Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Exp Eye Res ; 187: 107755, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31408630

RESUMEN

Dark-rearing has been found to slow the rate of retinal degeneration in albino P23H but not S334ter mutant rhodopsin transgenic (Tg) rats. Since eye pigmentation has the same protective slowing effect as dark-rearing in RCS rats, we examined whether eye pigmentation has a comparable slowing effect in the different mutant rhodopsin Tg rats. Different lines of albino P23H and S334ter Tg rats on the Sprague-Dawley (SD) background were bred to Long-Evans (LE) rats to produce pigmented Tg rats. These were compared to albino Tg rats at postnatal days of different ages using the outer nuclear layer (ONL) as a morphological measure of photoreceptor number and electroretinogram (ERG) a- and b-wave amplitudes as a measure of retinal function. When compared to albino P23H rats, pigmented P23H rats had a slower rate of degeneration as measured by greater ONL thicknesses and greater ERG a- and b-wave amplitudes. By contrast, pigmented S334ter rats showed no difference in ONL thicknesses or ERG a- and b-wave amplitudes when compared to their albino equivalents. Thus, degeneration of photoreceptors in P23H Tg rats is slowed by eye pigmentation as measured by ONL thickness, while it is not in the S334ter Tg rats. Eye pigmentation also protects functional changes in ERG a- and b-waves for the P23H lines, but not for the S334ter lines.


Asunto(s)
Color del Ojo/genética , Retina/fisiopatología , Degeneración Retiniana/genética , Degeneración Retiniana/fisiopatología , Rodopsina/genética , Animales , Electrorretinografía , Mutación , Fenotipo , Células Fotorreceptoras de Vertebrados/patología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Ratas Transgénicas
2.
Adv Exp Med Biol ; 1185: C1, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32274704

RESUMEN

The title of the chapter is "Melatonin as the Possible Link Between Age-Related Retinal Degeneration and the Disrupted Circadian Rhythm in Elderly" but degeneration was incorrectly published as regeneration. Now this has been corrected to degeneration.

3.
Exp Eye Res ; 167: 56-90, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29122605

RESUMEN

We produced 8 lines of transgenic (Tg) rats expressing one of two different rhodopsin mutations in albino Sprague-Dawley (SD) rats. Three lines were generated with a proline to histidine substitution at codon 23 (P23H), the most common autosomal dominant form of retinitis pigmentosa in the United States. Five lines were generated with a termination codon at position 334 (S334ter), resulting in a C-terminal truncated opsin protein lacking the last 15 amino acid residues and containing all of the phosphorylation sites involved in rhodopsin deactivation, as well as the terminal QVAPA residues important for rhodopsin deactivation and trafficking. The rates of photoreceptor (PR) degeneration in these models vary in proportion to the ratio of mutant to wild-type rhodopsin. The models have been widely studied, but many aspects of their phenotypes have not been described. Here we present a comprehensive study of the 8 Tg lines, including the time course of PR degeneration from the onset to one year of age, retinal structure by light and electron microscopy (EM), hemispheric asymmetry and gradients of rod and cone degeneration, rhodopsin content, gene dosage effect, rapid activation and invasion of the outer retina by presumptive microglia, rod outer segment disc shedding and phagocytosis by the retinal pigmented epithelium (RPE), and retinal function by the electroretinogram (ERG). The biphasic nature of PR cell death was noted, as was the lack of an injury-induced protective response in the rat models. EM analysis revealed the accumulation of submicron vesicular structures in the interphotoreceptor space during the peak period of PR outer segment degeneration in the S334ter lines. This is likely due to the elimination of the trafficking consensus domain as seen before as with other rhodopsin mutants lacking the C-terminal QVAPA. The 8 rhodopsin Tg lines have been, and will continue to be, extremely useful models for the experimental study of inherited retinal degenerations.


Asunto(s)
Modelos Animales de Enfermedad , Células Fotorreceptoras de Vertebrados/patología , Mutación Puntual , Retina/fisiología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Rodopsina/genética , Animales , Electrorretinografía , Microscopía , Microscopía Electrónica , Fenotipo , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Degeneración Retiniana/fisiopatología
4.
PLoS Genet ; 11(12): e1005723, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26656104

RESUMEN

Inherited photoreceptor degenerations (IPDs) are the most genetically heterogeneous of Mendelian diseases. Many IPDs exhibit substantial phenotypic variability, but the basis is usually unknown. Mutations in MERTK cause recessive IPD phenotypes associated with the RP38 locus. We have identified a murine genetic modifier of Mertk-associated photoreceptor degeneration, the C57BL/6 (B6) allele of which acts as a suppressor. Photoreceptors degenerate rapidly in Mertk-deficient animals homozygous for the 129P2/Ola (129) modifier allele, whereas animals heterozygous for B6 and 129 modifier alleles exhibit an unusual intermixing of degenerating and preserved retinal regions, with females more severely affected than males. Mertk-deficient mice homozygous for the B6 modifier allele display degeneration only in the far periphery, even at 8 months of age, and have improved retinal function compared to animals homozygous for the 129 allele. We genetically mapped the modifier to an approximately 2-megabase critical interval that includes Tyro3, a paralog of Mertk. Tyro3 expression in the outer retina varies with modifier genotype in a manner characteristic of a cis-acting expression quantitative trait locus (eQTL), with the B6 allele conferring an approximately three-fold higher expression level. Loss of Tyro3 function accelerates the pace of photoreceptor degeneration in Mertk knockout mice, and TYRO3 protein is more abundant in the retinal pigment epithelium (RPE) adjacent to preserved central retinal regions of Mertk knockout mice homozygous for the B6 modifier allele. Endogenous human TYRO3 protein co-localizes with nascent photoreceptor outer segment (POS) phagosomes in a primary RPE cell culture assay, and expression of murine Tyro3 in cultured cells stimulates phagocytic ingestion of POS. Our findings demonstrate that Tyro3 gene dosage modulates Mertk-associated retinal degeneration, provide strong evidence for a direct role for TYRO3 in RPE phagocytosis, and suggest that an eQTL can modify a recessive IPD.


Asunto(s)
Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Degeneración Retiniana/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Fagocitosis , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Retina/metabolismo , Retina/patología , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/patología , Tirosina Quinasa c-Mer
5.
Exp Eye Res ; 165: 175-181, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28974356

RESUMEN

Retinal degenerations, including age-related macular degeneration and the retinitis pigmentosa family of diseases, are among the leading causes of legal blindness in the United States. We previously found that Stanniocalcin-1 (STC-1) reduced photoreceptor loss in the S334ter-3 and Royal College of Surgeons rat models of retinal degeneration. The results were attributed in part to a reduction in oxidative stress. Herein, we tested the hypothesis that long-term delivery of STC-1 would provide therapeutic rescue in more chronic models of retinal degeneration. To achieve sustained delivery, we produced an adeno-associated virus (AAV) construct to express STC-1 (AAV-STC-1) under the control of a retinal ganglion cell targeting promoter human synapsin 1 (hSYN1). AAV-STC-1 was injected intravitreally into the P23H-1 and S334ter-4 rhodopsin transgenic rats at postnatal day 10. Tissues were collected at postnatal day 120 for confirmation of STC-1 overexpression and histologic and molecular analysis. Electroretinography (ERG) was performed in a cohort of animals at that time. Overexpression of STC-1 resulted in a significant preservation of photoreceptors as assessed by outer nuclear thickness in the P23H-1 (P < 0.05) and the S334ter-4 (P < 0.005) models compared to controls. Additionally, retinal function was significantly improved in the P23H-1 model with overexpressed STC-1 as assessed by ERG analysis (scotopic b-wave P < 0.005 and photopic b-wave P < 0.05). Microarray analysis identified common downstream gene expression changes that occurred in both models. Genes of interest based on their function were selected for validation by quantitative real-time PCR and were significantly increased in the S334ter-4 model.


Asunto(s)
Dependovirus , Glicoproteínas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Retinitis Pigmentosa/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Glicoproteínas/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Células Fotorreceptoras de Vertebrados/patología , Ratas , Ratas Transgénicas , Retinitis Pigmentosa/patología
6.
J Lipid Res ; 57(5): 818-31, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26947037

RESUMEN

Retinal degeneration (RD) affects millions of people and is a major cause of ocular impairment and blindness. With a wide range of mutations and conditions leading to degeneration, targeting downstream processes is necessary for developing effective treatments. Ceramide and sphingosine-1-phosphate, a pair of bioactive sphingolipids, are involved in apoptosis and its prevention, respectively. Apoptotic cell death is a potential driver of RD, and in order to understand the mechanism of degeneration and potential treatments, we studied rhodopsin mutant RD model, P23H-1 rats. Investigating this genetic model of human RD allows us to investigate the association of sphingolipid metabolites with the degeneration of the retina in P23H-1 rats and the effects of a specific modulator of sphingolipid metabolism, FTY720. We found that P23H-1 rat retinas had altered sphingolipid profiles that, when treated with FTY720, were rebalanced closer to normal levels. FTY720-treated rats also showed protection from RD compared with their vehicle-treated littermates. Based on these data, we conclude that sphingolipid dysregulation plays a secondary role in retinal cell death, which may be common to many forms of RDs, and that the U.S. Food and Drug Administration-approved drug FTY720 or related compounds that modulate sphingolipid metabolism could potentially delay the cell death.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Distrofias Retinianas/metabolismo , Esfingolípidos/metabolismo , Animales , Vías Biosintéticas , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Clorhidrato de Fingolimod/uso terapéutico , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Ratas Sprague-Dawley , Distrofias Retinianas/tratamiento farmacológico , Esfingomielina Fosfodiesterasa/metabolismo
7.
Hum Genet ; 135(3): 327-43, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26825853

RESUMEN

MERTK is an essential component of the signaling network that controls phagocytosis in retinal pigment epithelium (RPE), the loss of which results in photoreceptor degeneration. Previous proof-of-concept studies have demonstrated the efficacy of gene therapy using human MERTK (hMERTK) packaged into adeno-associated virus (AAV2) in treating RCS rats and mice with MERTK deficiency. The purpose of this study was to assess the safety of gene transfer via subretinal administration of rAAV2-VMD2-hMERTK in subjects with MERTK-associated retinitis pigmentosa (RP). After a preclinical phase confirming the safety of the study vector in monkeys, six patients (aged 14 to 54, mean 33.3 years) with MERTK-related RP and baseline visual acuity (VA) ranging from 20/50 to <20/6400 were entered in a phase I open-label, dose-escalation trial. One eye of each patient (the worse-seeing eye in five subjects) received a submacular injection of the viral vector, first at a dose of 150 µl (5.96 × 10(10)vg; 2 patients) and then 450 µl (17.88 × 10(10)vg; 4 patients). Patients were followed daily for 10 days at 30, 60, 90, 180, 270, 365, 540, and 730 days post-injection. Collected data included (1) full ophthalmologic examination including best-corrected VA, intraocular pressure, color fundus photographs, macular spectral domain optical coherence tomography and full-field stimulus threshold test (FST) in both the study and fellow eyes; (2) systemic safety data including CBC, liver and kidney function tests, coagulation profiles, urine analysis, AAV antibody titers, peripheral blood PCR and ASR measurement; and (3) listing of ophthalmological or systemic adverse effects. All patients completed the 2-year follow-up. Subretinal injection of rAAV2-VMD2-hMERTK was associated with acceptable ocular and systemic safety profiles based on 2-year follow-up. None of the patients developed complications that could be attributed to the gene vector with certainty. Postoperatively, one patient developed filamentary keratitis, and two patients developed progressive cataract. Of these two patients, one also developed transient subfoveal fluid after the injection as well as monocular oscillopsia. Two patients developed a rise in AAV antibodies, but neither patient was positive for rAAV vector genomes via PCR. Three patients also displayed measurable improved visual acuity in the treated eye following surgery, although the improvement was lost by 2 years in two of these patients. Gene therapy for MERTK-related RP using careful subretinal injection of rAAV2-VMD2-hMERTK is not associated with major side effects and may result in clinical improvement in a subset of patients.


Asunto(s)
Terapia Genética/métodos , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Adolescente , Adulto , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Determinación de Punto Final , Femenino , Estudios de Seguimiento , Vectores Genéticos , Humanos , Macaca , Masculino , Persona de Mediana Edad , Mutación , Complicaciones Posoperatorias/terapia , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Líquido Subretiniano , Tomografía de Coherencia Óptica , Resultado del Tratamiento , Agudeza Visual , Adulto Joven , Tirosina Quinasa c-Mer
8.
Adv Exp Med Biol ; 854: 487-93, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26427450

RESUMEN

MERTK-associated retinal degenerations are thought to have defects in phagocytosis of shed outer segment membranes by the retinal pigment epithelium (RPE), as do the rodent models of these diseases. We have subretinally injected an RPE-specific AAV2 vector, AAV2-VMD2-hMERTK, to determine whether this would provide long-term photoreceptor rescue in the RCS rat, which it did for up to 6.5 months, the longest time point examined. Moreover, we found phagosomes in the RPE in the rescued regions of RCS retinas soon after the onset of light. The same vector also had a major protective effect in Mertk-null mice, with a concomitant increase in ERG response amplitudes in the vector-injected eyes. These findings suggest that planned clinical trials with this vector will have a favorable outcome.


Asunto(s)
Terapia Genética/métodos , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Degeneración Retiniana/genética , Degeneración Retiniana/terapia , Animales , Bestrofinas , Canales de Cloruro/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Electrorretinografía , Proteínas del Ojo/genética , Vectores Genéticos/genética , Humanos , Ratones Noqueados , Fagocitosis/genética , Fagocitosis/fisiología , Fagosomas/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/metabolismo , Ratas Mutantes , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/deficiencia , Proteínas Tirosina Quinasas Receptoras/metabolismo , Degeneración Retiniana/fisiopatología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/fisiopatología , Resultado del Tratamiento , Tirosina Quinasa c-Mer
9.
Adv Exp Med Biol ; 854: 185-91, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26427410

RESUMEN

RHO (Rod opsin) encodes a G-protein coupled receptor that is expressed exclusively by rod photoreceptors of the retina and forms the essential photopigment, rhodopsin, when coupled with 11-cis-retinal. Many rod opsin disease -mutations cause rod opsin protein misfolding and trigger endoplasmic reticulum (ER) stress, leading to activation of the Unfolded Protein Response (UPR) signal transduction network. Chop is a transcriptional activator that is induced by ER stress and promotes cell death in response to chronic ER stress. Here, we examined the role of Chop in transgenic mice expressing human P23H rhodopsin (hP23H Rho Tg) that undergo retinal degeneration. With the exception of one time point, we found no significant induction of Chop in these animals and no significant change in retinal degeneration by histology and electrophysiology when hP23H Rho Tg animals were bred into a Chop (-/-) background. Our results indicate that Chop does not play a significant causal role during retinal degeneration in these animals. We suggest that other modules of the ER stress-induced UPR signaling network may be involved photoreceptor disease induced by P23H rhodopsin.


Asunto(s)
Degeneración Retiniana/genética , Células Fotorreceptoras Retinianas Bastones/metabolismo , Rodopsina/genética , Factor de Transcripción CHOP/genética , Animales , Supervivencia Celular/genética , Electrorretinografía , Expresión Génica , Humanos , Ratones Noqueados , Ratones Transgénicos , Degeneración Retiniana/metabolismo , Degeneración Retiniana/fisiopatología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rodopsina/metabolismo , Factor de Transcripción CHOP/deficiencia , Transgenes/genética
11.
Adv Exp Med Biol ; 801: 585-92, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24664747

RESUMEN

Rhodopsin mutations cause many types of heritable retinitis pigmentosa (RP). Biochemical and in vitro studies have demonstrated that many RP-linked mutant rhodopsins produce misfolded rhodopsin proteins, which are prone to aggregation and retention within the endoplasmic reticulum, where they cause endoplasmic reticulum stress and activate the Unfolded Protein Response signaling pathways. Many vertebrate models of retinal degeneration have been created through expression of RP-linked rhodopsins in photoreceptors including, but not limited to, VPP/GHL mice, P23H Rhodopsin frogs, P23H rhodopsin rats, S334ter rhodopsin rats, C185R rhodopsin mice, T17M rhodopsin mice, and P23H rhodopsin mice. These models have provided many opportunities to test therapeutic strategies to prevent retinal degeneration and also enabled in vivo investigation of cellular and molecular mechanisms responsible for photoreceptor cell death. Here, we examine and compare the contribution of endoplasmic reticulum stress to retinal degeneration in several vertebrate models of RP generated through expression of mutant rhodopsins.


Asunto(s)
Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/fisiología , Degeneración Retiniana/genética , Degeneración Retiniana/fisiopatología , Rodopsina/genética , Animales , Humanos , Ratones , Ratones Transgénicos , Ratas , Ratas Transgénicas , Especificidad de la Especie , Respuesta de Proteína Desplegada/fisiología , Vertebrados , Xenopus laevis
13.
Mol Ther ; 20(4): 788-97, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22294148

RESUMEN

Oxidative stress and photoreceptor apoptosis are prominent features of many forms of retinal degeneration (RD) for which there are currently no effective therapies. We previously observed that mesenchymal stem/stromal cells reduce apoptosis by being activated to secrete stanniocalcin-1 (STC-1), a multifunctional protein that reduces oxidative stress by upregulating mitochondrial uncoupling protein-2 (UCP-2). Therefore, we tested the hypothesis that intravitreal injection of STC-1 can rescue photoreceptors. We first tested STC-1 in the rhodopsin transgenic rat characterized by rapid photoreceptor loss. Intravitreal STC-1 decreased the loss of photoreceptor nuclei and transcripts and resulted in measurable retinal function when none is otherwise present in this rapid degeneration. We then tested STC-1 in the Royal College of Surgeons (RCS) rat characterized by a slower photoreceptor degeneration. Intravitreal STC-1 reduced the number of pyknotic nuclei in photoreceptors, delayed the loss of photoreceptor transcripts, and improved function of rod photoreceptors. Additionally, STC-1 upregulated UCP-2 and decreased levels of two protein adducts generated by reactive oxygen species (ROS). Microarrays from the two models demonstrated that STC-1 upregulated expression of a similar profile of genes for retinal development and function. The results suggested that intravitreal STC-1 is a promising therapy for various forms of RD including retinitis pigmentosa and atrophic age-related macular degeneration (AMD).


Asunto(s)
Glicoproteínas/farmacología , Degeneración Retiniana/tratamiento farmacológico , Animales , Electrorretinografía , Ensayo de Inmunoadsorción Enzimática , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Degeneración Macular/tratamiento farmacológico , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Degeneración Retiniana/metabolismo , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinitis Pigmentosa/tratamiento farmacológico , Proteína Desacopladora 2
14.
Proc Natl Acad Sci U S A ; 107(13): 5961-6, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20231467

RESUMEN

The P23H mutation within the rhodopsin gene (RHO) causes rhodopsin misfolding, endoplasmic reticulum (ER) stress, and activates the unfolded protein response (UPR), leading to rod photoreceptor degeneration and autosomal dominant retinitis pigmentosa (ADRP). Grp78/BiP is an ER-localized chaperone that is induced by UPR signaling in response to ER stress. We have previously demonstrated that BiP mRNA levels are selectively reduced in animal models of ADRP arising from P23H rhodopsin expression at ages that precede photoreceptor degeneration. We have now overexpressed BiP to test the hypothesis that this chaperone promotes the trafficking of P23H rhodopsin to the cell membrane, reprograms the UPR favoring the survival of photoreceptors, blocks apoptosis, and, ultimately, preserves vision in ADRP rats. In cell culture, increasing levels of BiP had no impact on the localization of P23H rhodopsin. However, BiP overexpression alleviated ER stress by reducing levels of cleaved pATF6 protein, phosphorylated eIF2alpha and the proapoptotic protein CHOP. In P23H rats, photoreceptor levels of cleaved ATF6, pEIF2alpha, CHOP, and caspase-7 were much higher than those of wild-type rats. Subretinal delivery of AAV5 expressing BiP to transgenic rats led to reduction in CHOP and photoreceptor apoptosis and to a sustained increase in electroretinogram amplitudes. We detected complexes between BiP, caspase-12, and the BH3-only protein BiK that may contribute to the antiapoptotic activity of BiP. Thus, the preservation of photoreceptor function resulting from elevated levels of BiP is due to suppression of apoptosis rather than to a promotion of rhodopsin folding.


Asunto(s)
Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Visión Ocular/genética , Visión Ocular/fisiología , Sustitución de Aminoácidos , Animales , Apoptosis , Secuencia de Bases , Dependovirus/genética , Modelos Animales de Enfermedad , Electrorretinografía , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Células HeLa , Humanos , Ratones , Modelos Biológicos , Complejos Multiproteicos , Mutación Missense , Células Fotorreceptoras de Vertebrados/patología , Células Fotorreceptoras de Vertebrados/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Retina/patología , Retina/fisiopatología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/fisiopatología , Retinitis Pigmentosa/terapia , Rodopsina/química , Estrés Fisiológico , Factor de Transcripción CHOP/metabolismo , Transfección , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/fisiología
15.
Exp Eye Res ; 104: 65-73, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23036564

RESUMEN

The aim of this study was to examine the temporal relationship between behaviorally measured visual thresholds, photoreceptor degeneration and dysfunction, synaptic and neuronal morphology changes in the retina in the S334ter line 4 rat. Specifically, we examined the optokinetic tracking (OKT) behavior in S334ter rats daily and found that OKT thresholds reflected normal values at eye opening but quickly reduced to a non-response level by postnatal day (P) 22. By contrast, the scotopic electroretinogram (ERG) showed a much slower degeneration, with substantial scotopic function remaining after P90 as previously demonstrated for this line of rats. Photopic b-wave amplitudes revealed functional levels between 70 and 100% of normal between P30 and P90. Histological evidence demonstrated that photoreceptor degeneration occurred over many months, with an outer nuclear layer (ONL) roughly half the thickness of a normal age-matched control at P90. Immunohistochemical analysis revealed a number of changes in retinal morphology in the Tg S334ter line 4 rat that occur at or before P40 including: elevated levels of rod opsin expression in the ONL, cone photoreceptor morphology changes, glial cell activation, inner retinal neuron sprouting, and microglial cell activation. Many of these changes were evident at P30 and in some cases as early as eye opening (P15). Thus, the morphological changes occurred in concert with or before the very rapid loss of the behavioral (OKT) responses, and significantly before the loss of photoreceptors and photoreceptor function.


Asunto(s)
Mutación , Nistagmo Optoquinético/fisiología , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/fisiopatología , Rodopsina/genética , Animales , Biomarcadores/metabolismo , Supervivencia Celular , Electrorretinografía , Técnica del Anticuerpo Fluorescente Indirecta , Microscopía Confocal , Neuroglía/metabolismo , Neuroglía/patología , Células Fotorreceptoras de Vertebrados/metabolismo , Ratas , Ratas Long-Evans , Ratas Transgénicas , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Umbral Sensorial/fisiología , Percepción Visual/fisiología
16.
Mol Vis ; 16: 1004-18, 2010 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-20577653

RESUMEN

PURPOSE: In a previous study, several quantitative trait loci (QTL) that influence age-related degeneration (ageRD) were identified in a cross between the albino strains B6(Cg)-Tyr(c-2J)/J (B6a) and BALB/cByJ (C). The Chromosome (Chr) 6 and Chr 10 QTL were the strongest and most highly significant loci and both involved B6a protective alleles. The QTL were responsible for 21% and 9% of the variance in phenotypes, respectively. We focused on these two QTL to identify candidate genes. METHODS: DNA microarrays were used for the two mouse strains at four and eight months of age to identify genes that are differentially regulated and map to either QTL. Gene Ontology (GO) analysis of the differentially expressed genes was performed to identify possible processes and pathways associated with ageRD. To identify additional candidates, database analyses (Positional Medline or PosMed) were used. Based on differential expression, PosMed, and the presence of reported polymorphisms, five genes per QTL were selected for further study by sequencing analysis and qRT-PCR. Tumor necrosis factor, alpha- induced protein 3 (Tnfaip3; on a C57BL/6J (B6) background) was phenotypically tested. Single nucleotide polymorphisms (SNPs) flanking this gene were correlated with outer nuclear layer thickness (ONL), and eight-month-old Tnfaip3(+/-) mice were tested for ageRD. RESULTS: Polymorphisms were found in the coding regions of eight genes. Changes in gene expression were identified by qRT-PCR for Hexokinase 2 (Hk2) and Docking protein 1 (Dok1) at four months and for Dok1 and Tnfaip3 at eight months. Tnfaip3 was selected for phenotypic testing due to differential expression and the presence of two nonsynonymous mutations. However, when ONL thickness was compared in eight-month-old congenic Tnfaip3(+/-) and Tnfaip3(+/+) mice, no differences were found, suggesting that Tnfaip3 is not the quantitative trait gene (QTG) for the Chr 10 QTL. The GO analysis revealed that GO terms associated with stress and cell remodeling are overrepresented in the ageRD-sensitive C strain compared with the B6a strain with age (eight months). In the ageRD-resistant B6a strain, compared with the C strain, GO terms associated with antioxidant response and the regulation of blood vessel size are overrepresented with age. CONCLUSIONS: The analyses of differentially expressed genes and the PosMed database yielded candidate genes for the Chr 6 and Chr 10 QTL. HtrA serine peptidase 2 (Htra2), Dok1, and Tnfaip3 were deemed most promising because of their known roles in apoptosis and our finding of nonsynonymous substitutions between B6a and C strains. While Tnfaip3 was excluded as the QTG for the Chr 10 QTL, Dok1 and Htra2 remain good candidates for the Chr 6 QTL. Finally, the GO term analysis further supports the general hypothesis that oxidative stress is involved in ageRD.


Asunto(s)
Mapeo Cromosómico , Estudios de Asociación Genética , Sitios de Carácter Cuantitativo , Degeneración Retiniana/genética , Animales , Cisteína Endopeptidasas/genética , Proteínas de Unión al ADN/genética , Femenino , Serina Peptidasa A2 que Requiere Temperaturas Altas , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos , Ratones Noqueados , Análisis por Micromatrices , Proteínas Mitocondriales/genética , Estrés Oxidativo , Fenotipo , Fosfoproteínas/genética , Polimorfismo de Nucleótido Simple , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina Endopeptidasas/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
17.
Adv Exp Med Biol ; 664: 115-21, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20238009

RESUMEN

Many mutations associated with retinal degeneration lead to the production of misfolded proteins by cells of the retina. Emerging evidence suggests that these abnormal proteins cause cell death by activating the Unfolded Protein Response, a set of conserved intracellular signaling pathways that detect protein misfolding within the endoplasmic reticulum and control protective and proapoptotic signal transduction pathways. Here, we review the misfolded proteins associated with select types of retinitis pigmentosa, Stargadt-like macular degeneration, and Doyne Honeycomb Retinal Dystrophy and discuss the role that endoplasmic reticulum stress and UPR signaling play in their pathogenesis. Last, we review new therapies for these diseases based on preventing protein misfolding in the retina.


Asunto(s)
Pliegue de Proteína , Deficiencias en la Proteostasis/metabolismo , Distrofias Retinianas/metabolismo , Animales , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Humanos , Deficiencias en la Proteostasis/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Degeneración Retiniana/prevención & control , Distrofias Retinianas/patología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
18.
Mol Cell Neurosci ; 38(3): 431-43, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18511296

RESUMEN

BDNF signaling through its TrkB receptor plays a pivotal role in activity-dependent refinement of synaptic connectivity of retinal ganglion cells. Additionally, studies using TrkB knockout mice have suggested that BDNF/TrkB signaling is essential for the development of photoreceptors and for synaptic communication between photoreceptors and second order retinal neurons. Thus the action of BDNF on refinement of synaptic connectivity of retinal ganglion cells could be a direct effect in the inner retina, or it could be secondary to its proposed role in rod maturation and in the formation of rod to bipolar cell synaptic transmission. To address this matter we have conditionally eliminated TrkB within the retina. We find that rod function and synaptic transmission to bipolar cells is not compromised in these conditional knockout mice. Consistent with previous work, we find that inner retina neural development is regulated by retinal BDNF/TrkB signaling. Specifically we show here also that the complexity of neuronal processes of dopaminergic cells is reduced in conditional TrkB knockout mice. We conclude that retinal BDNF/TrkB signaling has its primary role in the development of inner retinal neuronal circuits, and that this action is not a secondary effect due to the loss of visual signaling in the outer retina.


Asunto(s)
Receptor trkB/fisiología , Retina/crecimiento & desarrollo , Retina/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Red Nerviosa/crecimiento & desarrollo , Red Nerviosa/metabolismo , Receptor trkB/deficiencia , Receptor trkB/genética , Segmento Externo de la Célula en Bastón/metabolismo , Segmento Externo de la Célula en Bastón/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
19.
J Neurosci ; 27(27): 7245-55, 2007 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-17611277

RESUMEN

Glutamatergic neurotransmission requires vesicular glutamate transporters (VGLUTs) to sequester glutamate into synaptic vesicles. Generally, VGLUT1 and VGLUT2 isoforms show complementary expression in the CNS and retina. However, little is known about whether isoform-specific expression serves distinct pathways and physiological functions. Here, by examining visual functions in VGLUT1-null mice, we demonstrate that visual signaling from photoreceptors to retinal output neurons requires VGLUT1. However, photoentrainment and pupillary light responses are preserved. We provide evidence that melanopsin-containing, intrinsically photosensitive retinal ganglion cells (RGCs), signaling via VGLUT2 pathways, support these non-image-forming functions. We conclude that VGLUT1 is essential for transmitting visual signals from photoreceptors to second- and third-order neurons, but VGLUT1 is not necessary for intrinsic visual functions. Furthermore, melanopsin and VGLUT2 expression in a subset of RGCs immediately after birth strongly supports the idea that intrinsic vision can function well before rod- and cone-mediated signaling has matured.


Asunto(s)
Células Fotorreceptoras/fisiología , Transducción de Señal/fisiología , Sinapsis/fisiología , Proteína 1 de Transporte Vesicular de Glutamato/fisiología , Visión Ocular/fisiología , Animales , Potenciales Evocados Visuales/fisiología , Ratones , Ratones Noqueados , Estimulación Luminosa/métodos , Isoformas de Proteínas/fisiología , Ratas , Ratas Long-Evans , Células Ganglionares de la Retina/fisiología
20.
Exp Eye Res ; 87(6): 561-70, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18848932

RESUMEN

The early loss of photoreceptors in some retinal degenerations in mice has been shown to have a profound effect on vascular development of the retina. To better characterize this relationship, we have examined the formation of retinal blood vessels during the first month of life in 8 lines of transgenic rats with different ages of onset and rates of photoreceptor cell loss mediated by the expression of mutant rhodopsin (P23H and S334ter). The number of capillary profiles in the superficial plexus (SP) and deep capillary plexus (DCP) of the retina were quantified in retinal sections taken at postnatal day (P) 8, 10, 12, 15 and 30. In normal wild-type rats, the SP and DCP had mostly established mature, adult patterns by P15, as previously shown. In the transgenic rats, the loss of photoreceptors had relatively little effect on the SP. By contrast, the loss of photoreceptors during vascular development had a major impact on the DCP. In the two lines with early and most rapid photoreceptor loss, S334ter-7 and S334ter-3, where about 90% and 65%, respectively, of the photoreceptors were already lost by P15, the DCP either failed to form (S334ter-7) or the number of capillary profiles was less than 7% of controls (S334ter-3). In lines where almost all photoreceptors were still present at P15 (S334ter-4, S334ter-9, P23H-2 and P23H-3), the number of profiles in the DCP were the same as in wild-type controls at P30. In two lines with an intermediate rate of degeneration (S334ter-5 and P23H-1), where only about 25% of the photoreceptors were lost by P15, there was an intermediate number of vascular profiles in the DCP at P30. Thus, a very close relationship between the number of photoreceptors and vessel profiles in the DCP during its development exists in the transgenic rats, and the loss of photoreceptors results in the failure or inhibition of the DCP to develop. Several mechanisms may explain this relationship including changes in the level of physiological oxygen tension or alteration in the release of angiogenic factors that normally drive vessel development. Analysis of older transgenic retinas up to 1 year of age revealed that (1) vascular profiles are lost from the DCP in essentially all lines once fewer than about 30-33% of photoreceptors remain; (2) in those lines where the DCP essentially did not develop (S334ter-7 and S334ter-3), the effect of photoreceptor absence was permanent, and there was no late vascularization of the DCP; (3) the number of capillary profiles in the SP remained no different from controls in any of the lines, despite long-standing loss of photoreceptors; and (4) neovascularization of the RPE by retinal capillaries occurred with a latency of 60-180 days after the loss of photoreceptors, except in S334ter-7 rats, where neovascularization essentially did not occur. Analysis of RCS rats was carried out for comparison.


Asunto(s)
Mutación , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/patología , Vasos Retinianos/crecimiento & desarrollo , Rodopsina/genética , Envejecimiento/patología , Animales , Neovascularización Patológica/patología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Degeneración Retiniana/fisiopatología , Epitelio Pigmentado de la Retina/irrigación sanguínea , Epitelio Pigmentado de la Retina/patología , Segmento Externo de la Célula en Bastón/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA