Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 80(8): 214, 2023 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-37466729

RESUMEN

Mutations in the photoreceptor-specific tetraspanin gene peripherin-2 (PRPH2) lead to widely varying forms of retinal degeneration ranging from retinitis pigmentosa to macular dystrophy. Both inter- and intra-familial phenotypic heterogeneity has led to much interest in uncovering the complex pathogenic mechanisms of PRPH2-associated disease. Majority of disease-causing mutations in PRPH2 reside in the second intradiscal loop, wherein seven cysteines control protein folding and oligomerization. Here, we utilize knockin models to evaluate the role of three D2 loop cysteine mutants (Y141C, C213Y and C150S), alone or in combination. We elucidated how these mutations affect PRPH2 properties, including oligomerization and subcellular localization, and contribute to disease processes. Results from our structural, functional and molecular studies revealed that, in contrast to our understanding from prior investigations, rods are highly affected by PRPH2 mutations interfering with oligomerization and not merely by the haploinsufficiency associated with these mutations. On the other hand, cones are less affected by the toxicity of the mutant protein and significantly reduced protein levels, suggesting that knockdown therapeutic strategies may sustain cone functionality for a longer period. This observation provides useful data to guide and simplify the current development of effective therapeutic approaches for PRPH2-associated diseases that combine knockdown with high levels of gene supplementation needed to generate prolonged rod improvement.


Asunto(s)
Degeneración Macular , Degeneración Retiniana , Retinitis Pigmentosa , Humanos , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/patología , Retinitis Pigmentosa/metabolismo , Degeneración Macular/patología , Tetraspaninas/metabolismo , Mutación/genética
2.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33526685

RESUMEN

We previously reported a model of progressive retinal degeneration resulting from the knockout of the retina-specific riboflavin binding protein, retbindin (Rtbdn-/- ). We also demonstrated a reduction in neural retinal flavins as a result of the elimination of RTBDN. Given the role of flavins in metabolism, herein we investigated the underlying mechanism of this retinal degeneration by performing metabolomic analyses on predegeneration at postnatal day (P) 45 and at the onset of functional degeneration in the P120 retinas. Metabolomics of hydrophilic metabolites revealed that individual glycolytic products accumulated in the P45 Rtbdn-/- neural retinas along with the elevation of pentose phosphate pathway, while TCA cycle intermediates remained unchanged. This was confirmed by using 13C-labeled flux measurements and immunoblotting, revealing that the key regulatory step of phosphoenolpyruvate to pyruvate was inhibited via down-regulation of the tetrameric pyruvate kinase M2 (PKM2). Separate metabolite assessments revealed that almost all intermediates of acylcarnitine fatty acid oxidation, ceramides, sphingomyelins, and multiple toxic metabolites were significantly elevated in the predegeneration Rtbdn-/- neural retina. Our data show that lack of RTBDN, and hence reduction in flavins, forced the neural retina into repurposing glucose for free-radical mitigation over ATP production. However, such sustained metabolic reprogramming resulted in an eventual metabolic collapse leading to neurodegeneration.


Asunto(s)
Proteínas del Ojo/genética , Piruvato Quinasa/genética , Retina/metabolismo , Degeneración Retiniana/genética , Animales , Ciclo del Ácido Cítrico/genética , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Flavinas/metabolismo , Glucólisis/genética , Homeostasis , Humanos , Ratones , Piruvato Quinasa/metabolismo , Retina/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología
3.
FASEB J ; 36(5): e22284, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35344225

RESUMEN

Prph2 is a photoreceptor-specific tetraspanin with an essential role in the structure and function of photoreceptor outer segments. PRPH2 mutations cause a multitude of retinal diseases characterized by the degeneration of photoreceptors as well as defects in neighboring tissues such as the RPE. While extensive research has analyzed photoreceptors, less attention has been paid to these secondary defects. Here, we use different Prph2 disease models to evaluate the damage of the RPE arising from photoreceptor defects. In Prph2 disease models, the RPE exhibits structural abnormalities and cell loss. Furthermore, RPE functional defects are observed, including impaired clearance of phagocytosed outer segment material and increased microglia activation. The severity of RPE damage is different between models, suggesting that the different abnormal outer segment structures caused by Prph2 disease mutations lead to varying degrees of RPE stress and thus influence the clinical phenotype observed in patients.


Asunto(s)
Periferinas , Enfermedades de la Retina , Tetraspaninas , Humanos , Mutación , Periferinas/genética , Células Fotorreceptoras , Enfermedades de la Retina/genética , Epitelio Pigmentado de la Retina , Tetraspaninas/genética
4.
Adv Exp Med Biol ; 1415: 277-281, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37440045

RESUMEN

The light-sensitive outer segment organelle of photoreceptor cells contains a stack of hundreds of flat, disc-shaped membranes called discs. The rims of these discs contain a photoreceptor-specific tetraspanin protein peripherin-2 (also known as rds or PRPH2). Mutations in the PRPH2 gene lead to a wide variety of inherited retinal degenerations in humans. The vast majority of these mutations occur within a large, intradiscal loop of peripherin-2, known as the D2 loop. The D2 loop mediates well-established intermolecular interactions of peripherin-2 molecules among themselves and a homologous protein ROM1. These interactions lead to the formation of large, highly ordered oligomers. In this chapter, we discuss the supramolecular organization of peripherin-2/ROM1 complexes and their contribution to the process of outer segment disc morphogenesis and enclosure.


Asunto(s)
Degeneración Retiniana , Tetraspaninas , Humanos , Periferinas/genética , Tetraspaninas/genética , Degeneración Retiniana/genética , Mutación , Morfogénesis , Proteínas del Ojo/genética
5.
Proc Natl Acad Sci U S A ; 117(34): 20615-20624, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32778589

RESUMEN

Trafficking of photoreceptor membrane proteins from their site of synthesis in the inner segment (IS) to the outer segment (OS) is critical for photoreceptor function and vision. Here we evaluate the role of syntaxin 3 (STX3), in trafficking of OS membrane proteins such as peripherin 2 (PRPH2) and rhodopsin. Photoreceptor-specific Stx3 knockouts [Stx3f/f(iCre75) and Stx3f/f(CRX-Cre) ] exhibited rapid, early-onset photoreceptor degeneration and functional decline characterized by structural defects in IS, OS, and synaptic terminals. Critically, in the absence of STX3, OS proteins such as PRPH2, the PRPH2 binding partner, rod outer segment membrane protein 1 (ROM1), and rhodopsin were mislocalized along the microtubules to the IS, cell body, and synaptic region. We find that the PRPH2 C-terminal domain interacts with STX3 as well as other photoreceptor SNAREs, and our findings indicate that STX3 is an essential part of the trafficking pathway for both disc (rhodopsin) and rim (PRPH2/ROM1) components of the OS.


Asunto(s)
Periferinas/metabolismo , Proteínas Qa-SNARE/metabolismo , Segmento Interno de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Rodopsina/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Ratones , Células Fotorreceptoras de Vertebrados/fisiología , Transporte de Proteínas , Proteínas Qa-SNARE/genética , Segmento Interno de las Células Fotorreceptoras Retinianas/ultraestructura , Segmento Externo de las Células Fotorreceptoras Retinianas/ultraestructura , Proteínas SNARE/metabolismo
6.
J Neurosci ; 41(16): 3588-3596, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33707293

RESUMEN

Mutations in the PRPH2 gene encoding the photoreceptor-specific protein PRPH2 (also known as peripherin-2 or rds) cause a broad range of autosomal dominant retinal diseases. Most of these mutations affect the structure of the light-sensitive photoreceptor outer segment, which is composed of a stack of flattened "disc" membranes surrounded by the plasma membrane. The outer segment is renewed on a daily basis in a process whereby new discs are added at the outer segment base and old discs are shed at the outer segment tip. New discs are formed as serial membrane evaginations, which eventually enclose through a complex process of membrane remodeling (completely in rods and partially in cones). As disc enclosure proceeds, PRPH2 localizes to the rims of enclosed discs where it forms oligomers which fortify the highly curved membrane structure of these rims. In this study, we analyzed the outer segment phenotypes of mice of both sexes bearing a single copy of either the C150S or the Y141C PRPH2 mutation known to prevent or increase the degree of PRPH2 oligomerization, respectively. Strikingly, both mutations increased the number of newly forming, not-yet-enclosed discs, indicating that the precision of disc enclosure is regulated by PRPH2 oligomerization. Without tightly controlled enclosure, discs occasionally over-elongate and form large membranous "whorls" instead of disc stacks. These data show that the defects in outer segment structure arising from abnormal PRPH2 oligomerization are manifested at the stage of disc enclosure.SIGNIFICANCE STATEMENT The light-sensitive photoreceptor outer segment contains a stack of flattened "disc" membranes that are surrounded, or "enclosed," by the outer segment membrane. Disc enclosure is an adaptation increasing photoreceptor light sensitivity by facilitating the diffusion of the second messenger along the outer segment axes. However, the molecular mechanisms by which photoreceptor discs enclose within the outer segment membrane remain poorly understood. We now demonstrate that oligomers of the photoreceptor-specific protein peripherin-2, or PRPH2, play an active role in this process. We further propose that defects in disc enclosure because of abnormal PRPH2 oligomerization result in major structural abnormalities of the outer segment, ultimately leading to loss of visual function and cell degeneration in PRPH2 mutant models and human patients.


Asunto(s)
Periferinas/fisiología , Células Fotorreceptoras de Vertebrados/fisiología , Animales , Membrana Celular/genética , Membrana Celular/ultraestructura , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Células Fotorreceptoras de Vertebrados/ultraestructura , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Segmento Externo de la Célula en Bastón/ultraestructura
7.
Hum Mol Genet ; 29(16): 2708-2722, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32716032

RESUMEN

Peripherin 2 (PRPH2) is a retina-specific tetraspanin protein essential for the formation of rod and cone photoreceptor outer segments (OS). Patients with mutations in PRPH2 exhibit severe retinal degeneration characterized by vast inter- and intra-familial phenotypic heterogeneity. To help understand contributors to this within-mutation disease variability, we asked whether the PRPH2 binding partner rod OS membrane protein 1 (ROM1) could serve as a phenotypic modifier. We utilized knockin and transgenic mouse models to evaluate the structural, functional and biochemical effects of eliminating one allele of Rom1 (Rom1+/-) in three different Prph2 models which mimic human disease: C213Y Prph2 (Prph2C/+), K153Del Prph2 (Prph2K/+) and R172W (Prph2R172W). Reducing Rom1 in the absence of Prph2 mutations (Rom1+/-) had no effect on retinal structure or function. However, the effects of reducing Rom1 in the presence of Prph2 mutations were highly variable. Prph2K/+/Rom1+/- mice had improved rod and cone function compared with Prph2K/+ as well as amelioration of K153Del-associated defects in PRPH2/ROM1 oligomerization. In contrast, Prph2R172W/Rom1+/- animals had worsened rod and cone function and exacerbated retinal degeneration compared with Prph2R172W animals. Removing one allele of Rom1 had no effect in Prph2C/+. Combined, our findings support a role for non-pathogenic ROM1 null variants in contributing to phenotypic variability in mutant PRPH2-associated retinal degeneration. Since the effects of Rom1 reduction are variable, our data suggest that this contribution is specific to the type of Prph2 mutation.


Asunto(s)
Proteínas del Ojo/genética , Periferinas/genética , Degeneración Retiniana/genética , Tetraspaninas/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Mutación/genética , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/patología
8.
FASEB J ; 34(1): 1211-1230, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914632

RESUMEN

Mutations in peripherin 2 (PRPH2) have been associated with retinitis pigmentosa (RP) and macular/pattern dystrophies, but the origin of this phenotypic variability is unclear. The majority of Prph2 mutations are located in the large intradiscal loop (D2), a region that contains seven cysteines involved in intra- and intermolecular disulfide bonding and protein folding. A mutation at cysteine 213, which is engaged in an intramolecular disulfide bond, leads to butterfly-shaped pattern dystrophy in humans, in sharp contrast to mutations in the adjacent cysteine at position 214 which result in RP. To help understand this unexpected phenotypic variability, we generated a knockin mouse line carrying the C213Y disease mutation. The mutant Prph2 protein lost the ability to oligomerize with rod outer segment membrane protein 1 (Rom1), but retained the ability to form homotetramers. C213Y heterozygotes had significantly decreased overall Prph2 levels as well as decreased rod and cone function. Critically, supplementation with extra wild-type Prph2 protein elicited improvements in Prph2 protein levels and rod outer segment structure, but not functional rescue in rods or cones. These findings suggest that not all interruptions of D2 loop intramolecular disulfide bonding lead to haploinsufficiency-related RP, but rather that more subtle changes can lead to mutant proteins stable enough to exert gain-of-function defects in rods and cones. This outcome highlights the difficulty in targeting Prph2-associated gain-of-function disease and suggests that elimination of the mutant protein will be a pre-requisite for any curative therapeutic strategy.


Asunto(s)
Degeneración Macular , Mutación Missense , Periferinas , Células Fotorreceptoras Retinianas Conos , Células Fotorreceptoras Retinianas Bastones , Retinitis Pigmentosa , Sustitución de Aminoácidos , Animales , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Humanos , Degeneración Macular/genética , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones , Ratones Transgénicos , Periferinas/genética , Periferinas/metabolismo , Multimerización de Proteína , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Tetraspaninas/genética , Tetraspaninas/metabolismo
9.
Hum Mol Genet ; 27(20): 3507-3518, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29961824

RESUMEN

Mutations in peripherin 2 (PRPH2, also known as Rds), a tetraspanin protein found in photoreceptor outer segments (OSs), cause retinal degeneration ranging from rod-dominant retinitis pigmentosa (RP) to cone-dominant macular dystrophy (MD). Understanding why some Prph2 mutants affect rods while others affect cones remains a critical unanswered question. Prph2 is essential for OS structure and function and exhibits a very specific pattern of oligomerization with its homolog Rom1. Non-covalent Prph2/Rom1 homo- and hetero-tetramers assemble into higher-order covalently linked complexes held together by an intermolecular disulfide bond at Prph2-C150/Rom1-C153. Here we disrupt this crucial bond using a C150S-Prph2 knockin mouse line to study the role of Prph2 higher-order complex formation. We find that C150S-Prph2 traffics to the OS, interacts with Rom1 and forms non-covalent tetramers, but alone cannot support normal OS structure and function. However, C150S-Prph2 supports the initiation or elaboration of OS disc structures, and improves rod OS ultrastructure in the presence of wild-type (WT) Prph2 (i.e. Prph2C150S/+ versus Prph2+/-). Prph2C150S/+ animals exhibit haploinsufficiency in rods, but a dominant-negative phenotype in cones, suggesting cones have a different requirement for large Prph2 complexes than rods. Importantly, cone but not rod function can be improved by the addition of one Prph2Y141C allele, a mutation responsible for pattern dystrophy owing to the extra cysteine. Combined these findings show that covalently linked Prph2 complexes are essential for OS formation, but not for Prph2 targeting to the OS, and that cones are especially sensitive to having a broad distribution of Prph2 complex types (i.e. tetramers and large complexes).


Asunto(s)
Proteínas del Ojo/metabolismo , Proteínas de la Membrana/metabolismo , Mutación , Periferinas/metabolismo , Multimerización de Proteína , Degeneración Retiniana/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Periferinas/genética , Dominios y Motivos de Interacción de Proteínas , Degeneración Retiniana/genética , Degeneración Retiniana/fisiopatología , Segmento Externo de las Células Fotorreceptoras Retinianas/fisiología , Tetraspaninas
10.
Int J Mol Sci ; 21(21)2020 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-33138244

RESUMEN

The large number of inherited retinal disease genes (IRD), including the photopigment rhodopsin and the photoreceptor outer segment (OS) structural component peripherin 2 (PRPH2), has prompted interest in identifying common cellular mechanisms involved in degeneration. Although metabolic dysregulation has been shown to play an important role in the progression of the disease etiology, identifying a common regulator that can preserve the metabolic ecosystem is needed for future development of neuroprotective treatments. Here, we investigated whether retbindin (RTBDN), a rod-specific protein with riboflavin binding capability, and a regulator of riboflavin-derived cofactors flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), is protective to the retina in different IRD models; one carrying the P23H mutation in rhodopsin (which causes retinitis pigmentosa) and one carrying the Y141C mutation in Prph2 (which causes a blended cone-rod dystrophy). RTBDN levels are significantly upregulated in both the rhodopsin (Rho)P23H/+ and Prph2Y141C/+ retinas. Rod and cone structural and functional degeneration worsened in models lacking RTBDN. In addition, removing Rtbdn worsened other phenotypes, such as fundus flecking. Retinal flavin levels were reduced in RhoP23H/+/Rtbdn-/- and Prph2Y141C/+/Rtbdn-/- retinas. Overall, these findings suggest that RTBDN may play a protective role during retinal degenerations that occur at varying rates and due to varying disease mechanisms.


Asunto(s)
Proteínas del Ojo/fisiología , Mutación , Periferinas/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/patología , Degeneración Retiniana/patología , Proteínas de Unión al GTP rho/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Periferinas/genética , Retina/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Proteínas de Unión al GTP rho/genética
12.
Adv Exp Med Biol ; 1185: 335-339, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31884634

RESUMEN

Diabetic retinopathy (DR) is a multifaceted disease, combining the deleterious effects of hyperglycemia and the propensity for accumulation of reactive oxygen species. Studies indicate that auto-oxidation of glucose, reduced antioxidant activity, and metabolic aberrations contribute to the pathogenesis of DR. These abnormalities stem from a fundamental imbalance between ROSs and antioxidant scavengers. To correct this imbalance and downstream effects, we propose that superoxide dismutase 3 (SOD3) is a viable therapeutic target for DR.


Asunto(s)
Retinopatía Diabética/fisiopatología , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Antioxidantes/metabolismo , Diabetes Mellitus , Humanos , Especies Reactivas de Oxígeno/metabolismo
13.
Adv Exp Med Biol ; 1185: 495-499, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31884660

RESUMEN

Peripherin 2 (also known as RDS/Prph2) is localized to the rims of rod and cone outer segment (OS) discs. The C-terminus of Prph2 is a critical functional domain, but its exact role is still unknown. In this mini review, we describe work on the Prph2 C-terminus, highlighting its role as a regulator of protein trafficking, membrane curvature, ectosome secretion, and membrane fusion. Evidence supports a role for the Prph2 C-terminus in these processes and demonstrates that it is necessary for the initiation of OS morphogenesis.


Asunto(s)
Periferinas/fisiología , Segmento Externo de las Células Fotorreceptoras Retinianas/fisiología , Segmento Externo de la Célula en Bastón/fisiología , Humanos , Morfogénesis , Transporte de Proteínas , Retina/crecimiento & desarrollo
14.
Adv Exp Med Biol ; 1185: 575-579, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31884673

RESUMEN

The retina and RPE together constitute the most metabolically active ecosystem in the body, harboring high levels of flavins. Although diabetic patients have been reported to suffer from riboflavin deficiency and use of flavins as nutritional interventions to combat diabetic insult on other tissues have been investigated, such attempts have never been tested for the retina to avoid diabetic retinopathy. Furthermore, the role of flavins in pathophysiology of the retina and RPE has mostly been overlooked. Herein, we review the impact of flavins on various clinical manifestations of diabetic retinopathy and discuss possible ways to address them.


Asunto(s)
Retinopatía Diabética/fisiopatología , Flavinas/fisiología , Epitelio Pigmentado de la Retina/patología , Diabetes Mellitus , Humanos
15.
J Biol Chem ; 292(51): 21023-21034, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29079576

RESUMEN

The interface between the neural retina and the retinal pigment epithelium (RPE) is critical for several processes, including visual pigment regeneration and retinal attachment to the RPE. One of its most important functions is the exchange of metabolites between the photoreceptors and RPE because photoreceptor cells have very high energy demands, largely satisfied by oxidative metabolism. The riboflavin (RF) cofactors, flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN), are two key cofactors involved in oxidative metabolism. We have previously shown that retbindin is a photoreceptor-specific RF-binding protein exclusively expressed in the rods and present in the interphotoreceptor matrix at the interface between the RPE and photoreceptor outer segments. Here, we show that retbindin ablation in mice causes a retinal phenotype characterized by time- and dose-dependent declines in rod and cone photoreceptor functions as early as 120 days of age. Whereas minor retinal ultrastructural defects were observed at all ages examined, a significant decline occurred in photoreceptor nuclei at 240 days of age (∼36.8% rods and ∼19.9% cones). Interestingly, significant reductions in FAD and FMN levels were observed before the onset of degeneration (∼46.1% FAD and ∼45% FMN). These findings suggest that the reduced levels of these flavins result in the disruption of intracellular mechanisms, leading to photoreceptor cell death. Altogether, our results suggest that retbindin is a key player in the acquisition and retention of flavins in the neural retina, warranting future investigation into retbindin's role in photoreceptor cell death in models of retinal degenerative disorders.


Asunto(s)
Proteínas del Ojo/metabolismo , Flavinas/metabolismo , Degeneración Retiniana/etiología , Animales , Proteínas del Ojo/antagonistas & inhibidores , Proteínas del Ojo/genética , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Retina/metabolismo , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
16.
Adv Exp Med Biol ; 1074: 275-280, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721953

RESUMEN

Fibulins 2 and 5 are part of a seven-member family of proteins integral to the retinal extracellular matrix. Our study aimed to further explore the roles of both fibulins in retinal function. We obtained knockout mouse models of both fibulins and performed immunohistochemistry, electroretinography, and histology to investigate the outcome of eliminating these proteins. Immunohistochemical analysis showed that both fibulins are localized to the RPE, choroid, and Bruch's membrane. Functional testing showed a significantly reduced scotopic A response at 1 month of age, when compared to their wild-type counterpart. This functional reduction remained constant throughout the age of the animal and only declined as a result of normal aging. The functional decline was associated with reduced number of photoreceptor cells. The results presented clearly demonstrate that fibulins 2 and 5, as extracellular proteins, are necessary for normal retinal development.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Proteínas de la Matriz Extracelular/fisiología , Proteínas del Ojo/fisiología , Degeneración Macular/metabolismo , Retina/crecimiento & desarrollo , Envejecimiento/patología , Animales , Lámina Basal de la Coroides/química , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Coroides/química , Electrorretinografía , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Proteínas del Ojo/genética , Degeneración Macular/genética , Ratones , Ratones Noqueados , Visión Nocturna , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Proteínas Recombinantes/genética , Retina/metabolismo , Retina/fisiopatología , Epitelio Pigmentado de la Retina/química
17.
Adv Exp Med Biol ; 1074: 485-490, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721980

RESUMEN

Retbindin (Rtbdn) is a novel protein of unknown function found exclusively in the retina. Recently, our group has suggested, from in silico analysis of the peptide sequence and in vitro binding data, that Rtbdn could function to bind riboflavin (RF) and its derivatives flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN), collectively known as flavins. Here we confirm that Rtbdn is capable of flavin binding and that this characteristic can protect photoreceptors from flavin-sensitized light damage.


Asunto(s)
Proteínas del Ojo/farmacología , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Animales , Células COS , Muerte Celular , Línea Celular Tumoral , Chlorocebus aethiops , Medios de Cultivo Condicionados , Humanos , Ratones , Fármacos Sensibilizantes a Radiaciones/farmacología , Proteínas Recombinantes/farmacología , Células Fotorreceptoras Retinianas Conos/efectos de la radiación , Riboflavina/farmacología
18.
Adv Exp Med Biol ; 1074: 109-115, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721934

RESUMEN

Considerable progress has been made in the design and delivery of non-viral gene therapy vectors, but, like their viral counterparts, therapeutic levels of transgenes have not met the requirements for successful clinical applications so far. The biggest advantage of polymer-based nanoparticle vectors is the ease with which they can be modified to increase their ability to penetrate the cell membrane and target specific cells by simply changing the formulation of the nanoparticle compaction. We took advantage of this characteristic to improve transfection rates of our particles to meet the transgene levels which will be needed for future treatment of patients. For this study, we successfully investigated the possibility of our established pegylated polylysine particles to be administered via intravitreal rather than subretinal route to ease the damage during injection. We also demonstrated that our particles are flexible enough to sustain changes in the formulation to accommodate additional targeting sequences without losing their efficiency in transfecting neuronal cells in the retina. Together, these results give us the opportunity to even further improve our particles.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Inyecciones Intraoculares/métodos , Nanopartículas/administración & dosificación , Polilisina/administración & dosificación , Epitelio Pigmentado de la Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , ADN Recombinante/administración & dosificación , Inyecciones Intravítreas , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Tamaño de la Partícula , Polietilenglicoles/administración & dosificación , Epitelio Pigmentado de la Retina/citología , Transgenes
19.
Hum Mol Genet ; 24(10): 2709-23, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25637522

RESUMEN

Rhodopsin, a G-protein coupled receptor, most abundant protein in retinal rod photoreceptors, is glycosylated at asparagines-2 and 15 on its N-terminus. To understand the role of rhodopsin's glycosylation in vivo, we generated and characterized a transgenic mouse model that expresses a non-glycosylated form of rhodopsin. We show that lack of glycosylation triggers a dominant form of progressive retinal degeneration. Electron microscopic examination of retinas at postnatal day 17 revealed the presence of vacuolar structures that distorted rod photoreceptor outer segments and became more prominent with age. Expression of non-glycosylated rhodopsin alone showed that it is unstable and is regulated via ubiquitin-mediated proteasomal degradation at the base of outer segments. We observed similar vacuolization in outer segments of transgenic mice expressing human rhodopsin with a T17M mutation (hT17M), suggesting that the mechanism responsible for the degenerative process in mice expressing the non-glycosylated rhodopsin and the RHO(hT17M) mice is likely the cause of phenotype observed in retinitis pigmentosa patients carrying T17M mutation.


Asunto(s)
Rodopsina/metabolismo , Segmento Externo de la Célula en Bastón/metabolismo , Animales , Modelos Animales de Enfermedad , Expresión Génica , Glicosilación , Humanos , Ratones , Ratones Transgénicos , Microscopía Electrónica , Mutación Missense , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/fisiopatología , Rodopsina/genética , Segmento Externo de la Célula en Bastón/fisiología , Ubiquitinación
20.
J Biol Chem ; 290(8): 5041-5052, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25542898

RESUMEN

Retbindin is a novel retina-specific protein of unknown function. In this study, we have used various approaches to evaluate protein expression, localization, biochemical properties, and function. We find that retbindin is secreted by the rod photoreceptors into the inter-photoreceptor matrix where it is maintained via electrostatic forces. Retbindin is predominantly localized at the interface between photoreceptors and retinal pigment epithelium microvilli, a region critical for retinal function and homeostasis. Interestingly, although it is associated with photoreceptor outer segments, retbindin's expression is not dependent on their presence. In vitro, retbindin is capable of binding riboflavin, thus implicating the protein as a metabolite carrier between the retina and the retinal pigment epithelium. Altogether, our data show that retbindin is a novel photoreceptor-specific protein with a unique localization and function. We hypothesize that retbindin is an excellent candidate for binding retinal flavins and possibly participating in their transport from the extracellular space to the photoreceptors. Further investigations are warranted to determine the exact function of retbindin in retinal homeostasis and disease.


Asunto(s)
Proteínas del Ojo/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Riboflavina/metabolismo , Segmento Externo de la Célula en Bastón/metabolismo , Animales , Humanos , Ratones , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA