Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Neurochem ; 158(5): 1131-1150, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34265077

RESUMEN

Retinitis pigmentosa (RP) is a group of inherited photoreceptor degeneration diseases that causes blindness without effective treatment. The pathogenesis of retinal degeneration involves mainly oxidative stress and inflammatory responses. Zeaxanthin dipalmitate (ZD), a wolfberry-derived carotenoid, has anti-inflammatory and anti-oxidative stress effects. Here we investigated whether these properties of ZD can delay the retinal degeneration in rd10 mice, a model of RP, and explored its underlying mechanism. One shot of ZD or control vehicle was intravitreally injected into rd10 mice on postnatal day 16 (P16). Retinal function and structure of rd10 mice were assessed at P25, when rods degenerate substantially, using a visual behavior test, multi-electrode-array recordings and immunostaining. Retinal pathogenic gene expression and regulation of signaling pathways by ZD were explored using transcriptome sequencing and western blotting. Our results showed that ZD treatment improved the visual behavior of rd10 mice and delayed the degeneration of retinal photoreceptors. It also improved the light responses of photoreceptors, bipolar cells and retinal ganglion cells. The expression of genes that are involved in inflammation, apoptosis and oxidative stress were up-regulated in rd10 mice, and were reduced by ZD. ZD further reduced the activation of two key factors, signal transducer and activator of transcription 3 and chemokine (C-C motif) ligand 2, down-regulated the expression of the inflammatory factor GFAP, and inhibited extracellular signal regulated protein kinases and P38, but not the JNK pathways. In conclusion, ZD delays the degeneration of the rd10 retina both morphologically and functionally. Its anti-inflammatory function is mediated primarily through the signal transducer and activator of transcription 3, chemokine (C-C motif) ligand 2 and MAPK pathways. Thus, ZD may serve as a potential clinical candidate to treat RP.


Asunto(s)
Quimiocina CCL2/antagonistas & inhibidores , Lycium , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Palmitatos/uso terapéutico , Degeneración Retiniana/prevención & control , Retinitis Pigmentosa/prevención & control , Factor de Transcripción STAT3/antagonistas & inhibidores , Xantófilas/uso terapéutico , Animales , Quimiocina CCL2/metabolismo , Femenino , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Palmitatos/aislamiento & purificación , Palmitatos/farmacología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Factor de Transcripción STAT3/metabolismo , Xantófilas/aislamiento & purificación , Xantófilas/farmacología
2.
Eur J Neurosci ; 43(11): 1509-22, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27037829

RESUMEN

Heterotrimeric G-proteins couple metabotropic receptors to downstream effectors. In retinal ON bipolar cells, Go couples the metabotropic receptor mGluR6 to the TRPM1 channel and closes it in the dark, thus hyperpolarizing the cell. Light, via GTPase-activating proteins, deactivates Go , opens TRPM1 and depolarizes the cell. Go comprises Gαo1 , Gß3 and Gγ13; all are necessary for efficient coupling. In addition, Gß3 contributes to trafficking of certain cascade proteins and to maintaining the synaptic structure. The goal of this study was to determine the role of Gαo1 in maintaining the cascade and synaptic integrity. Using mice lacking Gαo1 , we quantified the immunostaining of certain mGluR6-related components. Deleting Gαo1 greatly reduced staining for Gß3, Gγ13, Gß5, RGS11, RGS7 and R9AP. Deletion of Gαo1 did not affect mGluR6, TRPM1 or PCP2. In addition, deleting Gαo1 reduced the number of rod bipolar dendrites that invaginate the rod terminal, similar to the effect seen in the absence of mGluR6, Gß3 or the matrix-associated proteins, pikachurin, dystroglycan and dystrophin, which are localized presynaptically to the rod bipolar cell. We therefore tested mice lacking mGluR6, Gαo1 and Gß3 for expression of these matrix-associated proteins. In all three genotypes, staining intensity for these proteins was lower than in wild type, suggesting a retrograde trans-synaptic effect. We propose that the mGluR6 macromolecular complex is connected to the presynaptic rod terminal via a protein chain that includes the matrix-associated proteins. When a component of the macromolecular chain is missing, the chain may fall apart and loosen the dendritic tip adherence within the invagination.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Sinapsis/ultraestructura , Animales , Dendritas/metabolismo , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Masculino , Ratones , Ratones Noqueados , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/ultraestructura , Células Fotorreceptoras Retinianas Bastones/ultraestructura , Transducción de Señal , Canales Catiónicos TRPM/metabolismo
3.
J Physiol ; 593(7): 1531-50, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25416620

RESUMEN

Heterotrimeric G-proteins (comprising Gα and Gßγ subunits) are critical for coupling of metabotropic receptors to their downstream effectors. In the retina, glutamate released from photoreceptors in the dark activates metabotropic glutamate receptor 6 (mGluR6) receptors in ON bipolar cells; this leads to activation of Go , closure of transient receptor potential melastatin 1 channels and hyperpolarization of these cells. Go comprises Gαo , Gß3 and a Gγ. The best Gγ candidate is Gγ13, although functional data to support this are lacking. Thus, we tested Gγ13 function by generating Gng13(-/-) knockout (KO) mice, recording electroretinograms (ERG) and performing immunocytochemical staining. The amplitude of scotopic ERG b-waves in KO mice was lower than in wild-type (WT) mice. Furthermore, in both KO and WT mice, the ERG b-wave decreased with age; this decrease was much more pronounced in KO mice. By contrast, the photopic ERG b-waves in KO mice were hardly affected at any age. In KO mice retinas, immunostaining for Gß3 and for the GTPase activating proteins RGS7, RGS11, R9AP and Gß5 decreased significantly in rod bipolar cells but not in ON cone bipolar cells. Staining for Gαo and certain other cascade elements decreased only slightly. Analysis of our ON bipolar cDNA library showed that these cells express mRNAs for Gγ5, Gγ10 and Gγ11. Quantitative RT-PCR of retinal cDNA showed greater values for these transcripts in retinas of KO mice, although the difference was not significant. Our results suggest that Gγ13 contributes to mGluR6 signalling in rod bipolar cells more than in ON cone bipolar cells, and that this contribution includes both coupling the receptor and maintaining a stable localization of the mGluR6-related cascade elements.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Células Bipolares de la Retina/fisiología , Animales , Electrorretinografía , Femenino , Proteínas de Unión al GTP Heterotriméricas/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
4.
J Neurosci ; 33(12): 5182-94, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23516284

RESUMEN

Mammalian cones respond to light by closing a cGMP-gated channel via a cascade that includes a heterotrimeric G-protein, cone transducin, comprising Gαt2, Gß3 and Gγt2 subunits. The function of Gßγ in this cascade has not been examined. Here, we investigate the role of Gß3 by assessing cone structure and function in Gß3-null mouse (Gnb3(-/-)). We found that Gß3 is required for the normal expression of its partners, because in the Gnb3(-/-) cone outer segments, the levels of Gαt2 and Gγt2 are reduced by fourfold to sixfold, whereas other components of the cascade remain unaltered. Surprisingly, Gnb3(-/-) cones produce stable responses with normal kinetics and saturating response amplitudes similar to that of the wild-type, suggesting that cone phototransduction can function efficiently without a Gß subunit. However, light sensitivity was reduced by approximately fourfold in the knock-out cones. Because the reduction in sensitivity was similar in magnitude to the reduction in Gαt2 level in the cone outer segment, we conclude that activation of Gαt2 in Gnb3(-/-) cones proceeds at a rate approximately proportional to its outer segment concentration, and that activation of phosphodiesterase and downstream cascade components is normal. These results suggest that the main role of Gß3 in cones is to establish optimal levels of transducin heteromer in the outer segment, thereby indirectly contributing to robust response properties.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/genética , Células Fotorreceptoras Retinianas Conos/fisiología , Transducina/genética , Visión Ocular/fisiología , Animales , Color , Femenino , Proteínas Transportadoras de GABA en la Membrana Plasmática/genética , Proteínas Fluorescentes Verdes/genética , Proteínas de Unión al GTP Heterotriméricas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Neurológicos , Estimulación Luminosa , Segmento Externo de las Células Fotorreceptoras Retinianas/fisiología , Transducina/fisiología , Rayos Ultravioleta
5.
J Biol Chem ; 288(10): 7420-9, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23339194

RESUMEN

Kir2.4, a strongly rectifying potassium channel that is localized to neurons and is especially abundant in retina, was fished with yeast two-hybrid screen using a constitutively active Gαo1. Here, we wished to determine whether and how Gαo affects this channel. Using transfected HEK 293 cells and retinal tissue, we showed that Kir2.4 interacts with Gαo, and this interaction is stronger with the GDP-bound form of Gαo. Using two-electrode voltage clamp, we recorded from oocytes that were injected with Kir2.4 mRNA and a combination of G-protein subunit mRNAs. We found that the wild type and the inactive mutant of Gαo reduce the Kir2.4 basal current, whereas the active mutant has little effect. Other pertussis-sensitive Gα subunits also reduce this current, whereas Gαs increases it. Gßγ increases the current, whereas m-phosducin, which binds Gßγ without affecting the state of Gα, reduces it. We then tested the effect of G-protein subunits on the surface expression of the channel fused to cerulean by imaging the plasma membranes of the oocytes. We found that the surface expression is affected, with effects paralleling those seen with the basal current. This suggests that the observed effects on the current are mainly indirect and are due to surface expression. Similar results were obtained in transfected HEK cells. Moreover, we show that in retinal ON bipolar cells lacking Gß3, localization of Kir2.4 in the dendritic tips is reduced. We conclude that Gßγ targets Kir2.4 to the plasma membrane, and Gαo slows this down by binding Gßγ.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Activación del Canal Iónico/fisiología , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Células HEK293 , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Activación del Canal Iónico/genética , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Oocitos/metabolismo , Oocitos/fisiología , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/genética , Unión Proteica , Retina/metabolismo , Técnicas del Sistema de Dos Híbridos , Xenopus
6.
Biomolecules ; 14(6)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38927058

RESUMEN

The retina, a tissue of the central nervous system, is vital for vision as its photoreceptors capture light and transform it into electrical signals, which are further processed before they are sent to the brain to be interpreted as images. The retina is unique in that it is continuously exposed to light and has the highest metabolic rate and demand for energy amongst all the tissues in the body. Consequently, the retina is very susceptible to oxidative stress. VDAC, a pore in the outer membrane of mitochondria, shuttles metabolites between mitochondria and the cytosol and normally protects cells from oxidative damage, but when a cell's integrity is greatly compromised it initiates cell death. There are three isoforms of VDAC, and existing evidence indicates that all three are expressed in the retina. However, their precise localization and function in each cell type is unknown. It appears that most retinal cells express substantial amounts of VDAC2 and VDAC3, presumably to protect them from oxidative stress. Photoreceptors express VDAC2, HK2, and PKM2-key proteins in the Warburg pathway that also protect these cells. Consistent with its role in initiating cell death, VDAC is overexpressed in the retinal degenerative diseases retinitis pigmentosa, age related macular degeneration (AMD), and glaucoma. Treatment with antioxidants or inhibiting VDAC oligomerization reduced its expression and improved cell survival. Thus, VDAC may be a promising therapeutic candidate for the treatment of these diseases.


Asunto(s)
Retina , Canales Aniónicos Dependientes del Voltaje , Humanos , Canales Aniónicos Dependientes del Voltaje/metabolismo , Retina/metabolismo , Animales , Estrés Oxidativo , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Mitocondrias/metabolismo , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
7.
J Neurosci ; 32(33): 11343-55, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22895717

RESUMEN

Heterotrimeric G-proteins, comprising Gα and Gßγ subunits, couple metabotropic receptors to various downstream effectors and contribute to assembling and trafficking receptor-based signaling complexes. A G-protein ß subunit, Gß(3), plays a critical role in several physiological processes, as a polymorphism in its gene is associated with a risk factor for several disorders. Retinal ON bipolar cells express Gß(3), and they provide an excellent system to study its role. In the ON bipolar cells, mGluR6 inverts the photoreceptor's signal via a cascade in which glutamate released from photoreceptors closes the TRPM1 channel. This cascade is essential for vision since deficiencies in its proteins lead to complete congenital stationary night blindness. Here we report that Gß(3) participates in the G-protein heterotrimer that couples mGluR6 to TRPM1. Gß(3) deletion in mouse greatly reduces the light response under both scotopic and photopic conditions, but it does not eliminate it. In addition, Gß(3) deletion causes mislocalization and downregulation of most cascade elements and modulators. Furthermore, Gß(3) may play a role in synaptic maintenance since in its absence, the number of invaginating rod bipolar dendrites is greatly reduced, a deficit that was not observed at 3 weeks, the end of the developmental period.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Regulación de la Expresión Génica/genética , Células Bipolares de la Retina/metabolismo , Sinapsis/fisiología , Animales , Colina O-Acetiltransferasa/metabolismo , Dendritas/ultraestructura , Estimulación Eléctrica , Electrorretinografía , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Subunidades beta de la Proteína de Unión al GTP/deficiencia , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Inmunoprecipitación , Técnicas In Vitro , Luz , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Placa-Clamp , Estimulación Luminosa , Propionatos/farmacología , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/deficiencia , Receptores de Glutamato Metabotrópico/genética , Retina/citología , Células Bipolares de la Retina/efectos de los fármacos , Células Bipolares de la Retina/ultraestructura , Células Fotorreceptoras Retinianas Conos/metabolismo , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/ultraestructura , Canales Catiónicos TRPM/metabolismo , Vías Visuales/fisiología
8.
J Neurosci ; 31(8): 2855-67, 2011 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-21414907

RESUMEN

Decoding the wiring diagram of the retina requires simultaneous observation of activity in identified neuron populations. Available recording methods are limited in their scope: electrodes can access only a small fraction of neurons at once, whereas synthetic fluorescent indicator dyes label tissue indiscriminately. Here, we describe a method for studying retinal circuitry at cellular and subcellular levels combining two-photon microscopy and a genetically encoded calcium indicator. Using specific viral and promoter constructs to drive expression of GCaMP3, we labeled all five major neuron classes in the adult mouse retina. Stimulus-evoked GCaMP3 responses as imaged by two-photon microscopy permitted functional cell type annotation. Fluorescence responses were similar to those measured with the small molecule dye OGB-1. Fluorescence intensity correlated linearly with spike rates >10 spikes/s, and a significant change in fluorescence always reflected a significant change in spike firing rate. GCaMP3 expression had no apparent effect on neuronal function. Imaging at subcellular resolution showed compartment-specific calcium dynamics in multiple identified cell types.


Asunto(s)
Potenciales de Acción/fisiología , Neuronas/fisiología , Retina/fisiología , Visión Ocular/fisiología , Imagen de Colorante Sensible al Voltaje/métodos , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/clasificación , Neuronas/citología , Técnicas de Cultivo de Órganos , Retina/citología
9.
J Neurosci ; 31(11): 3962-7, 2011 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-21411639

RESUMEN

Melanoma-associated retinopathy (MAR) is characterized by night blindness, photopsias, and a selective reduction of the electroretinogram b-wave. In certain cases, the serum contains autoantibodies that react with ON bipolar cells, but the target of these autoantibodies has not been identified. Here we show that the primary target of autoantibodies produced in MAR patients with reduced b-wave is the TRPM1 cation channel, the newly identified transduction channel in ON bipolar cells. Sera from two well characterized MAR patients, but not from a control subject, stained human embryonic kidney cells transfected with the TRPM1 gene, and Western blots probed with these MAR sera showed the expected band size (∼180 kDa). Staining of mouse and primate retina with MAR sera revealed immunoreactivity in all types of ON bipolar cells. Similar to staining for TRPM1, staining with the MAR sera was strong in dendritic tips and somas and was weak or absent in axon terminals. This staining colocalized with GFP in Grm6-GFP transgenic mice, where GFP is expressed in all and only ON bipolar cells, and also colocalized with Gα(o), a marker for all types of ON bipolar cells. The staining in ON bipolar cells was confirmed to be specific to TRPM1 because MAR serum did not stain these cells in a Trpm1(-/-) mouse. Evidence suggests that the recognized epitope is likely intracellular, and the sera can be internalized by retinal cells. We conclude that the vision of at least some patients with MAR is compromised due to autoantibody-mediated inactivation of the TRPM1 channel.


Asunto(s)
Autoanticuerpos/metabolismo , Melanoma/inmunología , Células Bipolares de la Retina/metabolismo , Enfermedades de la Retina/inmunología , Canales Catiónicos TRPM/metabolismo , Animales , Autoanticuerpos/inmunología , Western Blotting , Células HEK293 , Humanos , Masculino , Melanoma/complicaciones , Ratones , Ratones Transgénicos , Enfermedades de la Retina/etiología , Canales Catiónicos TRPM/genética , Transfección
10.
J Neurophysiol ; 107(3): 948-57, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22131384

RESUMEN

In darkness, glutamate released from photoreceptors activates the metabotropic glutamate receptor 6 (mGluR6) on retinal ON bipolar cells. This activates the G protein G(o), which then closes transient receptor potential melastatin 1 (TRPM1) channels, leading to cells' hyperpolarization. It has been generally assumed that deleting mGluR6 would render the cascade inactive and the ON bipolar cells constitutively depolarized. Here we show that the rod bipolar cells in mGluR6-null mice were hyperpolarized. The slope conductance of the current-voltage curves and the current noise were smaller than in wild type. Furthermore, while in wild-type rod bipolar cells, TRPM1 could be activated by local application of capsaicin; in null cells, it did not. These results suggest that the TRPM1 channel in mGluR6-null rod bipolar cells is inactive. To explore the reason for this lack of activity, we tested if mGluR6 deletion affected expression of cascade components. Immunostaining for G protein subunit candidates Gα(o), Gß(3), and Gγ(13) showed no significant changes in their expression or distribution. Immunostaining for TRPM1 in the dendritic tips was greatly reduced, but the channel was still present in the soma and primary dendrites of mGluR6-null bipolar cells, where a certain fraction of TRPM1 appears to localize to the plasma membrane. Consequently, the lack of TRPM1 activity in the null retina is unlikely to be due to failure of the channels to localize to the plasma membrane. We speculate that, to be constitutively active, TRPM1 channels in ON bipolar cells have to be in a complex, or perhaps require an unidentified factor.


Asunto(s)
Receptores de Glutamato Metabotrópico/fisiología , Células Bipolares de la Retina/fisiología , Canales Catiónicos TRPM/fisiología , Animales , Capsaicina/farmacología , Dendritas/química , Dendritas/fisiología , Eliminación de Gen , Proteínas de Unión al GTP Heterotriméricas/análisis , Proteínas de Unión al GTP Heterotriméricas/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Glutamato Metabotrópico/genética , Células Bipolares de la Retina/efectos de los fármacos , Fármacos del Sistema Sensorial/farmacología , Canales Catiónicos TRPM/análisis
11.
Transl Neurodegener ; 11(1): 58, 2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36578022

RESUMEN

BACKGROUND: Alzheimer's disease (AD) exhibits mitochondrial dysfunctions associated with dysregulated metabolism, brain inflammation, synaptic loss, and neuronal cell death. As a key protein serving as the mitochondrial gatekeeper, the voltage-dependent anion channel-1 (VDAC1) that controls metabolism and Ca2+ homeostasis is positioned at a convergence point for various cell survival and death signals. Here, we targeted VDAC1 with VBIT-4, a newly developed inhibitor of VDAC1 that prevents its pro-apoptotic activity, and mitochondria dysfunction. METHODS: To address the multiple pathways involved in AD, neuronal cultures and a 5 × FAD mouse model of AD were treated with VBIT-4. We addressed multiple topics related to the disease and its molecular mechanisms using immunoblotting, immunofluorescence, q-RT-PCR, 3-D structural analysis and several behavioral tests. RESULTS: In neuronal cultures, amyloid-beta (Aß)-induced VDAC1 and p53 overexpression and apoptotic cell death were prevented by VBIT-4. Using an AD-like 5 × FAD mouse model, we showed that VDAC1 was overexpressed in neurons surrounding Aß plaques, but not in astrocytes and microglia, and this was associated with neuronal cell death. VBIT-4 prevented the associated pathophysiological changes including neuronal cell death, neuroinflammation, and neuro-metabolic dysfunctions. VBIT-4 also switched astrocytes and microglia from being pro-inflammatory/neurotoxic to neuroprotective phenotype. Moreover, VBIT-4 prevented cognitive decline in the 5 × FAD mice as evaluated using several behavioral assessments of cognitive function. Interestingly, VBIT-4 protected against AD pathology, with no significant change in phosphorylated Tau and only a slight decrease in Aß-plaque load. CONCLUSIONS: The study suggests that mitochondrial dysfunction with its gatekeeper VDAC1 is a promising target for AD therapeutic intervention, and VBIT-4 is a promising drug candidate for AD treatment.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Proteínas Mitocondriales , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Mitocondrias/metabolismo
12.
Sci Rep ; 11(1): 9376, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33931669

RESUMEN

Regulator of G-protein signaling 7 (RGS7) is predominately present in the nervous system and is essential for neuronal signaling involving G-proteins. Prior studies in cultured cells showed that RGS7 is regulated via proteasomal degradation, however no protein is known to facilitate proteasomal degradation of RGS7 and it has not been shown whether this regulation affects G-protein signaling in neurons. Here we used a knockout mouse model with conditional deletion of arginyltransferase (Ate1) in the nervous system and found that in retinal ON bipolar cells, where RGS7 modulates a G-protein to signal light increments, deletion of Ate1 raised the level of RGS7. Electroretinographs revealed that lack of Ate1 leads to increased light-evoked response sensitivities of ON-bipolar cells, as well as their downstream neurons. In cultured mouse embryonic fibroblasts (MEF), RGS7 was rapidly degraded via proteasome pathway and this degradation was abolished in Ate1 knockout MEF. Our results indicate that Ate1 regulates RGS7 protein level by facilitating proteasomal degradation of RGS7 and thus affects G-protein signaling in neurons.


Asunto(s)
Aminoaciltransferasas/fisiología , Fibroblastos/metabolismo , Luz , Sistema Nervioso/metabolismo , Proteínas RGS/metabolismo , Células Bipolares de la Retina/metabolismo , Animales , Femenino , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sistema Nervioso/patología , Sistema Nervioso/efectos de la radiación , Proteínas RGS/genética , Células Bipolares de la Retina/patología , Células Bipolares de la Retina/efectos de la radiación , Transducción de Señal
13.
Eur J Neurosci ; 31(4): 685-96, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20384812

RESUMEN

Certain bipolar cells in most species immunostain for GABA or its synthesizing enzyme glutamic acid decarboxylase. However, it is unknown whether they actually release GABA and, if so, from which cellular compartment and by what release mechanism. We investigated these questions in monkey retina where rod bipolar cells immunostain for GABA. We found that rod bipolar cells immunostain for one isoform of GAD (GAD65) in their somas, dendrites and axon terminals. Near the fovea, the somatic stain of rod bipolar cells is weaker than that of horizontal cells but, at the periphery, it is stronger. Staining for the vesicular GABA transporter in monkey rod bipolar cells is negative. However, staining for the GABA transporter GAT3 is positive in the soma and primary dendrites (but not in the axon terminals). Staining for GAT3 is also positive in horizontal cells. Double staining of rod bipolar cells and the alpha subunit of the GABAA receptor reveals scarce GABAA puncta that appose rod bipolar dendrites. We conclude that monkey rod bipolar cells use GABA and discuss the possibility that they tonically release GABA from their dendrites using a reverse action of GAT3.


Asunto(s)
Receptores de GABA-A/metabolismo , Células Bipolares de la Retina/metabolismo , Transmisión Sináptica , Ácido gamma-Aminobutírico/metabolismo , Animales , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Glutamato Descarboxilasa/metabolismo , Inmunohistoquímica , Macaca fascicularis , Macaca mulatta , Fragmentos de Péptidos/metabolismo , Proteína Quinasa C/metabolismo , Células Horizontales de la Retina/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
14.
Nat Neurosci ; 9(1): 85-92, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16341211

RESUMEN

The cellular mechanisms underlying axogenesis and dendritogenesis are not completely understood. The axons and dendrites of retinal bipolar cells, which contact their synaptic partners within specific laminae in the inner and outer retina, provide a good system for exploring these issues. Using transgenic mice expressing enhanced green fluorescent protein (GFP) in a subset of bipolar cells, we determined that axonal and dendritic arbors of these interneurons develop directly from apical and basal processes attached to the outer and inner limiting membranes, respectively. Selective stabilization of processes contributed to stratification of axonal and dendritic arbors within the appropriate synaptic layer. This unusual mode of axogenesis and dendritogenesis from neuroepithelial-like processes may act to preserve neighbor-neighbor relationships in synaptic wiring between the outer and inner retina.


Asunto(s)
Axones/fisiología , Dendritas/fisiología , Células Epiteliales/fisiología , Neuronas/fisiología , Retina/citología , Retina/crecimiento & desarrollo , Animales , Femenino , Genes Reporteros/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Glutamato Metabotrópico/genética
15.
J Neurosci ; 28(36): 8873-84, 2008 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-18768681

RESUMEN

PCP2, a member of the GoLoco domain-containing family, is present exclusively in cerebellar Purkinje cells and retinal ON bipolar cells. Its function in these tissues is unknown. Biochemical and expression system studies suggest that PCP2 is a guanine nucleotide dissociation inhibitor, although a guanine nucleotide exchange factor has also been suggested. Here, we studied the function of PCP2 in ON bipolar cells because their light response depends on Galpha(o1), which is known to interact with PCP2. We identified a new splice variant of PCP2 (Ret-PCP2) and localized it to rod bipolar and ON cone bipolar cells. Electroretinogram recordings from PCP2-null mice showed a normal a-wave but a slower falling phase of the b-wave (generated by the activity of ON bipolar cells) relative to the wild type. Whole-cell recordings from rod bipolar cells showed, both under Ames medium and after blocking GABA(A/C) and glycine receptors, that PCP2-null rod bipolar cells were more depolarized than wild-type cells with greater inward current when clamped to -60 mV. Also under both conditions, the rise time of the response to intense light was slower by 28% (Ames) and 44% (inhibitory blockers) in the null cells. Under Ames medium, we also observed >30% longer decay time in the PCP2-null rod bipolar cells. We conclude that PCP2 facilitates cation channels closure in the dark, shortens the rise time of the light response directly, and accelerates the decay time indirectly via the inhibitory network. These data can most easily be explained if PCP2 serves as a guanine nucleotide exchange factor.


Asunto(s)
ADN Recombinante/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Luz , Neuropéptidos/metabolismo , Células Bipolares de la Retina/fisiología , Células Bipolares de la Retina/efectos de la radiación , Animales , Células Cultivadas , Electrorretinografía/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factores de Intercambio de Guanina Nucleótido/deficiencia , Humanos , Espectrometría de Masas/métodos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Potenciales de la Membrana/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/deficiencia , Técnicas de Placa-Clamp/métodos , Estimulación Luminosa/métodos , Picrotoxina/farmacología , Tiempo de Reacción/genética , Tiempo de Reacción/efectos de la radiación , Retina/citología , Estricnina/farmacología , Transfección/métodos
16.
Nat Neurosci ; 7(8): 826-33, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15235608

RESUMEN

Traditional models of synaptic transmission hold that release sites within an active zone operate independently. Although the release of multiple vesicles (multivesicular release; MVR) from single active zones occurs at some central synapses, MVR is not thought to require coordination among release sites. Ribbon synapses seem to be optimized to release many vesicles over an extended period, but the dynamics of MVR at ribbon synapses is unknown. We examined MVR at a ribbon synapse in a retinal slice preparation using paired recordings from presynaptic rod bipolar and postsynaptic AII amacrine cells. When evoked release was highly desynchronized, discrete postsynaptic events were larger than quantal miniature excitatory postsynaptic currents (mEPSCs) but had the same time course. The amplitude of these multiquantal mEPSCs, which seem to arise from the essentially simultaneous release of multiple vesicles, was reduced by lowering release probability. The release synchrony reflected in these multivesicular events suggests that release within an active zone is coordinated during MVR.


Asunto(s)
Vesículas Citoplasmáticas/fisiología , Exocitosis/fisiología , Retina/fisiología , Sinapsis/fisiología , Animales , Vesículas Citoplasmáticas/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores , Inmunohistoquímica , Método de Montecarlo , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Ratas , Retina/efectos de los fármacos , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
17.
Invest Ophthalmol Vis Sci ; 59(1): 597-611, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29372259

RESUMEN

Purpose: As an active component in wolfberry, lycium barbarum polysaccharides (LBP) are capable of protecting retinal neurons in several animal disease models. Here, we asked whether LBP rescues the retinal morphology and function in rd1 mouse, a photoreceptor fast-degenerating animal model of retinitis pigmentosa, and in particular focused on LBP's effects on the function of retinal ganglion cells (RGCs) during photoreceptor degeneration. Methods: An equal volume of LBP or control vehicle was daily intraperitoneal (i.p.) injected in rd1 mice from postnatal day 4 (P4) to P14, P20, or P24 when photoreceptors completely degenerate. Immunostaining, electroretinogram (ERG), visual behavior tests and multielectrode array (MEA) recordings were assessed to determine the structure and function of the treated retina. Results: LBP treatment greatly promoted photoreceptor survival, enhanced ERG responses, and improved visual behaviors in rd1 mice. MEA data showed that LBP treatment in general decreased the abnormally high spontaneous spiking that occurs in rd1 mice, and increased the percentage of light-responsive RGCs as well as their light-evoked response, light sensitivity, signal-to-noise ratio, and response speed. Interestingly, LBP treatment affected ON and OFF responses differently. Conclusions: LBP improves retinal morphology and function in rd1 mice, and delays the functional decay of RGCs during photoreceptor degeneration. This is the first study that has examined in detail the effects of LBP on RGC responses. Our data suggest that LBP may help extend the effective time window before more invasive RP therapeutic approaches such as retinoprosthesis are applied.


Asunto(s)
Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Retinitis Pigmentosa/prevención & control , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Electrorretinografía , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Células Fotorreceptoras de Vertebrados/fisiología , Retinitis Pigmentosa/fisiopatología , Visión Ocular/fisiología
18.
Neuropharmacology ; 139: 137-149, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29940208

RESUMEN

Retinitis pigmentosa (RP) is a hereditary blinding disease characterized by neurodegeneration of photoreceptors. Retinal ganglion cells (RGCs) in animal models of RP exhibit an abnormally high spontaneous activity that interferes with signal processing. Blocking AMPA/Kainate receptors by bath application of CNQX decreases the spontaneous firing, suggesting that inhibiting these receptors in vivo may help maintain the function of inner retinal neurons in rd10 mice experiencing photoreceptor degeneration. To test this, rd10 mice were i.p. injected with CNQX or GYKI 52466 (an AMPA receptor antagonist) for 1-2 weeks, and examined for their retinal morphology (by immunocytochemistry), function (by MEA recordings) and visual behaviors (using a black/white box). Our data show that iGluRs were up-regulated in the inner plexiform layer (IPL) of rd10 retinas. Application of CNQX at low doses both in vitro and in vivo, attenuated the abnormal spontaneous spiking in RGCs, and increased the light-evoked response of ON RGCs, whereas GYKI 52466 had little effect. CNQX application also improved the behavioral performance. Interestingly, in vivo administration of CNQX delayed photoreceptor degeneration, evidenced by the increased cell number and restored structure. CNQX also improved the structure of bipolar cells. Together, we demonstrated that during photoreceptor degeneration, blockade of the non-NMDA iGluRs decelerates the progression of RGCs dysfunction, possibly by dual mechanisms including slowing photoreceptor degeneration and modulating signal processing within the IPL. Accordingly, this strategy may effectively extend the time window for treating RP.


Asunto(s)
6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Fármacos Neuroprotectores/farmacología , Receptores Ionotrópicos de Glutamato/antagonistas & inhibidores , Degeneración Retiniana/prevención & control , Potenciales de Acción/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/patología , Células Bipolares de la Retina/efectos de los fármacos , Células Bipolares de la Retina/fisiología , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/fisiología , Técnicas de Cultivo de Tejidos , Visión Ocular/efectos de los fármacos
20.
J Comp Neurol ; 499(1): 132-43, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16958091

RESUMEN

The developmental switch of GABA's action from excitation to inhibition is likely due to a change in intracellular chloride concentration from high to low. Here we determined if the GABA switch correlates with the developmental expression patterns of KCC2, the chloride extruder K+-Cl- cotransporter, and NKCC, the chloride accumulator Na+-K+-Cl- cotransporter. Immunoblots of ferret retina showed that KCC2 upregulated in an exponential manner similar to synaptophysin (a synaptic marker). In contrast, NKCC, which was initially expressed at a constant level, upregulated quickly between P14 and P28, and finally downregulated to an adult level that was greater than the initial phase. At the cellular level, immunocytochemistry showed that in the inner plexiform layer KCC2's density increased gradually and its localization within ganglion cells shifted from being primarily in the cytosol (between P1-13) to being in the plasma membrane (after P21). In the outer plexiform layer, KCC2 was detected as soon as this layer started to form and increased gradually. Interestingly, however, KCC2 was initially restricted to photoreceptor terminals, while in the adult it was restricted to bipolar dendrites. Thus, the overall KCC2 expression level in ferret retina increases with age, but the time course differs between cell types. In ganglion cells the upregulation of KCC2 by itself cannot explain the relatively fast switch in GABA's action; additional events, possibly KCC2's integration into the plasma membrane and downregulation of NKCC, might also contribute. In photoreceptors the transient expression of KCC2 suggests a role for this transporter in development.


Asunto(s)
Diferenciación Celular/fisiología , Membrana Celular/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Simportadores/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Western Blotting/métodos , Bromodesoxiuridina/metabolismo , Hurones , Inmunohistoquímica/métodos , Neuronas/clasificación , Neuronas/citología , Neuronas/metabolismo , Fenómenos Fisiológicos Oculares , Retina/citología , Retina/crecimiento & desarrollo , Retina/metabolismo , Vías Visuales/citología , Vías Visuales/crecimiento & desarrollo , Vías Visuales/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Cotransportadores de K Cl
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA