Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
bioRxiv ; 2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-37034790

RESUMEN

Lack of non-muscle ß -actin gene (Actb) leads to early embryonic lethality in mice, however mice with ß - to γ -actin replacement develop normally and show no detectable phenotypes at young age. Here we investigated the effect of this replacement in the retina. During aging, these mice have accelerated de-generation of retinal structure and function, including elongated microvilli and defective mitochondria of retinal pigment epithelium (RPE), abnormally bulging photoreceptor outer segments (OS) accompanied by reduced transducin concentration and light sensitivity, and accumulation of autofluorescent microglia cells in the subretinal space between RPE and OS. These defects are accompanied by changes in the F-actin binding of several key actin interacting partners, including ezrin, myosin, talin, and vinculin known to play central roles in modulating actin cytoskeleton and cell adhesion and mediating the phagocytosis of OS. Our data show that ß -actin protein is essential for maintaining normal retinal structure and function.

2.
Transl Neurodegener ; 11(1): 20, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35395956

RESUMEN

BACKGROUND: Alpha-synuclein (α-syn) exhibits pathological misfolding in many human neurodegenerative disorders. We previously showed that α-syn is arginylated in the mouse brain and that lack of arginylation leads to neurodegeneration in mice. METHODS: Here, we tested α-syn arginylation in human brain pathology using newly derived antibodies in combination with Western blotting, biochemical assays, and experiments in live neurons. RESULTS: We found that α-syn was arginylated in the human brain on E46 and E83, two sites previously implicated in α-syn pathology and familial cases of Parkinson's disease. The levels of arginylation in different brain samples ranged between ~ 3% and ~ 50% of the total α-syn pool, and this arginylation nearly exclusively concentrated in the subcellular α-syn fraction that sedimented at low centrifugation speeds and appeared to be simultaneously targeted by multiple posttranslational modifications. Arginylated α-syn was less susceptible to S129 phosphorylation and pathological aggregation in neurons. The arginylation level inversely correlated with the overall α-syn levels and with patient age, suggesting a possible causal relationship between arginylation decline and α-syn-dependent neuropathology. CONCLUSION: We propose that α-syn arginylation constitutes a potential neuroprotective mechanism that prevents its abnormal accumulation during neurodegeneration and aging in the human brain.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Sinucleinopatías , Animales , Encéfalo/metabolismo , Humanos , Ratones , Enfermedad de Parkinson/patología , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
3.
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
4.
Front Cell Dev Biol ; 9: 807345, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35127722

RESUMEN

Arginylation is a post-translational modification mediated by the arginyltransferase (Ate1). We recently showed that conditional deletion of Ate1 in the nervous system leads to increased light-evoked response sensitivities of ON-bipolar cells in the retina, indicating that arginylation regulates the G-protein signaling complexes of those neurons and/or photoreceptors. However, none of the key players in the signaling pathway were previously shown to be arginylated. Here we show that Gαt1, Gß1, RGS6, and RGS7 are arginylated in the retina and RGS6 and RGS7 protein levels are elevated in Ate1 knockout, suggesting that arginylation plays a direct role in regulating their protein level and the G-protein-mediated responses in the retina.

5.
Sci Rep ; 7(1): 11323, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28900170

RESUMEN

Alpha synuclein (α-syn) is a central player in neurodegeneration, but the mechanisms triggering its pathology are not fully understood. Here we found that α-syn is a highly efficient substrate for arginyltransferase ATE1 and is arginylated in vivo by a novel mid-chain mechanism that targets the acidic side chains of E46 and E83. Lack of arginylation leads to increased α-syn aggregation and causes the formation of larger pathological aggregates in neurons, accompanied by impairments in its ability to be cleared via normal degradation pathways. In the mouse brain, lack of arginylation leads to an increase in α-syn's insoluble fraction, accompanied by behavioral changes characteristic for neurodegenerative pathology. Our data show that lack of arginylation in the brain leads to neurodegeneration, and suggests that α-syn arginylation can be a previously unknown factor that facilitates normal α-syn folding and function in vivo.


Asunto(s)
Arginina/metabolismo , Encéfalo/fisiología , Enfermedades Neurodegenerativas/metabolismo , alfa-Sinucleína/metabolismo , Secuencia de Aminoácidos , Aminoaciltransferasas/genética , Aminoaciltransferasas/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Espectrometría de Masas , Ratones , Ratones Noqueados , Modelos Biológicos , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/prevención & control , Neuronas/metabolismo , Neuronas/patología , Péptidos/química , Péptidos/metabolismo , Agregado de Proteínas , Agregación Patológica de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , Proteínas Recombinantes , Especificidad por Sustrato , alfa-Sinucleína/química
6.
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
7.
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
8.
Invest Ophthalmol Vis Sci ; 55(3): 1483-92, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24519419

RESUMEN

PURPOSE: L-type voltage gated calcium channels in retina localize primarily at the presynaptic active zones of photoreceptors and bipolar cells where they modulate glutamate release. However, the pore forming subunit Cacna1s of certain L-type channels is also expressed postsynaptically at the tips of ON bipolar cell dendrites where it colocalizes with mGluR6, but has an unknown function. At these dendritic tips, the components of the mGluR6 signaling cascade cluster together in a macromolecular complex, and each one's localization often depends on that of the others. Thus, we explored if Cacna1s is part of the mGluR6 complex. METHODS: We determined Cacna1s expression by PCR using an ON bipolar library, by Western blotting, and by standard immunohistochemistry. RESULTS: The PCR amplification confirmed expression of the transcript in ON bipolar cells, and Western blotting showed the expected bands. Immunostaining for Cacna1s was stronger in the dendritic tips of rod bipolar cells than in those of ON cone bipolar cells. This staining severely decreased in mice missing various mGluR6 cascade elements (Grm6(-/-), Gnao1(-/-), Gnb3(-/-), Gng13(-/-), and Trpm1(-/-)). During development, the ratio of the number of Cacna1s puncta to the number of presynaptic ribbons followed a sigmoidal pattern, rising rapidly from P13 to P17. The mGluR6 expression preceded that of Cacna1s and RGS11. CONCLUSIONS: Our results show that the localization and stability of Cacna1s depend on the expression of mGluR6 and its cascade components, and they suggest that Cacna1s is part of the mGluR6 complex. We hypothesize that Cacna1s contributes to light adaptation by permeating calcium.


Asunto(s)
Canales de Calcio/genética , ADN/genética , Dendritas/metabolismo , Regulación de la Expresión Génica , Receptores de Glutamato Metabotrópico/genética , Células Bipolares de la Retina/metabolismo , Animales , Western Blotting , Canales de Calcio/biosíntesis , Canales de Calcio Tipo L , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/biosíntesis , Células Bipolares de la Retina/citología , Transducción de Señal
9.
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
10.
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
11.
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
12.
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
13.
J Histochem Cytochem ; 59(12): 1076-86, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22034516

RESUMEN

To study mGluR6 expression, the authors investigated two transgenic mouse lines that express enhanced green fluorescent protein (GFP) under control of mGluR6 promoter. In retina, GFP was expressed exclusively in all ON bipolar cell types, either uniformly across all cells of this class (line 5) or in a mosaic (patchy) fashion (line 1). In brain, GFP was found in certain cortical areas, superior colliculus, axons of the corpus callosum, accessory olfactory bulb, and cells of the subcommissural organ. Outside the nervous system, GFP was seen in the corneal endothelium, testis, the kidney's medulla, collecting ducts and parietal layer that surround the glomeruli, and B lymphocytes. Furthermore, RT-PCR showed that most tissues that expressed GFP in the transgenic mouse also transcribed two splice variants of mGluR6 in the wild-type mouse. The alternate variant was lacking exon 8, predicting a protein product of 545 amino acids that lacks the 7-transmembrane domains of the receptor. In cornea, immunostaining for mGluR6 gave strong staining in the endothelium, and this was stronger in wild-type than in mGluR6-null mice. Furthermore, calcium imaging with Fura-2 showed that application of L-AP4, an agonist for group III metabotropic glutamate receptors including mGluR6, elevated calcium in endothelial cells.


Asunto(s)
Receptores de Glutamato Metabotrópico/metabolismo , Empalme Alternativo , Animales , Encéfalo/metabolismo , Calcio/metabolismo , Córnea/metabolismo , Células Endoteliales/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Riñón/metabolismo , Tejido Linfoide/metabolismo , Masculino , Ratones , Ratones Transgénicos , Especificidad de Órganos , Regiones Promotoras Genéticas , Propionatos/farmacología , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/genética , Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/metabolismo
14.
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
15.
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
16.
J Comp Neurol ; 518(7): 1098-112, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20127818

RESUMEN

Purkinje cell protein 2 (PCP2), a member of the family of guanine dissociation inhibitors and a strong interactor with the G-protein subunit G alpha(o), localizes to retinal ON bipolar cells. The retina-specific splice variant of PCP2, Ret-PCP2, accelerates the light response of rod bipolar cells by modulating the mGluR6 transduction cascade. All ON cone bipolar cells express mGluR6 and G alpha(o), but only a subset expresses Ret-PCP2. Here we test the hypothesis that Ret-PCP2 contributes to shaping the various temporal bandwidths of ON cone bipolar cells in monkey retina. We found that the retinal splice variants in monkey and mouse are similar and longer than the cerebellar variants. Ret-PCP2 is strongly expressed by diffuse cone bipolar type 4 cells (DB4; marked with anti-PKCalpha) and weakly expressed by midget bipolar dendrites (labeled by antibodies against G alpha(o), G gamma 13, or mGluR6). Ret-PCP2 is absent from diffuse cone bipolar type 6 (DB6; marked with anti-CD15) and blue cone bipolar cells (marked with anti-CCK precursor). Thus, cone bipolar cells that terminate in stratum 3 of the inner plexiform layer (DB4) express more Ret-PCP2 than those that terminate in strata 3 + 4 (midget bipolar cells), and these in turn express more than those that terminate in stratum 5 (DB6 and blue cone bipolar cells). This expression pattern approximates the arborization of ganglion cells (GC) with different temporal bandwidths: parasol GCs stratifying near stratum 3 are faster than midget GCs stratifying in strata 3 + 4, and these are probably faster than the sluggish GCs that arborize in stratum 5.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C-alfa/metabolismo , Células Bipolares de la Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Animales , Secuencia de Bases , Western Blotting , ADN Recombinante , Inmunohistoquímica , Antígeno Lewis X/metabolismo , Macaca fascicularis , Macaca mulatta , Proteínas del Tejido Nervioso/genética , Células de Purkinje/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Retina/citología , Retina/metabolismo , Células Bipolares de la Retina/clasificación , Células Fotorreceptoras Retinianas Conos/clasificación , Células Ganglionares de la Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Comp Neurol ; 510(5): 484-96, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18671302

RESUMEN

Retinal ON bipolar cells make up about 70% of all bipolar cells. Glutamate hyperpolarizes these cells by binding to the metabotropic glutamate receptor mGluR6, activating the G-protein G(o1), and closing an unidentified cation channel. To facilitate investigation of ON bipolar cells, we here report on the production of a transgenic mouse (Grm6-GFP) in which enhanced green fluorescent protein (EGFP), under control of mGluR6 promoter, was expressed in all and only ON bipolar cells. We used the mouse to determine density of ON bipolar cells, which in central retina was 29,600 cells/mm(2). We further sorted the fluorescent cells and created a pure ON bipolar cDNA library that was negative for photoreceptor unique genes. With this library, we determined expression of 27 genes of interest. We obtained positive transcripts for G(o) interactors: regulators of G-protein signaling (RGS), Ret-RGS1 (a variant of RGS20), RGS16, RGS7, purkinje cell protein 2 (PCP2, also called L7 or GPSM4), synembryn (RIC-8), LGN (GPSM2), RAP1GAP, and Gbeta5; cGMP modulators: guanylyl cyclase (GC) 1alpha1, GC1beta1, phosphodiesterase (PDE) 1C, and PDE9A; and channels: inwardly rectifying potassium channel Kir2.4, transient receptor potential TRPC2, and sperm-specific cation channels CatSper 2-4. The following transcripts were not found in our library: AGS3 (GPSM1), RGS10, RGS19 (GAIP), calbindin, GC1alpha2, GC1beta2, PDE5, PDE2A, amiloride-sensitive sodium channel ACCN4, and CatSper1. We then localized Kir2.4 to several cell types and showed that, in ON bipolar cells, the channel concentrates in their dendritic tips. The channels and modulators found in ON bipolar cells likely shape their light response. Additional uses of the Grm6-GFP mouse are also discussed.


Asunto(s)
Ratones Transgénicos , Células Bipolares de la Retina/metabolismo , Animales , Secuencia de Bases , Línea Celular , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Perfilación de la Expresión Génica , Biblioteca de Genes , Humanos , Ratones , Datos de Secuencia Molecular , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Regiones Promotoras Genéticas , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Bipolares de la Retina/citología , Transducina/genética , Transducina/metabolismo
18.
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