosis of 661w cells obviously increased together with autophagy levels increasing and peaking at 8 hours after hypoxia. Upon coculturing with BMSCs, hypoxic 661w cells had a better morphology and fewer apoptosis. After autophagy was inhibited, the apoptotic 661w cells under the hypoxia increased, and the cell viability was reduced, even in the presence of transplanted BMSCs. In retina-detached eyes transplanted with BMSCs, the retinal ONL thickness was closer to that of the normal retina. After transplantation, apoptosis decreased significantly and retinal autophagy was activated in the BMSC-treated retinas. Increased autophagy in the early stage could facilitate the survival of 661w cells under hypoxic stress. Coculturing with BMSCs protects 661w cells from hypoxic damage, possibly due to autophagy activation. In retinal detachment models, BMSC transplantation can significantly reduce photoreceptor cell death and preserve retinal structure. The capacity of BMSCs to reduce retinal cell apoptosis and to initiate autophagy shortly after transplantation may facilitate the survival of retinal cells in the low-oxygen and nutrition-restricted milieu after retinal detachment.


Asunto(s)
Hipoxia de la Célula/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Fotorreceptoras Retinianas Conos/citología , Desprendimiento de Retina/patología , Animales , Apoptosis/fisiología , Autofagia/fisiología , Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Línea Celular , Supervivencia Celular , Técnicas de Cocultivo , Femenino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratas , Ratas Wistar , Desprendimiento de Retina/terapia , Rodopsina/biosíntesis
13.
Dev Biol ; 445(2): 245-255, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30502325

RESUMEN

The ascidian larva has a pigmented ocellus comprised of a cup-shaped array of approximately 30 photoreceptor cells, a pigment cell, and three lens cells. Morphological, physiological and molecular evidence has suggested evolutionary kinship between the ascidian larval photoreceptors and vertebrate retinal and/or pineal photoreceptors. Rx, an essential factor for vertebrate photoreceptor development, has also been suggested to be involved in the development of the ascidian photoreceptor cells, but a recent revision of the photoreceptor cell lineage raised a crucial discrepancy between the reported expression patterns of Rx and the cell lineage. Here, we report spatio-temporal expression patterns of Rx at single-cell resolution along with mitotic patterns up to the final division of the photoreceptor-lineage cells in Ciona. The expression of Rx commences in non-photoreceptor a-lineage cells on the right side of the anterior sensory vesicle at the early tailbud stage. At the mid tailbud stage, Rx begins to be expressed in the A-lineage photoreceptor cell progenitors located on the right side of the posterior sensory vesicle. Thus, Rx is specifically but not exclusively expressed in the photoreceptor-lineage cells in the ascidian embryo. Two cis-regulatory modules are shown to be important for the photoreceptor-lineage expression of Rx. The cell division patterns of the photoreceptor-lineage cells rationally explain the generation of the cup-shaped structure of the pigmented ocellus. The present findings demonstrate the complete cell lineage of the ocellus photoreceptor cells and provide a framework elucidating the molecular and cellular mechanisms of photoreceptor development in Ciona.


Asunto(s)
Ciona intestinalis/crecimiento & desarrollo , Ciona intestinalis/genética , Proteínas de Homeodominio/genética , Células Fotorreceptoras de Invertebrados/citología , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Sitios de Unión/genética , Linaje de la Célula/genética , Ciona intestinalis/citología , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Larva/citología , Larva/genética , Larva/crecimiento & desarrollo , Mitosis/genética , Secuencias Reguladoras de Ácido Ribonucleico , Análisis Espacio-Temporal
14.
Biochem Biophys Res Commun ; 524(3): 542-548, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32014251

RESUMEN

ES1 homologs are conserved among prokaryotes and eukaryotes, and the gene expression of ES1 homologs has been confirmed in diverse mammalian tissues. However, the localization and function of mammalian ES1 proteins remain poorly understood. ES1 protein was found specifically expressed in the cone cells of zebrafish and was proposed to contribute to the formation of mega mitochondria. We also observed mega mitochondria in the cone cells of porcine retinas, which raised the question regarding the localization of the porcine ES1. Therefore, in the present study, we aimed to determine the localization of ES1 in porcine retinas. We prepared a rabbit polyclonal antibody against the ES1 C-terminal and performed western blotting analysis and immunoelectron microscopy. The ES1 was found to be localized mainly in the mitochondrial intermembrane space of the porcine retinal cells. Immunopositive signals for ES1 were observed in the mitochondria of almost all retinal cells, and not specifically in cone cells. Our results and the ES1 sequences indicated that the glyoxalase III activity of ES1 might contribute to the stable functionality of the active mitochondria in a protective manner.


Asunto(s)
Proteínas del Ojo/metabolismo , Membranas Mitocondriales/metabolismo , Retina/citología , Homología de Secuencia de Aminoácido , Porcinos/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas del Ojo/química , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Células Fotorreceptoras de Vertebrados/citología , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/ultraestructura , Retina/ultraestructura , Solubilidad
15.
Exp Eye Res ; 199: 108166, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32771499

RESUMEN

Inherited retinal degenerative disorders such as retinitis pigmentosa and Usher syndrome are characterized by progressive death of photoreceptor cells. To restore vision to patients blinded by these diseases, a stem cell-based photoreceptor cell replacement strategy will likely be required. Although retinal stem cell differentiation protocols suitable for generating photoreceptor cells exist, they often yield a rather heterogenous mixture of cell types. To enrich the donor cell population for one or a few cell types, scientists have traditionally relied upon the use of antibody-based selection approaches. However, these strategies are quite labor intensive and require animal derived reagents and equipment that are not well suited to current good manufacturing practices (cGMP). The purpose of this study was to develop and evaluate a microfluidic cell sorting device capable of exploiting the physical and mechanical differences between retinal cell types to enrich specific donor cell populations such as Retinal Pigment Epithelial (RPE) cells and photoreceptor cells. Using this device, we were able to separate a mixture of RPE and iPSC-derived photoreceptor precursor cell lines into two substantially enriched fractions. The enrichment factor of the RPE fraction was 2 and that of the photoreceptor precursor cell fraction was 2.7. Similarly, when human retina, obtained from 3 independent donors, was dissociated and passed through the sorting device, the heterogeneous mixture could be reliably sorted into RPE and photoreceptor cell rich fractions. In summary, microfluidic cell sorting is a promising approach for antibody free enrichment of retinal cell populations.


Asunto(s)
Microfluídica/métodos , Células Fotorreceptoras/patología , Degeneración Retiniana/diagnóstico , Epitelio Pigmentado de la Retina/patología , Animales , Diferenciación Celular , Línea Celular , Humanos , Microscopía de Fuerza Atómica , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
16.
Proc Natl Acad Sci U S A ; 114(20): 5259-5264, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28468800

RESUMEN

Retinitis pigmentosa (RP) is an inherited neurodegenerative disease, in which the death of mutant rod photoreceptors leads secondarily to the non-cell autonomous death of cone photoreceptors. Gene therapy is a promising treatment strategy. Unfortunately, current methods of gene delivery treat only a fraction of diseased cells, yielding retinas that are a mosaic of treated and untreated rods, as well as cones. In this study, we created two RP mouse models to test whether dying, untreated rods negatively impact treated, rescued rods. In one model, treated and untreated rods were segregated. In the second model, treated and untreated rods were diffusely intermixed, and their ratio was controlled to achieve low-, medium-, or high-efficiency rescue. Analysis of these mosaic retinas demonstrated that rescued rods (and cones) survive, even when they are greatly outnumbered by dying photoreceptors. On the other hand, the rescued photoreceptors did exhibit long-term defects in their outer segments (OSs), which were less severe when more photoreceptors were treated. In summary, our study suggests that even low-efficiency gene therapy may achieve stable survival of rescued photoreceptors in RP patients, albeit with OS dysgenesis.


Asunto(s)
Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Animales , Muerte Celular , Modelos Animales de Enfermedad , Terapia Genética/métodos , Ratones , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Rodopsina/metabolismo
17.
Proc Natl Acad Sci U S A ; 114(21): 5437-5442, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28484015

RESUMEN

Most vertebrate retinas contain a single type of rod for scotopic vision and multiple types of cones for photopic and color vision. The retinas of certain amphibian species uniquely contain two types of rods: red rods, which express rhodopsin, and green rods, which express a blue-sensitive cone pigment (M1/SWS2 group). Spontaneous activation of rhodopsin induced by thermal isomerization of the retinal chromophore has been suggested to contribute to the rod's background noise, which limits the visual threshold for scotopic vision. Therefore, rhodopsin must exhibit low thermal isomerization rate compared with cone visual pigments to adapt to scotopic condition. In this study, we determined whether amphibian blue-sensitive cone pigments in green rods exhibit low thermal isomerization rates to act as rhodopsin-like pigments for scotopic vision. Anura blue-sensitive cone pigments exhibit low thermal isomerization rates similar to rhodopsin, whereas Urodela pigments exhibit high rates like other vertebrate cone pigments present in cones. Furthermore, by mutational analysis, we identified a key amino acid residue, Thr47, that is responsible for the low thermal isomerization rates of Anura blue-sensitive cone pigments. These results strongly suggest that, through this mutation, anurans acquired special blue-sensitive cone pigments in their green rods, which could form the molecular basis for scotopic color vision with normal red rods containing green-sensitive rhodopsin.


Asunto(s)
Ambystoma mexicanum/fisiología , Visión de Colores , Visión Nocturna , Opsinas/química , Xenopus/fisiología , Adaptación Biológica , Sustitución de Aminoácidos , Animales , Evolución Molecular , Opsinas/genética
18.
Zhonghua Yan Ke Za Zhi ; 56(2): 149-154, 2020 Feb 11.
Artículo en Zh | MEDLINE | ID: mdl-32074826

RESUMEN

The outer retinal tubular structure (ORT) indicates a round or oval-shaped structure with a highly reflective border and a relatively low reflective lumen on spectrum-domain coherence tomography. ORTs are located in the outer nuclear layer accompanied by disruption and curling of the external limiting membrane and retinal pigment epithelial atrophy. Histopathological researches have revealed that ORTs are some kind of remodeling of the outer retina especially photoreceptors under pathological conditions, representing an advanced damage to the outer retina and a common final pathway of various retinal degenerative diseases. ORTs are common in neovascular age-related macular degeneration. They show some similarities in morphology with inter retinal fluid or cystoid edema on spectrum-domain coherence tomography, but they are not an indication for neovascular activity and retreatment. Their response to anti-vascular endothelial growth factor is poor, and the visual prognosis is not optimistic. Therefore, raising awareness of ORTs is very important for guiding clinical treatment and judging prognosis. (Chin J Ophthalmol, 2020, 56: 149-154).


Asunto(s)
Degeneración Retiniana , Tomografía de Coherencia Óptica , Humanos , Células Fotorreceptoras de Vertebrados , Degeneración Retiniana/diagnóstico por imagen , Degeneración Retiniana/patología , Estudios Retrospectivos
19.
Neurobiol Dis ; 121: 1-16, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30218757

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder characterized by accumulation of misfolded α-synuclein within the central nervous system (CNS). Visual problems in PD patients are common, although retinal pathology associated with PD is not well understood. The purpose of this study was to investigate retinal pathology in a transgenic mouse model (TgM83) expressing the human A53T α-synuclein mutation and assess the effect of α-synuclein "seeding" on the development of retinal pathology. Two-month-old TgM83 mice were intracerebrally inoculated with brain homogenate from old (12-18 months) TgM83 mice. Retinas were then analyzed at 5 months of age. We analyzed retinas from 5-month-old and 8-month-old uninoculated healthy TgM83 mice, and old (12-18 months) mice that were euthanized following the development of clinical signs. Retinas of B6C3H mice (genetic background of the TgM83 mouse) served as control. We used immunohistochemistry and western blot analysis to detect accumulation of α-synuclein, pTauThr231, inflammation, changes in macroautophagy, and cell death. Raman spectroscopy was used to test the potential to differentiate between retinal tissues of healthy mice and diseased mice. This work demonstrates retinal changes associated with the A53T mutation. Retinas of non-inoculated TgM83 mice had accumulation of α-synuclein, "pre-tangle" tau, activation of retinal glial cells, and photoreceptor cell loss by 8 months of age. The development of these changes is accelerated by inoculation with brain homogenate from clinically ill TgM83 mice. Compared to non-inoculated 5-month-old TgM83 mice, retinas of inoculated 5-month-old mice had increased accumulation of α-synuclein (pSer129) and pTauThr231 proteins, upregulated microglial activation, and dysregulated macroautophagy. Raman spectroscopic analysis was able to discriminate between healthy and diseased mice. This study describes retinal pathology resulting from the A53T mutation. We show that seeding with brain homogenates from old TgM83 mice accelerates retinal pathology. We demonstrate that Raman spectroscopy can be used to accurately identify a diseased retina based on its biochemical profile, and that α-synuclein accumulation may contribute to accumulation of pTauThr231 proteins, neuroinflammation, metabolic dysregulation, and photoreceptor cell death. Our work provides insight into retinal changes associated with Parkinson's disease, and may contribute to a better understanding of visual symptoms experienced by patients.


Asunto(s)
Autofagia , Encefalitis/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Retina/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Animales , Modelos Animales de Enfermedad , Encefalitis/complicaciones , Ratones Transgénicos , Neuroglía/metabolismo , Enfermedad de Parkinson/complicaciones , Fosforilación , Retina/patología
20.
J Membr Biol ; 252(4-5): 413-423, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31286171

RESUMEN

Rhodopsin is the light receptor in photoreceptor cells of the retina and a prototypical G protein-coupled receptor. Two types of quaternary structures can be adopted by rhodopsin. If rhodopsin folds and attains a proper tertiary structure, it can then form oligomers and nanodomains within the photoreceptor cell membrane. In contrast, if rhodopsin misfolds, it cannot progress through the biosynthetic pathway and instead will form aggregates that can cause retinal degenerative disease. In this review, emerging views are highlighted on the supramolecular organization of rhodopsin within the membrane of photoreceptor cells and the aggregation of rhodopsin that can lead to retinal degeneration.


Asunto(s)
Membrana Celular , Células Fotorreceptoras de Vertebrados , Pliegue de Proteína , Degeneración Retiniana , Rodopsina , Animales , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/patología , Humanos , Células Fotorreceptoras de Vertebrados/química , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Dominios Proteicos , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Rodopsina/química , Rodopsina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA