Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Neurosci ; 17: 1306006, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38099150

RESUMEN

Introduction: Rod bipolar cells (RBCs) faithfully transmit light-driven signals from rod photoreceptors in the outer retina to third order neurons in the inner retina. Recently, significant work has focused on the role of leucine-rich repeat (LRR) proteins in synaptic development and signal transduction at RBC synapses. We previously identified trophoblast glycoprotein (TPBG) as a novel transmembrane LRR protein localized to the dendrites and axon terminals of RBCs. Methods: We examined the effects on RBC physiology and retinal processing of TPBG genetic knockout in mice using immunofluorescence and electron microscopy, electroretinogram recording, patch-clamp electrophysiology, and time-resolved membrane capacitance measurements. Results: The scotopic electroretinogram showed a modest increase in the b-wave and a marked attenuation in oscillatory potentials in the TPBG knockout. No effect of TPBG knockout was observed on the RBC dendritic morphology, TRPM1 currents, or RBC excitability. Because scotopic oscillatory potentials primarily reflect RBC-driven rhythmic activity of the inner retina, we investigated the contribution of TPBG to downstream transmission from RBCs to third-order neurons. Using electron microscopy, we found shorter synaptic ribbons in TPBG knockout axon terminals in RBCs. Time-resolved capacitance measurements indicated that TPBG knockout reduces synaptic vesicle exocytosis and subsequent GABAergic reciprocal feedback without altering voltage-gated Ca2+ currents. Discussion: TPBG is required for normal synaptic ribbon development and efficient neurotransmitter release from RBCs to downstream cells. Our results highlight a novel synaptic role for TPBG at RBC ribbon synapses and support further examination into the mechanisms by which TPBG regulates RBC physiology and circuit function.

2.
J Comp Neurol ; 528(10): 1660-1671, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31891182

RESUMEN

We recently identified the leucine-rich repeat (LRR) adhesion protein, trophoblast glycoprotein (TPBG), as a novel PKCα-dependent phosphoprotein in retinal rod bipolar cells (RBCs). Since TPBG has not been thoroughly examined in the retina, this study characterizes the localization and expression patterns of TPBG in the developing and adult mouse retina using two antibodies, one against the N-terminal LRR domain and the other against the C-terminal PDZ-interacting motif. Both antibodies labeled RBC dendrites in the outer plexiform layer and axon terminals in the IPL, as well as a putative amacrine cell with their cell bodies in the inner nuclear layer (INL) and a dense layer in the middle of the inner plexiform layer (IPL). In live transfected HEK293 cells, TPBG was localized to the plasma membrane with the N-terminal LRR domain facing the extracellular space. TPBG immunofluorescence in RBCs was strongly altered by the loss of TRPM1 in the adult retina, with significantly less dendritic and axon terminal labeling in TRPM1 knockout compared to wild type, despite no change in total TPBG detected by immunoblotting. During retinal development, TPBG expression increases dramatically just prior to eye opening with a time course closely correlated with that of TRPM1 expression. In the retina, LRR proteins have been implicated in the development and maintenance of functional bipolar cell synapses, and TPBG may play a similar role in RBCs.


Asunto(s)
Antígenos de Superficie/metabolismo , Glicoproteínas de Membrana/metabolismo , Neurogénesis/fisiología , Células Bipolares de la Retina/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Canales Catiónicos TRPM/metabolismo
3.
J Proteomics ; 206: 103423, 2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31255707

RESUMEN

Adjusting to a wide range of light intensities is an essential feature of retinal rod bipolar cell (RBC) function. While persuasive evidence suggests this modulation involves phosphorylation by protein kinase C-alpha (PKCα), the targets of PKCα phosphorylation in the retina have not been identified. PKCα activity and phosphorylation in RBCs was examined by immunofluorescence confocal microscopy using a conformation-specific PKCα antibody and antibodies to phosphorylated PKC motifs. PKCα activity was dependent on light and expression of TRPM1, and RBC dendrites were the primary sites of light-dependent phosphorylation. PKCα-dependent retinal phosphoproteins were identified using a phosphoproteomics approach to compare total protein and phosphopeptide abundance between phorbol ester-treated wild type and PKCα knockout (PKCα-KO) mouse retinas. Phosphopeptide mass spectrometry identified over 1100 phosphopeptides in mouse retina, with 12 displaying significantly greater phosphorylation in WT compared to PKCα-KO samples. The differentially phosphorylated proteins fall into the following functional groups: cytoskeleton/trafficking (4 proteins), ECM/adhesion (2 proteins), signaling (2 proteins), transcriptional regulation (3 proteins), and homeostasis/metabolism (1 protein). Two strongly differentially expressed phosphoproteins, BORG4 and TPBG, were localized to the synaptic layers of the retina, and may play a role in PKCα-dependent modulation of RBC physiology. Data are available via ProteomeXchange with identifier PXD012906. SIGNIFICANCE: Retinal rod bipolar cells (RBCs), the second-order neurons of the mammalian rod visual pathway, are able to modulate their sensitivity to remain functional across a wide range of light intensities, from starlight to daylight. Evidence suggests that this modulation requires the serine/threonine kinase, PKCα, though the specific mechanism by which PKCα modulates RBC physiology is unknown. This study examined PKCα phosophorylation patterns in mouse rod bipolar cells and then used a phosphoproteomics approach to identify PKCα-dependent phosphoproteins in the mouse retina. A small number of retinal proteins showed significant PKCα-dependent phosphorylation, including BORG4 and TPBG, suggesting a potential contribution to PKCα-dependent modulation of RBC physiology.


Asunto(s)
Fosfoproteínas/metabolismo , Proteína Quinasa C-alfa/metabolismo , Procesamiento Proteico-Postraduccional , Proteoma/metabolismo , Retina/metabolismo , Animales , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/análisis , Fosforilación/genética , Proteína Quinasa C-alfa/genética , Procesamiento Proteico-Postraduccional/genética , Proteoma/análisis , Células Bipolares de la Retina/química , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/fisiología , Células Fotorreceptoras Retinianas Bastones/química , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/fisiología , Canales Catiónicos TRPM/genética
4.
Invest Ophthalmol Vis Sci ; 60(6): 2330-2335, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31117125

RESUMEN

Purpose: Melanoma-associated retinopathy (MAR) is a paraneoplastic syndrome associated with cutaneous malignant melanoma (CMM). Visual symptoms include night blindness, photopsia, and reduced-contrast sensitivity. An abnormal ERG b-wave and the presence of anti-bipolar cell autoantibodies, including autoantibodies reacting with the ON-bipolar cell TRPM1 channel, help to confirm the diagnosis. The goal of this study was to determine if CMM patients without visual symptoms also express anti-TRPM1 autoantibodies. Methods: Serum samples from 15 CMM patients were tested using three assays: immunofluorescent labeling of TRPM1-transfected HEK cells, immunofluorescent labeling of retinal sections from wild-type and TRPM1 knockout mice, and immunoblot detection of a bacterially produced recombinant TRPM1 peptide. Results: Serum specimens from 5 of the 15 CMM patients without declared visual symptoms were positive for anti-TRPM1 autoantibodies in at least one of the three assays. One of 50 control sera from patients not known to have cancer was also weakly reactive with the TRPM1 peptide. Conclusions: Autoantibodies against TRPM1 are present in CMM patient sera without self-reported visual symptoms. Most patients had advanced (stage III and IV) disease and were undergoing aggressive treatments, including immunotherapy. It is unknown if immunotherapy affects the expression of TRPM1 autoantibodies. The presence of TRPM1 autoantibodies may predispose patients for MAR.


Asunto(s)
Autoanticuerpos/metabolismo , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Canales Catiónicos TRPM/inmunología , Animales , Estudios de Casos y Controles , Células Cultivadas , Humanos , Melanoma/inmunología , Ratones , Síndromes Paraneoplásicos Oculares/inmunología , Neoplasias Cutáneas/inmunología , Melanoma Cutáneo Maligno
5.
Methods Mol Biol ; 1622: 225-239, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28674812

RESUMEN

Adenosine is an important modulator of metabolic activity with powerful tissue and cell protective functions. Adenosine kinase (ADK), the major adenosine-regulating enzyme, is critical to adapt its intra- and extracellular levels in response to environmental changes. Lentiviral RNAi-mediated downregulation of ADK in human mesenchymal stem cells (hMSCs) has therefore been considered an effective tool for engineering therapeutically effective adenosine-releasing cell grafts that could constitute patient-identical autologous implants for clinical application. We constructed lentiviral vectors that co-express miRNA directed against ADK and an emerald green fluorescent protein (EmGFP) reporter gene. Following lentiviral transduction of hMSCs, we demonstrated up to 80% downregulation of ADK and 98% transduction efficiency. Transduced hMSCs continued to express EmGFP after four to six consecutive passages, and EmGFP-positive hMSC grafts survived in the hippocampal fissure of mouse brains and provided efficient adenosine-dependent neuroprotection in a mouse model of seizure-induced cell loss.


Asunto(s)
Adenosina/metabolismo , Ingeniería Genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Actinas/metabolismo , Adenosina Quinasa/genética , Adenosina Quinasa/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ingeniería Genética/métodos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Lentivirus/genética , Trasplante de Células Madre Mesenquimatosas , Interferencia de ARN , Transducción Genética
6.
Invest Ophthalmol Vis Sci ; 58(5): 2732-2738, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28549093

RESUMEN

Purpose: Melanoma-associated retinopathy (MAR) is a paraneoplastic syndrome associated with malignant melanoma and the presence of anti-retinal autoantibodies, including autoantibodies against transient receptor potential melanopsin 1 (TRPM1), a cation channel expressed by both melanocytes and retinal bipolar cells. The goal of this study was to further map the antigenic epitope. Methods: Patient sera were tested by immunofluorescence and Western blotting on HEK293 cells transfected with enhanced green fluorescent protein (EGFP)-TRPM1 fusion constructs and mouse retina sections. Results: The epitope recognized by MAR patient sera was mapped to a region encoded by exons 9 and 10 of the human TRPM1 gene. This region of TRPM1 is highly conserved with TRPM3, and indeed MAR sera were found to cross-react with TRPM3, a closely related channel expressed in the retinal pigment epithelium (RPE). Conclusions: These results indicate that TRPM1 autoantibodies in MAR patient sera recognize a short, intracellular segment of TRPM1. Cross-reactivity with TRPM3 in the RPE may account for other visual symptoms that are experienced by some MAR patients such as retinal and RPE detachments. We propose that TRPM1 autoantibodies are generated in response to abnormal TRPM1 polypeptides encoded by an alternate mRNA splice variant expressed by malignant melanocytes.


Asunto(s)
Autoanticuerpos/sangre , Epítopos , Síndromes Paraneoplásicos Oculares/inmunología , Canales Catiónicos TRPM/inmunología , Secuencia de Aminoácidos , Animales , Western Blotting , Reacciones Cruzadas , Exones/genética , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas Fluorescentes Verdes , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/genética , Canales Catiónicos TRPM/genética , Transfección
7.
Nucleic Acids Res ; 42(5): 3119-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371280

RESUMEN

DNA mismatches that occur between vector homology arms and chromosomal target sequences reduce gene targeting frequencies in several species; however, this has not been reported in human cells. Here we demonstrate that even a single mismatched base pair can significantly decrease human gene targeting frequencies. In addition, we show that homology arm polymorphisms can be used to direct allele-specific targeting or to improve unfavorable vector designs that introduce deletions.


Asunto(s)
Marcación de Gen , Polimorfismo Genético , Disparidad de Par Base , Línea Celular Tumoral , Cromosomas/química , Sitios Genéticos , Vectores Genéticos/química , Humanos , Polimorfismo de Nucleótido Simple
8.
Mol Ther ; 20(1): 204-13, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22031238

RESUMEN

Osteogenesis imperfecta (OI) is caused by dominant mutations in the type I collagen genes. In principle, the skeletal abnormalities of OI could be treated by transplantation of patient-specific, bone-forming cells that no longer express the mutant gene. Here, we develop this approach by isolating mesenchymal cells from OI patients, inactivating their mutant collagen genes by adeno-associated virus (AAV)-mediated gene targeting, and deriving induced pluripotent stem cells (iPSCs) that were expanded and differentiated into mesenchymal stem cells (iMSCs). Gene-targeted iMSCs produced normal collagen and formed bone in vivo, but were less senescent and proliferated more than bone-derived MSCs. To generate iPSCs that would be more appropriate for clinical use, the reprogramming and selectable marker transgenes were removed by Cre recombinase. These results demonstrate that the combination of gene targeting and iPSC derivation can be used to produce potentially therapeutic cells from patients with genetic disease.


Asunto(s)
Colágeno/biosíntesis , Colágeno/genética , Terapia Genética , Células Madre Pluripotentes Inducidas/trasplante , Osteogénesis Imperfecta/terapia , Osteogénesis/genética , Adolescente , Diferenciación Celular , Niño , Preescolar , Orden Génico , Marcación de Gen/métodos , Técnicas de Transferencia de Gen , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/citología , Osteogénesis Imperfecta/genética , Transgenes
9.
Methods Mol Biol ; 650: 225-40, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20686955

RESUMEN

Adenosine is an important modulator of metabolic activity with powerful tissue- and cell-protective functions. Adenosine kinase (ADK), the major adenosine-regulating enzyme, is critical to adapt its intra- and extra-cellular levels in response to environmental changes. Lentiviral RNAi-mediated down-regulation of ADK in human mesenchymal stem cells (hMSCs) has therefore been considered an effective tool for engineering therapeutically effective adenosine-releasing cell grafts that could constitute patient-identical autologous implants for clinical application. We constructed lentiviral vectors that coexpress miRNA directed against ADK and an emerald green fluorescent protein (EmGFP) reporter gene. Following lentiviral transduction of hMSCs, we demonstrated up to 80% down-regulation of ADK and 98% transduction efficiency. Transduced hMSCs continued to express EmGFP after 4-6 consecutive passages and EmGFP-positive hMSC grafts survived in the hippocampal fissure of mouse brains and provided efficient adenosine-dependent neuroprotection in a mouse model of seizure-induced cell loss.


Asunto(s)
Adenosina/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Adenosina Quinasa/genética , Adenosina Quinasa/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN/fisiología , Convulsiones/patología , Convulsiones/terapia
10.
Epilepsy Res ; 84(2-3): 238-41, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19217263

RESUMEN

A novel generation of silk-based brain implants engineered to release adenosine was recently shown to provide robust seizure suppression in kindled rats. As a first step to develop stem cell-coated silk-based 3D-scaffolds for the therapeutic long-term delivery of adenosine we engineered human mesenchymal stem cells (hMSCs) to release adenosine. Here we demonstrate reduction of chronic seizures in a mouse model of CA3-selective epileptogenesis after infrahippocampal transplantation of adenosine-releasing hMSCs.


Asunto(s)
Adenosina/uso terapéutico , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Convulsiones/tratamiento farmacológico , Ingeniería de Tejidos/métodos , Adenosina/biosíntesis , Adenosina Quinasa/antagonistas & inhibidores , Adenosina Quinasa/genética , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Electroencefalografía/métodos , Proteínas Fluorescentes Verdes/genética , Hipocampo/cirugía , Humanos , Indoles , Ácido Kaínico , Ratones , MicroARNs/genética , Convulsiones/inducido químicamente , Transfección/métodos
11.
J Clin Invest ; 118(2): 571-82, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18172552

RESUMEN

Astrogliosis is a pathological hallmark of the epileptic brain. The identification of mechanisms that link astrogliosis to neuronal dysfunction in epilepsy may provide new avenues for therapeutic intervention. Here we show that astrocyte-expressed adenosine kinase (ADK), a key negative regulator of the brain inhibitory molecule adenosine, is a potential predictor and modulator of epileptogenesis. In a mouse model of focal epileptogenesis, in which astrogliosis is restricted to the CA3 region of the hippocampus, we demonstrate that upregulation of ADK and spontaneous focal electroencephalographic seizures were both restricted to the affected CA3. Furthermore, spontaneous seizures in CA3 were mimicked in transgenic mice by overexpression of ADK in this brain region, implying that overexpression of ADK without astrogliosis is sufficient to cause seizures. Conversely, after pharmacological induction of an otherwise epileptogenesis-precipitating acute brain injury, transgenic mice with reduced forebrain ADK were resistant to subsequent epileptogenesis. Likewise, ADK-deficient ES cell-derived brain implants suppressed astrogliosis, upregulation of ADK, and spontaneous seizures in WT mice when implanted after the epileptogenesis-precipitating brain injury. Our findings suggest that astrocyte-based ADK provides a critical link between astrogliosis and neuronal dysfunction in epilepsy.


Asunto(s)
Adenosina Quinasa/metabolismo , Epilepsias Parciales/genética , Convulsiones/genética , Adenosina Quinasa/análisis , Adenosina Quinasa/genética , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Astrocitos/patología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Epilepsias Parciales/inducido químicamente , Epilepsias Parciales/patología , Ácido Kaínico/toxicidad , Masculino , Ratones , Ratones Transgénicos , Pronóstico , Convulsiones/inducido químicamente , Convulsiones/patología
12.
Exp Neurol ; 208(1): 26-37, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17716659

RESUMEN

Cell therapies based on focal delivery of the inhibitory neuromodulator adenosine were previously shown to provide potent seizure suppression in animal models of epilepsy. However, hitherto used therapeutic cells were derived from rodents and thus not suitable for clinical applications. Autologous patient-derived adenosine-releasing cell implants would constitute a major therapeutic advance to avoid both xenotransplantation and immunosuppression. Here we describe a novel approach based on lentiviral RNAi mediated downregulation of adenosine kinase (ADK), the major adenosine-removing enzyme, in human mesenchymal stem cells (hMSCs), which would be compatible with autologous cell grafting in patients. Following lentiviral transduction of hMSCs with anti-ADK miRNA expression cassettes we demonstrate up to 80% downregulation of ADK and a concentration of 8.5 ng adenosine per ml of medium after incubating 10(5) cells for 8 h. hMSCs with a knockdown of ADK or cells expressing a scrambled control sequence were transplanted into hippocampi of mice 1 week prior to the intraamygdaloid injection of kainic acid (KA). While mice with control implants expressing a scrambled miRNA sequence or sham treated control animals were characterized by KA-induced status epilepticus and subsequent CA3 neuronal cell loss, animals with therapeutic ADK knockdown implants displayed a 35% reduction in seizure duration and 65% reduction in CA3 neuronal cell loss, when analyzed 24 h after KA-injection. We conclude that lentiviral expression of anti-ADK miRNA constitutes a versatile tool to generate therapeutically effective adenosine releasing hMSCs, thus representing a model system to generate patient identical autologous adult stem cell grafts.


Asunto(s)
Adenosina Quinasa/metabolismo , Lentivirus/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/enzimología , Interferencia de ARN , Estado Epiléptico/cirugía , Adenosina/metabolismo , Adenosina Quinasa/genética , Animales , Regulación hacia Abajo , Agonistas de Aminoácidos Excitadores , Vectores Genéticos , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Ácido Kaínico , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Estado Epiléptico/inducido químicamente , Estado Epiléptico/patología , Estado Epiléptico/fisiopatología , Transducción Genética
13.
J Clin Immunol ; 27(2): 181-92, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17235687

RESUMEN

Autoantibodies against alpha-enolase, a glycolytic enzyme, have been frequently associated with visual loss and retinal degeneration in patients with autoimmune and cancer-associated retinopathy; however their role in the pathogenicity of retinopathy has not been fully explained. Thus, we examined the causative role of anti-enolase antibodies on retinal cells. In the in vitro studies reported here, we found that Enol-1 monoclonal antibody against alpha-enolase significantly inhibited the catalytic function of enolase, which resulted in the depletion of glycolytic ATP. Enol-1 significantly increased intracellular Ca(2+), which led to Bax translocation to the mitochondria, and the release of cytochrome c into the cytoplasm--events that correlated with the initiation of apoptosis. Normal IgG did not induce intracellular calcium or reduce cytosolic ATP. L-type voltage-gated calcium channel blockers (nifedipine, D-cis-diltiazem, and verapamil) were effective in blocking the Ab-induced intracellular Ca(2+) rise and induction of Bax. Based on these findings we propose that chronic access of autoantibodies to the retina results in the inhibition of enolase catalytic function, depletion of ATP, and elevation in intracellular Ca(2+), leading to deregulation of glycolysis in retinal neurons and their destruction.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Neuronas/metabolismo , Fosfopiruvato Hidratasa/inmunología , Enfermedades de la Retina/inmunología , Adenosina Trifosfato/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Apoptosis/fisiología , Western Blotting , Calcio/metabolismo , Línea Celular , Glucólisis , Ratas
14.
Eur J Neurosci ; 23(5): 1163-71, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16553780

RESUMEN

Complete X-linked congenital stationary night blindness (CSNB1) is a hereditary visual disease characterized by abnormalities in both the dark- and light-adapted electroretinogram, consistent with a defect in synaptic transmission between photoreceptors and ON-bipolar cells. The gene responsible for CSNB1, NYX, encodes a novel, leucine-rich repeat protein, nyctalopin. Consistent with its predicted glycosylphosphatidylinositol linkage, we show that recombinant nyctalopin is targeted to the extracellular cell surface in transfected HEK293 cells. Within the retina, strong nyctalopin immunoreactivity is present in the outer plexiform layer, the site of the photoreceptor to bipolar cell synapses. Double labelling of nyctalopin and known synaptic proteins in the outer plexiform layer indicate that nyctalopin is associated with the ribbon synapses of both rod and cone terminals. In the inner plexiform layer, nyctalopin immunoreactivity is associated with rod bipolar cell terminals. Our findings support a role for nyctalopin in synaptic transmission and/or synapse formation at ribbon synapses in the retina.


Asunto(s)
Proteoglicanos/análisis , Retina/química , Secuencia de Aminoácidos , Animales , Línea Celular , Humanos , Macaca , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteoglicanos/genética , Conejos , Ratas , Ratas Sprague-Dawley , Retina/citología , Sinapsis/química , Sinapsis/ultraestructura , Transmisión Sináptica
15.
Vis Neurosci ; 22(5): 561-8, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16332266

RESUMEN

The genetic locus for incomplete congenital stationary night blindness (CSNB2) has been identified as the CACNA1f gene, encoding the alpha 1F calcium channel subunit, a member of the L-type family of calcium channels. The electroretinogram associated with CSNB2 implicates alpha 1F in synaptic transmission between retinal photoreceptors and bipolar cells. Using a recently developed monoclonal antibody to alpha 1F, we localize the channel to ribbon active zones in rod photoreceptor terminals of the mouse retina, supporting a role for alpha 1F in mediating glutamate release from rods. Detergent extraction experiments indicate that alpha 1F is part of a detergent-resistant active zone complex, which also includes the synaptic ribbons. Comparison of native mouse rod calcium currents with recombinant alpha 1F currents reveals that the current-voltage relationship for the native current is shifted approximately 30 mV to more hyperpolarized potentials than for the recombinant alpha 1F current, suggesting modulation of the native channel by intracellular factors. Lastly, we present evidence for L-type alpha 1D calcium channel subunits in cone terminals of the mouse retina. The presence of alpha 1D channels in cones may explain the residual visual abilities of individuals with CSNB2.


Asunto(s)
Canales de Calcio/fisiología , Ceguera Nocturna/genética , Ceguera Nocturna/fisiopatología , Células Fotorreceptoras Retinianas Conos/fisiología , Oxidorreductasas de Alcohol , Animales , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales/farmacología , Western Blotting , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/efectos de los fármacos , Canales de Calcio/genética , Canales de Calcio Tipo L , Proteínas Co-Represoras , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Electrofisiología , Inmunohistoquímica , Ratones , Microscopía Confocal , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Recombinantes/farmacología , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Sinaptotagminas/genética , Sinaptotagminas/metabolismo
16.
J Autoimmun ; 23(2): 161-7, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15324934

RESUMEN

Autoantibodies against alpha-enolase are often associated with visual loss in patients with autoimmune retinopathy. Anti-recoverin autoantibodies have been the most extensively studied for their pathologic association with cancer-associated retinopathy (CAR). It has been shown that anti-recoverin antibodies penetrate retinal layers corresponding to the cellular location of recoverin and cause the death of photoreceptors and bipolar cells. However, the pathogenic effects of anti-alpha-enolase antibodies have not been studied. In this study, we tested the labeling and apoptotic effects of such autoantibodies on retinal cells. Serum antibodies against alpha-enolase from patients with autoimmune retinopathy were tested ex vivo and in vivo in Sprague-Dawley rats. Autoantibodies to alpha-enolase specifically labeled the retinal ganglion cells and inner nuclear layer cells. Using ex vivo experiments and intravitreal injections, we observed that antibodies were capable of penetrating retinal tissue to target ganglion cell and inner nuclear layers and, consequently, were able to induce cell death through an apoptotic process. The apoptotic nuclei detected by a DNA fragmentation assay and caspase 3-positive cells were co-localized in the ganglion cell layer and inner nuclear layer. The results showed that antibodies against alpha-enolase target antigens in these layers and induce the apoptotic death of sensitive cells. Rat retinal explants and the intravitreal injection of antibodies provide us with a good model to identify antibody pathogenic targets in the retina. Such identification may help explain the complex of clinical symptoms for autoimmune retinopathy mediated by autoantibody and may help guide treatment strategies.


Asunto(s)
Autoanticuerpos/inmunología , Fosfopiruvato Hidratasa/inmunología , Retinitis Pigmentosa/inmunología , Animales , Antígenos Heterófilos/inmunología , Apoptosis , Autoanticuerpos/análisis , Estudios de Casos y Controles , Caspasa 3 , Caspasas/análisis , Fragmentación del ADN , Humanos , Técnicas In Vitro , Ratas , Ratas Sprague-Dawley , Retina/citología , Retina/inmunología , Células Ganglionares de la Retina/inmunología
17.
BMC Ophthalmol ; 4: 5, 2004 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-15180904

RESUMEN

BACKGROUND: Autoimmune retinal degeneration may occur in patients who present with sudden or, less commonly, subacute loss of vision of retinal origin, associated with an abnormal ERG, through the action of autoantibodies against retinal proteins. Often the patients are initially diagnosed with or suspected of having a paraneoplastic retinopathy (PR), such as cancer-associated retinopathy (CAR). However, there is limited information on the occurrence, the specificity of autoantibodies in these patients, and their association with clinical symptoms. METHODS: Sera were obtained from 193 retinopathy patients who presented with clinical symptoms resembling PR or autoimmune retinopathy (AR), including sudden painless loss of vision, typically associated with visual field defects and photopsias, and abnormal rod and/or cone responses on the electroretinogram (ERG). Sera were tested for the presence of anti-retinal autoantibodies by Western blot analysis using proteins extracted from human retina and by immunohistochemistry. Autoantibody titers against recoverin and enolase were measured by ELISA. RESULTS: We identified a higher prevalence of anti-retinal autoantibodies in retinopathy patients. Ninety-one patients' sera (47.1%) showed autoantibodies of various specificities with a higher incidence of antibodies present in retinopathy patients diagnosed with cancer (33/52; 63.5%; p = 0.009) than in retinopathy patients without cancer (58/141; 41.1%). The average age of PR patients was 62.0 years, and that of AR patients was 55.9 years. Autoantibodies against recoverin (p23) were only present in the sera of PR patients, autoantibodies against unknown p35 were more common in patients with AR, while anti-enolase (anti-p46) autoantibodies were nearly equally distributed in the sera of patients with PR and those with AR. In the seropositive patients, the autoantibodies persisted over a long period of time--from months to years. A rebound in anti-recoverin autoantibody titer was found to be associated with exacerbations in visual symptoms but not in the recurrence of cancer. When compared to sera from healthy subjects, autoantibodies against retinal proteins from both groups of patients were cytotoxic to retinal cells, indicating their pathogenic potential. CONCLUSIONS: These studies showed that patients with sudden or subacute, unexplained loss of vision of retinal origin have anti-retinal antibodies in a broad range of specificity and indicate the need for autoantibody screening. Follow-up tests of antibody levels may be useful as a biomarker of disease activity associated with worsening of vision. Moreover, the heterogeneity in autoantibody specificity may explain the variation and complexity of clinical symptoms in retinopathy patients.


Asunto(s)
Autoanticuerpos/sangre , Enfermedades Autoinmunes/inmunología , Proteínas del Ojo/inmunología , Síndromes Paraneoplásicos/inmunología , Retina/inmunología , Enfermedades de la Retina/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Autoantígenos/inmunología , Western Blotting , Pruebas Inmunológicas de Citotoxicidad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Técnicas para Inmunoenzimas , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...