Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Exp Eye Res ; 242: 109872, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38514024

RESUMEN

X-linked retinoschisis (XLRS) is an early onset degenerative retinal disease characterized by cystic lesions in the middle layers of the retina. These structural changes are accompanied by a loss of visual acuity and decreased contrast sensitivity. XLRS is caused by mutations in the gene Rs1 which encodes the secreted protein Retinoschisin 1. Young Rs1-mutant mouse models develop key hallmarks of XLRS including intraretinal schisis and abnormal electroretinograms. The electroretinogram (ERG) comprises activity of multiple cellular generators, and it is not known how and when each of these is impacted in Rs1 mutant mice. Here we use an ex vivo ERG system and pharmacological blockade to determine how ERG components generated by photoreceptors, ON-bipolar, and Müller glial cells are impacted in Rs1 mutants and to determine the time course of these changes. We report that ERG abnormalities begin near eye-opening and that all ERG components are involved.


Asunto(s)
Moléculas de Adhesión Celular , Modelos Animales de Enfermedad , Electrorretinografía , Proteínas del Ojo , Retinosquisis , Animales , Retinosquisis/genética , Retinosquisis/fisiopatología , Ratones , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Ratones Endogámicos C57BL , Mutación , Células Ependimogliales/patología , Células Ependimogliales/metabolismo , Masculino , Células Bipolares de la Retina/patología , Células Bipolares de la Retina/metabolismo
2.
Exp Eye Res ; 212: 108770, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34562437

RESUMEN

PURPOSE: Cancer-associated retinal ON bipolar cell dysfunction (CARBD), which includes melanoma-associated retinopathy (MAR), has been reported to be caused by autoantibodies against the molecules expressed in ON bipolar cells, including TRPM1. The purpose of this study was to determine the antigenic regions of the autoantibodies against TRPM1 in the sera of CARBD patients, in whom we previously detected anti-TRPM1 autoantibodies. METHODS: The antigenic regions against TRPM1 in the sera of eight CARBD patients were examined by Western blots using HEK293T cells transfected with the plasmids expressing FLAG-tagged TRPM1 fragments. The clinical course of these patients was also documented. RESULTS: The clinical course differed among the patients. The electroretinograms (ERGs) and symptoms were improved in three patients, deteriorated in one patient, remained unchanged for a long time in one patient, and were not followable in three patients. Seven of the eight sera possessed multiple antigenic regions: two sera contained at least four antigen recognition regions, and three sera had at least three regions. The antigen regions were spread over the entire TRPM1 protein: five sera in the N-terminal intracellular domain, six sera in the transmembrane-containing region, and six sera in the C-terminal intracellular domain. No significant relationship was observed between the location of the antigen epitope and the patients' clinical course. CONCLUSIONS: The antigenic regions of anti-TRPM1 autoantibodies in CARBD patients were present not only in the N-terminal intracellular domain, which was reported in an earlier report, but also in the transmembrane-containing region and in the C-terminal intracellular domain. In addition, the antigenic regions for TRPM1 were found to vary among the CARBD patients examined, and most of the sera had multiple antigenic regions.


Asunto(s)
Autoanticuerpos/sangre , Síndromes Paraneoplásicos Oculares/inmunología , Células Bipolares de la Retina/metabolismo , Canales Catiónicos TRPM/inmunología , Anciano , Western Blotting , Electrorretinografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Síndromes Paraneoplásicos Oculares/metabolismo , Síndromes Paraneoplásicos Oculares/patología , Células Bipolares de la Retina/patología , Estudios Retrospectivos , Células Tumorales Cultivadas
3.
Int J Mol Sci ; 22(4)2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33671472

RESUMEN

To reveal the pathomechanisms of glaucoma, a common cause of blindness, suitable animal models are needed. As previously shown, retinal ganglion cell and optic nerve degeneration occur in ßB1-CTGF mice. Here, we aimed to determine possible apoptotic mechanisms and degeneration of different retinal cells. Hence, retinae were processed for immunohistology (n = 5-9/group) and quantitative real-time PCR analysis (n = 5-7/group) in 5- and 10-week-old ßB1-CTGF and wildtype controls. We noted significantly more cleaved caspase 3+ cells in ßB1-CTGF retinae at 5 (p = 0.005) and 10 weeks (p = 0.02), and a significant upregulation of Casp3 and Bax/Bcl2 mRNA levels (p < 0.05). Furthermore, more terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL+) cells were detected in transgenic mice at 5 (p = 0.03) and 10 weeks (p = 0.02). Neurofilament H staining (p = 0.01) as well as Nefh (p = 0.02) and Tubb3 (p = 0.009) mRNA levels were significantly decreased at 10 weeks. GABAergic synapse intensity was lower at 5 weeks, while no alterations were noted at 10 weeks. The glutamatergic synapse intensity was decreased at 5 (p = 0.007) and 10 weeks (p = 0.01). No changes were observed for bipolar cells, photoreceptors, and macroglia. We conclude that apoptotic processes and synapse loss precede neuronal death in this model. This slow progression rate makes the ßB1-CTGF mice a suitable model to study primary open-angle glaucoma.


Asunto(s)
Apoptosis , Factor de Crecimiento del Tejido Conjuntivo/genética , Animales , Recuento de Células , Ratones Transgénicos , Modelos Animales , Proteínas de Neurofilamentos/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Células Bipolares de la Retina/patología , Células Ganglionares de la Retina/patología , Sinapsis/patología
4.
Int J Mol Sci ; 22(8)2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33919796

RESUMEN

Guanylate cyclase-activating protein 1 (GCAP1) is involved in the shutdown of the phototransduction cascade by regulating the enzymatic activity of retinal guanylate cyclase via a Ca2+/cGMP negative feedback. While the phototransduction-associated role of GCAP1 in the photoreceptor outer segment is widely established, its implication in synaptic transmission to downstream neurons remains to be clarified. Here, we present clinical and biochemical data on a novel isolate GCAP1 variant leading to a double amino acid substitution (p.N104K and p.G105R) and associated with cone dystrophy (COD) with an unusual phenotype. Severe alterations of the electroretinogram were observed under both scotopic and photopic conditions, with a negative pattern and abnormally attenuated b-wave component. The biochemical and biophysical analysis of the heterologously expressed N104K-G105R variant corroborated by molecular dynamics simulations highlighted a severely compromised Ca2+-sensitivity, accompanied by minor structural and stability alterations. Such differences reflected on the dysregulation of both guanylate cyclase isoforms (RetGC1 and RetGC2), resulting in the constitutive activation of both enzymes at physiological levels of Ca2+. As observed with other GCAP1-associated COD, perturbation of the homeostasis of Ca2+ and cGMP may lead to the toxic accumulation of second messengers, ultimately triggering cell death. However, the abnormal electroretinogram recorded in this patient also suggested that the dysregulation of the GCAP1-cyclase complex further propagates to the synaptic terminal, thereby altering the ON-pathway related to the b-wave generation. In conclusion, the pathological phenotype may rise from a combination of second messengers' accumulation and dysfunctional synaptic communication with bipolar cells, whose molecular mechanisms remain to be clarified.


Asunto(s)
Calcio/metabolismo , Distrofia del Cono/genética , Distrofia del Cono/fisiopatología , Proteínas Activadoras de la Guanilato-Ciclasa/genética , Mutación/genética , Células Bipolares de la Retina/patología , Células Fotorreceptoras Retinianas Bastones/patología , Transmisión Sináptica , Atrofia , Cationes , Distrofia del Cono/diagnóstico por imagen , Progresión de la Enfermedad , Electrorretinografía , Femenino , Fondo de Ojo , Guanilato Ciclasa/metabolismo , Proteínas Activadoras de la Guanilato-Ciclasa/química , Heterocigoto , Humanos , Hidrodinámica , Interacciones Hidrofóbicas e Hidrofílicas , Persona de Mediana Edad , Simulación de Dinámica Molecular , Fenotipo , Agregado de Proteínas , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Células Bipolares de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Tomografía de Coherencia Óptica
5.
J Neurochem ; 148(3): 400-412, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30548540

RESUMEN

Phospholipids are asymmetrically distributed across the mammalian plasma membrane, with phosphatidylserine (PS) and phosphatidylethanolamine concentrated in the cytoplasmic leaflet of the membrane bilayer and phosphatidylcholine in the exoplasmic leaflet. This asymmetric distribution is dependent on a group of P4 ATPases called PS flippases. The proper transport and function of PS flippases require a ß-subunit transmembrane protein 30A (TMEM30A). Disruption of PS flippases leads to several human diseases. Tmem30a is essential for photoreceptor survival. However, the roles of Tmem30a in the retinal rod bipolar cells (RBC) remain elusive. To investigate the role of Tmem30a in the RBCs, we generated a RBC-specific Tmem30a knockout (cKO) mouse model using PCP2-Cre line. The Tmem30a cKO mice exhibited defect in RBC function and progressive RBC death. PKCα staining of retinal cryosections from cKO mice revealed a remarkable dendritic sprouting of rod bipolar cells during the early degenerative process. Immunostaining analysis of PSD95 and mGluT6 expression demonstrated that rod bipolar cells in Tmem30a cKO retinas exhibited aberrant dendritic sprouting as a result of impaired synaptic efficacy, which implied a crucial role for Tmem30a in synaptic transmission in the retina. In addition, loss of Tmem30a led to reactive gliosis with increased expression of glial fibrillary acidic protein and CD68. TUNEL staining suggested that apoptotic cell death occurred in the retinal inner nuclear layer (INL). Our data show that loss of Tmem30a in RBCs results in dendritic sprouting of rod bipolar cells, increased astrogliosis and RBC death. Taken together, our studies demonstrate an essential role for Tmem30a in the retinal bipolar cells. Cover Image for this issue: doi: 10.1111/jnc.14492.


Asunto(s)
Proteínas de la Membrana/metabolismo , Células Bipolares de la Retina/metabolismo , Degeneración Retiniana/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Bipolares de la Retina/patología , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/patología , Transmisión Sináptica/fisiología
6.
Retina ; 39(12): 2410-2418, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30260920

RESUMEN

PURPOSE: To report the clinical course of eyes with paraneoplastic retinopathy caused by an autoantibody against transient receptor potential cation channel, subfamily M, member 1 (TRPM1). METHODS: Ten paraneoplastic retinopathy patients with retinal ON-bipolar cell dysfunction, including six melanoma-associated retinopathy, from eight institutions in Japan were evaluated for the presence of an anti-TRPM1 antibody. The results of ophthalmic examinations and the presence of anti-TRPM1 antibody were analyzed. RESULTS: Five patients were positive for the anti-TRPM1 antibody. These patients had similar clinical findings in both eyes at the time of diagnosis; relatively preserved best-corrected visual acuity, absence of fundus and optical coherence tomography abnormalities, and specific abnormalities of the electroretinography (ERG); and negative-type ERGs with bright stimulus flashes. One patient whose retinal ON-bipolar cells remained dysfunctional for the entire testing period, although the anti-TRPM1 antibody had disappeared. On the other hand, the ERGs recovered in 2 cases within 2 years after the onset. One case progressed to additional impairment of the photoreceptors with deterioration of ERGs. One case died and the clinical course was unavailable. CONCLUSION: Paraneoplastic retinopathy patients with retinal ON-bipolar cell dysfunction possess autoantibodies against TRPM1 at the onset of the disease process; however, the clinical course of these eyes can be different.


Asunto(s)
Autoanticuerpos/sangre , Síndromes Paraneoplásicos Oculares/inmunología , Canales Catiónicos TRPM/inmunología , Anciano , Pueblo Asiatico/etnología , Electrorretinografía , Femenino , Angiografía con Fluoresceína , Humanos , Japón/epidemiología , Masculino , Persona de Mediana Edad , Oftalmoscopía , Síndromes Paraneoplásicos Oculares/diagnóstico , Síndromes Paraneoplásicos Oculares/etnología , Células Bipolares de la Retina/patología , Estudios Retrospectivos , Tomografía de Coherencia Óptica
7.
Sensors (Basel) ; 19(3)2019 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-30696016

RESUMEN

This paper presents a fully integrated photodiode-based low-power and low-mismatch stimulator for a subretinal prosthesis. It is known that a subretinal prosthesis achieves 1600-pixel stimulators on a limited single-chip area that is implanted beneath the bipolar cell layer. However, the high-density pixels cause high power dissipation during stimulation and high fabrication costs because of special process technologies such as the complementary metal-oxide semiconductor CMOS image sensor process. In addition, the many residual charges arising from the high-density pixel stimulation have deleterious effects, such as tissue damage and electrode corrosion, on the retina tissue. In this work, we adopted a switched-capacitor current mirror technique for the single-pixel stimulator (SPStim) that enables low power consumption and low mismatch in the subretinal device. The customized P+/N-well photodiode used to sense the incident light in the SPStim also reduces the fabrication cost. The 64-pixel stimulators are fabricated in a standard 0.35-µm CMOS process along with a global digital controller, which occupies a chip area of 4.3 × 3.2 mm² and are ex-vivo demonstrated using a dissected pig eyeball. According to measured results, the SPStim accomplishes a maximum biphasic pulse amplitude of 143 µA, which dissipates an average power of 167 µW in a stimulation period of 5 ms, and an average mismatch of 1.12 % between the cathodic and anodic pulses.


Asunto(s)
Implantación de Prótesis/métodos , Retina/fisiopatología , Células Bipolares de la Retina/patología , Prótesis Visuales , Animales , Técnicas Biosensibles , Humanos , Luz , Células Bipolares de la Retina/fisiología , Semiconductores , Porcinos
8.
Cell Tissue Res ; 371(2): 237-249, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29064077

RESUMEN

Previous studies have revealed a loss of retinal ganglion cells (RGCs) and optic nerve fibers after immunization with the S100B protein. Addition of heat shock protein 27 (HSP27) also leads to a decrease of RGCs. Our present aim has been to analyze various retinal cell types after immunization with S100B or S100B + HSP27 (S100 + HSP). After 28 days, retinas were processed for immunohistology and Western blot. RGCs, immunostained for NeuN, were significantly decreased in the S100 and the S100 + HSP groups. Significantly fewer ChAT+ cells were noted in both groups, whereas parvalbumin+ cells were only affected in the S100 + HSP group. Western blot results also revealed fewer ChAT signals in both immunized groups. No changes were noted with regard to PKCα+ rod bipolar cells, whereas a significant loss of recoverin+ cone bipolar cells was observed in both groups via immunohistology and Western blot. The presynaptic marker Bassoon and the postsynaptic marker PSD95 were significantly reduced in the S100 + HSP group. Opsin+ and rhodopsin+ photoreceptors revealed no changes in either group. Thus, the inner retinal layers are affected by immunization. However, the combination of S100 and HSP27 has a stronger additive effect on the retinal synapses and AII amacrine cells.


Asunto(s)
Células Amacrinas/patología , Autoinmunidad , Glaucoma/inmunología , Glaucoma/patología , Proteínas de Choque Térmico HSP27/inmunología , Inmunización , Proteínas S100/metabolismo , Sinapsis/patología , Células Amacrinas/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratas Endogámicas Lew , Retina/metabolismo , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Sinapsis/metabolismo
9.
J Hum Genet ; 63(8): 893-900, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29760528

RESUMEN

The macula is a unique structure in higher primates, where cone and rod photoreceptors show highest density in the fovea and the surrounding area, respectively. The hereditary macular dystrophies represent a heterozygous group of rare disorders characterized by central visual loss and atrophy of the macula and surrounding retina. Here we report an atypical absence of ON-type bipolar cell response in a Japanese patient with autosomal dominant macular dystrophy (adMD). To identify a causal genetic mutation for the adMD, we performed whole-exome sequencing (WES) on four affected and four-non affected members of the family for three generations, and identified a novel p.C538Y mutation in a post-synaptic gene, LRRTM4. WES analysis revealed seven rare genetic variations in patients. We further referred to our in-house WES data from 1360 families with inherited retinal diseases, and found that only p.C538Y mutation in LRRTM4 was associated with adMD-affected patients. Combinatorial filtration using public database of single-nucleotide polymorphism frequency and genotype-phenotype annotated database identified novel mutation in atypical adMD.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Degeneración Macular/patología , Proteínas de la Membrana/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Células Bipolares de la Retina/patología , Adulto , Secuencia de Aminoácidos , Animales , Pueblo Asiatico , Preescolar , Electrorretinografía , Familia , Femenino , Genes Dominantes , Haplorrinos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proteínas del Tejido Nervioso/química , Linaje , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Secuenciación del Exoma
10.
Adv Exp Med Biol ; 1074: 167-173, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721941

RESUMEN

Noninvasive ocular imaging platforms are undeniably useful in identifying retinal abnormalities. The purpose of this study was to investigate a novel method for integrating information acquired from two independent imaging platforms, AF-SLO and SDOCT, in order to demonstrate retinal perturbations as a result of genetic or pharmacological manipulation. Two cohorts of mice were investigated, Nyx nob and C57BL/6 J. In Nyx nob mice, SLO revealed an atypical but variable amount of autofluorescent foci (AFF); SDOCT showed altered photoreceptor outer segment architecture. Naïve Nyx nob had significantly more AFF than C57BL/6 J, suggesting that Nyx nob have some predisposition for developing AFF. Interestingly, both findings were significantly ameliorated in diabetic Nyx nob mice as compared to the controls. These data were incorporated into a novel analysis plot comparing AF-SLO and SDOCT results. The integration of the qualitative changes and accompanying quantitative analysis approach described herein provide a sensitive means for detecting whether a mouse model is susceptible to degeneration before other hallmark indicators are observed.


Asunto(s)
Microscopía Confocal/métodos , Oftalmoscopía/métodos , Imagen Óptica/métodos , Retina/patología , Enfermedades de la Retina/patología , Tomografía de Coherencia Óptica/métodos , Animales , Diabetes Mellitus Experimental/patología , Retinopatía Diabética/patología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Proteoglicanos , Distribución Aleatoria , Células Bipolares de la Retina/patología , Enfermedades de la Retina/genética , Enfermedades de la Retina/terapia , Epitelio Pigmentado de la Retina/patología , Estreptozocina
11.
Proc Natl Acad Sci U S A ; 112(8): 2593-8, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25675503

RESUMEN

Glaucoma is the second leading cause of blindness in the United States and the world, characterized by progressive degeneration of the optic nerve and retinal ganglion cells (RGCs). Glaucoma patients exhibit an early diffuse loss of retinal sensitivity followed by focal loss of RGCs in sectored patterns. Recent evidence has suggested that this early sensitivity loss may be associated with dysfunctions in the inner retina, but detailed cellular and synaptic mechanisms underlying such sensitivity changes are largely unknown. In this study, we use whole-cell voltage-clamp techniques to analyze light responses of individual bipolar cells (BCs), AII amacrine cells (AIIACs), and ON and sustained OFF alpha-ganglion cells (ONαGCs and sOFFαGCs) in dark-adapted mouse retinas with elevated intraocular pressure (IOP). We present evidence showing that elevated IOP suppresses the rod ON BC inputs to AIIACs, resulting in less sensitive AIIACs, which alter AIIAC inputs to ONαGCs via the AIIAC→cone ON BC→ONαGC pathway, resulting in lower ONαGC sensitivity. The altered AIIAC response also reduces sOFFαGC sensitivity via the AIIAC→sOFFαGC chemical synapses. These sensitivity decreases in αGCs and AIIACs were found in mice with elevated IOP for 3-7 wk, a stage when little RGC or optic nerve degeneration was observed. Our finding that elevated IOP alters neuronal function in the inner retina before irreversible structural damage occurs provides useful information for developing new diagnostic tools and treatments for glaucoma in human patients.


Asunto(s)
Glaucoma/fisiopatología , Presión Intraocular , Fotofobia , Neuronas Retinianas/fisiología , Potenciales de Acción/efectos de la radiación , Células Amacrinas/metabolismo , Células Amacrinas/patología , Animales , Cationes , Canales de Cloruro/metabolismo , Modelos Animales de Enfermedad , Glaucoma/patología , Humanos , Luz , Ratones Endogámicos C57BL , Modelos Biológicos , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Sinapsis/metabolismo
12.
J Neurophysiol ; 118(2): 845-854, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28490646

RESUMEN

GRM6 encodes the metabotropic glutamate receptor 6 (mGluR6) used by retinal depolarizing bipolar cells (DBCs). Mutations in GRM6 lead to DBC dysfunction and underlie the human condition autosomal recessive complete congenital stationary night blindness. Mouse mutants for Grm6 are important models for this condition. Here we report a new Grm6 mutant, identified in an electroretinogram (ERG) screen of mice maintained at The Jackson Laboratory. The Grm6nob8 mouse has a reduced-amplitude b-wave component of the ERG, which reflects light-evoked DBC activity. Sequencing identified a missense mutation that converts a highly conserved methionine within the ligand binding domain to leucine (p.Met66Leu). Consistent with prior studies of Grm6 mutant mice, the laminar size and structure in the Grm6nob8 retina were comparable to control. The Grm6nob8 phenotype is distinguished from other Grm6 mutants that carry a null allele by a reduced but not absent ERG b-wave, decreased but present expression of mGluR6 at DBC dendritic tips, and mislocalization of mGluR6 to DBC somas. Consistent with a reduced but not absent b-wave, there were a subset of retinal ganglion cells whose responses to light onset have times to peak within the range of those in control retinas. These data indicate that the p.Met66Leu mutant mGluR6 is trafficked less than control. However, the mGluR6 that is localized to the DBC dendritic tips is able to initiate DBC signal transduction. The Grm6nob8 mouse extends the Grm6 allelic series and will be useful for elucidating the role of mGluR6 in DBC signal transduction and in human disease.NEW & NOTEWORTHY This article describes a mouse model of the human disease complete congenital stationary night blindness in which the mutation reduces but does not eliminate GRM6 expression and bipolar cell function, a distinct phenotype from that seen in other Grm6 mouse models.


Asunto(s)
Enfermedades Hereditarias del Ojo/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Mutación Missense , Miopía/metabolismo , Ceguera Nocturna/metabolismo , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Células Bipolares de la Retina/metabolismo , Visión Ocular/fisiología , Animales , Dendritas/metabolismo , Dendritas/patología , Dendritas/efectos de la radiación , Modelos Animales de Enfermedad , Electrorretinografía , Proteínas de Escherichia coli , Enfermedades Hereditarias del Ojo/genética , Enfermedades Hereditarias del Ojo/patología , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Miopía/genética , Miopía/patología , Ceguera Nocturna/genética , Ceguera Nocturna/patología , Células Bipolares de la Retina/patología , Factores de Transcripción
13.
J Neurosci ; 35(13): 5422-33, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25834065

RESUMEN

Diabetes leads to dysfunction of the neural retina before and independent of classical microvascular diabetic retinopathy, but previous studies have failed to demonstrate which neurons and circuits are affected at the earliest stages. Here, using patch-clamp recording and two-photon Ca(2+) imaging in rat retinal slices, we investigated diabetes-evoked changes in a microcircuit consisting of rod bipolar cells and their dyad postsynaptic targets, AII and A17 amacrine cells, which play an essential role in processing scotopic visual signals. AII amacrines forward their signals to ON- and OFF-cone bipolar cells and A17 amacrines provide GABAergic feedback inhibition to rod bipolar cells. Whereas Ca(2+)-permeable AMPA receptors mediate input from rod bipolar cells to both AII and A17 amacrines, diabetes changes the synaptic receptors on A17, but not AII amacrine cells. This was expressed as a change in pharmacological properties and single-channel conductance of the synaptic receptors, consistent with an upregulation of the AMPA receptor GluA2 subunit and reduced Ca(2+) permeability. In addition, two-photon imaging revealed reduced agonist-evoked influx of Ca(2+) in dendritic varicosities of A17 amacrine cells from diabetic compared with normal animals. Because Ca(2+)-permeable receptors in A17 amacrine cells mediate synaptic release of GABA, the reduced Ca(2+) permeability of these receptors in diabetic animals leads to reduced release of GABA, followed by disinhibition and increased release of glutamate from rod bipolar cells. This perturbation of neuron and microcircuit dynamics can explain the decreased dynamic range and sensitivity of scotopic vision that has been observed in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Vías Nerviosas/patología , Células Fotorreceptoras Retinianas Bastones/patología , Adamantano/análogos & derivados , Adamantano/farmacología , Células Amacrinas/efectos de los fármacos , Células Amacrinas/metabolismo , Animales , Calcio/metabolismo , Diabetes Mellitus Experimental/metabolismo , Potenciales Postsinápticos Excitadores , Femenino , Ácido Glutámico/metabolismo , Ratas , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/biosíntesis , Receptores AMPA/metabolismo , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Regulación hacia Arriba , Ácido gamma-Aminobutírico/metabolismo
14.
Genes Cells ; 20(5): 408-26, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25757744

RESUMEN

Mef2 transcription factors play a crucial role in cardiac and skeletal muscle differentiation. We found that Mef2d is highly expressed in the mouse retina and its loss causes photoreceptor degeneration similar to that observed in human retinitis pigmentosa patients. Electroretinograms (ERGs) were severely impaired in Mef2d-/- mice. Immunohistochemistry showed that photoreceptor and bipolar cell synapse protein levels severely decreased in the Mef2d-/- retina. Expression profiling by microarray analysis showed that Mef2d is required for the expression of various genes in photoreceptor and bipolar cells, including cone arrestin, Guca1b, Pde6h and Cacna1s, which encode outer segment and synapse proteins. We also observed that Mef2d synergistically activates the cone arrestin (Arr3) promoter with Crx, suggesting that functional cooperation between Mef2d and Crx is important for photoreceptor cell gene regulation. Taken together, our results show that Mef2d is essential for photoreceptor and bipolar cell gene expression, either independently or cooperatively with Crx.


Asunto(s)
Diferenciación Celular , Factores de Transcripción MEF2/metabolismo , Células Fotorreceptoras de Vertebrados/citología , Células Fotorreceptoras de Vertebrados/metabolismo , Células Bipolares de la Retina/citología , Células Bipolares de la Retina/metabolismo , Animales , Arrestinas/genética , Diferenciación Celular/genética , Electrorretinografía , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Factores de Transcripción MEF2/genética , Ratones , Ratones Noqueados , Células Fotorreceptoras de Vertebrados/patología , Células Fotorreceptoras de Vertebrados/ultraestructura , Regiones Promotoras Genéticas , Unión Proteica , Células Bipolares de la Retina/patología , Células Bipolares de la Retina/ultraestructura , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Segmento Interno de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Interno de las Células Fotorreceptoras Retinianas/patología , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Sinapsis/genética , Sinapsis/metabolismo , Transactivadores/metabolismo , Transcripción Genética
15.
Mol Vis ; 22: 1468-1489, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28050121

RESUMEN

PURPOSE: Studies of neuronal development in the retina often examine the stages of proliferation, differentiation, and synaptic development, albeit independently. Our goal was to determine if a known neurotoxicant insult to a population of retinal progenitor cells (RPCs) would affect their eventual differentiation and synaptic development. To that end, we used our previously published human equivalent murine model of low-level gestational lead exposure (GLE). Children and animals with GLE exhibit increased scotopic electroretinogram a- and b-waves. Adult mice with GLE exhibit an increased number of late-born RPCs, a prolonged period of RPC proliferation, and an increased number of late-born rod photoreceptors and rod and cone bipolar cells (BCs), with no change in the number of late-born Müller glial cells or early-born neurons. The specific aims of this study were to determine whether increased and prolonged RPC proliferation alters the spatiotemporal differentiation and synaptic development of rods and BCs in early postnatal GLE retinas compared to control retinas. METHODS: C57BL/6N mouse pups were exposed to lead acetate via drinking water throughout gestation and until postnatal day 10, which is equivalent to the human gestation period for retinal neurogenesis. RT-qPCR, immunohistochemical analysis, and western blots of well-characterized, cell-specific genes and proteins were performed at embryonic and early postnatal ages to assess rod and cone photoreceptor differentiation, rod and BC differentiation and synaptic development, and Müller glial cell differentiation. RESULTS: Real-time quantitative PCR (RT-qPCR) with the rod-specific transcription factors Nrl, Nr2e3, and Crx and the rod-specific functional gene Rho, along with central retinal confocal studies with anti-recoverin and anti-rhodopsin antibodies, revealed a two-day delay in the differentiation of rod photoreceptors in GLE retinas. Rhodopsin immunoblots supported this conclusion. No changes in glutamine synthetase gene or protein expression, a marker for late-born Müller glial cells, were observed in the developing retinas. In the retinas from the GLE mice, anti-PKCα, -Chx10 (Vsx2) and -secretagogin antibodies revealed a two- to three-day delay in the differentiation of rod and cone BCs, whereas the expression of the proneural and BC genes Otx2 and Chx10, respectively, increased. In addition, confocal studies of proteins associated with functional synapses (e.g., vesicular glutamate transporter 1 [VGluT1], plasma membrane calcium ATPase [PMCA], transient receptor potential channel M1 [TRPM1], and synaptic vesicle glycoprotein 2B [SV2B]) revealed a two-day delay in the formation of the outer and inner plexiform layers of the GLE retinas. Moreover, several markers revealed that the initiation of the differentiation and intensity of the labeling of early-born cells in the retinal ganglion cell and inner plexiform layers were not different in the control retinas. CONCLUSIONS: Our combined gene, confocal, and immunoblot findings revealed that the onset of rod and BC differentiation and their subsequent synaptic development is delayed by two to three days in GLE retinas. These results suggest that perturbations during the early proliferative stages of late-born RPCs fated to be rods and BCs ultimately alter the coordinated time-dependent progression of rod and BC differentiation and synaptic development. These GLE effects were selective for late-born neurons. Although the molecular mechanisms are unknown, alterations in soluble neurotrophic factors and/or their receptors are likely to play a role. Since neurodevelopmental delays and altered synaptic connectivity are associated with neuropsychiatric and behavioral disorders as well as cognitive deficits, future work is needed to determine if similar effects occur in the brains of GLE mice and whether children with GLE experience similar delays in retinal and brain neuronal differentiation and synaptic development.


Asunto(s)
Diferenciación Celular , Plomo/toxicidad , Neurogénesis , Efectos Tardíos de la Exposición Prenatal/patología , Células Bipolares de la Retina/patología , Células Fotorreceptoras Retinianas Bastones/patología , Envejecimiento/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glutamato-Amoníaco Ligasa/metabolismo , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas Sprague-Dawley , Células Bipolares de la Retina/efectos de los fármacos , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Rodopsina/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
16.
Mol Ther ; 23(1): 7-16, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25095892

RESUMEN

Most inherited retinal dystrophies display progressive photoreceptor cell degeneration leading to severe visual impairment. Optogenetic reactivation of retinal neurons mediated by adeno-associated virus (AAV) gene therapy has the potential to restore vision regardless of patient-specific mutations. The challenge for clinical translatability is to restore a vision as close to natural vision as possible, while using a surgically safe delivery route for the fragile degenerated retina. To preserve the visual processing of the inner retina, we targeted ON bipolar cells, which are still present at late stages of disease. For safe gene delivery, we used a recently engineered AAV variant that can transduce the bipolar cells after injection into the eye's easily accessible vitreous humor. We show that AAV encoding channelrhodopsin under the ON bipolar cell-specific promoter mediates long-term gene delivery restricted to ON-bipolar cells after intravitreal administration. Channelrhodopsin expression in ON bipolar cells leads to restoration of ON and OFF responses at the retinal and cortical levels. Moreover, light-induced locomotory behavior is restored in treated blind mice. Our results support the clinical relevance of a minimally invasive AAV-mediated optogenetic therapy for visual restoration.


Asunto(s)
Ceguera/terapia , Dependovirus/genética , Terapia Genética/métodos , Células Bipolares de la Retina/metabolismo , Degeneración Retiniana/terapia , Animales , Conducta Animal , Ceguera/genética , Ceguera/patología , Channelrhodopsins , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Ingeniería Genética , Vectores Genéticos , Inyecciones Intravítreas , Luz , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Células Bipolares de la Retina/patología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Percepción Visual/genética , Cuerpo Vítreo
17.
Graefes Arch Clin Exp Ophthalmol ; 254(3): 497-503, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26743749

RESUMEN

BACKGROUND: Osmotic swelling of neurons and glial cells contributes to retinal edema and neurodegeneration. BDNF, a major neuroprotectant in the retina, was shown to inhibit osmotic swelling of glial (Müller) and bipolar cells in the rat retina; the effect of BDNF on the bipolar cell swelling is mediated by inducing a release of neuroprotective cytokines from Müller cells (Berk et al., Neuroscience 295:175-186, 2015). We determined whether BDNF-mediated cell volume regulation was altered after transient retinal ischemia. METHODS: Retinal slices from the eyes of rats that underwent a 1-h pressure-induced retinal ischemia and from control eyes were superfused with a hypoosmotic solution. RESULTS: Exogenous BDNF prevented osmotic swelling of Müller cells in both control and post-ischemic retinal slices. BDNF also prevented osmotic swelling of bipolar cells in the control retina, but not in the ischemic retina. On the other hand, exogenous bFGF prevented the swelling of both Müller and bipolar cells in the ischemic retina. Freshly isolated Müller cells of control retinas displayed immunoreactivity of truncated but not full-length TrkB. In contrast, Müller cells of post-ischemic retinas displayed immunoreactivity of both TrkB isoforms. Bipolar cells isolated from control and post-ischemic retinas were immunolabeled for both TrkB isoforms. CONCLUSIONS: The data may suggest that the ischemic abrogation of the BDNF effect in bipolar cells is related to altered BDNF receptor expression in Müller cells. Glial upregulation of full-length TrkB may support the survival of Müller cells in the ischemic retina, but may impair the BDNF-induced release of neuroprotective cytokines such as bFGF from Müller cells.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Tamaño de la Célula/efectos de los fármacos , Células Ependimogliales/metabolismo , Isquemia/metabolismo , Receptor trkB/metabolismo , Células Bipolares de la Retina/metabolismo , Vasos Retinianos/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Ependimogliales/patología , Femenino , Inmunohistoquímica , Isquemia/patología , Masculino , Presión Osmótica , Ratas , Ratas Long-Evans , Células Bipolares de la Retina/patología , Vasos Retinianos/patología , Transducción de Señal
18.
Graefes Arch Clin Exp Ophthalmol ; 254(10): 1951-1956, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27084085

RESUMEN

PURPOSE: To describe congenital stationary night blindness (CSNB) with negative electroretinogram, hypoplastic discs, nystagmus and thinning of the inner nuclear layer (INL). METHODS: Retinal structure was analyzed qualitatively with spectral domain optical coherence tomography and wide field imaging. Retinal function was evaluated with full-field electroretinography (ffERG). Molecular genetic testing included next-generation sequencing (NGS) of the known genes involved in CSNB. RESULTS: Patients presented with CSNB presented with nystagmus, high myopia, hypoplastic discs and negative ffERG with no measurable rod response. The retinas appeared normal and automated segmentation of retinal layers demonstrated a relative reduction of thickness of the INL. There was no significant change in the ffERG after prolonged 2 hour dark adaptation compared to standard 30 minute dark adaptation. Affected family members harboured the homozygous 1-bp deletion c.2394delC in exon 18 of the TRPM1 gene, whereas their unaffected parents were heterozygous carriers. CONCLUSIONS: This data expands the genotype and phenotype spectrum of CSNB. The lack of improvement of rod responses after prolonged dark adaptation, together with thinning of the INL, is compatible with postreceptoral transmission dysfunction in the bipolar cells. Such knowledge may prove useful in future development of treatment for outer retinal dystrophies, using opsin genes to restore light responses in survivor neurons in the inner retina.


Asunto(s)
Enfermedades en Gemelos/genética , Anomalías del Ojo/genética , Enfermedades Hereditarias del Ojo/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Miopía/genética , Ceguera Nocturna/genética , Disco Óptico/anomalías , Células Bipolares de la Retina/patología , Enfermedades de la Retina/genética , Canales Catiónicos TRPM/genética , Consanguinidad , Adaptación a la Oscuridad , Electrorretinografía , Anomalías del Ojo/diagnóstico por imagen , Anomalías del Ojo/fisiopatología , Enfermedades Hereditarias del Ojo/diagnóstico por imagen , Enfermedades Hereditarias del Ojo/fisiopatología , Mutación del Sistema de Lectura , Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico por imagen , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Genotipo , Humanos , Masculino , Miopía/diagnóstico , Miopía/diagnóstico por imagen , Miopía/fisiopatología , Ceguera Nocturna/diagnóstico por imagen , Ceguera Nocturna/fisiopatología , Nistagmo Patológico/diagnóstico , Nistagmo Patológico/genética , Nistagmo Patológico/fisiopatología , Linaje , Reacción en Cadena de la Polimerasa , Enfermedades de la Retina/diagnóstico por imagen , Enfermedades de la Retina/fisiopatología , Estudios Retrospectivos , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología , Adulto Joven
19.
Stem Cells ; 32(6): 1480-92, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24532057

RESUMEN

Human induced pluripotent stem cells (hiPSCs) have been shown to differentiate along the retinal lineage in a manner that mimics normal mammalian development. Under certain culture conditions, hiPSCs form optic vesicle-like structures (OVs), which contain proliferating progenitors capable of yielding all neural retina (NR) cell types over time. Such observations imply conserved roles for regulators of retinogenesis in hiPSC-derived cultures and the developing embryo. However, whether and to what extent this assumption holds true has remained largely uninvestigated. We examined the role of a key NR transcription factor, visual system homeobox 2 (VSX2), using hiPSCs derived from a patient with microphthalmia caused by an R200Q mutation in the VSX2 homeodomain region. No differences were noted between (R200Q)VSX2 and sibling control hiPSCs prior to OV generation. Thereafter, (R200Q)VSX2 hiPSC-OVs displayed a significant growth deficit compared to control hiPSC-OVs, as well as increased production of retinal pigmented epithelium at the expense of NR cell derivatives. Furthermore, (R200Q)VSX2 hiPSC-OVs failed to produce bipolar cells, a distinctive feature previously observed in Vsx2 mutant mice. (R200Q)VSX2 hiPSC-OVs also demonstrated delayed photoreceptor maturation, which could be overcome via exogenous expression of wild-type VSX2 at early stages of retinal differentiation. Finally, RNAseq analysis on isolated hiPSC-OVs implicated key transcription factors and extracellular signaling pathways as potential downstream effectors of VSX2-mediated gene regulation. Our results establish hiPSC-OVs as versatile model systems to study retinal development at stages not previously accessible in humans and support the bona fide nature of hiPSC-OV-derived retinal progeny.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Retina/embriología , Retina/metabolismo , Factores de Transcripción/metabolismo , Adulto , Sustitución de Aminoácidos , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Masculino , Ratones , Mutación/genética , Fenotipo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Retina/patología , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/patología , Epitelio Pigmentado de la Retina/embriología , Epitelio Pigmentado de la Retina/patología , Análisis de Secuencia de ARN , Transducción de Señal/genética , Factores de Transcripción/genética , Transcriptoma/genética
20.
Doc Ophthalmol ; 130(1): 71-6, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25391361

RESUMEN

PURPOSE: To report a patient with cancer-associated retinopathy and retinal ON-bipolar cell dysfunction who had a resolution of the electroretinograms (ERGs) after a resection of an ovarian cancer and chemotherapy. CASE REPORT: A 71-year-old Japanese female patient visited us complaining of night blindness and photopsia in both eyes for 6 months. Her visual acuity was 20/20 in both eyes, and fundus examination, fluorescence angiography, and optical coherence tomography showed no abnormalities in both eyes. The rod responses of the ERGs were absent and bright-flash ERGs were electronegative. The ON responses of the focal macular ERGs and full-field long-flash ERGs were absent. These ERG findings indicate an ON-bipolar cell dysfunction. A general physical examination revealed the presence of ovarian cancer. After resection of the ovarian cancer and adjuvant chemotherapy, the ERGs of the left eye completely recovered within 2 years and those of right eye recovered subsequently. The autoantibody against transient receptor potential melastatin 1 (TRPM1) was not detected in the serum. CONCLUSION: Our case demonstrates that retinal ON-bipolar dysfunction can be caused by ovarian cancer. Our case indicates that some autoantibodies against other than TRPM1 might cause transient dysfunction of retinal ON-bipolar cells.


Asunto(s)
Síndromes Paraneoplásicos Oculares/etiología , Células Bipolares de la Retina/patología , Células Ganglionares de la Retina/patología , Anciano , Quimioterapia Adyuvante , Electrorretinografía , Femenino , Angiografía con Fluoresceína , Humanos , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/cirugía , Síndromes Paraneoplásicos Oculares/fisiopatología , Estimulación Luminosa , Tomografía de Coherencia Óptica
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda